13.07.2013 Views

96. Jahrestagung der Deutschen Gesellschaft für Pathologie e. V ...

96. Jahrestagung der Deutschen Gesellschaft für Pathologie e. V ...

96. Jahrestagung der Deutschen Gesellschaft für Pathologie e. V ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

HCV-positive formalin-fixed and paraffin-embedded biopsies, sections<br />

were prepared and HCC were macrodissected. Subsequently, miR-125b<br />

was analyzed by real-time PCR. Putative miR-125b binding sites were fused<br />

to the luciferase reporter and reporter assays were carried out with<br />

miR-125b treated hepatoma cells.<br />

Results. During development of mouse HCC, the expression of miR-125b<br />

progressively decreased. In agreement miR-125b was reduced in human<br />

hepatoma cells in comparison to normal liver. Furthermore, miR-125b<br />

decrease depending on the progression of hepatocarcinogenesis was<br />

confirmed in human samples, showing significant lower levels in HCC<br />

high grades than in dysplastic foci or cirrhosis. Overexpression of miR-<br />

125b in Hep3B and Pop10 cells resulted in a pronounced reduction of cell<br />

growth. Screening of putative miR-125b target transcripts by various<br />

algorithm calculations identified various pathways involved in proliferation<br />

and apoptosis. Reporter assays of 3’-UTR-regions of the putative<br />

targets identified miR-125b binding sites in lin-28 mRNA. Since lin28 is<br />

known to effect synthesis of the mitogen IGF-II, the miR-125b/lin28 axis<br />

is suggested to be involved in HCC pathogenesis by IGF-II mediated regulation<br />

of cell growth.<br />

Conclusions. Expression of miR-125b is down-regulated during progression<br />

of hepatocarcinogenesis leading to up-regulation of lin-28 that in<br />

turn triggers enhanced cell growth and proliferation.<br />

DO-005<br />

The PI3K-AKT-mTOR axis contributes to the functional inactivation<br />

of p53 through stabilization of MDM4 in human hepatocellular<br />

carcinoma<br />

R . Pellegrino1 , O . Neumann1 , P . Schirmacher1 , T . Longerich1 1University Hospital Heidelberg/Institute of Pathology, Heidelberg<br />

Aims. Mutational inactivation of p53 gene is rare in Western hepatocellular<br />

carcinoma (HCC). MDM4, one of the main p53-regulating factors,<br />

is frequently upregulated in human HCC. This overexpression can be<br />

in part explained by chromosomal gains at 1q34.1. Here we investigated<br />

the role of the PI3K-AKT axis in the stabilization of the MDM4 protein.<br />

Methods. All experiments were performed in human HCC cell lines<br />

with different p53 gene status. PI3K and mTOR were specifically inhibited<br />

using chemical compounds in vitro; specific siRNAs were transiently<br />

transfected to target AKT1 and ATK2 and to validate the results from<br />

drug treatment. Realtime RT-PCR analysis was used to check for restored<br />

p53 transcriptional activity. In addition, combined cycloheximide<br />

and siRNAs treatment was performed to study the protein stability of<br />

MDM4 un<strong>der</strong> these experimental conditions.<br />

Results. Using a specific PI3K inhibitor or siRNAs targeting AKT1/2 in<br />

HCC cell lines, we observed a strong decrease of MDM4 protein levels,<br />

which resulted in the activation of p53 target genes (e.g. PUMA, BAX and<br />

p21) indicating restored p53 gene function in these lines. Cycloheximide<br />

treatment combined with siRNA-mediated AKT inhibition indicated<br />

that MDM4 is phosphorylated by AKT2 and that this phosphorylation<br />

is responsible for the stabilization of the protein via the protection from<br />

proteasomal degradation. This effect was independent from both MDM2<br />

and p53 gene status. Furthermore, we showed that the Eukaryotic translational<br />

Elongation Factor 1 alpha 2 (EEF1A2), which we reported upregulated<br />

in human HCC, is involved in the activation of the PI3K-AKT<br />

axis. Specific siRNA inhibition of EEF1A2 resulted in decreased pAKT<br />

and MDM4 protein levels in HCC cell lines. Moreover, treatment with<br />

the mTOR inhibitors, Rapamycin and PI-103, decreased MDM4 protein<br />

levels indicating that the PI3K-AKT-mTOR axis is involved in the<br />

MDM4 regulation in vitro.<br />

Conclusions. Our data demonstrate that the EEF1A2-PI3K-AKT-mTOR<br />

axis is involved in maintaining protumorigenic MDM4 levels in human<br />

HCC cell lines, which in turn promotes functional inactivation of<br />

p53. Moreover, we showed that the AKT-mediated phosphorylation of<br />

MDM4 is the crucial mechanism to prevent its proteasomal degradation.<br />

DO-006<br />

HSF1 is a downstream effector of Ras and AKT protooncogenes<br />

and contributes to hepatocellular carcinoma development and<br />

progression<br />

D .F . Calvisi1 , S . Mattu1 , S . Delogu1 , V . De Murtas1 , G . Gasparetti1 , G . Destefanis1 ,<br />

X . Chen2 , F . Dombrowski1 , M . Evert1 1University Medicine Greifswald, Institute for Pathology, Greifswald,<br />

2University of San Francisco, Liver Center, San Francisco, United States<br />

Aims. Recent evidence suggests an oncogenic role of heat shock transcription<br />

factor 1 (HSF1) in cancer, but its functional relevance in hepatocellular<br />

carcinoma (HCC) remains poorly delineated.<br />

Methods. We have investigated HSF1 function both via in vitro and in<br />

vivo approaches as well as in a collection of human HCC.<br />

Results. In human liver specimens, we found that HSF1 was progressively<br />

induced from non-tumorous surrounding livers to HCC, reaching the<br />

highest levels in tumors with a poorer outcome (as defined by the length<br />

of patient’s survival). In HCC cell lines, overexpression of HSF1 resulted<br />

in increased activity of MAPK and AKT/mTOR pathways and suppression<br />

of JNK cascade, leading to augmented proliferation and angiogenesis<br />

and reduced apoptosis in vitro. Conversely, suppression of HSF1 in<br />

HCC cell lines decreased MAPK and AKT/mTOR activity, and induced<br />

JNK-dependent apoptosis. Forced overexpression of either Ras or AKT<br />

protooncogenes triggered upregulation of HSF1 in HCC cell lines via the<br />

small RalA GTPase. Of note, HSF1-overexpressing cells were specifically<br />

sensitive to growth inhibition and induction of apoptosis following the<br />

treatment with either AMPK activators or hexokinase inhibitors. Finally,<br />

overexpression of a HSF1 dominant negative form by hydrodynamic<br />

gene delivery strongly reduced the oncogenic potential of activated Ras<br />

and AKT in a mouse model of aggressive liver cancer.<br />

Conclusions. Altogether, the present data indicate that activation of HSF1<br />

plays a major role in hepatocarcinogenesis by enhancing the activity of<br />

Ras and AKT, and might represent a valuable candidate for innovative<br />

targeted therapies against human HCC.<br />

DO-007<br />

Perturbation of hepatocytes metabolism by AKT contributes to<br />

growth in insulin-induced hepatocarcinogenesis and is reverted<br />

by the PI3K/mTOR dual inhibitor NVP-BEZ235<br />

M . Evert1 , D .F . Calvisi1 , K . Evert1 , V . De Murtas1 , G . Gasparetti1 , S . Mattu1 ,<br />

G . Destefanis1 , S . Thiel1 , A . Thiele1 , S . Ribback1 , F . Dombrowski1 1University Medicine Greifswald, Institute for Pathology, Greifswald<br />

Aims. Mounting evidence supports a role of insulin signaling <strong>der</strong>egulation<br />

and diabetes mellitus in human hepatocarcinogenesis. To study the<br />

oncogenic effect of chronically elevated secretion of insulin on hepatocytes<br />

in the presence of mild hyperglycemia, we developed a model of<br />

pancreatic islet transplantation into the liver.<br />

Methods. In this model, islets of a donor rat are transplanted into the<br />

liver of a recipient diabetic rat, with resulting local hyperinsulinism that<br />

leads to the development of preneoplastic lesions and hepatocellular carcinoma<br />

(HCC). Here, we investigated the metabolic and growth properties<br />

of the AKT pathway in this model of insulin-induced hepatocarcinogenesis.<br />

These findings were recapitulated in HCC cell lines in vitro.<br />

Results. We found that activation of insulin signaling triggers a strong<br />

induction of the AKT cascade that is paralleled by increased synthesis<br />

of fatty acids, cholesterol, and triglycerides, induction of glycolysis and<br />

decrease of fatty acid oxidation and gluconeogenesis in rat preneoplastic<br />

and neoplastic liver lesions when compared with normal liver. AKT-dependent<br />

metabolic effects of insulin on hepatocytes were recapitulated in<br />

vitro using human HCC cell lines. In these cells, suppression of lipogenesis,<br />

glycolysis, and the pentose phosphate pathway triggered a strong<br />

growth restraint despite insulin administration. Of note, metabolic abnormalities<br />

and proliferation driven by insulin were effectively reverted<br />

Der Pathologe · Supplement 1 · 2012 |<br />

17

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

Saved successfully!

Ooh no, something went wrong!