06.02.2013 Views

Tetracycline-inducible protein expression in pancreatic cancer cells

Tetracycline-inducible protein expression in pancreatic cancer cells

Tetracycline-inducible protein expression in pancreatic cancer cells

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

A<br />

Migration (relative units)<br />

Tonack S et al . Pancreatic <strong>cancer</strong> Tet-on cell clones; effects of CapG over<strong>expression</strong><br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0.0<br />

Control<br />

Dox<br />

P = 0.564<br />

B 2.4<br />

Control<br />

Motility <strong>in</strong>dex<br />

2.0<br />

1.6<br />

1.2<br />

0.8<br />

0.4<br />

0.0<br />

P = 0.032<br />

Panc-1 were all capable of stably harbour<strong>in</strong>g the pN1βact<strong>in</strong>rtTA2S-M2-IRES-EGFP<br />

vector. Moreover, <strong><strong>in</strong>ducible</strong> gene<br />

over<strong>expression</strong> was observed for GST-P, CYP2E1, and<br />

S100A6 <strong>in</strong> Suit-2 and for CapG <strong>in</strong> all three <strong>in</strong>vestigated cell<br />

l<strong>in</strong>es. This <strong>in</strong>dicates that the system functions for a variety<br />

of different <strong>prote<strong>in</strong></strong>s. Equally, the method was efficient with<br />

80% of pN1βact<strong>in</strong>-rtTA2S-M2-IRES-EGFP transfected<br />

Suit-2 clones and 60% of MiaPaca-2 clones show<strong>in</strong>g greater<br />

than five-fold luciferase <strong>in</strong>duction. Only Panc-1 <strong>cells</strong> showed<br />

relatively low efficiency, with only 14.4% of the selected cell<br />

clones show<strong>in</strong>g a greater than five-fold luciferase <strong>in</strong>crease.<br />

A clear advantage of the Tet-on system is the time and<br />

dose-dependent activation of <strong>expression</strong> of the gene of<br />

<strong>in</strong>terest. Investigation of stable CapG overexpress<strong>in</strong>g Suit-2<br />

cell clones showed that CapG <strong>prote<strong>in</strong></strong> <strong>expression</strong> was dox<strong><strong>in</strong>ducible</strong><br />

<strong>in</strong> a time dependent manner. The level of CapG<br />

<strong>in</strong>creased from 12 h after <strong>in</strong>duction, and was ma<strong>in</strong>ta<strong>in</strong>ed up<br />

to 72 h after <strong>in</strong>duction. A similar time-dependent <strong>in</strong>duction<br />

of gene <strong>expression</strong> <strong>in</strong> the liver carc<strong>in</strong>oma <strong>cells</strong>, HepG2 has<br />

been described previously [14] . However, we did not observe<br />

a l<strong>in</strong>ear correlation between dox concentration and <strong>prote<strong>in</strong></strong><br />

<strong>expression</strong>, as was reported <strong>in</strong> the HepG2 <strong>cells</strong> [14] . Instead,<br />

we detected little <strong>in</strong>crease <strong>in</strong> CapG <strong>prote<strong>in</strong></strong> concentration<br />

between 0-10 ng/mL dox, and over<strong>expression</strong> of CapG<br />

WJG|www.wjgnet.com<br />

P = 0.0171<br />

TET29 C35 C43<br />

Dox<br />

P = 0.4311<br />

P = 0.0006<br />

P = 0.031<br />

TET29 C35 C43<br />

Figure 9 Wound heal<strong>in</strong>g capacity (A) and motility (B) of CapG-overexpress<strong>in</strong>g<br />

<strong>cells</strong>. A: Bars represent the migration <strong>in</strong> relative units for Sβtet29Cap35<br />

(C35), Sβtet29Cap43 (C43) and the control Sβtet29 (TET29) <strong>cells</strong> treated with<br />

doxycycl<strong>in</strong>e (dox, grey bar) or PBS (Control, black bar). Error bars present the<br />

SE for three experiments carried out <strong>in</strong> triplicate; B: The motility <strong>in</strong>vestigated by a<br />

Boyden Chamber assay of Sβtet29Cap35 (C35), Sβtet29Cap43 (C43) and the<br />

control Sβtet29 (TET29) <strong>cells</strong> treated with doxycycl<strong>in</strong>e (dox, gray bar) or PBS<br />

(Control, black bar) is shown. Each experiment was performed <strong>in</strong> triplicate with<br />

three replicates.<br />

with concentrations of 50 ng/mL and greater (up to<br />

1000 ng/mL). Possible reasons for these different results<br />

could be related to the particular <strong>prote<strong>in</strong></strong> overexpressed or<br />

the different promoter used to drive the rtTA2S-M2 <strong>in</strong> both<br />

studies. The HepG2 Tet-on clones harboured CMV driven<br />

rtTA2S-M2, unlike the pancreas clones <strong>in</strong> this study.<br />

Concerns have been raised about the toxicity of dox,<br />

the agent used to activate gene <strong>expression</strong> <strong>in</strong> the Teton<br />

vector system [18] . The cell growth of PC-12 <strong>cells</strong> was<br />

reduced after 96h <strong>in</strong>cubation with concentrations rang<strong>in</strong>g<br />

from as low as 0.2 to 100 μg/mL. Growth perturbations<br />

were observed with WI-38 VA-13 <strong>cells</strong>, but only with<br />

concentrations of dox greater than 2 μg/mL [18] . It appears<br />

that the concentration of dox which mediates a toxic effect<br />

is cell-l<strong>in</strong>e-dependent. At the concentrations of dox<br />

used <strong>in</strong> this study, no loss of proliferation was observed<br />

48 h after dox treatment. These data were supported by<br />

cell cycle analyses profiles, which showed no change <strong>in</strong><br />

cell cycle follow<strong>in</strong>g dox treatment for 24 h and 72 h.<br />

The function of CapG <strong>in</strong> the cytoplasm is well described.<br />

CapG caps and therefore blocks rapidly grow<strong>in</strong>g<br />

act<strong>in</strong> filaments, promot<strong>in</strong>g the elongation of shorter<br />

act<strong>in</strong> filaments. It thus contributes to cell motility [19] and<br />

membrane ruffl<strong>in</strong>g [20] . Increased levels of CapG <strong>prote<strong>in</strong></strong><br />

have been described <strong>in</strong> several tumors, <strong>in</strong>clud<strong>in</strong>g <strong>pancreatic</strong><br />

ductal adenocarc<strong>in</strong>oma [12,21] and glioblastomas [22] .<br />

Transient CapG knockdown experiments <strong>in</strong> <strong>pancreatic</strong><br />

<strong>cancer</strong> cell l<strong>in</strong>es Suit-2, MiaPaca-2, and Panc-1 cell l<strong>in</strong>es<br />

led to a significant decrease <strong>in</strong> wound heal<strong>in</strong>g capacity<br />

and cell motility [12] . Furthermore, Van den Abbeele et al [23]<br />

showed that downregulation of CapG <strong>in</strong> the breast <strong>cancer</strong><br />

<strong>cells</strong> MDA-MB 231 and the prostate <strong>cancer</strong> <strong>cells</strong> PC-3<br />

also decreased <strong>in</strong>vasion and motility. Here, we show for<br />

the first time that over<strong>expression</strong> of CapG <strong>in</strong> a <strong>pancreatic</strong><br />

<strong>cancer</strong> cell l<strong>in</strong>e caused a significant <strong>in</strong>crease <strong>in</strong> motility and<br />

a modest, but significant, <strong>in</strong>crease <strong>in</strong> wound heal<strong>in</strong>g capacity.<br />

This is consistent with a previous report of an <strong>in</strong>crease<br />

<strong>in</strong> <strong>in</strong>vasion after CapG over<strong>expression</strong> <strong>in</strong> Mad<strong>in</strong>-Darby<br />

Can<strong>in</strong>e Kidney Epithelial Cells MDCK <strong>cells</strong> [24] . Interest<strong>in</strong>gly,<br />

it has been reported that active nuclear import of<br />

CapG is necessary for CapG to promote <strong>in</strong>vasion [24] . Prevention<br />

of nuclear accumulation of CapG <strong>in</strong> MDCK <strong>cells</strong><br />

abolished collagen <strong>in</strong>vasion, whereas restor<strong>in</strong>g the nuclear<br />

import also restored collagen <strong>in</strong>vasion of these <strong>cells</strong>. We<br />

found no evidence for elevated nuclear levels of CapG<br />

follow<strong>in</strong>g over<strong>expression</strong> <strong>in</strong> Suit-2 derived <strong>cells</strong>. Immunohistochemical<br />

sta<strong>in</strong><strong>in</strong>g localized CapG to the cytoplasm<br />

<strong>in</strong> Suit-2 <strong>cells</strong>. Dox treatment of stable <strong><strong>in</strong>ducible</strong> CapG<br />

expressors led to an <strong>in</strong>crease <strong>in</strong> cytoplasmic sta<strong>in</strong><strong>in</strong>g, with<br />

<strong>in</strong>creased CapG around the nuclear membrane, but CapG<br />

did not accumulate <strong>in</strong> the nucleus. Subcellular fractionation<br />

further validated this f<strong>in</strong>d<strong>in</strong>g, show<strong>in</strong>g <strong>in</strong>creased<br />

CapG <strong>prote<strong>in</strong></strong> levels <strong>in</strong> the cytoplasmic fraction after dox<br />

treatment. We observed <strong>in</strong>creased motility, suggest<strong>in</strong>g that<br />

elevated cytoplasmic CapG may be sufficient for <strong>in</strong>creased<br />

<strong>cancer</strong> cell motility.<br />

CapG has been shown to <strong>in</strong>teract with microtubuledependent<br />

organelles dur<strong>in</strong>g the cell cycle [25] , possibly<br />

mediat<strong>in</strong>g cross-talk between the act<strong>in</strong> cytoskeleton and<br />

1958 April 21, 2011|Volume 17|Issue 15|

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

Saved successfully!

Ooh no, something went wrong!