16.07.2013 Views

10 1 Full Volume (PDF)(jcmb.halic.edu.tr) - Journal of Cell and ...

10 1 Full Volume (PDF)(jcmb.halic.edu.tr) - Journal of Cell and ...

10 1 Full Volume (PDF)(jcmb.halic.edu.tr) - Journal of Cell and ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

independent <strong>of</strong> changes in the related mRNA level<br />

(Figures 3 <strong>and</strong> 5). The increased activity <strong>of</strong><br />

caspase-3 was negated via overexpression <strong>of</strong> DFF-<br />

45. DFF-45 is the natural inhibitor <strong>of</strong> DFF-40<br />

(CAD) (Liu et al., 1997). In addition, the increased<br />

expression <strong>of</strong> DFF-45, along with a modest<br />

increase (for sulfabenzamide) <strong>and</strong> a significant<br />

decrease (for doxorubicin) in DFF-40 expression<br />

(Figure 5), indicated that the increased activity <strong>of</strong><br />

caspase-3 could be blocked by the increased<br />

expression ratio <strong>of</strong> DFF-45/DFF-40. This r<s<strong>tr</strong>ong>edu</s<strong>tr</strong>ong>ces<br />

the level <strong>of</strong> active DFF-40 to <strong>tr</strong>igger DNA<br />

fragmentation <strong>and</strong> appearance <strong>of</strong> apoptotic<br />

symptoms. Furthermore, it has been reported that<br />

caspases are activated during autophagy in dying<br />

cells <strong>and</strong> are suppressed for apoptosis induction<br />

(Martin et al., 2004; Yu et al., 2004). Therefore, it<br />

can be d<s<strong>tr</strong>ong>edu</s<strong>tr</strong>ong>ced that during autophagy, the effects<br />

<strong>of</strong> activated caspase-3 on their downs<strong>tr</strong>eam<br />

subs<strong>tr</strong>ates (like DFF-40) should be suppressed by<br />

special factors (e.g. DFF-45 in T-47D cells) only in<br />

those cellular routes which are involved in the<br />

appearance <strong>of</strong> apoptosis symptoms (e.g. DNA<br />

fragmentation).<br />

<strong>Cell</strong> cycle arrest, an important cellular target<br />

affected by sulfabenzamide <strong>and</strong> doxorubicin, was<br />

analyzed using flow cytome<strong>tr</strong>y. Incubation <strong>of</strong> T-<br />

47D cells with 0.33 µM doxorubicin resulted in a<br />

significant accumulation <strong>of</strong> cells in S phase, <strong>and</strong> to<br />

a lesser extent in G2/M phase <strong>of</strong> the cell cycle<br />

(Figure 4B <strong>and</strong> Table 2). Thus, doxorubicin exerts<br />

its antiproliferative action mainly through cell cycle<br />

arrest. Induced mitotic catas<strong>tr</strong>ophe following<br />

increased activation <strong>of</strong> cyclinB1/Cdc2 may occur<br />

while cells are delayed, particularly in G2 phase <strong>of</strong><br />

the cell cycle (Lindqvist et al., 2007). The induced<br />

G2/M arrest along with down regulation <strong>of</strong><br />

cyclinB1 expression confirmed that anticancer<br />

activity <strong>of</strong> doxorubicin is not via mitotic<br />

catas<strong>tr</strong>ophe (Figure 5 <strong>and</strong> Table 2). In con<strong>tr</strong>ast to<br />

doxorubicin, minimal cell cycle arrest in S <strong>and</strong><br />

G2/M phases (totally 18%) was observed in T-47D<br />

cells incubated with <s<strong>tr</strong>ong>10</s<strong>tr</strong>ong>.8 mM sulfabenzamide<br />

(Figure 4B <strong>and</strong> Table2). Thus, cell cycle arrest<br />

could not be mainly responsible for a 50%<br />

r<s<strong>tr</strong>ong>edu</s<strong>tr</strong>ong>ction in cell viability in the presence <strong>of</strong><br />

sulfabenzamide.<br />

As we know, when apoptosis is blocked or<br />

delayed autophagy <strong>tr</strong>iggered <strong>and</strong> vice versa. These<br />

possibilities are consistent with our findings<br />

regarding lack <strong>of</strong> apoptosis in drug <strong>tr</strong>eated cells <strong>and</strong><br />

induction <strong>of</strong> autophagy. The induced<br />

overexpression <strong>of</strong> ATG5 supported that autophagy<br />

Sulfabenzamide promotes autophagic cell death 51<br />

<strong>tr</strong>iggered in the presence <strong>of</strong> sulfabenzamide (Figure<br />

5). This could be probably occurred through the<br />

increase in Bax activity working on mitochondrial<br />

membrane to result in activation <strong>of</strong> caspase-3 for<br />

PARP1 <strong>and</strong> DNA-PK deactivation <strong>and</strong> autophagy<br />

induction. It has been reported that induction <strong>of</strong><br />

autophagy by PUMA (the p53-inducible BH3-only<br />

protein) depends on Bax/Bak <strong>and</strong> can be<br />

reproduced by overexpression <strong>of</strong> Bax (Yee et al.,<br />

2009). Here, in doxorubicin <strong>tr</strong>eatment, increase in<br />

Bax activity could be occurred in parallel with the<br />

increment <strong>of</strong> Bax <strong>tr</strong>anscripts affecting on the cells<br />

for caspase-3 activation <strong>and</strong> changing the cell's<br />

destiny toward autophagy (Figure 5). This notion<br />

could be also supplied in sulfabenzamide <strong>tr</strong>eatment<br />

aside from the mild decrease in Bax expression<br />

because activation <strong>of</strong> the existed Bax molecules in<br />

the cells could be happened for caspase-3 activation<br />

<strong>and</strong> autophagy induction (Figure 5). It has been<br />

also reported that proteolytic cleavage <strong>of</strong> PARP1,<br />

performed by caspase-3, produces specific<br />

proteolytic cleavage fragments which are involved<br />

in the cell’s decision to change its fate from<br />

apoptosis toward autophagy (Munoz-Gamez et al.,<br />

2009; Chaitanya et al., 20<s<strong>tr</strong>ong>10</s<strong>tr</strong>ong>). Induction <strong>of</strong><br />

autophagic cell death is dependent on DNA-PK<br />

inhibition (Daido et al., 2005). Thus, the increased<br />

activity <strong>of</strong> caspase-3 could finally deactivate<br />

PARP1 (has a decreased <strong>and</strong> constant expression<br />

level in sulfabenzamide <strong>and</strong> doxorubicin<br />

<strong>tr</strong>eatments, respectively) <strong>and</strong> DNA-PK (has an<br />

increased <strong>and</strong> invariable expression level in<br />

sulfabenzamide <strong>and</strong> doxorubicin <strong>tr</strong>eatments,<br />

respectively) until apoptosis was blocked <strong>and</strong><br />

autophagy induced (Figures 3 <strong>and</strong> 5).<br />

Despite the existence <strong>of</strong> some con<strong>tr</strong>oversies<br />

regarding the possible role <strong>of</strong> autophagy in tumor<br />

progression by promoting cell survival, autophagy<br />

can exist as a backup mechanism promoting<br />

cellular death when other mortality mechanisms are<br />

not functional. Hyperactivation <strong>of</strong> autophagy above<br />

the threshold point leads to unlimited self-eating <strong>of</strong><br />

the cells causing autophagy or type II programmed<br />

cell death (Hoyer-Hansen et al., 2005; Maiuri et al.,<br />

20<s<strong>tr</strong>ong>10</s<strong>tr</strong>ong>). Based on our data, downregulation <strong>of</strong> AKT1<br />

<strong>and</strong> AKT2 as well as upregulation <strong>of</strong> PTEN in<br />

sulfabenzamide <strong>tr</strong>eated cells indicated that cell<br />

survival pathways were slowed down (Figure 5). In<br />

addition, down regulation <strong>of</strong> bcl-2 was happened<br />

along with the induction <strong>of</strong> autophagy (Figure 5). It<br />

has been reported that targeted silencing <strong>of</strong> bcl-2<br />

expression (an anti-autophagic gene) in human<br />

breast cancer cells with RNA-interference has

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

Saved successfully!

Ooh no, something went wrong!