학술논문

Protein kinase C delta inhibits Caco-2 cell proliferation by selective changes in cell cycle and cell death regulators
Document Type
Academic Journal
Source
Oncogene. May 25, 2006 25(22):3123-3138
Subject
Language
English
ISSN
0950-9232
Abstract
PKC-δ is a serine/threonine kinase that mediates diverse signal transduction pathways. We previously demonstrated that overexpression of PKC-δ slowed the G1 progression of Caco-2 colon cancer cells, accelerated apoptosis, and induced cellular differentiation. In this study, we further characterized the PKC-δ dependent signaling pathways involved in these tumor suppressor actions in Caco-2 cells overexpressing PKC-δ using a Zn inducible expression vector. Consistent with a G1 arrest, increased expression of PKC-δ caused rapid and significant downregulation of cyclin D1 and cyclin E proteins (50% decreases, P<0.05), while mRNA levels remained unchanged. The PKC agonist, phorbol 12-myristate 13-acetate (TPA, 100 nM, 4 h), induced two-fold higher protein and mRNA levels of p21, a cyclin-dependent kinase (cdk) inhibitor in PKC-δ transfectants compared with empty vector (EV) transfected cells, whereas the PKC-δ specific inhibitor rottlerin (3 μM) or knockdown of this isoenzyme with specific siRNA oligonucleotides blocked p21 expression. Concomitantly, compared to EV control cells, PKC-δ upregulation decreased cyclin D1 and cyclin E proteins co-immunoprecipitating with cdk6 and cdk2, respectively. In addition, overexpression of PKC-δ increased binding of cdk inhibitor p27 to cdk4. These alterations in cyclin-cdks and their inhibitors are predicted to decrease G1 cyclin kinase activity. As an independent confirmation of the direct role PKC-δ plays in cell growth and cell cycle regulation, we knocked down PKC-δ using specific siRNA oligonucleotides. PKC-δ specific siRNA oligonucleotides, but not irrelevant control oligonucleotides, inhibited PKC-δ protein by more than 80% in Caco-2 cells. Moreover, PKC-δ knockdown enhanced cell proliferation (∼1.4-2-fold, P<0.05) and concomitantly increased cyclin D1 and cyclin E expression (∼1.7-fold, P<0.05). This was a specific effect, as nontargeted PKC-ζ was not changed by PKC-δ siRNA oligonucleotides. Consistent with accelerated apoptosis in PKC-δ transfectants, compared to EV cells, PKC-δ upregulation increased proapoptotic regulator Bax two-fold at mRNA and protein levels, while antiapoptotic Bcl-2 protein was decreased by 50% at a post-transcriptional level. PKC-δ specific siRNA oligonucleotides inhibited Bax protein expression by more than 50%, indicating that PKC-δ regulates apoptosis through Bax. Taken together, these results elucidate two critical mechanisms regulated by PKC-δ that inhibit cell cycle progression and enhance apoptosis in colon cancer cells. We postulate these antiproliferative pathways mediate an important tumor suppressor function for PKC-δ in colonic carcinogenesis.