In mediating drug resistance, PKCα

In mediating drug resistance, PKCα translocates from the cytoplasm to the membrane, phosphorylates the linker region of P-gp, activates the pump (P-gp), and subsequently causes reduction of intracellular

drug accumulation. In this respect, the membrane-associated PKCα should be considered as the functional form that coordinates with P-gp. TGF-β1 inhibits the growth of PC3 (a prostate cancer cell line with wild-type Smad4) by decreasing the membrane-associated PKCα, not by altering the total level of PKCα [37]. Another study showed that TGF-β1 suppressed PTEN expression in Smad4-null pancreatic cancer cells by activating Thiazovivin mouse PKCα [38]. These data suggest that the existence of Smad4 may repress the Smad4-independent pathway of TGF-β1 by inhibiting functions of several modulators (such as PKCα). Therefore, we propose that a Smad4-independent TGF-β1 pathway may promote the drug resistant phenotype in pancreatic cancer through PKCα and P-gp. Studies have shown that the MAPK and ERK pathway may be the downstream signaling pathways activated by TGF-β1. Several studies showed that

p38 and ERK pathways might mediate Smad4-independent click here TGF-β1 responses [39–41]. Our data show that TGF-β1 treatment induces phosphorylation of p38 but not ERK1/2. We believe that in absence of Smad4 (BxPC3 cells lack of Smad4 expression) TGF-β1 activates p38 but not ERK1/2 as a transient mediator in its signaling cascades. Indeed, we found that inhibition of PKCα or silence of TβRII reverses the resistance of BxPC3 cells to

the selleck kinase inhibitor chemotherapeutic drugs gemcitabine and cisplatin, suggesting that the PKCα inhibitor Gö6976 is a potential sensitizer to chemotherapy. Inhibition of PKCα function has been shown to effectively restore the drug-sensitive phenotype of cancer cells [42]. The PKCα inhibitor used in this study is a small molecule that has been reported to effectively abrogate DNA damage-induced cell cycle arrest and induce apoptosis [43]. In addition, we found that targeting TβRII Celecoxib by using siRNA did not achieve the same effect as Gö6976; it merely helped reverse gemcitabine resistance to a certain extent. However, tumor cells still remained tolerant to gemcitabine treatment. Another study demonstrated that the blockade of TβRII could not completely shut down the pathway, which may be because TβRI itself may be sufficient to transmit the TGF-β1 signal [43]. All of these findings suggest reasons why the PKCα inhibitor might be more effective in re-sensitizing cancer cells to cisplatin than that of TβRII silencing. In summary, we have demonstrated that TGF-β1-induced drug resistance in pancreatic cancer was mediated by upregulation of both PKCα and P-gp expression and by induction of the epithelial-to-mesenchymal transition. The PKCα inhibitor Gő6976, but not TβRII silencing, restores the sensitivity of pancreatic cancer cells to cisplatin or gemcitabine.

Comments are closed.