Skip to main content

TRAIL sensitize MDR cells to MDR-related drugs by down-regulation of P-glycoprotein through inhibition of DNA-PKcs/Akt/GSK-3β pathway and activation of caspases

Abstract

Background

The development of new modulator possessing high efficacy, low toxicity and high selectivity is a pivotal approach to overcome P-glycoprotein (P-gp) mediated multidrug resistance (MDR) in cancer treatment. In this study, we suggest a new molecular mechanism that TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) down-regulates P-glycoprotein (P-gp) through inhibition of DNA-PKcs/Akt/GSK-3β pathway and activation of caspases and thereby sensitize MDR cells to MDR-related drugs.

Results

MDR variants, CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells, with gradually increased levels of P-gp derived from human lymphoblastic leukemia CEM cells, were gradually more susceptible to TRAIL-induced apoptosis and cytotoxicity than parental CEM cells. The P-gp level of MDR variants was positively correlated with the levels of DNA-PKcs, pAkt, pGSK-3β and c-Myc as well as DR5 and negatively correlated with the level of c-FLIPs. Hypersensitivity of CEM/VLB100 cells to TRAIL was accompanied by the activation of mitochondrial apoptotic pathway as well as the activation of initiator caspases. In addition, TRAIL-induced down-regulation of DNA-PKcs/Akt/GSK-3β pathway and c-FLIP and up-regulation of cell surface expression of death receptors were associated with the increased susceptibility to TRAIL of MDR cells. Moreover, TRAIL inhibited P-gp efflux function via caspase-3-dependent degradation of P-gp as well as DNA-PKcs and subsequently sensitized MDR cells to MDR-related drugs such as vinblastine and doxorubicin. We also found that suppression of DNA-PKcs by siRNA enhanced the susceptibility of MDR cells to vincristine as well as TRAIL via down-regulation of c-FLIP and P-gp expression and up-regulation of DR5.

Conclusion

This study showed for the first time that the MDR variant of CEM cells was hypersensitive to TRAIL due to up-regulation of DR5 and concomitant down-regulation of c-FLIP, and degradation of P-gp and DNA-PKcs by activation of caspase-3 might be important determinants of TRAIL-induced sensitization of MDR cells to MDR-related drugs. Therefore, combination of TRAIL and chemotherapeutic drugs may be a good strategy for treatment of cancer with multidrug resistance.

Background

Acquired resistance to chemotherapeutic agents remains a major obstacle for the effective treatment of many advanced and metastatic cancers. Several mechanisms are thought to be involved in the development of multidrug resistance (MDR), defined by simultaneous cross-resistance to a variety of anticancer drugs that differ in their chemical structures, modes of action, and molecular targets [13]. Emergence of MDR is often associated with over-expression of the MDR1 gene product, P-glycoprotein (P-gp) [4]. In certain cancers, such as chronic or acute myeloid leukemia and breast cancer, over-expression of MDR1 gene is a prognostic indicator for clinical outcome and correlates with a poor response to chemotherapy [58]. Therefore, inhibition of P-gp function or expression can reverse P-gp-mediated MDR and improve the efficacy of chemotherapy [9].

Previously, we have reported that an increased expression of DNA-dependent protein kinase (DNA-PK) participates in the development of MDR, and inhibition of DNA-PK leads to increase of drug sensitivity in MDR cells [10]. DNA-PK comprises a catalytic subunit (DNA-PKcs) with a DNA- binding Ku70 and Ku80 heterodimer acting as the regulatory element. It has been proposed that DNA-PK is a molecular sensor for DNA damage that enhances the signal via phosphorylation of many downstream targets [11]. Recently, it has been demonstrated that DNA-PKcs-catalyzed RNA Helicase A phosphorylation enhanced the transcription of the MDR1 gene through the CAAT-like element of the MDR1 gene promoter and thus DNA-PKcs played an important role in regulation of P-gp expression by MDR1 promoter activation [12].

The phosphoinositide 3-kinase (PI3K)/Akt pathway is also frequently implicated in tumorigenesis and chemotherapeutic resistance [13]. Recent studies have shown that there is a significant correlation between the phosphorylated, activated Akt and P-gp expression, and inhibition of the PI3K/Akt signaling pathway can reverse P-gp-mediated MDR [1416]. Akt phosphorylation on Ser473 (S473) is required for activation of Akt, and a major Akt S473 kinase activity was found to be DNA-PK, a member of the PI3K-related kinase subfamily of protein kinases [17]. DNA-PKcs has been shown to colocalize with Akt and enhance Akt phosphorylation [18]. One of the downstream targets of pro-survival Akt is GSK-3β, which is inactivated by phosphorylation on Ser9 by Akt. The inactivation of GSK-3β through Akt-mediated phosphorylation leads to down-regulate its pro-apoptotic activity and inhibit the induction of cell death. Death receptor-induced extrinsic apoptotic signaling is also modulated by GSK-3β activity [19].

Recently, it has been shown that the extrinsic death receptor pathway represents a suitable target for cancer treatment [20]. Since TNF-related apoptosis inducing ligand (TRAIL) has been shown to induce apoptosis in various tumor cells, but only rarely in non-transformed cells, TRAIL is currently assessed in clinical trials [21]. The extrinsic apoptotic signaling cascade is a vital process initiated by activation of death receptors, DR4/DR5. Stimulation of these death receptors causes receptor trimerization, followed by recruitment of FADD (Fas associated with death domain protein) and caspase-8 (or caspase-10) to form the death-inducing signaling complex (DISC). DISC formation promotes autoactivation of caspase-8/10 and the subsequent activation of effector caspases, primarily caspase-3, -6 and -7, which implement the cell death program. Cellular FLICE inhibitory protein (c-FLIP) is expressed as long form (c-FLIPL) and short form (c-FLIPS) and inhibits caspase-8 binding to FADD and prevents DISC formation and apoptosis and splice forms [22]. Elevated Akt activity up-regulates c-FLIP and inhibits TRAIL-induced apoptosis in cancer cells [23].

It has been reported that MDR cells over-expressing P-gp are more susceptible to TRAIL than their drug-sensitive counterparts through various mechanisms such as a reduced expression of endogenous Akt [24] or enhancement of TRAIL binding to DR5 by P-gp [25]. However, the mechanism underlying the increased susceptibility of MDR cells to TRAIL mediated cell death was not understood well. Here, we demonstrated that TRAIL sensitized MDR cells to MDR-related drugs by inhibition of DNA-PKcs/Akt/GSK-3β pathway, activation of caspases and subsequent down-regulation of P-gp.

Results

The high susceptibility of MDR cells to TRAIL is associated with up-regulation of DR5 and down-regulation of c-FLIPs in the cells

Many studies have focused on overcoming MDR through modulation of P-gp function [4, 26]. Since TRAIL as a targeted cancer therapy preferentially kills cancer versus normal cells [20], we compared the cytotoxic and apoptotic effects of TRAIL between CEM human lymphoblastic leukemic cell line and its multidrug resistant (MDR) variants, including CEM/VLB10-2, CEM/VLB55-8, and CEM/VLB100 isolated from parental CEM cells. In a model of CEM cells with gradually increasing expression of multidrug-resistance 1 (MDR1) gene, the higher expression level of MDR1 gene in MDR variants, the higher cytotoxicity to TRAIL as shown in MTT assay (Figure 1A) and flow cytometric analysis of apoptosis (Figure 1B), demonstrating that the MDR cells were more susceptible to TRAIL-induced cell death than TRAIL-insensitive parental cells. Since TRAIL acts through the TRAIL receptors (DR5 and DR4) to induce apoptosis [20], we compared the mRNA levels of DR5 and DR4 between CEM and its MDR variants (Figure 1C). The level of DR5 but not DR4 in MDR variants was significantly increased as compared with that in CEM cells. This result suggests that an increased level of DR5 in MDR cells plays an important role in the sensitization of the cells to TRAIL. To understand further the factors that contribute to the sensitization of MDR cells to TRAIL, we determined the expression of c-FLIPL/S, an important downstream regulator of death receptor-mediated apoptosis [27], in MDR variants. We found that the mRNA level of c-FLIPS, but not c-FLIPL, was significantly decreased in MDR variants as compared to that in CEM cells. Therefore, our data indicated that TRAIL susceptibility of MDR cells was closely associated with up-regulation of DR5 and down-regulation of c-FLIPs in the cells.

Figure 1
figure 1

High susceptibility of MDR cells to TRAIL and up-regulation of death receptor and down-regulation of c-FLIP in the cells. (A) mRNA levels of mdr1 in CEM cells and its MDR variants, CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells were determined by RT-PCR (insert). To determine the growth inhibitory effect of TRAIL, CEM cells and its multidrug-riesistant variants were treated with graded single doses of TRAIL, and the percentage of cell survival was determined after 5 days incubation using the MTT assay. (B) CEM cells and its MDR variants were treated with or without TRAIL (10 ng/ml) for 24 h. Thereafter, the percentage of apoptotic cells in each cell population was determined by Annexin V staining and flow cytometry. Each bar represents the mean ± S.D. of triplicate experiments. *p < 0.05, **p < 0.01, ***p < 0.005 versus TRAIL untreated control cells. (C) mRNA levels of DR4/DR5 and c-FLIPL/S in CEM cells and its MDR variants were determined by RT-PCR analysis. β-Actin (Actin) was used as a loading control.

TRAIL-induced apoptosis in MDR cells is mediated through caspase activation and mitochondrial pathway

We determined if the increased susceptibility to TRAIL-induced apoptosis of CEM/VLB100 cells was accompanied by activation of caspases and the mitochondrial apoptotic pathway (Figure 2). Consistent with the more sensitive response of CEM/VLB100 cells to TRAIL-induced cell death, cleavage and activation of procaspase-8 and -10, which are initiator caspases linked to receptor mediated apoptotic pathway, occurred remarkably in CEM/VLB100 cells as compared to CEM cells. In case of caspase-9 that has been linked to the mitochondrial death pathway, treatment of CEM/VLB100 cells with TRAIL resulted in significant induction of proteolytic processing of caspase-9 to its active form compared to CEM cells, which did not show proteolytic cleavage of procaspase-9. Finally, procaspase-3, an executioner caspase, and PARP, a hallmark of caspase activation, were cleaved profoundly in CEM/VLB100 cells compared with CEM cells.

Figure 2
figure 2

Sensitization of MDR cells to TRAIL is mediated through caspase-dependent mitochondrial apoptotic pathway. The cell lysates obtained from CEM or CEM/VLB100 cells after exposure to graded doses of TRAIL (1~10 ng/ml) for 24 h were subjected to western blot analysis to monitor levels of caspase-8, -10, -9 and -3, Bid, truncated Bid (tBid), Bax and Bcl-2. The levels of PARP and its cleavage fragment (CF) in TRAIL-treated cells were also determined.

Meanwhile, we investigated whether the modulation of Bcl-2 family proteins is involved in TRAIL-induced apoptosis of MDR cells. Down-regulation of Bcl-2 and up-regulation of Bax apparently occurred after treatment with TRAIL in CEM/VLB100 cells but not in CEM cells and these results were followed by TRAIL-induced truncation of Bid in CEM/VLB100 cells but not in CEM cells, consistent with activation status of procaspase-9 in CEM/VLB100 cells and CEM cells after treatment with TRAIL. Therefore, these data indicated that TRAIL-induces apoptosis occurred in the MDR cells through caspase-dependent mitochondrial pathway as well as receptor-mediated apoptotic pathway.

The increased expression of DNA-PKcs is associated with up-regulation of P-gp and c-Myc expression via Akt/GSK-3β pathway in MDR cells

It has been reported DNA-PKcs regulates c-Myc stability via phosphorylation of Akt on Ser473, which in turn inactivates GSK-3β by the phosphorylation of GSK-3b on Ser9, resulting in stabilization of c-Myc [28]. The c-Myc is known to be involved in regulating expression of P-gp, the product of MDR1 gene [29, 30] and renders cells sensitive to TRAIL-induced apoptosis [31]. Since the increased activity of DNA-PK participates in the development of MDR phenotype [10], we determined the relationships among P-gp, c-Myc and DNA-PKcs/Akt/GSK-3β molecules in MDR variants (Figure 3). We found that the gradually increased level of P-gp in CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells was well correlated with the level of c-Myc in the each MDR variant. Therefore, we examined whether the basal levels of DNA-PKcs, phosphorylated Akt (pAkt) and phosphorylated GSK-3β (pGSK-3β) in MDR variants were compared with those in parental CEM cells. The basal level of DNA-PKcs was gradually higher in CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells than in CEM cells, which was followed by gradual increase in pAkt and pGSK-3β levels without change in total Akt (tAkt) and GSK-3α/β levels. We also observed that the basal level of DR5 but not DR4 in MDR variants was significantly increased as compared with those in CEM cells. These results suggested that the increased expression of DNA-PKcs in MDR cells might lead to up-regulation of P-gp and c-Myc expression via phosphorylation of Akt and GSK-3β proteins.

Figure 3
figure 3

Expression of p-gp in MDR cells is associated with the enhanced expression of c-Myc and DR5 and activation of DNA-PKcs/Akt/GSK-3β pathway. The protein levels of P-gp, c-Myc, DNA-PKcs, pAkt, GSK-3β, phosphorylated Akt (pAkt) at Ser 473, total Akt (tAkt), phosphorylated GSK-3β (pSK3β) at Ser9, total GSK-3α/β, DR4 and DR5 were determined by western analysis. Actin was used as a loading control.

TRAIL-induced down-regulation of DNA-PKcs/Akt/GSK-3β pathway and c-FLIP and up-regulation of DR4/DR5 cell surface expression are associated with the susceptibility to TRAIL of MDR cells

Since our data showed that the level of DR5 was well correlated with the activity of the DNA-PKcs/Akt/GSK-3β pathway in MDR cells, we determined whether the levels of DNA-PKcs, Akt as a downstream target of DNA-PKcs, and GSK-3b, a downstream target of the Akt pathway, are modulated after treatment of CEM and CEM/VLB100 cells with TRAIL (Figure 4A). When both CEM and CEM/VLB100 cells were treated with TRAIL, the levels of DNA-PKcs, pAkt and pGSK-3b as well as P-gp were significantly decreased in CEM/VLB100 cells as compared with those of CEM cells in a dose-dependent manner. The decrease of pAkt was not followed by decrease of tAkt, but the decrease of pGSK-3b seemed to be associated with decrease of total GSK-3b. These results were followed by down-regulation of Mcl-1, one of the anti-apoptotic Bcl-2 family, that plays a key role in acquired TRAIL resistance [32]. These results suggest that down-regulation of both P-gp and DNA-PKcs/Akt/GSK-3β pathway by TRAIL might be played an important role in death receptor-mediating TRAIL-induced apoptosis in the P-gp over-expressing MDR cells with high expression of c-Myc. Meanwhile, treatment of CEM/VLB100 cells with TRAIL led to a significant suppression of mRNA level of c-FLIPL and c-FLIPS and an increase in surface expression of DR4 and DR5 (Figure 4B). These results suggest that an increase in cell surface expression of DR4/DR5 and down-regulation of c-FLIP and DNA-PKcs/Akt/GSK-3β pathway by TRAIL play an important role in death receptor-mediating TRAIL-induced apoptosis in the MDR cells.

Figure 4
figure 4

TRAIL inhibited P-gp expression and DNA-PKcs/Akt/GSK-3β signaling pathway and up-regulation of death receptors and down-regulation of c-FLIP in MDR cells. (A) Cell lysates obtained from CEM or CEM/VLB100 cells treated with indicated dose of TRAIL for 24 h were subjected to western blot analysis to monitor levels of P-glycoprotein (P-gp), DNA-PKcs, pAkt (Ser473), tAkt, pGSK3β (Ser 9), GSK-3α and -3β, Mcl-1. (B) CEM/VLB100 cells treated with TRAIL (10 ng/ml) for 24 h were incubated on ice in the presence of DR4- and DR5-specific antibodies (1:500), and subsequently labeled with FITC-conjugated secondary antibody (1:1000). The fluorescence intensity was analyzed with flow cytometry. The thin line indicates that the cells only incubated with Goat IgG2a that was used as a control isotype antibody; the thick line indicates the specific labeling (left). The cells treated with or without TRAIL, and changed mRNA level of c-FLIPL/S was determined by RT-PCR analysis (right).

TRAIL inhibited P-gp efflux function via degradation of P-gp and potentiated the cytotoxicity of MDR-related drug in MDR cells

Since P-gp cleavage is dependent on caspase-3 activity following treatment with apoptosis inducers [33], we hypothesized that TRAIL-induced caspase-3 activation in CEM/VLB100 cells may lead to the degradation of P-gp. When the MDR cells were treated with TRAIL, the cleavage of P-gp was accompanied with induction of proteolytic cleavage of procaspase-3 to the active forms. In addition, the cleavage of DNA-PKcs and PARP, well known endogenous substrates of caspase-3 [34, 35], was observed in TRAIL-treated CEM/VLB100 cells (Figure 5A, left). These results were followed by prevention of cleavage of P-gp as well as DNA-PKcs and PARP by pretreatment with Z-DEVD-FMK, a caspase-3-specific inhibitor (Figure 5A, right). Therefore, we determined whether TRAIL inhibited P-gp efflux function of the MDR cells, using a flow cytometric functional efflux assay based on the extrusion of the fluorescent substrate, rhodamine 123 (Figure 5B). P-gp functions optimally near 37°C, but it is inactive at 4°C. When P-gp expressing CEM/VLB100 cells are preloaded with rhodamine 123 and incubated at 4°C, they retain the dye and consequently have high fluorescence levels. But, CEM/VLB100 cells incubated at 37°C more readily efflux the dye and show reduced fluorescence. Treatment with TRAIL significantly reduced rhodamine123 efflux and thereby increased the accumulation of rhodamine123 in CEM/VLB100 cells. When we performed this functional assay also on CEM cells, which does not express P-gp, there were no differences in the fluorescence intensity between the three experimental conditions. Therefore, this result suggests that the inhibitory effect of TRAIL on P-gp efflux function might be due to caspase-3-dependent P-gp cleavage.

Figure 5
figure 5

TRAIL inhibited P-gp efflux function in MDR cells by P-gp cleavage via caspase-3 activation. (A) The cell lysates obtained from CEM/VLB100 cells treated with or without TRAIL (10 ng/ml) for 3 ~ 9 h (left) and the cell lysates of the MDR cells treated with TRAIL (5 ng/ml) for 6 h or pretreated with 50 μM Z-DEVD-FMK, a specific caspase-3 inhibitor, for 3 h and then with TRAIL for 6 h (right) were subjected to western blot analysis to monitor levels of P-gp, DNA-PKcs, caspase-3 and PARP and their its cleavage fragment (CF). (B) flow cytometric assay of P-gp efflux activity in TRAIL-treated MDR cells was based on extrusion of the fluorescent P-gp substrate, rhodamine123 (Rho123). The efflux activity of P-gp is highly temperature sensitive because functions optimally 37°C but is inactive at 4°C. Cell suspension from CEM and CEM/VLB100 cells treated with or without 10 ng/ml TRAIL for 6 h was incubated with Rho 123 and further incubated at 37°C for 3 h (TREATED 37°C as TRAIL-treated cells or CTRL37°C as TRAIL-untreated control) to allow P-gp-mediated drug efflux or on ice as control (CTRL 4°C).

We next examined whether the cytotoxicity of the MDR-related drugs such vinblastine (VLB) and doxorubicin (DOX) could be enhanced by pretreatment of TRAIL (Figure 6A). We found that the cytotoxicity of VLB and DOX was significantly enhanced in CEM/VLB100 cells by pre-treatment with low dose of TRAIL (1 ng/ml). These results suggest that inactivation of P-gp by TRAIL may be a cause of sensitization of CEM/VLB100 cells to MDR-related drugs, and thus the use of TRAIL in combination with MDR-related drug for growth inhibition in MDR cells might be overcome the drug resistance of the MDR cells. In addition, to reveal the synergistic cytotoxic mechanisms of the combined treatment of TRAIL with MDR-related drug against MDR cells, we determined the change of activity of caspase-3 and expression of P-gp, DNA-PKcs and PARP in CEM/VLB100 cells after the combined treatment with MDR-related drug and TRAIL (Figure 6B). As expected, the combined treatment with DOX and TRAIL was more effective than either treatment alone to down-regulate P-gp and DNA-PKcs and to increase subsequent PARP cleavage via caspase-3 activation in the MDR cells. Therefore, these results suggest that TRAIL might be effective for overcoming the MDR phenotype of cancer cells by combination with MDR-related drug.

Figure 6
figure 6

TRAIL enhanced the cytotoxicity of MDR-related drug in MDR cells by the down-regulation of DNA-PKcs and P-gp via caspase-3 activation. (A) CEM/VLB100 cells were treated with graded single doses of doxorubicin (DOX) or vinblastine (VLB) after pretreatment of low dose of TRAIL (1 ng/ml) for 6 h. The percentage of cell survival after combined treatment of TRAIL with MDR-related drug was determined after 5 days incubation using the MTT assay. Each bar represents the mean ± S.D. of triplicate experiments. ***p < 0.005 versus TRAIL alone treated cells at the same dose point. (B) The cell lysates obtained from CEM/VLB100 cells co-treated with TRAIL (1 ng/ml) and indicated dose of DOX were subjected to western blot analysis to monitor levels of P-gp, DNA-PKcs, caspase-3, PARP, and actin as a loading control.

Suppression of DNA-PKcs by siRNA enhanced the susceptibility of MDR cells to MDR-related drug as well as TRAIL via up-regulation of DR5 and down-regulation of c-FLIP and P-gp expression

To determine the role of DNA-PKcs on the expression of c-FLIPL/S and DR4/DR5, which are major determinants of responsiveness to TRAIL, and MDR1, we silenced DNA-PKcs in CEM/VLB100 cells using small interfering RNA (siRNA) and determined the changed mRNA levels of the genes using RT-PCR analysis (Figure 7A). The apparent increase in the mRNA level of DR5 was observed in the MDR cells after transfection with DNA-PKcs siRNA compared with scrambled siRNA, and DR4 also increased slightly. Conversely, the mRNA level of c-FLIPS but not c-FLIPL in CEM/VLB100 cells was significantly reduced after transfection with DNA-PKcs siRNA. We also found that siRNA-mediated silencing of DNA-PKcs significantly down-regulated the expression of MDR1 gene in the MDR cells. Moreover, the increased transcription of DR5 gene was followed by increased cell surface expression of DR5 in CEM/VLB100 cells after transfection with DNA-PKcs siRNA (Figure 7B). These results suggest that the down-regulated DNA-PKcs after treatment with TRAIL may play important roles in the regulation of death receptors and c-FLIP as well as MDR1 gene expression, and the inhibition of DNA-PKcs in MDR cells may enhance the susceptibility to TRAIL as well as MDR drugs via up-regulation of DR5 and down-regulation of c-FLIP and P-gp expression, respectively. In addition, we demonstrated that the inhibition of DNA-PKcs by transfection with DNA-PKcs siRNA caused the reduction of pAkt, pGSK-3β and P-gp levels in CEM/VLB100 cells (Figure 8A). The reduction of pAkt level was not followed by the reduction of total Akt, while the reduction of pGSK-3β level was associated with the reduction of total GSK-3. In addition, suppression of DNA-PKcs led to the decrease in P-gp and c-FLIPs and a concurrent increase in cleaved PARP, which was accelerated by TRAIL. These results suggest that suppression of DNA-PKcs would lead to increased susceptibility to TRAIL-induced cytotoxicity in MDR cells. Therefore, we next examined whether siRNA-mediated suppression of DNA-PKcs affects the susceptibility of CEM/VLB100 cells to TRAIL-induced cytotoxicity. After transfection with DNA-PKcs siRNA or scrambled siRNA, the transfected cells were treated with indicated doses of TRAIL for 5 days. The susceptibility to TRAIL-induced cytotoxicity of CEM/VLB100 cells was significantly increased after transfection with DNA-PKcs siRNA (Figure 8B, left). Furthermore, we also confirmed whether siRNA-mediated suppression of DNA-PKcs affects the susceptibility of CEM/VLB100 cells to vincristine (VCR) (Figure 8B, right). These results suggest that targeting of DNA-PKcs could enhance the susceptibility of MDR-related drug as well as TRAIL on P-gp over-expressing MDR cells with high expression of DNA-PKcs.

Figure 7
figure 7

Supprerssion of DNA-PKcs up-regulated surface expression of DR5 and down-regulated the expression of c-FLIPs. (A) CEM/VLB100 cells were transfected with a siRNA against DNA-PKcs or scrambled siRNA as a control. After 48 h, the total RNA extracted from transfectant of CEM/VLB100 cells performed RT-PCR analysis to monitor the mRNA levels of DNA-PKcs, DR4/5, c-FLIPL/S , MDR1, and actin as a loading control. (B) The transfectant incubated with an anti-DR4 or -DR5 (1:500), and subsequently labeled with FITC-conjugated secondary antibodies (1:1000) to determine the surface expression of DR4 and DR5. Goat IgG2a was also used as control isotype antibody.

Figure 8
figure 8

Potentiated suppression of DNA-PKcs expression by TRAIL leads to a severe inhibition of both Akt/GSK-3β phosphorylation and P-gp and c-FLIPs expression and enhances the cytotoxicity of TRAIL and of MDR-related drug in MDR cells. (A) CEM/VLB100 cells were transfected with a siRNA against DNA-PKcs or scrambled siRNA as a control. After 48 h, the transfectant was treated with or without TRAIL (1- and 10 ng/ml for 24 h), and the changed level of DNA-PKcs, pAkt (Ser473), tAkt, pGSK3β (Ser9), GSK-3α/β, P-gp, PARP, c-FLIPL/S and actin was performed by western blot analysis. (B) Each transfectant was treated with indicated doses of TRAIL or vincristine (VCR). After 5 days, the percentage of growth inhibition was determined incubation using the MTT assay. Data represent means ± S.D. of triplicate experiments. **p < 0.001, ***p < 0.005 versus cells transfected with scrambled siRNA at the same dose point.

Discussion

Although targeted drugs are being developed or used in some leukemia, chemotherapeutic drugs are still useful for the treatment of leukemia. However, acquired resistance against MDR-related drugs is a serious problem in the management of leukemic patients. Altered expression of various kinds of protein and enzymes could be seen in MDR-type cancer cells [2, 9]. In the present study, we suggest a new molecular mechanism that TRAIL down-regulates P-gp through inhibition of DNA-PKcs/Akt/GSK-3β pathway and activation of caspases and thereby sensitize MDR cells to MDR-related drugs.

TRAIL is emerging as most promising agent for cancer therapy, because it induces apoptosis in a variety of cancers and transformed cells without any toxicity to normal cells [20]. But, it has been reported that a majority of human leukemic cells such as CEM, K562 and Molt-4 cells are relatively resistant to TRAIL-induced apoptosis [36, 37]. In our study, interestingly, MDR variants derived from human lymphoblastic leukemia CEM cells showed a hypersensitive response to TRAIL compared with parental CEM cells. MDR variants, CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells with gradually increased levels of P-gp were gradually more susceptible to TRAIL-induced apoptosis and cytotoxicity than CEM cells. This result was supported by the findings that the expression of DR5 was gradually up-regulated in the CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells, and conversely, the expression of c-FLIPs was gradually down-regulated in the MDR variants as compared with those of CEM cells. Therefore, modulation of TRAIL receptor pathway including up-regulation of DR5 and down-regulation of c-FLIPs might contribute to TRAIL sensitization of MDR cells. It has been reported that TRAIL responsiveness correlates with a reduced expression of endogenous Akt in MDR-U2OS human osteosarcoma cell line [24], and P-gp enhances TRAIL-triggered apoptosis by interacting with the death receptor DR5 in the MDR1-transfected MCF-7 breast cancer cell line [25]. Therefore, it could be suggested that a marked sensitivity to TRAIL of MDR cells might be mediated by complex mechanisms, not a single mechanism. In the present study, hypersensitivity to TRAIL of CEM/VLB100 cells, MDR variant of CEM cells, was accompanied by the activation of the mitochondrial apoptotic pathway by the cleavage of bid as well as the activation of caspase-8 and -10, which are apoptotic characteristics of the type II cells and caspase-3 and -9 [38]. We also observed an increase in cell surface expression of DR4/DR5 and down-regulation of c-FLIP by TRAIL in MDR-variant of CEM cells. These results suggest that there might be a positive feedback regulation in TRAIL receptor signaling leading to intensification of sensitivity to TRAIL in MDR-variant of CEM cells.

Oncogene c-Myc is known to act as an important regulator for TRAIL sensitivity in cancer cells. It has been shown that c-Myc induces and represses the transcription of DR5 [39] and c-FLIP [40], respectively, therefore enhancing the sensitivity of cancer cells to TRAIL-induced apoptosis. Recently, it has been reported that abnormal overexpression of DNA-PKcs may contribute to cell proliferation and even oncogenic transformation by stabilizing the c-Myc oncoprotein via at least the Akt/GSK3 pathway [28]. Previously, we have demonstrated that the increased expression of DNA-PKcs is associated with the development of drug resistance in MDR variants of CEM cells [10]. In addition, the c-Myc is known to be involved in regulating expression of P-gp, the product of MDR1 gene [29, 30]. It has been reported that elevated P-gp expression in MDR cells is accompanied by increased level of pAkt [41]. Once phosphorylated, activated Akt inactivate GSK-3β through phosphorylation at Ser9, resulting in stabilization and activation of β-catenin that enhanced P-gp expression [42]. In the present study, the gradually increased level of P-gp, was well correlated with the gradually increased levels of c-Myc, DNA-PKcs, pAkt and pGSK-3β in MDR variants, CEM/VLB10-2, CEM/VLB55-8 and CEM/VLB100 cells, suggesting that the molecular changes are not dependent on the each subline type, but implicate the causal relationships between the molecules, which have been changed during the process of MDR acquisition. And the increased level of DR5 and decreased level of c-FLIPs in the MDR-variants of CEM cells also might be associated with the up-regulated c-Myc since it has been reported that c-Myc up-regulated the DR5 receptor and down-regulated c-FLIP [39, 40]. We also found that the expression of up-regulated molecules in CEM/VLB100 cells including P-gp, DNA-PKcs, pAkt and pGSK-3β were suppressed after treatment with TRAIL. Akt and GSK-3β are signaling molecules downstream to DNA-PKcs. We showed that the phosphorylated form of Akt and GSK-3β would be decreased in TRAIL-treated CEM/VLB100 cells since DNA-PKcs was down-regulated by TRAIL treatment. Therefore, our data indicated that TRAIL caused the down-regulation of P-gp in MDR cells by the inactivation of DNA-PKcs/Akt/GSK-3b pathway. Since these molecules are related with drug-resistance, down-regulation of P-gp, DNA-PKcs, pAkt, and pGSK-3β after treatment with TRAIL might lead to the hypersensitivity to MDR-related drugs of MDR-variant of CEM cells. Indeed, inhibition of Akt enhances susceptibility to TRAIL by up-regulation of death receptors [43] and down-regulation of c-FLIP [44] and down-regulates P-gp expression in multidrug-resistant human T-acute leukemia [14].

Our study also showed that anti-apoptotic Bcl-2 and Mcl-1 proteins were over-expressed in CEM/VBL100 cells and the levels of these proteins and Bax were significantly decreased and increased by Bcl-2 and Mcl-1, the antiapoptotic Bcl-2 family proteins, were over-expressed in CEM/VBL100 cells in comparison with CEM cells, and the levels of these anti-apoptotic proteins and Bax were significantly decreased and increased by treatment of TRAIL in CEM/VBL100 cells, respectively, suggesting that TRAIL-induced apoptosis of MDR cells was mediated through mitochondria-dependent pathway as well as caspase activation. Bcl-2 and Mcl-1 are often highly expressed in chemotherapy-resistant cancers and prevents apoptosis by inactivating pro-apoptotic Bax and Bak [45, 46]. The increased expression of Bcl-2 or Bcl-xL was the common feature of P-gp-related drug-resistant human leukemic ce1l lines [47]. Over-expression of Mcl-1 decreased sensitivity of leukemia cells to cytotoxic chemotherapeutic agents [45] and specific down-regulation of Mcl-1 via RNA interference sensitized multidrug-resistant leukemia cells towards chemotherapy and induced apoptosis [48]. Therefore, the reduction of Bcl-2 and Mcl-1 after exposure to TRAIL may be in part a cause of TRAIL-induced sensitization of CEM/VLB100 cells to MDR-related drugs.

Moreover, our data showed the cleavage of P-gp and DNA-PKcs by treatment with TRAIL. Recently, it has been shown that the cleavage of P-gp (170 kDa) is dependent on caspase-3 during apoptotic cell death induced by LY294002, H2O2, and Z-LEHD-FMK in MDR variant of CEM cells [33]. DNA-PKcs is also a substrate of caspase-3 [35]. Here, we demonstrated that the degradation of P-gp and DNA-PKcs during treatment of CEM/VLB100 cells with TRAIL was a caspase-3 dependent manner. This result was followed by the significant reduction of rhodamine123 efflux and the increased sensitivity to MDR-related drugs such as VLB and DOX after exposure to TRAIL in CEM/VLB100 cells, suggesting that the degradation of P-gp as well as the down-regulation of Bcl-2 and Mcl-1 could be involved in sensitization of MDR cells to MDR-related drug after treatment with TRAIL. Since the expression of DNA-PKcs regulates MDR1 gene [12] and cellular c-Myc protein levels [28], which can affect TRAIL sensitivity in cancer cells as described above, we showed that knockdown of DNA-PKcs with specific siRNA led to the increased expression of DR5 and the decreased expression of c-FLIP and caused the reduction of pAkt, pGSK-3β and P-gp levels in CEM/VBL100 cells. These results resembled the effects of TRAIL on the CEM/VBL100 cells. Therefore, DNA-PKcs may play an important role on TRAIL sensitivity in MDR-variant of CEM cells.

Conclusions

In conclusion, this study showed for the first time that the MDR-variants of CEM cells with an increased P-gp and c-Myc were hypersensitive to TRAIL and that the degradation of both P-gp and DNA-PKcs after exposure to TRAIL might be an important determinant of susceptibility to TRAIL-induced apoptosis in MDR cells. In addition, the treatment with TRAIL caused severe down-regulation of the up-regulated P-gp and DNA-PKcs in MDR cells and consequently sensitized the MDR cells to MDR-related drugs. Therefore, combination of TRAIL and chemotherapeutic drugs may be a good strategy for treatment of cancer with multidrug resistance, and DNA-PKcs/Akt/GSK-3β signaling pathway may be an important target to overcome multidrug resistance.

Methods

Cell culture and Reagents

Human lymphoblastic leukemia CCRF-CEM (CEM) line and its the multidrug-resistant sublines, CEM/VLB10-2, CEM/VLB55-8, and CEM/VLB100 [10], were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS, GIBCO BRL, Life Technologies, Inc.). The recombinant human soluble TRAIL was purchased from R&D System (Minneapolis, MN). Vinblastine, vincristine, doxorubicin and Rho123 were obtained from Sigma-Aldrich (St. Louis, MO).

Cell Proliferation Assay

Cell proliferation was measured either by counting viable cells by using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma Chemical Company, St. Louis, MO) colorimetric dye-reduction method. Exponentially growing cells (5 × 103 cells/well) were plated in 96 well and incubated in growth medium treated with indicated condition of TRAIL and/or drug at 37°C. After 5 days, the medium was aspirated using centrifugation and MTT-formazan crystals solubilized in 100 μl DMSO. The optical density of each sample at 570 nm was measured using ELISA reader. The optical density of the media was proportional to the number of viable cells. Inhibition of proliferation was evaluated as a percentage of control growth (no drug in the sample). All experiments were repeated at least two experiments in triplicate.

Flow cytometric analysis of TRAIL receptors

CEM/VLB100 cells (2 × 106 cells) from the culture media were spun down at 500 × g, washed with phosphate-buffered saline (PBS) and resuspended in 500 μl PBS. The cells were then incubated with 5 μl of goat IgG2a, anti-DR4 or anti-DR5 polyclonal goat antibody (1:100, R&D, Minneapolis, MN) for 1 h. After washing with PBS, FITC-conjugated rabbit anti-goat polyclonal antibody (1:200, Sigma-Aldrich Co., St. Louis, MO) was added to the cell suspension and incubated for 1 h on ice. After rinsing with PBS, the samples were analyzed with a FACSort flow cytometer (Becton Dickinson, San Jose, CA). The data were analyzed using the CellQuest program.

RT-PCR analysis

Total cellular RNA was isolated using RNeasy Mini Kit (Qiagen, Hilden, Germany) according to the manufacturer's protocol and the levels of RNA transcripts were assessed with The Titan One Tube RT-PCR System (MJ research Inc, NV, USA). One mg of total cellular RNA was reverse transcribed using Maloney murine leukemia virus reverse transcriptase (Invitrogen, Paisley, UK) with each dNTP and 1 μg oligo dT. Amplification of 1 μl of these cDNA by PCR was performed using the following gene-specific primers: DR4 (forward), 5'-CTGAGCAACGCAGACTCGCTGTCCAC-3' and (reverse), 5'-AAGGACACGGCAGAGCCT GTGCCAT-3'; DR5 (forward), 5'-CTGAAAGGCATCTGCTCAGGTG-3' and (reverse), 5'-CA GAGTCTGCATTACCTTCTA G-3'; FLIPL (forward), 5'-TTCCAGGCTTT CGGTTTCTT-3' and (reverse), 5'-GTCCGAAACAAGGTGAGGGT-3'; FLIPS (forward), 5'-ACCCTCACCTTG TTTCGGAC-3' and (reverse), 5'-CTTTTGGATTGCTGCTTGGA-3'; β-actin (forward), 5'-CAGAGCAAGAGAGGCATCCT-3' and (reverse), 5'-TTGAAGGTCTC AAACATGAT-3.'

The resulting total cDNA was used in PCR performed in total volume of 20 μl using Taq polymerase (Solgent Co., Korea) at 94°C for denaturation for 60 sec, 60°C for annealing for 60 sec, and 72°C for amplification for 90 sec for 30 cycles, followed by a final extension at 72°C for 12 min. The amplified fragments were separated on 1.5% agarose gel and visualized with ethidium bromide staining.

Western blot analysis

Protein samples were separated by SDS-PAGE and blotted to nitrocellulose membrane (Hybond-ECL, GE Healthcare). The membrane was incubated with antibody as specified, followed by secondary antibody conjugated with horseradish peroxidase. Specific antigen-antibody complexes were detected by enhanced chemiluminescence (PerkinElmer, Life science). Western blot analysis was performed with the following antibodies: anti-Bax, anti-caspase-3, anti-PARP, anti-Bcl-2 (Santa Cruz Biotechnology, CA), anti-Akt, anti-phospho-Akt (Ser 473), anti-caspase-8, anti-caspase-9 (Cell signal, Danvers, MA), anti-DNA-PKcs (Thermo Fisher Scientific, CA), anti-DR5, anti-caspase-10 (Calbiochem, Germany), anti-pGSK-3β (Ser8), anti-GSK-3β, anti-GSK-3α, anti-c-Myc (Epitomics, CA), anti-DR4 (R&D Systems, MN) and anti β-actin (Sigma-Aldrich) antibodies, Secondary antibodies were obtained from GE Healthcare.

Preparation of siRNA Transfection

The siRNA used for targeted silencing of DNA-PKcs were (5'-CAGUCUUAGUCCGGAUCAUdTdT-3'). CEM/VLB100 cells were transfected with 0.1 uM siRNA for 48 h by oligofectamine according to the manufacture's protocol (InVitrogen, Carlsbad, CA). In brief, CEM/VLB100 (2 × 105 cells/well) were seeded of 6-well plates and added to the siRNA/oligofectamine complex. Cells were incubated for 4 h at 37°C in serum free RPMI medium and then FBS was added. After 48 h, the cells were treated with TRAIL for another 24 h and collected for western blot analysis to determine the levels of DNA-PKcs and other indicated proteins.

Apoptosis assay

Cells (2 × 105 cells/ml) were treated with or without TRAIL and/or indicated drug for 24 h and the cells were centrifuged and resuspended in 500 μl of the staining solution containing Annexin V fluorescein (FITC Apoptosis detection kit; BD ParMingen San Diego, CA) and propidium iodide in PBS. After incubation at room temperature for 15 min, cells were analyzed by flow cytometry. Annexin V binds to those cells that express phosphatidyl serine on the outer layer of the cell membrane, and propidium iodide stains the cellular DNA of those cells that have a compromised cell membrane. This allows for the discrimination of live cells (unstained with either fluorochrome) from apoptotic cells (stained only with Annexin V) and necrotic cells (stained with both Annexin V and propidium iodide).

Flow-cytometric dye-efflux assay for multidrug resistance

The accumulation of rhodamine 123, a fluorescent substrate of P-gp, in CEM and CEM/VLB100 cells treated with or without TRAIL was measured using a FACS flowcytometer (FACScalibur, BD Biosciences, San Jose, CA) equipped with an ultraviolet argon laser (excitation at 488 nm and emission at 530 ± 15 nm). Cell suspension (500 μl) from CEM and CEM/VLB100 cells treated with or without 10 ng/ml TRAIL for 6 h was incubated with rhodamine 123 (0.5 μg/ml) at 37°C for 30 min. After incubation, the cells were washed with ice-cold PBS and further incubated at 37°C for 3 h to allow P-gp-mediated drug efflux or on ice (4°C) as control. Cells were pelleted by centrifugation at 500 × g and resuspended in PBS containing. Cellular fluorescence was analyzed immediately by using Flow cytometer.

Statistical analysis

The results obtained were expressed as the mean ± S.E. of at least three independent experiments. The statistical significance of differences assessed using the Student's t-test. *p < 0.05, **p < 0.01, ***p < 0.005 was considered statistically significant in all experiments.

Abbreviations

P-GP:

P-GlycoProtein, tumor necrosis factor-related apoptosis-inducing ligand

TRAIL:

C-FLIP, cellular FLICE inhibitory protein

MDR:

multidrug resistance

PI3K:

phosphoinositide 3-kinase

PAKT:

DNA-PKcs, DNA-dependent protein kinase catalytic subunit, phosphorylated Akt, tAkt, total Akt

GSK-3α/β:

glycogen synthase kinase-3α/β

PGSK-3β:

phosphorylated GSK-3β, siRNA: small interfering RNA

CEM:

human lymphoblastic leukemia

VCR:

vincristine: VLB: vinblastine

DOX:

doxorubicin.

References

  1. Gottesman MM, Fojo T, Bates SE: Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002, 2: 48-58. 10.1038/nrc706

    Article  CAS  PubMed  Google Scholar 

  2. Tsuruo T, Naito M, Tomida A, Fujita N, Mashima T, Sakamoto H, Haga N: Molecular targeting therapy of cancer: drug resistance, apoptosis and survival signal. Cancer Sci. 2003, 94: 15-21. 10.1111/j.1349-7006.2003.tb01345.x

    Article  CAS  PubMed  Google Scholar 

  3. Tan B, Piwnica-Worms D, Ratner L: Multidrug resistance transporters and modulation. Curr Opin Oncol. 2000, 12: 450-458. 10.1097/00001622-200009000-00011

    Article  CAS  PubMed  Google Scholar 

  4. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM: Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006, 5: 219-234. 10.1038/nrd1984

    Article  CAS  PubMed  Google Scholar 

  5. Trock BJ, Leonessa F, Clarke R: Multidrug resistance in breast cancer: a meta-analysis of MDR1/gp170 expression and its possible functional significance. J Natl Cancer Inst. 1997, 89: 917-931. 10.1093/jnci/89.13.917

    Article  CAS  PubMed  Google Scholar 

  6. Norgaard JM, Bukh A, Langkjer ST, Clausen N, Palshof T, Hokland P: MDR1 gene expression and drug resistance of AML cells. Br J Haematol. 1998, 100: 534-540. 10.1046/j.1365-2141.1998.00593.x

    Article  CAS  PubMed  Google Scholar 

  7. Steinbach D, Legrand O: ABC transporters and drug resistance in leukemia: was P-gp nothing but the first head of the Hydra?. Leukemia. 2007, 21: 1172-1176. 10.1038/sj.leu.2404692

    Article  CAS  PubMed  Google Scholar 

  8. Ivy SP, Olshefski RS, Taylor BJ, Patel KM, Reaman GH: Correlation of P-glycoprotein expression and function in childhood acute leukemia: a children's cancer group study. Blood. 1996, 88: 309-318.

    CAS  PubMed  Google Scholar 

  9. Li X, Li JP, Yuan HY, Gao X, Qu XJ, Xu WF, Tang W: Recent advances in P-glycoprotein-mediated multidrug resistance reversal mechanisms. Methods Find Exp Clin Pharmacol. 2007, 29: 607-617. 10.1358/mf.2007.29.9.1139054

    Article  CAS  PubMed  Google Scholar 

  10. Kim SH, Um JH, Dong-Won B, Kwon BH, Kim DW, Chung BS, Kang CD: Potentiation of chemosensitivity in multidrug-resistant human leukemia CEM cells by inhibition of DNA-dependent protein kinase using wortmannin. Leuk Res. 2000, 24: 917-925. 10.1016/S0145-2126(00)00061-8

    Article  CAS  PubMed  Google Scholar 

  11. Bolderson E, Richard DJ, Zhou BB, Khanna KK: Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair. Clin Cancer Res. 2009, 15: 6314-6320. 10.1158/1078-0432.CCR-09-0096

    Article  CAS  PubMed  Google Scholar 

  12. Zhong X, Safa AR: Phosphorylation of RNA helicase A by DNA-dependent protein kinase is indispensable for expression of the MDR1 gene product P-glycoprotein in multidrug-resistant human leukemia cells. Biochemistry. 2007, 46: 5766-5775. 10.1021/bi700063b

    Article  CAS  PubMed  Google Scholar 

  13. West KA, Castillo SS, Dennis PA: Activation of the PI3K/Akt pathway and chemotherapeutic resistance. Drug Resist Updat. 2002, 5: 234-248. 10.1016/S1368-7646(02)00120-6

    Article  CAS  PubMed  Google Scholar 

  14. Chiarini F, Del Sole M, Mongiorgi S, Gaboardi GC, Cappellini A, Mantovani I, Follo MY, McCubrey JA, Martelli AM: The novel Akt inhibitor, perifosine, induces caspase-dependent apoptosis and downregulates P-glycoprotein expression in multidrug-resistant human T-acute leukemia cells by a JNK-dependent mechanism. Leukemia. 2008, 22: 1106-1116. 10.1038/leu.2008.79

    Article  CAS  PubMed  Google Scholar 

  15. Choi BH, Kim CG, Lim Y, Shin SY, Lee YH: Curcumin down-regulates the multidrug-resistance mdr1b gene by inhibiting the PI3K/Akt/NF kappa B pathway. Cancer Lett. 2008, 259: 111-118. 10.1016/j.canlet.2007.10.003

    Article  CAS  PubMed  Google Scholar 

  16. Zhang Y, Qu X, Hu X, Yang X, Hou K, Teng Y, Zhang J, Sada K, Liu Y: Reversal of P-glycoprotein-mediated multi-drug resistance by the E3 ubiquitin ligase Cbl-b in human gastric adenocarcinoma cells. J Pathol. 2009, 218: 248-255. 10.1002/path.2533

    Article  CAS  PubMed  Google Scholar 

  17. Bozulic L, Surucu B, Hynx D, Hemmings BA: PKBalpha/Akt1 acts downstream of DNA-PK in the DNA double-strand break response and promotes survival. Mol Cell. 2008, 30: 203-213. 10.1016/j.molcel.2008.02.024

    Article  CAS  PubMed  Google Scholar 

  18. Feng J, Park J, Cron P, Hess D, Hemmings BA: Identification of a PKB/Akt hydrophobic motif Ser-473 kinase as DNA-dependent protein kinase. J Biol Chem. 2004, 279: 41189-41196. 10.1074/jbc.M406731200

    Article  CAS  PubMed  Google Scholar 

  19. Beurel E, Jope RS: The paradoxical pro- and anti-apoptotic actions of GSK3 in the intrinsic and extrinsic apoptosis signaling pathways. Prog Neurobiol. 2006, 79: 173-189. 10.1016/j.pneurobio.2006.07.006

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Srivastava RK: TRAIL/Apo-2L: mechanisms and clinical applications in cancer. Neoplasia. 2001, 3: 535-546. 10.1038/sj.neo.7900203

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Falschlehner C, Emmerich CH, Gerlach B, Walczak H: TRAIL signalling: decisions between life and death. Int J Biochem Cell Biol. 2007, 39: 1462-1475. 10.1016/j.biocel.2007.02.007

    Article  CAS  PubMed  Google Scholar 

  22. Day TW, Huang S, Safa AR: c-FLIP knockdown induces ligand-independent DR5-, FADD-, caspase-8-, and caspase-9-dependent apoptosis in breast cancer cells. Biochem Pharmacol. 2008, 76: 1694-1704. 10.1016/j.bcp.2008.09.007

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  23. Nam SY, Amoscato AA, Lee YJ: Low glucose-enhanced TRAIL cytotoxicity is mediated through the ceramide-Akt-FLIP pathway. Oncogene. 2002, 21: 337-346. 10.1038/sj.onc.1205068

    Article  CAS  PubMed  Google Scholar 

  24. Cenni V, Maraldi NM, Ruggeri A, Secchiero P, Del Coco R, De Pol A, Cocco L, Marmiroli S: Sensitization of multidrug resistant human ostesarcoma cells to Apo2 Ligand/TRAIL-induced apoptosis by inhibition of the Akt/PKB kinase. Int J Oncol. 2004, 25: 1599-1608.

    PubMed  Google Scholar 

  25. Park SJ, Wu CH, Choi MR, Najafi F, Emami A, Safa AR: P-glycoprotein enhances TRAIL-triggered apoptosis in multidrug resistant cancer cells by interacting with the death receptor DR5. Biochem Pharmacol. 2006, 72: 293-307. 10.1016/j.bcp.2006.04.024

    Article  CAS  PubMed  Google Scholar 

  26. Krishna R, Mayer LD: Multidrug resistance (MDR) in cancer. Mechanisms, reversal using modulators of MDR and the role of MDR modulators in influencing the pharmacokinetics of anticancer drugs. Eur J Pharm Sci. 2000, 11: 265-283. 10.1016/S0928-0987(00)00114-7

    Article  CAS  PubMed  Google Scholar 

  27. Safa AR, Day TW, Wu CH: Cellular FLICE-like inhibitory protein (C-FLIP): a novel target for cancer therapy. Curr Cancer Drug Targets. 2008, 8: 37-46. 10.2174/156800908783497087

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. An J, Yang DY, Xu QZ, Zhang SM, Huo YY, Shang ZF, Wang Y, Wu DC, Zhou PK: DNA-dependent protein kinase catalytic subunit modulates the stability of c-Myc oncoprotein. Mol Cancer. 2008, 7: 32- 10.1186/1476-4598-7-32

    Article  PubMed Central  PubMed  Google Scholar 

  29. Nakamura Y, Sato H, Motokura T: Development of multidrug resistance due to multiple factors including P-glycoprotein overexpression under K-selection after MYC and HRAS oncogene activation. Int J Cancer. 2006, 118: 2448-2454. 10.1002/ijc.21691

    Article  CAS  PubMed  Google Scholar 

  30. He Y, Zhang J, Yuan Y: The role of c-myc in regulating mdr1 gene expression in tumor cell line KB. Chin Med J (Engl). 2000, 113: 848-851.

    CAS  Google Scholar 

  31. Klefstrom J, Verschuren EW, Evan G: c-Myc augments the apoptotic activity of cytosolic death receptor signaling proteins by engaging the mitochondrial apoptotic pathway. J Biol Chem. 2002, 277: 43224-43232. 10.1074/jbc.M206967200

    Article  CAS  PubMed  Google Scholar 

  32. Zhang J, Hawkins SF, Glenn MA, Lin K, Johnson GG, Carter A, Cawley JC, Pettitt AR: Akt is activated in chronic lymhocytic leukemia cells and delivers a pro-survival signal: the therapeutic potential of Akt inhibition. Haematologica. 2010, 95: 110-118. 10.3324/haematol.2009.010272

    Article  Google Scholar 

  33. Mantovani I, Cappellini A, Tazzari PL, Papa V, Cocco L, Martelli AM: Caspase-dependent cleavage of 170-kDa P-glycoprotein during apoptosis of human T-lymphoblastoid CEM cells. J Cell Physiol. 2006, 207: 836-844. 10.1002/jcp.20628

    Article  CAS  PubMed  Google Scholar 

  34. Affar EB, Germain M, Winstall E, Vodenicharov M, Shah RG, Salvesen GS, Poirier GG: Caspase-3-mediated processing of poly(ADP-ribose) glycohydrolase during apoptosis. J Biol Chem. 2001, 276: 2935-2942. 10.1074/jbc.M007269200

    Article  CAS  PubMed  Google Scholar 

  35. Graham KL, Gustin KE, Rivera C, Kuyumcu-Martinez NM, Choe SS, Lloyd RE, Sarnow P, Utz PJ: Proteolytic cleavage of the catalytic subunit of DNA-dependent protein kinase during poliovirus infection. J Virol. 2004, 78: 6313-6321. 10.1128/JVI.78.12.6313-6321.2004

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Park SJ, Kim MJ, Kim HB, Sohn HY, Bae JH, Kang CD, Kim SH: Cotreatment with apicidin overcomes TRAIL resistance via inhibition of Bcr-Abl signaling pathway in K562 leukemia cells. Exp Cell Res. 2009, 315: 1809-1818. 10.1016/j.yexcr.2009.02.024

    Article  CAS  PubMed  Google Scholar 

  37. Lee NS, Cheong HJ, Kim SJ, Kim SE, Kim CK, Lee KT, Park SK, Baick SH, Hong DS, Park HS, Won JH: Ex vivo purging of leukemia cells using tumor-necrosis-factor-related apoptosis-inducing ligand in hematopoietic stem cell transplantation. Leukemia. 2003, 17: 1375-1383. 10.1038/sj.leu.2402960

    Article  CAS  PubMed  Google Scholar 

  38. Nakamura H, Kawakami A, Iwamoto N, Ida H, Koji T, Eguchi K: Rapid and significant induction of TRAIL-mediated type II cells in apoptosis of primary salivary epithelial cells in primary Sjogren's syndrome. Apoptosis. 2008, 13: 1322-1330. 10.1007/s10495-008-0261-2

    Article  CAS  PubMed  Google Scholar 

  39. Wang Y, Engels IH, Knee DA, Nasoff M, Deveraux QL, Quon KC: Synthetic lethal targeting of MYC by activation of the DR5 death receptor pathway. Cancer Cell. 2004, 5: 501-512. 10.1016/S1535-6108(04)00113-8

    Article  CAS  PubMed  Google Scholar 

  40. Ricci MS, Jin Z, Dews M, Yu D, Thomas-Tikhonenko A, Dicker DT, El-Deiry WS: Direct repression of FLIP expression by c-myc is a major determinant of TRAIL sensitivity. Mol Cell Biol. 2004, 24: 8541-8555. 10.1128/MCB.24.19.8541-8555.2004

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Han Z, Hong L, Han Y, Wu K, Han S, Shen H, Li C, Yao L, Qiao T, Fan D: Phospho Akt mediates multidrug resistance of gastric cancer cells through regulation of P-gp, Bcl-2 and Bax. J Exp Clin Cancer Res. 2007, 26: 261-268.

    CAS  PubMed  Google Scholar 

  42. Lim JC, Kania KD, Wijesuriya H, Chawla S, Sethi JK, Pulaski L, Romero IA, Couraud PO, Weksler BB, Hladky SB, Barrand MA: Activation of beta-catenin signalling by GSK-3 inhibition increases p-glycoprotein expression in brain endothelial cells. J Neurochem. 2008, 106: 1855-1865.

    PubMed Central  CAS  PubMed  Google Scholar 

  43. David E, Sinha R, Chen J, Sun SY, Kaufman JL, Lonial S: Perifosine synergistically enhances TRAIL-induced myeloma cell apoptosis via up-regulation of death receptors. Clin Cancer Res. 2008, 14: 5090-5098. 10.1158/1078-0432.CCR-08-0016

    Article  CAS  PubMed  Google Scholar 

  44. Nam SY, Jung GA, Hur GC, Chung HY, Kim WH, Seol DW, Lee BL: Upregulation of FLIP(S) by Akt, a possible inhibition mechanism of TRAIL-induced apoptosis in human gastric cancers. Cancer Sci. 2003, 94: 1066-1073. 10.1111/j.1349-7006.2003.tb01402.x

    Article  CAS  PubMed  Google Scholar 

  45. Gul O, Basaga H, Kutuk O: Apoptotic blocks and chemotherapy resistance: strategies to identify Bcl-2 protein signatures. Brief Funct Genomic Proteomic. 2008, 7: 27-34. 10.1093/bfgp/eln002

    Article  CAS  PubMed  Google Scholar 

  46. Del Poeta G, Bruno A, Del Principe MI, Venditti A, Maurillo L, Buccisano F, Stasi R, Neri B, Luciano F, Siniscalchi A: Deregulation of the mitochondrial apoptotic machinery and development of molecular targeted drugs in acute myeloid leukemia. Curr Cancer Drug Targets. 2008, 8: 207-222. 10.2174/156800908784293640

    Article  CAS  PubMed  Google Scholar 

  47. Nuessler V, Stotzer O, Gullis E, Pelka-Fleischer R, Pogrebniak A, Gieseler F, Wilmanns W: Bcl-2, bax and bcl-xL expression in human sensitive and resistant leukemia cell lines. Leukemia. 1999, 13: 1864-1872. 10.1038/sj/leu/2401571

    Article  CAS  PubMed  Google Scholar 

  48. Ji M, Li J, Yu H, Ma D, Ye J, Sun X, Ji C: Simultaneous targeting of MCL1 and ABCB1 as a novel strategy to overcome drug resistance in human leukaemia. Br J Haematol. 2009, 145: 648-656. 10.1111/j.1365-2141.2009.07678.x

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by Mid-career Researcher Program through NRF grant funded by the MEST (No. 2009-0084444).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Chi-Dug Kang or Sun-Hee Kim.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

SBS, JGH, MJK, JWL, HBK and JHB designed and conducted experiments as well data analysis. DWK participated in discussion of the data and draft of the manuscript. SHK and CD K equally participated in experimental design, coordination, data analysis and draft of the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Seo, SB., Hur, JG., Kim, MJ. et al. TRAIL sensitize MDR cells to MDR-related drugs by down-regulation of P-glycoprotein through inhibition of DNA-PKcs/Akt/GSK-3β pathway and activation of caspases. Mol Cancer 9, 199 (2010). https://doi.org/10.1186/1476-4598-9-199

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1476-4598-9-199

Keywords