Inhibition of autophagy by chloroquine enhances the antitumor activity of gemcitabine for gallbladder cancer
Fang‑Tao Wang · Hui Wang1 · Qi‑Wei Wang · Mu‑Su Pan1 · Xin‑Ping Li1 · Wei Sun · Yue‑Zu Fan1
Abstract
Gemcitabine (GEM), as an anti-metabolic nucleoside analog, has been shown to have anticancer effects in various tumors, but its chemotherapy resistance is still an important factor leading to poor prognosis of cancer patient. A large number of studies in recent years have shown that autophagy plays an important role in the chemotherapy sensitivity of many tumors, including pancreatic, non-small cell lung, and bladder cancer. However, whether GEM causes autophagy in gallbladder cancer (GBC) and whether it is related to chemotherapy resistance is unknown. In the present study, we demonstrated that GEM induced apoptosis and protective autophagy in GBC cells, which may be related to the AKT/mTOR signaling pathway, and GEM in combination with autophagy inhibitor chloroquine can strengthen the cytotoxic effect of GEM on GBC in vitro and in vivo. These findings showed that both autophagy and AKT/mTOR signals were engaged in GBC cell death evoked by GEM, GBC patients might benefit from this new treatment strategy, and molecular targeted treatment in combination with autophagy inhibitors shows promise as a treatment improvement.
Keywords Gallbladder neoplasm · Gemcitabine · Autophagy · Apoptosis
Introduction
Gallbladder cancer (GBC) is the most common malignant tumor of the biliary tract, and its incidence shows an obvious increasing trend [1, 2]. The early diagnosis of GBC is difficult because of its concealed symptoms. Most patients are diagnosed with advanced tumors and lost the best time for surgery [3]. Even if some patients have the opportunity to undergo surgery, recurrence is very easy after surgery, leading to a poor prognosis of GBC patients. Therefore, for patients with advanced GBC who have lost the opportunity for surgery or recurred after surgery, palliative adjuvant therapy such as chemotherapy is often chosen.
Recently, the National Comprehensive Cancer Network guidelines recommend gemcitabine (GEM) as a first-line chemotherapy drug for the treatment of unresectable hepatobiliary tumors, and also as adjuvant therapy for resectable tumors [4]. GEM is a deoxypyrimidine analog with anti-metabolic effects and inhibition of DNA synthesis. It has been approved as a clinical anticancer drug in many countries [4] and is widely used to treat various advanced malignancies that have lost the opportunity for surgery, including advanced pancreatic cancer, advanced non-small cell lung cancer, and metastatic or local bladder cancer.
[5–7]. Based on recent clinical trials, GEM combined with cisplatin (GEMCIS) has a better effect on biliary tract cancers than GEM alone. The median overall survival (OS) and progression-free survival (PFS) can reach 11.7 months and 8.1 months, respectively [8], so GEMCIS has been recommended as a first-line chemotherapy drug for unresectable biliary tract tumors. However, it has been reported that the prognosis and response to chemotherapy of GBC are poor compared with those of other biliary tract cancer subtypes (extrahepatic bile duct cancer) [9]. A phase II clinical survey involving only unresectable GBC showed that these patients had a median OS and PFS of only 6.2 and 3.1 months, respectively [10]. It has also been reported that GEM combined with tumor angiogenesis inhibitors (bevacizumab or sorafenib) in the treatment of advanced biliary tract cancer had no significant improvement compared with GEM alone [11, 12]. In the clinical observation of adjuvant chemotherapy for resectable biliary tract cancer, compared with GEM alone, GEMCIS did not show a significant advantage, either [13]. Clinical practice has shown that chemotherapy resistance is an important factor affecting the efficacy of GEM in the treatment of advanced tumors [7, 14]. These results also indicate that chemotherapy resistance attenuates the therapeutic effect of GEM on GBC. Therefore, it is urgent to further elucidate the molecular mechanism of chemotherapy resistance in GBC to improve the effect of chemotherapy.
In recent years, a large number of studies have shown that autophagy plays two distinct roles in tumor chemotherapy resistance and chemotherapy sensitization. On the one hand, moderate autophagy can increase the resistance of tumor cells to chemotherapeutic drugs; on the other hand, excessive autophagy can inhibit the formation of drug resistance in tumor cells, thereby accelerating cell death [15]. These results provide a new idea for guiding reasonable clinical chemotherapy. Many studies have pointed out that autophagy can enhance the resistance of pancreatic cancer cells to GEM through various molecular mechanisms [16–22], and GEM-induced autophagy can reduce its proapoptotic effect on lung cancer cells [23] and breast cancer cells [24]. These studies show that GEM can cause cytoprotective autophagy in a variety of tumors, increase tumor chemotherapy resistance, and affect the efficacy of chemotherapy [25]. It is interesting to note that many studies have pointed out that the combined application of autophagy inhibitors such as chloroquine (CQ) or interference with autophagy gene expression can significantly increase the induction of tumor cell apoptosis by GEM [26–28]. Cell autophagy and its autophagy gene suppression or application of autophagy inhibitors may be expected to solve the problem of chemotherapy resistance of tumors. However, it has not been fully investigated whether GEM can cause protective autophagy of GBC cells, whether it is related to the resistance of GBC to GEM, and whether the combined use of autophagy inhibitor CQ can improve the sensitivity of GEM to GBC chemotherapy.
In the present study, we confirmed that GEM can induce apoptosis and protective autophagy in GBC cells, which may be related to the AKT/mTOR signaling pathway. At the same time, GEM in combination with autophagy inhibitor CQ can strengthen the cytotoxic effect of GEM on GBC in vitro and in vivo. These findings provide new ideas and evidence for the study of chemotherapy resistance of GBC and the improvement of its efficacy.
Materials and methods
Cell culture and reagents
Human GBC cell lines GBC-SD and NOZ were purchased from Shanghai Institute of Cell Research, Chinese Academy of Sciences, and SGC-996 was donated by Professor Yaoqin Yang from the Cancer Cell Institute of Tongji University School of Medicine. The GBC-SD and SGC-996 were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Gibco, USA), while the NOZ was maintained in DMEM medium supplemented with 10% fetal bovine serum (Gibco, USA), at 37 °C in 5% C O2 (Shanghai Medical Instruments, China). GEM was obtained from Haosen Medicine Co. Ltd. (Jiangsu, China), dissolved in DMSO and diluted with RPMI-1640 or DMEM medium to the desired concentration for in vitro studies. CQ (Sigma, C6628, USA) was dissolved in PBS and diluted with RMPI-1640 or DMEM medium to achieve the desired concentration.
Cell viability assay
The cytotoxicity of GEM on human GBC cells was estimated using the CCK-8 assay (Cell Counting Kit-8, Dojindo, Japan) according to the manufacturer’s protocol. GBC-SD, SGC-996, and NOZ were plated in 96-well plates at 104 cells per well 24 h before the start of treatment. Then, cells were exposed to GEM at 0, 1, 5, 10, 20, 40 and 80 μM for 24, 48 and 72 h. At the test point, 100 μl Cell Counting Kit-8 (CCK8) solution was added into each well, and the plate was incubated at 37 °C for 2 h followed by OD detection using a spectrophotometer. Cell viability was calculated using the following formula: Cell viability (%) = (OD450 sampleOD450 blank)/(OD450 control-OD450 blank) × 100%.
Colony formation assay
For the colony formation assay, 500 cells were plated in 6-well plates and cultured with different treatment at 37 °C for 2 weeks. The number of colonies was visualized and quantified after staining with 0.1% crystal violet in methanol for 15 min. For each experimental condition, colonies were presented as the mean number ± SD from at least three independent experiments.
Apoptosis assay and cell cycle assay
An Annexin V-FITC Apoptosis Detection kit (BD Pharmingen, USA) was used to detect the apoptosis of cells according to the manufacturer’s protocol. In brief, both attached cells and floating cells were harvested after treatment at indicated intervals, then were washed twice with cold PBS and suspended in 100 μL Binding Buffer. Cells were incubated with 5 μL Annexin V-FITC and 10 μL PI at room temperature for 15 min in the dark. Finally, an additional 400 μL of binding buffer was added to each tube. The apoptosis rate was analyzed by flow cytometry using the FACSCalibur system (BD Biosciences, USA).
Cell cycle distribution was further analyzed by flow cytometry. After treatment, cells were harvested and washed twice with cold PBS. Single-cell suspensions were fixed with 70% ice-cold ethanol at 4 °C overnight. Samples were then resuspended with cold PBS and incubated with PI staining solution for 30 min in the dark and finally analyzed by flow cytometer. The percentage of cells at each phase of the cell cycle was quantified using the CELL QUEST software (Becton–Dickinson).
Western blotting
After treatment with GEM and/or CQ, cells were harvested and lysed with RIPA buffer supplemented with PMSF. The total protein concentration was quantified by BCA kit (Beyotime Biotechnology, China). The protein was transferred to PVDF membrane (Millipore, USA) after separated by SDS/ PAGE (Beyotime Biotechnology, China), and blots were probed with anti-p62/SQSTM-1, anti-LC3, anti-PARP (all from Cell Signaling Technology), anti-Bcl-2, anti-Bax, antiβ-actin, anti-AKT, anti-t-AKT, anti-mTOR, anti-t-mTOR (all from Abcam) overnight at 4 °C. The goat anti-rabbit or goat anti-mouse horseradish peroxidase-conjugated IgG (Beyotime Biotechnology, China) was used as secondary antibodies. Bands were visualized by an enhanced chemiluminescence reagent (Millipore, USA). The gray value and gray coefficient ratio of every band were analyzed and quantified using Image J analysis software (National Institutes of Health, USA).
In vivo studies
Male BALB/c nude mice (3–4 weeks old) were purchased from the Institute of Zoology, Chinese Academy of Sciences of Shanghai. All animal procedures were carried out in accordance with institutional guidelines after Tongji University School of Medicine Animal Care and Use Committee approved the study protocol. A suspension containing 2 × 106 SGC-996 cells in 200 μL PBS was subcutaneously injected into the right flanks of each nude mice. After 1 week, when the maximum diameter of the transplanted tumors reached approximately 5 mm, the mice were randomly divided into control (PBS only), CQ (CQ 60 mg/kg), GEM (GEM 20 mg/kg) and GEM + CQ (GEM 20 mg/kg + CQ 60 mg/kg) 4 groups, 4 mice in each group. Mice were administered intraperitoneally twice per week with these compounds. Tumor length and width were measured every 2 days, and the volume was calculated using the formula: volume = V = π/6 × (W2 × L). All mice were sacrificed after 22 days and tumors were removed and weighed.
Statistical analyses
SPSS 22.0 software (IBM Corp.) was used for statistical analysis. Data were expressed as x̄ ± SD. Student’s t test and one-way ANOVA were used for comparison among groups. Statistical significance was determined as P < 0.05.
Results
GEM inhibits the proliferation and colony formation of GBC cells in vitro
To determine the effect of GEM on the proliferation of GBC cells, a series of different concentrations of GEM (0, 1, 5, 10, 20, 40, and 80 μM) were used to treat GBC cell lines (GBC-SD, SGC-996, and NOZ) for 24 h, 48 h and 72 h. As shown in Supplementary Fig. 1, GEM significantly inhibited the in vitro proliferation of the above three GBC cell lines, and the activity of GBC cells decreased significantly with increasing drug concentration and prolonged action (Supplementary Fig. 1a), with the half inhibitory concentrations (IC50): 20.52 µM, 52.69 µM, 2.53 µM of GEM for GBC-SD, SGC-996, and NOZ for 72 h, respectively (Supplementary Fig. 1b). Plate clone formation assay was also performed to test for the contact-dependent proliferation of GBC cell lines. After GEM treatment, GBC-SD, SGC-996, and NOZ cell clone formation (cloning clusters) were significantly reduced, and the reduction of clone formation was more obvious as the drug concentration increased, showing a dose–response relationship (Supplementary Fig. 1c, d). Taken together, these data suggested that GEM inhibited the in vitro proliferation of GBC cells in a time- and dose-dependent manner.
GEM induces apoptosis and cycle arrest in GBC lines in vitro
Induction of apoptosis is the main cytotoxic pattern of many chemotherapy drugs. Studies have shown that GEM can induce apoptosis in a variety of tumor cells [29, 30]. In this experiment, flow cytometry was used to detect the apoptosisinducing effect of GEM on GBC cells. After 48 h of GEM treatment of GBC cells, compared with the control group, the apoptosis rate in the GEM (20 μM) group was significantly increased (GBC-SD: 21.94 ± 1.16% vs. 49.21 ± 2.14%, P < 0.01; SGC-996: 15.47 ± 0.87% vs. 23.86 ± 0.60%, P < 0.05; NOZ: 6.46 ± 0.29% vs. 11.46 ± 0.75%, P < 0.05; Supplementary Fig. 2a). Furthermore, western blotting was used to detect the effect of GEM on apoptosis-related proteins in GBC cells. Compared with the control group, the expressions of anti-apoptotic proteins Bcl-2 and PARP in the GBC-SD, SGC-996, and NOZ cells treated with GEM were significantly reduced, and the expressions of pro-apoptotic proteins BAX were significantly increased, which were related to the concentration of GEM, indicating that GEM induced GBC cells apoptosis in a dose-dependent manner (Supplementary Fig. 2b, c).
To further confirm the effect of GEM on the proliferation and apoptosis of GBC cells, this experiment also examined the effect of GEM on the cycle of GBC cells by flow cytometry. After GEM (20 μM) treatment of GBC cells for 48 h, compared with the control group, G0/G1 phase cells in GBC-SD and SGC-996 cells increased significantly (GBC-SD: 67.67 ± 1.45% vs. 83.33 ± 0.88%, P < 0.01; SGC-996: 69.33 ± 0.88% vs. 86.00 ± 1.16%, P < 0.01), S-phase cells were significantly reduced (GBCSD: 25.33 ± 0.88% vs. 16.67 ± 0.88%, P < 0.05; SGC-996: 23.00 ± 0.58% vs. 14.00 ± 1.16%, P < 0.05); G0/G1 phase cells in NOZ cells were significantly reduced (68.00 ± 1.00% vs. 48.00 ± 1.53%, P < 0.01), S-phase cells increased significantly (24.00 ± 0.58% vs. 45.33 ± 1.45%, P < 0.01; Supplementary Fig. 2d). These data revealed that GEM could effectively induce the cycle arrest of GBC cells.
GEM induces cell autophagy in GBC cell lines in vitro
Autophagy is involved in cell survival and cell death processes and can be induced by many cytotoxic compounds. Among them, the conversion of LC3-I to LC3-II is an important step in cell autophagy, and the expression level of LC3-II represents the overall level of autophagosome formation, so LC3-II is a good marker of autophagosome formation, and p62/SQSTM-1 is a known autophagydegrading scaffold protein. When cells undergo autophagy, LC3-II expression increases and p62/SQSTM-1 expression decreases [31]. To determine whether GEM induces autophagy of GBC cells, this experiment examined the effects of GEM on the expression of autophagosomeforming marker LC3-II and autophagy-degrading scaffold protein p62/SQSTM-1 in GBC cells. As the GEM concentration increased, the expression of LC3-II in GBC-SD, SGC-996, and NOZ cells increased significantly (LC3-II expression was positively correlated with GEM concentration), and p62/SQSTM-1 expression was significantly reduced (P62/SQSTM-1 expression is inversely related to GEM concentration) (Fig. 1a, b). Therefore, it was confirmed that GEM induced autophagy flux in GBC cells in vitro.
Autophagy inhibitor CQ synergistically enhances the inhibition of GEM on the proliferation and colony formation in GBC cells in vitro
Autophagy promotes cell survival and also induces cell death. To further observe the effect of GEM-induced autophagy on GBC cells, in this study we applied a late autophagy inhibitor CQ, which can prevent autophagy by blocking autophagosomal–lysosomal fusion events [32]. As shown in Fig. 4, when CQ concentration is 10–40 μM, its effect on cell viability is not obvious, but the concentration is greater than 40 μM, the cytotoxicity is significantly increased (Fig. 2a). The CQ working concentration is thus set to 10 μM, so as not to cause significant cell death. Moreover, because autophagy is a dynamic process, upregulation of autophagy and high-level inhibition of late autophagy can cause autophagy to accumulate, thereby increasing the expression level of autophagosome-forming marker LC3-II [33], the expression of LC3-II and p62/ SQSTM-1 proteins were, therefore, detected. It is shown that the expression of LC3-II and p62/SQSTM-1 in GBCSD, SGC-996, and NOZ cells in the GEM combined CQ group was significantly increased compared with GEM alone (Fig. 2b, c).
To observe the effect of autophagy inhibition in combination with GEM on the proliferation of GBC cells, CQ (10 μM) was then used to block the autophagy of GBC cells in this study. The results showed that the proliferation (GBC-SD: 36.34 ± 0.93% vs. 18.49 ± 1.61%, P < 0.01; SGC-996: 62.31 ± 1.84% vs. 52.49 ± 0.81%, P < 0.05; NOZ: 59.42 ± 3.41% vs. 47.91 ± 2.73%, P < 0.05; Fig. 3a) and clone formation (GBC-SD: 39.32 ± 3.17% vs. 4.13 ± 0.39%, P < 0.01; SGC-996: 36.56 ± 2.38% vs. 7.36 ± 1.45%, P < 0.01; NOZ: 66.37 ± 1.48% vs. 8.70 ± 0.89%, P < 0.01; Fig. 3b, c) of GBC cells in the GEM + CQ group were significantly inhibited compared with the single GEM group. These results revealed that GEM could induce protective autophagy of GBC cells, and inhibition of autophagy could significantly enhance the growth inhibitory effect of GEM on GBC cells.
Autophagy inhibitor CQ synergistically enhances the induction of apoptosis and cycle arrest of GBC cells by GEM in vitro
To further observe the effect of autophagy inhibition in combination with GEM on the growth and GEM-induced autophagy of GBC cells, the apoptosis of GBC cells was detected by flow cytometry and western blotting. As shown in Fig. 4, the apoptosis rates in the GEM + CQ group (GBCSD: 54.34 ± 1.12% vs. 42.15 ± 2.10%, P < 0.05; SGC996: 14.31 ± 0.87% vs. 12.49 ± 0.96%, P < 0.05; NOZ: 11.42 ± 1.35% vs. 9.91 ± 1.57%, P < 0.05; flow cytometry; Fig. 4a, b) were significantly increased when compared with the single GEM group. Moreover, the expression of anti-apoptotic protein Bcl-2 and PARP was significantly decreased and the expression of pro-apoptotic protein BAX was increased in the GEM + CQ group compared with the single GEM group (Fig. 4c, d). Furthermore, we analyzed the effect of GEM in combination with CQ on the cell cycle of GBC cells by flow cytometry. As expected that compared with the single GEM, the cell cycle arrest in the GEM + CQ group was obvious: the percentage of G1/G0 phase cells in the GBC-SD and SGC-996 increased significantly (GBC-SD: 82.83 ± 1.73% vs. 93.5 ± 1.45%, P < 0.05; SGC-996: 82.16 ± 1.86% vs. 92.5 ± 0.88%, P < 0.05), and the percentage of S-phase cells decreased significantly (GBC-SD: 17.21 ± 1.45% vs. 6.87 ± 1.45%, P < 0.05; SGC996: 16.87 ± 1.45% vs. 7.87 ± 0.88%, P < 0.05); in NOZ, the percentage of cells in the S phase of the GEM + CQ group increased significantly (42.32 ± 1.86% vs. 61.99 ± 1.53%, P < 0.05), and the percentage of cells in the G1/G0 phase decreased significantly (48.5 ± 1.20% vs. 29.84 ± 0.88%, P < 0.01) (Fig. 4e, f). The results above suggested that inhibition of autophagy effectively enhanced the apoptosisinducing effect of GEM on GBC cells and significantly postponed the cell cycle of GBC cells following GEM application.
AKT/mTOR signaling pathway is involved in GEM‑induced autophagy in GBC cells in vitro
mTOR signaling pathway is a key signaling pathway for cell metabolism, growth, proliferation and survival. Recent studies have shown that the mTOR signaling pathway is also closely related to cell autophagy. The upstream AKTtargeted mTOR signaling pathway can inhibit cell autophagy [34]. AKT/mTOR signaling pathway is autophagy and negative regulatory mechanisms of apoptosis [35, 36]. To explore the molecular mechanism of autophagy induced by GEM in GBC cells, we here detect the expression levels of AKT and mTOR in GBC cells: GBC-SD, SGC-996, NOZ cells after GEM treatment using western blotting. The results showed that as the GEM concentration increased, the expression of p-AKT and p-mTOR in GBC-SD, SGC-996, and NOZ cells decreased significantly, despite the unchanged total amount of AKT and mTOR proteins (Fig. 5a, b). And as mentioned before, GEM induced autophagy and apoptosis in GBC cells. Thus, we believed that GEM may induce autophagy and apoptosis in GBC cells through inhibiting the AKT/mTOR signaling pathway.
Tumor suppression induced by GEM is enhanced by CQ in vivo
Based on the results above, xenograft models were used to further confirm the synergistic inhibitory effect of GEM in combination with autophagy inhibitor CQ on tumor growth in vivo. After SGC-996 cells were injected subcutaneously into the right back, nude mice were randomly divided into 4 groups: PBS (blank control group), CQ, GEM and GEM + CQ groups, the growth of subcutaneous GBC xenografts in nude mice in each group was observed for 3 weeks. After 3 weeks, the tumor volumes in CQ group did not show a significant difference compared with the control group. The GEM + CQ group exerted greater antitumor effects in GBC xenograft models compared with the drugs independently. As expected, the tumor xenografts in GEM + CQ group were significantly smaller than those in the GEM group (207.9 ± 46.86 mm3 vs. synergistically enhanced the inhibitory effect of GEM on GBC cells. a After treatment with GEM in the presence or absence of 10 µM CQ for 48 h, the cell viability of GBC cells was measured by CCK-8 assay. Results were expressed as mean ± SD representing at least three independent experiments. b, c The colony formation assay was used to assess the proliferation capacity of GBC cells treated with GEM in the presence or absence of 10 µM CQ for 48 h. Values were given as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, vs. control group 453.3 ± 70.1 mm3, P < 0.05; Fig. 6a, b). Consistent with the results of tumor volume, tumor weight could be suppressed by GEM monotherapy or CQ + GEM combination therapy. GEM in combination with CQ showed a more effective inhibition in GBC xenografts of nude mice (0.1667 ± 0.03333 g vs. 0.6 ± 0.05774 g, P < 0.05, Fig. 6c). Altogether, the data from in vitro and in vivo assays confirmed that inhibition of autophagy by CQ could significantly increase chemosensitivity to GEM for human GBC.
Discussion
Although the diagnosis and treatment technology of GBCs has made great progress in recent years, the overall survival rate and prognosis of the patients have not improved significantly. Chemotherapy is an alternative treatment for those patients who have lost their surgical opportunity or patients who have a recurrence after surgery. However, CQ synergistically enhanced GEM-induced apoptosis and cycle arrest in GBC cells. a, b GBC-SD, SGC-996, and NOZ cells treated by GEM (20 μM) implement with or without CQ (10 µM) for 48 h, the percentage of apoptotic cells was investigated using Annexin V-FITC and PI. (AnV +) (PI −) cells were considered early apoptotic and (AnV +) (PI +) cells were considered late apoptotic. The columns represent the mean ± SD of the three independent experiments. c, d GBC-SD, SGC-996, and NOZ cells were treated by GEM (20 μM) implement with or without CQ (10 µM) for 48 h, respectively. The PARP, Bcl-2, BAX, and β-actin expressions were detected by western blotting. Densitometry represents the expression of the proteins relative to β-actin. e, f Flow cytometric analysis of cell cycle progression in GBC cells treated by GEM (20 μM) with or without CQ (10 μM) for 48 h. The results were representative of three independent experiments. The percentages of cells in G1, S, and G2-M are shown as histograms. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, vs. control group traditional chemotherapy cannot significantly prolong the survival time of GBC patients due to the insensitivity or resistance of chemotherapy. GEM is an anti-metabolic nucleoside analog that has been shown to have anticancer effects in various tumors [37–40]. GEM or combined with other anticancer drugs has achieved certain effects in clinical trials of GBC treatment [9]. However, the increased chemotherapy resistance attenuates the chemotherapy effect of GEM on GBCs. Therefore, an in-depth understanding of the effects and molecular mechanisms in vitro and in vivo that reduce the efficacy of GEM chemotherapy may play an important role in improving the prognosis of GBC patients.
Autophagy is a dynamic process that involves extensive degradation of cytoplasm, organelles, and proteins. Based on the function of cell recycling, autophagy plays a key role in the quality control of cell components and provides nutrients and materials for newly constructed structures in cells under metabolic stress [41]. Recent studies have shown that autophagy not only plays an important regulatory role in the occurrence and development of malignant tumors but also plays an important role in the response of tumor cells to anticancer therapies [42, 43], which may be related to the increased demand for tumor tissue metabolism and biosynthesis during chemotherapy. Studies indicate that autophagy is upregulated in hypoxic tumor areas, which is important for the survival of tumor cells in them [44]. Although the genetic background to autophagy dependence in cancer is still poorly understood, autophagy has been shown to protect tumor cells during the antitumor process of various drugs because it can reduce cell death caused by DNA damage [45, 46]. In pancreatic cancer, GEM induces protective autophagy in pancreatic cancer cells through the AMPK/ mTOR signaling pathway, which affects its chemotherapy effect [22]. In addition, in human non-small cell lung cancer cells, triple-negative MDA-MB-231 breast cancer cells, and urothelial cancer cells, GEM can induce autophagy to reduce cancer cell apoptosis [23, 24, 47]. In this study, we found that after GEM treatment of GBC cells, the expression of autophagy marker proteins LC3-II/LC3-I increased significantly, and p62/SQSTM-1 expression decreased, indicating that GEM can induce autophagy in GBC cells, which is consistent with reports that GEM can induce autophagy in tumor cells such as pancreatic cancer [22–24, 47].
Recent studies have shown that autophagy and apoptotic signaling pathways can interact with each other [48–51]. Inhibition of autophagy can enhance apoptosis in pancreatic cancer cells [29, 30], and autophagy promotes apoptosis of mesenchymal stem cells in the inflammatory microenvironment [52]. Therefore, understanding the interaction between apoptosis and autophagy may help to explore new treatment strategies for GBC. Studies on the relationship between autophagy and apoptosis of pancreatic cancer cells induced by GEM have shown that inhibition of autophagy can significantly enhance the pro-apoptotic effect of GEM on pancreatic cancer [29, 30]. Based on these basic studies, a clinical study showed that the autophagy inhibitor hydroxy chloroquine (HCQ) combined with GEM in the treatment of patients with pancreatic cancer is biosafe and tolerated, and the results are encouraging. The tumor marker CA19-9 has been reduced to a certain extent, and the PFS and OS of patients have improved to varying degrees. These results support a larger number of more randomized clinical trials to verify the efficacy of GEM combined with autophagy inhibitors in the treatment of pancreatic cancer [53]. In addition, in non-small cell lung cancer cells and bladder cancer cells, inhibition of autophagy can increase the sensitivity of cancer cells to GEM [27, 54]. In this study, GEM induced autophagy and apoptosis in GBC cells. As an advanced autophagy inhibitor, CQ not only effectively eliminates GEM-induced autophagy in GBC cells but also enhances the anticancer effects of GEM on GBC by inhibiting autophagy in vitro and in vivo, including inhibition of proliferation, induction of apoptosis and cell cycle arrest. These results indicate that the interaction between cell autophagy and apoptosis plays an important role in the resistance of GBC treatment, and this new regulatory mechanism is conducive to the antitumor effect of GEM in the treatment of GBC.
In recent years, many studies have pointed out that autophagy and apoptosis affect the chemotherapy effect of malignant tumors through various molecular pathways. It has been reported that GEM can induce autophagy and apoptosis in pancreatic cancer cells through the AMPK/ mTOR signaling pathway, and blocking this pathway can inhibit autophagy to enhance the apoptosis-inducing effect of GEM on pancreatic cancer cells [22]. Endoplasmic reticulum-stress-mediated autophagy is also thought to be associated with chemotherapy resistance of GEM for pancreatic cancer [28]. In bladder cancer, CYLD-Livin is also a target for improving the therapeutic effect of GEM by inhibiting autophagy. Another study reported that non-coding RNAs play an important role in the sensitivity of breast cancer cells to chemotherapy by regulating autophagy and apoptosis induced by GEM [55]. In addition, the AKT signaling pathway is one of the main signaling pathways that regulate tumor cells [56]; and there is increasing evidence that the overactive AKT/mTOR pathway can promote tumor cell proliferation and survival [57–59]. As a downstream of VEGFR, the AKT/mTOR signaling pathway is involved in apoptosis of VEGFR-targeted therapy [60]. mTOR is also mainly related to apoptosis and autophagy. GEM has been reported to have VEGF-C inhibitory effects [61], which can inhibit the migration and invasion of GBC. Therefore, the AKT/mTOR signaling pathway may be closely related to the growth inhibitory effect of GEM in GBC. Our results show that GEM upregulates cell autophagy marker proteins LC3-II/LC3-I and pro-apoptotic protein BAX, downregulates p62/SQSTM-1, anti-apoptotic proteins Bcl-2, PARP, and inhibits the expression of p-AKT and p-mTOR protein, indicating that GEM may induce autophagy and apoptosis of GBC cells by inhibiting the AKT/mTOR signaling pathway. Therefore, cell autophagy may become a new potential therapeutic target for adjuvant therapy of GBC and autophagy inhibitors such as CQ as an adjuvant or potential synergist of GEM can increase the anticancer effect and efficacy of GEM, which could be a new strategy for adjuvant treatment of GBC.
In conclusion, our results confirm that GEM induces apoptosis and autophagy in GBC cell lines, which may be related to the AKT/mTOR pathway; GEM in combination with CQ, a late autophagy inhibitor, can enhance the antitumor effect on GBC cell lines in vitro and in vivo. These findings suggest that GEM in combination with autophagy inhibitors may be a potential new strategy for adjuvant treatment of GBC.
References
1. Hsing AW, Sakoda LC, Rashid A, Chen J, Shen MC, Han TQ, Wang BS, Gao YT (2008) Body size and the risk of biliary tract cancer: a population-based study in China. Br J Cancer 99(5):811–815
2. Hsing AW, Bai Y, Andreotti G, Rashid A, Deng J, Chen JB, Goldstein AM, Han TQ, Shen MC, Fraumeni JF et al (2007) Family history of gallstones and the risk of biliary tract cancer and gallstones: a population-based study in Shanghai, China. Int J Cancer 121(4):832–838
3. Wernberg JA, Lucarelli DD (2014) Gallbladder cancer. Surg Clin N Am 94(2):343–360
4. Zheng YH, Xu DY, Bu ZD (2016) Chinese version Chloroquine of NCCN clinical practice guidelines in oncology officially authorized by NCCN. Chin J Cancer Res 28(1):144–145
5. Pedersen KS, Kim GP, Foster NR, Wang-Gillam A, Erlichman C, McWilliams RR (2015) Phase II trial of gemcitabine and tanespimycin (17AAG) in metastatic pancreatic cancer: a Mayo Clinic Phase II Consortium study. Investig New Drugs 33(4):963–968
6. Chen YM, Perng RP, Tsai CM, Whang-Peng J (2006) A phase II trial of gemcitabine plus UFUR combination chemotherapy in non-small-cell lung cancer patients failing previous chemotherapy. Lung Cancer 52(3):333–338
7. Sun L, Lu J, Niu Z, Ding K, Bi D, Liu S, Li J, Wu F, Zhang H, Zhao Z, Ding S (2015) A potent chemotherapeutic strategy with Eg5 inhibitor against gemcitabine resistant bladder cancer. PLoS One 10(12):e0144484
8. Hidalgo M (2010) Pancreatic cancer. N Engl J Med 362(17):1605–1617
9. Valle J (2010) Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer (vol 362, pg 1273, 2010). New Engl J Med 363(2):198
10. Qu K, Liu SN, Chang HL, Liu C, Xu XS, Wang RT, Zhou L, Tian F, Wei JC, Tai MH et al (2012) Gallbladder cancer: a subtype of biliary tract cancer which is a current challenge in China. Asian Pac J Cancer Prev 13(4):1317–1320
11. Hirooka Y, Ishikawa T, Kawashima H, Ohno E, Nonogaki K, Kanamori A, Hirai T, Uchida H, Shirai O, Ishikawa H et al (2017) Prospective multicenter phase II study of gemcitabine plus cisplatin in patients with unresectable gallbladder cancer. Cancer Chemother Pharmacol 80(1):119–125
12. Iyer Renuka V, Pokuri Venkata K, Adrienne Groman et al (2018) A multicenter phase II study of gemcitabine, capecitabine, and bevacizumab for locally advanced or metastatic biliary tract cancer. Am J Clin Oncol 41:649–655
13. Moehler M, Maderer A, Schimanski C et al (2014) Gemcitabine plus sorafenib versus gemcitabine alone in advanced biliary tract cancer: a double-blind placebo-controlled multicentre phase II AIO study with biomarker and serum programme. Eur J Cancer 50:3125–3135
14. Siebenhüner Alexander R, Heike Seifert, Helga Bachmann et al (2018) Adjuvant treatment of resectable biliary tract cancer with cisplatin plus gemcitabine: a prospective single center phase II study. BMC Cancer 18:72
15. White E (2015) The role for autophagy in cancer. J Clin Investig 125(1):42–46
16. Zhang XF, Zhao P, Wang CH, Xin BR (2019) SNHG14 enhances gemcitabine resistance by sponging miR-101 to stimulate cell autophagy in pancreatic cancer. Biochem Biophys Res Commun 510(4):508–514
17. Xiong JJ, Wang D, Wei AL, Ke NW, Wang YC, Tang J, He SR, Hu WM, Liu XB (2017) MicroRNA-410-3p attenuates gemcitabine resistance in pancreatic ductal adenocarcinoma by inhibiting HMGB1-mediated autophagy. Oncotarget 8(64):107500–107512
18. Li CG, Zhao ZM, Zhou ZP, Liu R (2016) Linc-ROR confers gemcitabine resistance to pancreatic cancer cells via inducing autophagy and modulating the miR-124/PTBP1/PKM2 axis. Cancer Chemother Pharmacol 78(6):1199–1207
19. Song B, Bian Q, Zhang YJ, Shao CH, Li G, Liu AA, Jing W, Liu R, Zhou YQ, Jin G et al (2015) Downregulation of ASPP2 in pancreatic cancer cells contributes to increased resistance to gemcitabine through autophagy activation. Mol Cancer 14:177
20. Ma T, Chen W, Zhi X, Liu H, Zhou Y, Chen BW, Hu LQ, Shen J, Zheng XX, Zhang SF et al (2018) USP9X inhibition improves gemcitabine sensitivity in pancreatic cancer by inhibiting autophagy. Cancer Lett 436:129–138
21. Kaur S, Bafna S, Rachagani S, Ganti AK, Batra SK (2012) Mucin MUC4 resists gemcitabine-induced pancreatic cancer cell death by promoting autophagy. Pancreas 41(8):1374–1375
22. Zhu JH, Chen Y, Ji Y, Yu YQ, Jin Y, Zhang XX, Zhou JL (2018) Gemcitabine induces apoptosis and autophagy via the AMPK/ mTOR signaling pathway in pancreatic cancer cells. Biotechnol Appl Biochem 65(5):665–671
23. Wu HM, Shao LJ, Jiang ZF, Liu RY (2016) Gemcitabine-induced autophagy protects human lung cancer cells from apoptotic death. Lung 194(6):959–966
24. Chen M, He MY, Song YJ, Chen LQ, Xiao P, Wan XP, Dai F, Shen P (2014) The cytoprotective role of gemcitabine-induced autophagy associated with apoptosis inhibition in triple-negative MDA-MB-231 breast cancer cells. Int J Mol Med 34(1):276–282
25. Sui X, Chen R, Wang Z et al (2013) Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 4:e828
26. Donohue E, Thomas A, Maurer N, Manisali I, Zeisser-Labouebe M, Zisman N, Anderson HJ, Ng SSW, Webb M, Bally M et al (2013) The autophagy inhibitor verteporfin moderately enhances the antitumor activity of gemcitabine in a pancreatic ductal adenocarcinoma model. J Cancer 4(7):585–596
27. Yin L, Liu S, Li CS, Ding ST, Bi DB, Niu ZH, Han LP, Li WJ, Gao DX, Liu Z et al (2016) CYLD downregulates Livin and synergistically improves gemcitabine chemosensitivity and decreases migratory/invasive potential in bladder cancer: the effect is autophagy-associated. Tumor Biol 37(9):12731–12742
28. Fu Z, Cheng X, Kuang J, Feng H, Chen L, Liang J, Shen X, Yuen S, Peng C, Shen B et al (2018) CQ sensitizes human pancreatic cancer cells to gemcitabine through the lysosomal apoptotic pathway via reactive oxygen species. Mol Oncol 12(4):529–544
29. Papademetrio DL, Cavaliere V, Simunovich T, Costantino S, Campos MD, Lombardo T, Kaiser CMF, Alvarez E (2014) Interplay between autophagy and apoptosis in pancreatic tumors in response to gemcitabine. Target Oncol 9(2):123–134
30. Li XS, Yan J, Wang LS, Xiao FJ, Yang YF, Guo XZ, Wang H (2013) Beclin1 inhibition promotes autophagy and decreases gemcitabine-induced apoptosis in Miapaca2 pancreatic cancer cells. Cancer Cell Int 13:26
31. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T (2009) Monitoring autophagic degradation of P62/Sqstm1. Methods Enzymol Autophagy Mamm Syst 452(Pt B):181–197
32. Hasanain M, Bhattacharjee A, Pandey P, Ashraf R, Singh N, Sharma S, Vishwakarma AL, Datta D, Mitra K, Sarkar J (2015) Alpha-solanine induces ROS-mediated autophagy through activation of endoplasmic reticulum stress and inhibition of Akt/mTOR pathway. Cell Death Dis 6:e1860
33. Tanida I (2011) Autophagosome formation and molecular mechanism of autophagy. Antioxid Redox Signal 14(11):2201–2214
34. Saxton RA, Sabatini DM (2017) mTOR signaling in growth, metabolism, and disease (vol 168, pg 960, 2017). Cell 169(2):362
35. Nicholson KM, Anderson NG (2012) The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal 14:381–395
36. Majumder PK, Sellers WR (2005) Akt-regulated pathways in prostate cancer. Oncogene 24:7465–7474
37. Toyota Y, Iwama H, Kato K, Tani J, Katsura A, Miyata M, Fujiwara S, Fujita K, Sakamoto T, Fujimori T et al (2015) Mechanism of gemcitabine-induced suppression of human cholangiocellular carcinoma cell growth. Int J Oncol 47(4):1293–1302
38. Kilani R, Tamimi Y, Karmali S, Mackey J, Hanel E, Wong KK, Moore RB (2002) Selective cytotoxicity of gemcitabine in bladder cancer cell lines. Anticancer Drugs 13(6):557–566
39. Kang YW, Lee JE, Jung KH, Son MK, Shin SM, Kim SJ, Fang ZH, Yan HH, Park JH, Han B et al (2018) KRAS targeting antibody synergizes anti-cancer activity of gemcitabine against pancreatic cancer. Cancer Lett 438:174–186
40. Teng JP, Yang ZY, Zhu YM, Ni D, Zhu ZJ, Li XQ (2018) Gemcitabine and cisplatin for treatment of lung cancer in vitro and vivo. Eur Rev Med Pharmacol Sci 22(12):3819–3825
41. Tsuchihara K, Fujii S, Esumi H (2009) Autophagy and cancer: dynamism of the metabolism of tumor cells and tissues. Cancer Lett 278(2):130–138
42. Du HL, Yang WP, Chen L, Shi MM, Seewoo V, Wang JY, Lin AD, Liu ZR, Qiu WH (2012) Role of autophagy in resistance to oxaliplatin in hepatocellular carcinoma cells. Oncol Rep 27(1):143–150
43. Pardo R, Lo Re A, Archange C, Ropolo A, Papademetrio DL, Gonzalez CD, Alvarez EM, Iovanna JL, Vaccaro MI (2010) Gemcitabine induces the VMP1-mediated autophagy pathway to promote apoptotic death in human pancreatic cancer cells. Pancreatology 10(1):19–26
44. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen GH, Mukherjee C, Shi YF, Gelinas C, Fan YJ et al (2006) Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10(1):51–64
45. Abedin MJ, Wang D, McDonnell MA, Lehmann U, Kelekar A (2007) Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ 14(3):500–510
46. Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, Bartsch G, Michaelis M, Cinatl J, Haferkamp A et al (2015) Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (−)-Gossypol (AT-101). BMC Cancer 15:224
47. Ojha R, Jha V, Singh SK (2016) Gemcitabine and mitomycin induced autophagy regulates cancer stem cell pool in urothelial carcinoma cells. Biochim Biophys Acta-Mol Cell Res 1863(2):347–359
48. Tavassoly I, Parmar J, Shajahan-Haq AN, Clarke R, Baumann WT, Tyson JJ (2015) Dynamic modeling of the interaction between autophagy and apoptosis in mammalian cells. Cpt-Pharmacomet Syst Pharmacol 4(4):263–272
49. Mukhopadhyay S, Panda PK, Sinha N, Das DN, Bhutia SK (2014) Autophagy and apoptosis: where do they meet? Apoptosis 19(4):555–566
50. Moretti L, Cha YI, Niermann KJ, Lu B (2007) Switch between apoptosis and autophagy: radiation-induced endoplasmic reticulum stress? Cell Cycle 6(7):793–798
51. Jiang Q, Li F, Shi K, Wu P, An J, Yang Y, Xu C (2014) Involvement of p38 in signal switching from autophagy to apoptosis via the PERK/eIF2 alpha/ATF4 axis in selenite-treated NB4 cells. Cell Death Dis 5:e1270
52. Dang SP, Yu ZM, Zhang CY, Zheng J, Li KL, Wu Y, Qian LL, Yang ZY, Li XR, Zhang YY et al (2015) Autophagy promotes apoptosis of mesenchymal stem cells under inflammatory microenvironment. Stem Cell Res Ther 6:247
53. Boone BA, Bahary N, Zureikat AH, Moser AJ, Normolle DP, Wu WC, Singhi AD, Bao P, Bartlett DL, Liotta LA et al (2015) Safety and biologic response of pre-operative autophagy inhibition in combination with gemcitabine in patients with pancreatic adenocarcinoma. Ann Surg Oncol 22(13):4402–4410
54. Jiang YF, Han ZD, Wang Y, Hao WB (2018) Depletion of SIRT7 sensitizes human non-small cell lung cancer cells to gemcitabine therapy by inhibiting autophagy. Biochem Biophys Res Commun 506(1):266–271
55. Chen Y-M, Liu Y, Wei H-Y, Lv K-Z, Fu P-F (2016) Large intergenic non-coding RNA-ROR reverses gemcitabine-induced autophagy and apoptosis in breast cancer cells. Oncotarget 7(37):59604–59617
56. Blume-Jensen P, Hunter T (2001) Oncogenic kinase signalling. Nature 411(6835):355–365
57. Pene F, Claessens YE, Muller O, Viguie F, Mayeux P, Dreyfus F, Lacombe C, Bouscary D (2002) Role of the phosphatidylinositol 3-kinase/Akt and mTOR/P70S6-kinase pathways in the proliferation and apoptosis in multiple myeloma. Oncogene 21(43):6587–6597
58. LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA (2008) Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat 11(1–2):32–50
59. Hu HZ, Yang YB, Xu XD, Shen HW, Shu YM, Ren Z, Li XM, Shen HM, Zeng HT (2007) Oridonin induces apoptosis via PI3K/ Akt pathway in cervical carcinoma HeLa cell line. Acta Pharmacol Sin 28(11):1819–1826
60. Liu Y, Chen ZJ, Cheng HX, Chen J, Qian J (2017) Gremlin promotes retinal pigmentation epithelial (RPE) cell proliferation, migration and VEGF production via activating VEGFR2-AktmTORC2 signaling. Oncotarget 8(1):979–987
61. Yang MH, Lee KT, Yang S, Lee JK, Lee KH, Moon IH, Rhee JC (2012) Guggulsterone enhances antitumor activity of gemcitabine in gallbladder cancer cells through suppression of NF-kappa B. J Cancer Res Clin Oncol 138(10):1743–1751