Hydroxychloroquine reverses the drug resistance of leukemic K562/ADM cells by inhibiting autophagy

  • Authors:
    • Feifei Wang
    • Zhewen Zhang
    • Wing Ting Leung
    • Jing Chen
    • Juan Yi
    • Chunmei Ying
    • Minmin Yuan
    • Mingyan Wang
    • Na Zhang
    • Xuemin Qiu
    • Ling Wang
    • Hulai Wei
  • View Affiliations

  • Published online on: August 27, 2019     https://doi.org/10.3892/mmr.2019.10621
  • Pages: 3883-3892
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Autophagy is an essential metabolic pathway mediated by lysosomal degradation, which is involved in scavenging and recycling senescent or damaged organelles and biological macromolecules in eukaryotic cells. The present study explored the association between the autophagic activity and chemotherapy resistance of leukaemia cells, and the possibility of using autophagy inhibitors to combat leukemic drug resistance. It was found that the levels of basic autophagy in multidrug‑resistant leukaemia cells (K562/ADM) were significantly higher compared with sensitive cells (K562), and that Adriamycin (ADM) was capable of inducing autophagic activity in K562 and K562/ADM cells. K562 and K562/ADM cells were treated with a series of hydroxychloroquine (HCQ) concentrations to inhibit cellular autophagy and detect cell sensitivity to ADM. The results demonstrated that the sensitivity of K562 cells to ADM was mildly enhanced by HCQ, and that the sensitivity of K562/ADM cells to ADM was markedly strengthened by HCQ. In addition, more typical morphological changes associated with apoptosis emerged, and the ratio of Bax/Bcl‑2 and activity of caspase‑3 were markedly increased in K562/ADM cells treated with HCQ. Notably, the expression of mdr1 mRNA and P‑glycoprotein (P‑gp) in drug‑resistant K562/ADM cells was upregulated along with increasing autophagic activity induced by ADM. Furthermore, HCQ significantly reduced the increase in P‑gp expression by inhibiting autophagic activity. Collectively, these findings indicated that the inhibition of autophagy significantly promoted the sensitivity of K562/ADM cells to ADM by facilitating apoptosis. Furthermore, inhibition of autophagy attenuated the expression of P‑gp; therefore, P‑gp may be involved in autophagic regulation in drug‑resistant cells.

Introduction

Leukaemia, a clonal proliferative neoplasm derived from haematopoietic stem cells, is commonly caused by various genetic mutations (1). There are a variety of clinical therapies involving chemotherapy used to treat leukaemia (2). After prolonged exposure to chemotherapeutic drugs, some patients may develop multidrug resistance, which is defined by resistance to a wide range of functionally unrelated chemotherapeutic agents. Drug resistance hinders chemotherapeutic efficacy and leads to poor prognosis in patients with leukaemia (3). Therefore, there is an urgent requirement for more effective and less toxic drugs to manage chemotherapy-induced multidrug resistance.

The exact mechanism of multidrug resistance is not clear, and previous studies have suggested that it may be partially due to the overexpression of ATP-binding cassette (ABC) transporters, alteration of metabolised enzymes, impaired apoptosis and autophagy, and the existence of leukaemia stem cells (4). P-glycoprotein (P-gp) is one of the most studied ABC transporters (5), and functions as a transporter to move substrates across cellular membranes using the energy released from ATP hydrolysis (6). The mdr1 (multidrug-resistance) gene is located on chromosomal region 7q21 and encodes an amino acid sequence that forms P-gp (170 kDa) after glycosylation (7). P-gp is constitutively expressed in various adult tissues, such as those of the intestine, liver, kidney and brain, where it serves an important role in drug excretion and protects cells from external threats (8). Overexpression of P-gp in cancer cells often leads to multidrug resistance by pumping agents out of cells (9).

Apoptosis is a caspase-3-dependent programmed cell death process (10) that is regulated by a variety of elements such as the Bcl-2 protein family (11). The Bcl-2 family includes both pro-apoptotic proteins, such as Bax and Bcl-2 homologous antagonist killer (Bak), and anti-apoptotic proteins, such as Bcl-2 and Bcl-xL (12). Bax/Bak promotes apoptosis by forming homogenous or heterogeneous dimers, and enhancing the permeabilization of the outer mitochondrial membrane. Bcl-2/Bcl-xL can combine with Bax/Bak to suppress the formation of dimers, resulting in apoptosis inhibition (13,14). A number of studies have reported that the ratio of anti-apoptotic to pro-apoptotic protein levels, rather than a single Bcl-2 family protein, determines apoptosis susceptibility (15). Once cancer cells become unsusceptible to chemotherapy-induced apoptosis, they may acquire drug resistance (16).

Autophagy is an evolutionarily conserved process of eukaryotic cells. It enables cells to sequester cellular components by forming a double-membrane vacuole, called an autophagosome, and subsequently to degrade those components after the fusion of autophagosomes with lysosomes (17). The degradation products, including amino acids, fatty acids, nucleotides and ATP, are reused by cells to maintain the cell structure and metabolism (18). In addition to participating in physiological processes, autophagy also serves a vital role in various pathological conditions, such as neurodegenerative disorders, autoimmunity disease, inflammation and cancer (1922). Evidence suggests that autophagy emerges in a context-dependent role in cancer. On the one hand, autophagy inhibits tumour initiation by cleaning up oncogenic protein substrates, toxic misfolded proteins and dysfunctional organelles (23). On the other hand, once a tumour has been established, autophagy can facilitate tumour survival in an environment of nutrient depletion or hypoxia by catabolising unnecessary proteins into amino acids and generating the energy needed for tumour cell survival, which is associated with the drug resistance of cancer cells (24). However, it has been reported that hyperactive autophagy can induce apoptosis or degrade cytoplasmic contents, thereby resulting in the death of tumour cells (25,26).

In our previous study, it was determined that multidrug-resistant leukaemia cells (K562/ADM cells) demonstrated a higher level of autophagy than drug-sensitive leukaemia cells (K562 cells), both at the basic metabolic state and when under nutrient deprivation stress, implying that autophagy is associated with the drug resistance of leukaemia cells (27). To clarify the mechanism of drug resistance in leukaemia cells, hydroxychloroquine (HCQ), a classic autophagy inhibitor, was used to reverse the drug resistance of K562/ADM cells, increase their apoptosis level and inhibit their P-gp expression. This observation indicated the complicated relationship between autophagy and multidrug resistance, and suggested a novel target for leukaemia intervention.

Materials and methods

Chemicals and antibodies

HCQ was purchased from the Tokyo Chemical Industry Co., Ltd. Adriamycin (ADM) was obtained from the Kangbaotai Biochemical Industry Company. Newborn bovine serum was obtained from the Rongye Biotech Company (http://royabio.800400.net). RPMI-1640 medium was acquired from Gibco (Thermo Fisher Scientific, Inc.), MTT from Sigma-Aldrich (Merck KGaA); and TRIzol® was from Invitrogen (Thermo Fisher Scientific, Inc.). Specific primers for mdr1 and β-actin were synthesised by the Takara Bio, Inc. Antibodies against P62 (cat. no. 88588) and light chain (LC)3 (cat. no. 4108) were obtained from Cell Signaling Technology, Inc.; anti-β-actin antibody (cat. no. A1813) was from BioVision, Inc.; antibodies against Bax (cat. no. TA346891), Bcl-2 (cat. no. TA803003) and cleaved caspase-3 (cat. no. TA336455) were purchased from Beijing Zhongshan Jinqiao Biotechnology Co., Ltd.; anti-P-gp antibody (cat. no. BM4508), horseradish peroxidase (HRP)-linked anti-rabbit (cat. no. BA1054) and anti-mouse (cat. no. BA1050) IgG antibodies were from the Wuhan Boster Biological Technology, Ltd.

Cell lines and culture

The human ADM-resistant leukaemia cell line (K562/ADM cells) and its parental subline (K562 cells) were both provided by the Medical Experimental Center of Lanzhou University. K562/ADM and K562 cells were maintained in RPMI-1640 medium, supplemented with 10% inactivated newborn bovine serum and 2 mmol/l L-glutamine, at 37°C in a cell incubator with 5% CO2. Experiments were performed when the cells reached the mid-log phase.

Cell viability assay (MTT assay)

K562/ADM and K562 cells were seeded at a density of 1×105 cells/ml in 96-well plates. The cells were treated with 0, 2, 4, 8, 16, 20 and 40 µmol/l of HCQ for 24 h at 37°C. MTT solution (10 µl; 5 g/l) was added to each well before the cells were incubated at 37°C for a further 4 h. Then, 100 µl of 10% acidulated sodium dodecyl sulfate (SDS) were added to each well, which was incubated for 12 h at 37°C to dissolve the formazan crystals. Cell proliferation was then detected by MTT colorimetric assay. The optical density (OD) was measured at 570 nm using a Powerwave X plate reader (BioTek Instruments, Inc.). Cell proliferation inhibition rates were calculated using the following formula: Cell proliferation inhibition rate = [(ODcontrol-ODexperiment)/ODcontrol] × 100%. 4 and 16 µmol/l of HCQ were selected as the maximum non-toxic concentration to treat with K562 and K562/ADM cells in the following experiments. Cells were seeded in 96-well plates at a density of 1×105 cells/ml in triplicate and were cultured with ADM or/and HCQ at the indicated concentrations at 37°C for 12, 24 or 48 h. K562 cells were treated with 0, 0.09, 0.18, 0.375, 0.75, 1.5 µmol/l of ADM or pretreated with 4 µmol/l of HCQ for 3 h before exposure to ADM. K562/ADM cells were treated with 0, 1.5, 3, 6, 12, 24 µmol/l of ADM or pretreated with 16 µmol/l of HCQ for 3 h before exposure to ADM, and then MTT assay was executed as above. The half-maximal inhibitory concentration (IC50) was calculated using cell proliferation inhibition rate by SPSS 17.0 software (SPSS, Inc.). All samples were prepared in triplicate.

Acridine orange (AO) and ethidium bromide (EB) staining

AO and EB staining have been used to discriminate normal cells from those undergoing apoptosis and death (28). K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. Cells were harvested and washed twice with PBS. After being suspended in 100 µl of PBS, the cells were stained with 5 µl of EB (200 mg/l) and 5 µl of AO (200 mg/l) for 30 min at 37°C in the dark. Subsequently, the cells were rinsed with PBS three times, and a drop of suspension was placed on a glass slide. Cells were observed under a fluorescence microscope (Olympus Corporation) with a blue filter in fields with ×400 magnification.

Detection by transmission electron microscopy

K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. Cells were fixed with glutaraldehyde at 4°C overnight. The next day, after rinsing with PBS three times for 5 min, cells were fixed with 1% OsO4 at room temperature for 1.5 h. After dehydration, cells were embedded in epoxy resin and then solidified for 12 h at 45°C. After being sliced into 70 nm-thick sections, the cells were stained with 2% uranyl acetate for 30 min and 2% lead citrate for 15 min at 37°C. Finally, the ultrastructure of the cells was observed under a JEM1230 transmission electron microscope in fields with ×8,000 magnification (JEOL, Ltd.).

Flow cytometry

K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. For the determination of Bax and Bcl-2 levels, 1×106 cells were fixed with 200 µl of an acetone and glutaraldehyde mixture (1:1) for 20 min at 4°C. After rinsing with PBS, each sample was incubated with anti-Bax FITC (1:20, cat. no. DB1010, DB Biotech) or anti-Bcl-2 FITC (1:20, cat. no. DB1011) for 15 min in 100 µl of PBS in the dark at room temperature, and then suspended in 500 µl of PBS. For detection of P-gp expression, cells were collected and washed with PBS, and then incubated with antibodies against P-gp (1:20, cat. no. ab93590, Abcam) for 15 min in the dark. After rinsing with PBS, cells were suspended in 500 µl of PBS. To analyse caspase-3 activity, 1 µl of FITC-DEVD-FMK (1:100, cat. no. QIA911KIT, Sigma-Aldrich; Merck KGaA) was added to cell suspensions in 100 µl of washing buffer. After incubation for 30 min at 37°C, the cells were washed and suspended in 500 µl of washing buffer. After treatment, these samples were analysed using a flow cytometer (Beckman Coulter, Inc.) and analysed using Windows Multiple Document Interface for Flow Cytometry software (version 2.8; The Scripps Research Institute, La Jolla, CA, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. A total of 1×106 cells were collected, and then total RNA was extracted from cells with a TRIzol kit according to the manufacturer's protocols. Both the concentrations and purity of the extracted RNA samples were determined by spectrophotometry. Subsequently, 500 ng of each RNA sample was converted into cDNA using a Prime Script RT Master Mix (Takara Bio, Inc.). Temperature protocol: 70°C for 30 min, 37°C for 15 min, 95°C for 5 min. qPCR assays were performed on a Rotor-Gene 3000 quantitative PCR amplifier (Corbett, Australia) with a SYBR Premix Taq II kit (Takara) and primers. The following primers were used: mdr1 forward, 5′-CTCATAATTCCATTAGGACG-3′; mdr1 reverse, 5′-GCTCACGCTACAGGTCCTGT-3′; β-actin forward, 5′-TGCTCCTCCTGAGCGCAAGTA-3′; and β-actin reverse, 5′-CCACATCTGCTGGAAGGTGGA-3′. The conditions included an initial denaturing step at 95°C for 10 sec, then 40 cycles of denaturing at 95°C for 5 sec and annealing at 60°C for 30 sec. The relative expression of each mRNA was calculated by comparison to β-actin mRNA using the 2−ΔΔCq method (29). All samples were prepared in triplicate.

Western blotting

K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. Cells were lysed in RIPA lysis buffer with phenylmethanesulfonyl fluoride (PMSF; PMSF/RIPA=1:100) for 30 min on ice and then centrifuged at 16,000 × g and 4°C for 15 min. The protein concentration of the supernatant was measured using a bicinchoninic acid protein assay kit. Equal amounts of protein (40 µg) from cell extracts were separated via 10% SDS-PAGE and transferred onto PVDF membranes. After blocking with 5% defatted milk in PBST at 24°C for 1 h, the membranes were incubated with primary antibodies including anti-P62 (1:500), anti-LC3 (1:2,000), anti-Bax (1:1,000), anti-Bcl-2 (1:1,000), anti-cleaved Caspase-3 (1:1,000), anti-P-gp (1:500) and anti-β-actin (1:1,000) at 4°C overnight and then with HRP-conjugated secondary antibodies (1:10,000). Finally, the protein bands of the immunoblot were determined using a chemiluminescent approach in a dark room. Protein bands were visualized using enhanced chemiluminescence reagents (EMD Millipore). Western blots were scanned using an Infrared Imaging System (LI-COR Biosciences) and the bands were quantified using ImageJ software (version 1.45S; National Institutes of Health).

Statistical analysis

Student's t-test or one-way ANOVA was conducted to compare the differences in continuous data. Bonferroni correction was employed to determine the significance of differences between two groups. All statistical analyses were conducted by using SPSS 17.0 software (SPSS, Inc.). All statistical analyses were two-sided, and the results are all presented as the mean ± the standard deviation of at least three independent experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

Overexpression of P-gp and increased autophagic activity co-occur in K562/ADM cells

To investigate the differences in P-gp levels between K562/ADM and K562 cells, P-gp protein expression levels were detected via flow cytometric analysis. As shown in Fig. 1A, the positive rate and mean fluorescence intensity of P-gp were both markedly elevated in K562/ADM cells compared with K562 cells. Similar results were observed with a western blotting assay (Fig. 1B), which indicated that K562/ADM cells exhibited notably higher P-gp protein expression levels compared with K562 cells. Furthermore, the basic autophagic activities of K562/ADM and K562 cells were measured. After observation of autophagosomes in cells under a transmission electron microscope, more cytosolic contents-packaged autophagic vacuoles were identified in K562/ADM cells compared with K562 cells (Fig. 2A). To further verify the results described above, other autophagic indicators were examined, such as LC3 and P62 (also known as sequestosome-1). As we know, the ratio of LC3-II to LC3-I will become larger when the quantity of autophagosomes increases, and P62, a ubiquitination substrate, has a contrary relationship with autophagic activity (27). As presented in Fig. 2B and C, K562/ADM cells demonstrated a notably increased LC3-II/LC3-I ratio and decreased P62 levels compared with K562 cells, indicating that K562/ADM cells exhibited a higher level of autophagic flux.

HCQ enhances the cytotoxic effects of ADM in K562/ADM cells

Different concentrations of HCQ were used to investigate its cytotoxic effects on K562/ADM and K562 cells using an MTT assay. As presented in Fig. 1C, HCQ inhibited the viability of K562/ADM and K562 cells in a dose-dependent manner. K562 and K562/ADM cells demonstrated almost no cytotoxicity (cell viability >80%) following treatment with 4 and 16 µmol/l of HCQ, respectively, which were selected as the maximum non-toxic concentrations in the following experiments. Then, K562/ADM and K562 cells were treated with different concentrations of ADM alone or following pre-treatment with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM for 12, 24 or 48 h. It was identified that the IC50 values for ADM in K562/ADM cells following HCQ treatment for 12, 24 and 48 h were 45.66±5.08, 21.44±0.59 and 3.26±0.86 µmol/l, respectively, which were 0.84-, 0.46- and 0.21-fold less than those values after only ADM treatment (Fig. 1D). By contrast, HCQ slightly decreased the IC50 values for ADM in K562 cells (Fig. 1E). These results indicated that HCQ could effectively reverse the ADM resistance of K562/ADM cells.

HCQ reduces ADM-induced autophagy in K562 and K562/ADM cells

To investigate the effect of ADM on autophagy, K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM, respectively, for 24 h or pretreated with HCQ (16 and 4 µmol/l, respectively) for 3 h prior to exposure to ADM. Then, they were observed under a transmission electron microscope for autophagic vacuoles. As presented in Fig. 2A, more autophagic vacuoles (indicated by black arrows) were found in ADM-treated cells than in the controls, and HCQ increased the number of autophagic vacuoles in K562 and K562/ADM cells treated with ADM. To further validate these results, the levels of the autophagy markers LC3 and P62 were analysed using a western blotting assay. It was found that ADM induced autophagy in K562 and K562/ADM cells, as indicated by the increased LC3-II/LC3-I ratios (P=0.001 and 0.023, respectively; Fig. 2B and D) and a decrease in the P62 level (P=0.023 and 0.001, respectively; Fig. 2C and E). Furthermore, HCQ increased the accumulation of LC3-II (P=0.009 and 0.012, respectively) and P62 (P=0.012 and 0.022, respectively) in both K562 and K562/ADM cells exposed to ADM, indicating that HCQ reduced autophagy activity induced by ADM. Collectively, these findings indicated that HCQ can inhibit the ADM-induced autophagy response in K562 and K562/ADM cells by preventing the degradation of autophagic vacuoles.

HCQ potentiates ADM-induced apoptosis in K562/ADM cells

To explore the apoptosis response in two cell lines following ADM exposure, K562/ADM and K562 cells were treated with 35 and 0.75 µmol/l of ADM separately for 24 h and stained with AO/EB. Observed under a fluorescence microscope, the controls exhibited green-stained integral nuclei, whereas yellow or orange nuclei with condensed and fragmentary chromatin were found in cells treated with ADM, suggesting that ADM induced apoptosis in the two cell lines. Furthermore, when cells were pretreated with HCQ for 3 h prior to exposure to ADM, the proportion of apoptotic K562/ADM cells was notably increased compared with those treated only with ADM; however, the effects of HCQ pre-treatment on the apoptosis of K562 cells were not pronounced (Fig. 3A). Morphological changes indicated that HCQ upregulated ADM-induced apoptosis in K562/ADM.

To confirm these apparent changes in apoptosis, caspase-3 activity was evaluated using flow cytometry, and the expression of the cleaved forms of caspase-3 was detected via western blotting. As presented in Fig. 3B and C, ADM led to an increased mean fluorescence intensity for activated caspase-3 in K562 (P=0.032) and K562/ADM (P=0.001) cells. K562/ADM cells pre-treated with HCQ prior to exposure to ADM exhibited a significantly greater mean fluorescence intensity for activated caspase-3 compared with cells exposed to only ADM (P=0.007), whereas K562 cells were not significantly affected by HCQ pre-treatment. Similar results were also obtained from the western blot assay (Fig. 3D and E).

These results were verified by the determination of the protein expression levels of Bax/Bcl-2. Flow cytometry demonstrated that the ratio of Bax/Bcl-2 was upregulated in K562/ADM cells by ADM (P=0.001), which could be further enhanced following 3 h of HCQ pre-treatment (P=0.001; Fig. 4A-C). However, there were no notable effects on K562 cells. The results of the western blot assay of K562/ADM cells were also consistent with these observations, but the situations of K562 cells were incompatible between the flow cytometry and western blotting results (Fig. 4D and E). The change of K562 cells may be induced by other causes in addition to apoptosis. Collectively, these findings indicate that HCQ could sensitise K562/ADM cells to caspase-dependent apoptosis induced by ADM, thereby reversing multidrug resistance.

HCQ diminishes the expression of P-gp in K562/ADM cells exposed to ADM

To further clarify the role of autophagic activity in multidrug resistance, the present study modulated autophagy and observed alterations in the expression of the drug resistance-associated protein P-gp during the autophagy process. ADM (35 µmol/l) was used to induce the autophagic response, and 16 µmol/l HCQ was used to block autophagy in K562/ADM cells. As demonstrated by a western blot assay, P-gp expression in K562/ADM cells was increased in the presence of ADM (P=0.004), and 3 h HCQ treatment decreased ADM-induced P-gp expression to a level similar to that of control treatment (P=0.011; Fig. 5A and B). A flow cytometric assay validated this observation, as a slight increase in the mean fluorescence intensity for P-gp was found in K562/ADM cells following ADM incubation, which fell with HCQ treatment (Fig. 5C). In addition, the mRNA expression profile of the mdr1 gene in K562/ADM cells following the aforementioned treatments, as determined via RT-qPCR analysis, was consistent with P-gp expression (Fig. 5D). Collectively, these findings indicated that the inhibition of autophagy diminished the expression of P-gp, which may be involved in autophagic regulation in K562/ADM cells.

Discussion

Leukaemia is a highly malignant cancer of the blood system characterised by rapid onset, poor prognosis and high cost of treatment (30). It is known that leukaemia is a malignancy with one of the highest mortality rates (31). Although there are a variety of chemotherapy drugs for use against leukaemia, chemotherapy-induced multidrug resistance is still a challenging issue during drug treatment (31). Therefore, more effective clinical therapies are needed to conquer the drug resistance induced by chemotherapy.

K562/ADM cells are a multidrug-resistant leukaemia cell line with high expression of P-gp, and are acquired by exposing K562 cells to step-wise increasing concentrations of ADM (32). They are characterised by their resistance not only to ADM but also to other anticancer drugs with varying structures and functions (32). The results of the present study demonstrated that K562/ADM cells exhibited a high level of P-gp expression, while K562 cells expressed minimal levels of P-gp, indicating that K562/ADM cells are a multidrug-resistant cell line and that K562 is a sensitive cell strain. In addition, it was identified that basic autophagy activity in K562/ADM cells was distinctly higher than it was in K562 cells, which suggests that autophagy may be involved in multidrug resistance. This result is consistent with our previous study (27).

It is well known that autophagy serves a dual role in tumorigenesis. It has been suggested that, in cancer cells, certain chemotherapeutics are able to induce autophagic cell death (ACD), a cell death pathway distinct from apoptosis. Puissant et al (26) reported that resveratrol treatment could lead to ACD in chronic myelogenous leukaemia (CML) cells, and that autophagy inhibition decreased the sensitivity of CML cells to resveratrol. Li et al (33) suggested that 3-methyladenine, an autophagy inhibitor, could promote Raji cell proliferation by restraining the ACD induced by arsenic trioxide. However, in the majority of established tumours, autophagy protects against the survival of cancer cells and confers resistance to chemotherapy. Han et al (34) reported that daunorubicin initiated protective autophagy in acute myelocytic leukaemia (AML) via the mitogen-activated protein kinase (MAPK) kinase/ERK pathway, which resulted in the daunorubicin resistance of AML cells. This pro-survival role of autophagy has also been supported by other studies showing that autophagy inhibition elevated sensitivity to chemotherapy in multiple myeloma, breast cancer, colorectal cancer and prostate cancer cells (3538).

Chemotherapy drugs can induce autophagy in leukaemia cells. Li et al (39) indicated that macroautophagy activity in Raji cells was significantly enhanced by arsenic trioxide. Consistent with this result, the present study demonstrated that autophagy in K562/ADM and K562 cells was increased with ADM treatment. It was also found that HCQ could inhibit the ADM-induced autophagy response in K562 and K562/ADM cells by restraining the degradation of autophagic vacuoles. HCQ belongs to the 4-amino quinoline family, which is used for antimalarial medications (40). HCQ was employed to inhibit autophagy due to its well-known safety profile. Moreover, when cells were treated with HCQ at nontoxic concentrations, the IC50 values of ADM in K562/ADM cells at different time points were significantly reduced, indicating that HCQ could effectively reverse the ADM resistance of K562/ADM cells, and that increased autophagy is one of the mechanisms of multidrug resistance in K562/ADM cells.

In addition to autophagy, it has been suggested that apoptosis is involved in drug resistance (41). Over the years, evidence has accumulated demonstrating that apoptosis and autophagy may share the same signalling pathways, such as p38-MAPK and JNK (42), and there is an interconnected relationship between apoptosis and autophagy (43). For example, a previous study described that autophagy inhibited apoptosis by decreasing Bcl-2-associated death promoter and Bcl-2-like protein 11 expression in hepatocellular carcinoma cells (44). Another study suggested that autophagy was involved in the early stage of apoptosis, leading to the death of acute lymphocytic leukaemia cells (45). The data from the present study demonstrated that the proportion of apoptotic cells induced by ADM was prominently upregulated following pre-treatment with HCQ in K562/ADM cells, which was also confirmed by an increase in caspase-3 activity and the enhanced ratio of Bax/Bcl-2; two classic hallmark proteins for apoptosis. Therefore, autophagy inhibition by HCQ may potentiate ADM-induced apoptosis and contribute to overcoming ADM resistance in K562/ADM cells.

P-gp functions as an ATP-dependent drug ejector pump and decreases the intracellular concentrations of its substrates with different chemical structures (46). Consequently, the enhanced expression of P-gp may lead to resistance to a wide range of drugs (47). It has been suggested that P-gp may be involved in regulating molecular metabolism, proliferation and differentiation in cells (48), indicating that P-gp might potentially possess multiple physiological functions. A previous study demonstrated the synergistic roles of P-gp, autophagy and NF-κB pathways in the epirubicin resistance of triple-negative breast cancer (49). However, another study suggested that the role of P-gp in the development of multidrug resistance in breast cancer may be independent of autophagy (50). The present study preliminarily demonstrated an association between autophagy and P-gp expression. The results demonstrated that ADM treatment resulted in a significant augmentation of autophagy accompanied by increased mdr1/P-gp expression in K562/ADM cells. In addition, the autophagy inhibitor HCQ markedly inhibited the ADM-induced increase in autophagy, as well as enhancing mdr1/P-gp expression in the cells, and sensitised the drug-resistant cells to ADM. This may mean that inhibition of autophagy attenuates the expression of P-gp, and that P-gp may be involved in autophagic regulation in drug-resistant cells. The true co-regulation and interaction between autophagy and P-gp in modifying multidrug resistance requires confirmation in future investigations. In addition, Liu et al (51) suggested that nelfinavir could reverse ADM resistance in K562/ADR cells by inhibiting the P-gp efflux function without affecting P-gp protein and mRNA expression levels. However, the results of the present study indicated that the autophagy inhibitor HCQ might sensitise the drug-resistant cells to ADM by reducing mdr1/P-gp expression at the transcriptional level in cells.

Collectively, the present findings demonstrated that HCQ can reverse ADM resistance in K562/ADM cells by inhibiting autophagy, increasing K562/ADM cell apoptosis and decreasing P-gp expression. It is hypothesised that P-gp may potentially sustain drug resistance in leukaemia cells by participating in the regulation of autophagic activity. The present study revealed a potential novel target for leukaemia intervention and provides further insight into understanding the role of autophagy in the drug resistance of leukaemia cells.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China grant no. 81541025 (to HLW), the Fundamental Research Funds for the Central Universities grant no. lzujbky-2016-174 (to HLW) and the Natural Science Fund of Gansu grant no. 1208RJZA183 (to HLW). This work was also supported by the National Natural Science Foundation of China grant no. 31571196 (to LW).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

FFW performed the examinations, and was a major contributor in writing the manuscript. LW and HLW made substantial contributions to conception and design. WTL interpreted data, translated and revised the manuscript critically for important intellectual content. ZWZ, JC, JY, CMY, MMY, MYW, NZ and XMQ helped to analyze and interpret data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wouters BJ and Delwel R: Epigenetics and approaches to targeted epigenetic therapy in acute myeloid leukemia. Blood. 127:42–52. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Burnett A, Wetzler M and Lowenberg B: Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 29:487–494. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Gao W and Estey E: Moving toward targeted therapies in acute myeloid leukemia. Clin Adv Hematol Oncol. 13:748–754. 2015.PubMed/NCBI

4 

Ramos P and Bentires-Alj M: Mechanism-based cancer therapy: Resistance to therapy, therapy for resistance. Oncogene. 34:3617–3626. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Jones PM and George AM: The ABC transporter structure and mechanism: Perspectives on recent research. Cell Mol Life Sci. 61:682–699. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Borst P, Evers R, Kool M and Wijnholds J: A family of drug transporters: The multidrug resistance-associated proteins. J Natl Cancer Inst. 92:1295–1302. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Bodor M, Kelly EJ and Ho RJ: Characterization of the human MDR1 gene. AAPS J. 7:E1–E5. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Ambudkar SV, Dey S, Hrycyna CA, Ramachandra M, Pastan I and Gottesman MM: Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol. 39:361–398. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Abraham J, Salama NN and Azab AK: The role of P-glycoprotein in drug resistance in multiple myeloma. Leuk Lymphoma. 56:26–33. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Danial NN and Korsmeyer SJ: Cell death: Critical control points. Cell. 116:205–219. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Delbridge AR and Strasser A: The BCL-2 protein family, BH3-mimetics and cancer therapy. Cell Death Differ. 22:1071–1080. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Zheng JH, Viacava Follis A, Kriwacki RW and Moldoveanu T: Discoveries and controversies in BCL-2 protein-mediated apoptosis. FEBS J. 283:2690–2700. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Kroemer G, Galluzzi L and Brenner C: Mitochondrial membrane permeabilization in cell death. Physiol Rev. 87:99–163. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Ghibelli L and Diederich M: Multistep and multitask bax activation. Mitochondrion. 10:604–613. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Kvansakul M and Hinds MG: The Bcl-2 family: Structures, interactions and targets for drug discovery. Apoptosis. 20:136–150. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Indran IR, Tufo G, Pervaiz S and Brenner C: Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochim Biophys Acta. 1807:735–745. View Article : Google Scholar : PubMed/NCBI

17 

Zhi X, Feng W, Rong Y and Liu R: Anatomy of autophagy: From the beginning to the end. Cell Mol Life Sci. 75:815–831. 2017. View Article : Google Scholar : PubMed/NCBI

18 

He C and Klionsky DJ: Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 43:67–93. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Levy JMM, Towers CG and Thorburn A: Targeting autophagy in cancer. Nat Rev Cancer. 17:528–542. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Budini M, Buratti E, Morselli E and Criollo A: Autophagy and its impact on neurodegenerative diseases: New roles for TDP-43 and C9orf72. Front Mol Neurosci. 10:1702017. View Article : Google Scholar : PubMed/NCBI

21 

Morandi E, Jagessar SA, t Hart BA and Gran B: EBV infection empowers human b cells for autoimmunity: Role of autophagy and relevance to multiple sclerosis. J Immunol. 199:435–448. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Cosin-Roger J, Simmen S, Melhem H, Atrott K, Frey-Wagner I, Hausmann M, de Vallière C, Spalinger MR, Spielmann P, Wenger RH, et al: Hypoxia ameliorates intestinal inflammation through NLRP3/mTOR downregulation and autophagy activation. Nat Commun. 8:982017. View Article : Google Scholar : PubMed/NCBI

23 

Fulda S: Autophagy in cancer therapy. Front Oncol. 7:1282017. View Article : Google Scholar : PubMed/NCBI

24 

Gewirtz DA: The four faces of autophagy: Implications for cancer therapy. Cancer Res. 74:647–651. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Mizushima N, Levine B, Cuervo AM and Klionsky DJ: Autophagy fights disease through cellular self-digestion. Nature. 451:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Puissant A, Robert G, Fenouille N, Luciano F, Cassuto JP, Raynaud S and Auberger P: Resveratrol promotes autophagic cell death in chronic myelogenous leukemia cells via JNK-mediated p62/SQSTM1 expression and AMPK activation. Cancer Res. 70:1042–1052. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Wang F, Chen J, Zhang Z, Yi J, Yuan M, Wang M, Zhang N, Qiu X, Wei H and Wang L: Differences of basic and induced autophagic activity between K562 and K562/ADM cells. Intractable Rare Dis Res. 6:281–290. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Liu K, Liu PC, Liu R and Wu X: Dual AO/EB staining to detect apoptosis in osteosarcoma cells compared with flow cytometry. Med Sci Monit Basic Res. 21:15–20. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Daver N, Cortes J, Kantarjian H and Ravandi F: Acute myeloid leukemia: Advancing clinical trials and promising therapeutics. Expert Rev Hematol. 9:433–445. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Plasschaert SL, de Bont ES, Boezen M, vander Kolk DM, Daenen SM, Faber KN, Kamps WA, de Vries EG and Vellenga E: Expression of multidrug resistance-associated proteins predicts prognosis in childhood and adult acute lymphoblastic leukemia. Clin Cancer Res. 11:8661–8668. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Chen J, Wei H, Xie B, Wang B and Cheng J and Cheng J: Endoplasmic reticulum stress contributes to arsenic trioxide-induced apoptosis in drug-sensitive and -resistant leukemia cells. Leuk Res. 36:1526–1535. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Li CL, Wei HL, Chen J, Wang B, Xie B, Fan LL and Li LJ: Arsenic trioxide induces autophagy and antitumor effects in burkitt's lymphoma raji cells. Oncol Rep. 32:1557–1563. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Han W, Sun J, Feng L, Wang K, Li D, Pan Q, Chen Y, Jin W, Wang X, Pan H and Jin H: Autophagy inhibition enhances daunorubicin-induced apoptosis in K562 cells. PLoS One. 6:e284912011. View Article : Google Scholar : PubMed/NCBI

35 

Riz I, Hawley TS and Hawley RG: KLF4-SQSTM1/p62-associated prosurvival autophagy contributes to carfilzomib resistance in multiple myeloma models. Oncotarget. 6:14814–14831. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Sun Q, Liu T, Yuan Y, Guo Z, Xie G, Du S, Lin X, Xu Z, Liu M, Wang W, et al: MiR-200c inhibits autophagy and enhances radiosensitivity in breast cancer cells by targeting UBQLN1. Int J Cancer. 136:1003–1012. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Zhang H, Tang J, Li C, Kong J, Wang J, Wu Y, Xu E and Lai M: MiR-22 regulates 5-FU sensitivity by inhibiting autophagy and promoting apoptosis in colorectal cancer cells. Cancer Lett. 356:781–790. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Kim SH, Kim KY, Yu SN, Park SK, Choi HD, Ji JH and Ahn SC: Autophagy inhibition enhances silibinin-induced apoptosis by regulating reactive oxygen species production in human prostate cancer PC-3 cells. Biochem Biophys Res Commun. 468:151–156. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Li CL, Wei HL, Chen J, Wang B, Xie B, Fan LL and Li LJ: Ebb-and-flow of macroautophagy and chaperone-mediated autophagy in raji cells induced by starvation and arsenic trioxide. Asian Pac J Cancer Prev. 15:5715–5719. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Caminal-Montero L and Suárez-Díaz S: Hydroxychloroquine and antimalarials. J Rheumatol. 15:jrheum.190559. 2019.

41 

Zhang H, Han D, Lv T, Liu K, Yang Y, Xu X and Chen Y: Novel peptide myristoly-CM4 induces selective cytotoxicity in leukemia K562/MDR and Jurkat cells by necrosis and/or apoptosis pathway. Drug Des Devel Ther. 13:2153–2167. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, He C and Pan H: P38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 344:174–179. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Oral O, Akkoc Y, Bayraktar O and Gozuacik D: Physiological and pathological significance of the molecular cross-talk between autophagy and apoptosis. Histol Histopathol. 31:479–498. 2016.PubMed/NCBI

44 

Zhou Y, Sun K, Ma Y, Yang H, Zhang Y, Kong X and Wei L: Autophagy inhibits chemotherapy-induced apoptosis through downregulating bad and bim in hepatocellular carcinoma cells. Sci Rep. 4:53822014. View Article : Google Scholar : PubMed/NCBI

45 

Yuan N, Song L, Zhang S, Lin W, Cao Y, Xu F, Fang Y, Wang Z, Zhang H, Li X, et al: Bafilomycin A1 targets both autophagy and apoptosis pathways in pediatric B-cell acute lymphoblastic leukemia. Haematologica. 100:345–356. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Rees DC, Johnson E and Lewinson O: ABC transporters: The power to change. Nat Rev Mol Cell Biol. 10:218–227. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Breier A, Gibalova L, Seres M, Barancik M and Sulova Z: New insight into p-glycoprotein as a drug target. Anticancer Agents Med Chem. 13:159–170. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Silva R, Vilas-Boas V, Carmo H, Dinis-Oliveira RJ, Carvalho F, de Lourdes Bastos M and Remiao F: Modulation of P-glycoprotein efflux pump: Induction and activation as a therapeutic strategy. Pharmacol Ther. 149:1–123. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Zhang LH, Yang AJ, Wang M, Liu W, Wang CY, Xie XF, Chen X, Dong JF and Li M: Enhanced autophagy reveals vulnerability of P-gp mediated epirubicin resistance in triple negative breast cancer cells. Apoptosis. 21:473–488. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Sun WL, Lan D, Gan TQ and Cai ZW: Autophagy facilitates multidrug resistance development through inhibition of apoptosis in breast cancer cells. Neoplasma. 62:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Liu W, Meng Q, Sun Y, Wang C, Huo X, Liu Z, Sun P, Sun H, Ma X and Liu K: Targeting P-Glycoprotein: Nelfinavir reverses adriamycin resistance in K562/ADR cells. Cell Physiol Biochem. 51:1616–1631. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang F, Zhang Z, Leung WT, Chen J, Yi J, Ying C, Yuan M, Wang M, Zhang N, Qiu X, Qiu X, et al: Hydroxychloroquine reverses the drug resistance of leukemic K562/ADM cells by inhibiting autophagy. Mol Med Rep 20: 3883-3892, 2019
APA
Wang, F., Zhang, Z., Leung, W.T., Chen, J., Yi, J., Ying, C. ... Wei, H. (2019). Hydroxychloroquine reverses the drug resistance of leukemic K562/ADM cells by inhibiting autophagy. Molecular Medicine Reports, 20, 3883-3892. https://doi.org/10.3892/mmr.2019.10621
MLA
Wang, F., Zhang, Z., Leung, W. T., Chen, J., Yi, J., Ying, C., Yuan, M., Wang, M., Zhang, N., Qiu, X., Wang, L., Wei, H."Hydroxychloroquine reverses the drug resistance of leukemic K562/ADM cells by inhibiting autophagy". Molecular Medicine Reports 20.4 (2019): 3883-3892.
Chicago
Wang, F., Zhang, Z., Leung, W. T., Chen, J., Yi, J., Ying, C., Yuan, M., Wang, M., Zhang, N., Qiu, X., Wang, L., Wei, H."Hydroxychloroquine reverses the drug resistance of leukemic K562/ADM cells by inhibiting autophagy". Molecular Medicine Reports 20, no. 4 (2019): 3883-3892. https://doi.org/10.3892/mmr.2019.10621