Persistent STAT5-mediated ROS production and involvement of aberrant p53 apoptotic signaling in the resistance of chronic myeloid leukemia to imatinib

  • Authors:
    • Yanhong Cheng
    • Yingchan Hao
    • Aimei Zhang
    • Chaojie Hu
    • Xiaoxiao Jiang
    • Quan Wu
    • Xiucai Xu
  • View Affiliations

  • Published online on: October 20, 2017     https://doi.org/10.3892/ijmm.2017.3205
  • Pages: 455-463
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The persistent activation of signal transducer and activator of transcription 5 (STAT5) may principally be attributed to breakpoint cluster region (BCR)-Abelson murine leukemia viral oncogene homolog 1 (ABL1), and have multi-faceted effects in the development of chronic myeloid leukemia (CML). The p53 protein network regulates important mechanisms in DNA damage repair, cell cycle regulation/checkpoints, and cell senescence and apoptosis, as demonstrated by its ability to positively regulate the expression of various pro-apoptotic genes, including B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X protein (Bax). In the present study, it was observed that the mRNA levels of STAT5A and STAT5B were upregulated in patients with imatinib-resistant CML and in the imatinib-resistant K562/G CML cell line. In addition, increased expression of STAT5 was observed in the BCR-ABL1 mutation group, compared with that in the non-BCR-ABL1 mutation group, regardless of patient imatinib resistance state. Elevated levels of reactive oxygen species (ROS) and DNA double-strand breaks were identified in K562/G cells using flow cytometric and phosphorylated H2AX (γ-H2AX) foci immunofluorescence assays, respectively, compared with the imatinib-sensitive K562 cells. The levels of intracellular ROS and γ-H2AX were decreased by the ROS scavenger (N-acetylcysteine), and ROS levels were also markedly reduced by STAT5 inhibitor (SH-4-54). In addition, imatinib significantly inhibited the proliferation of K562 and K562/G cells, with half maximal inhibitory concentration values of 0.17±0.07 and 14.78±0.43 µM, respectively, and the levels of apoptosis were significantly different between K562 and K562/G cells following treatment with imatinib. The mRNA and protein levels of STAT5 and mouse double minute 2 homolog (MDM2) were upregulated, whereas those of Bax were downregulated in K562/G cells, as determined using western blot analysis. Additionally, although the two cell lines exhibited relatively low protein expression levels of p53, lower levels of p53 and TPp53BP1 transcripts were detected in the K562/G cells. Taken together, these findings suggest that the resistance of CML to the tyrosine kinase inhibitor, imatinib, may be associated with persistent STAT5-mediated ROS production, and the abnormality of the p53 pathway.

Introduction

Chronic myeloid leukemia (CML) is a malignant hematopoietic stem cell disorder characterized by the reciprocal translocation of chromosomes 9 and 22, resulting in the expression of a constitutively active breakpoint cluster region (BCR)-Abelson murine leukemia viral oncogene homolog 1 (ABL1) tyrosine kinase. The introduction of tyrosine kinase inhibitors (TKIs), including imatinib, nilotinib and dasatinib, has revolutionized the treatment of patients with CML and markedly improved their quality of life (13). A large proportion of patients with CML subjected to long-term TKI treatment exhibit high-level and stable molecular responses and, in certain cases, leads to undetectable minimal residual disease (4). However, up to 40% of patients show substantial resistance or intolerance to TKIs, typically during or following treatment, which seriously limits the attainment of treatment-free remission or a cure (5,6). The underlying mechanisms of drug resistance remain to be elucidated, although they are considered to be attributable mainly to mutations in the kinase domain (KD) of BCR-ABL1 (7) and to signaling pathways activated by BCR-ABL1, including RAS, phosphoinositide 3-kinase (PI3K), nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 5 (STAT5) (8). In particular, STAT5 has at least four key effects in the initiation and progression of CML (9), and may be considered a crucial modulator of imatinib responsiveness (10).

Reactive oxygen species (ROS) are generated as a by-product of the normal oxidative metabolism in eukaryotic cells which, if generated to excess, can cause damage to cellular molecules, including DNA, RNA and proteins, resulting in oxidative stress. It is well established that ROS are important in a variety of pathophysiological processes, including cell differentiation, host defense, oxygen sensing, and cell proliferation and apoptosis. The generation of ROS is regulated by the STAT5 transcription factor, which is crucial in a number of hematological diseases and commonly functions downstream of certain kinases, including BCR-ABL1 in CML (11). It is known that ROS can trigger genomic stress, secondary genome instability and the potential accumulation of gene mutations within the basal p53-mediated DNA damage response (DDR) pathway. Therefore, there is scientific support for a link between STAT5, ROS production and BCR-ABL1 mutations.

p53 is considered to be a universal sensor of genotoxic stress, and is involved in different DNA repair mechanisms and in cell cycle checkpoint regulation through various signaling pathways. Generally, p53 and its downstream signaling pathways are involved in ROS-induced apoptosis. Numerous studies have reported crosstalk between p53 signaling and ROS metabolism during normal physiological conditions, based on the feedback loop between ROS and p53, whereby mitochondria-generated ROS promote p53 translocation and, in turn, trigger oxidative stress. A previous study also demonstrated that the progression of CML was frequently accompanied by increased inactivation of p53, and that p53 mutations were seldom observed in chronic phase CML, but were detected in ~30% of patients presenting with myeloid blast crisis (12). However, the mechanism of the interaction between p53 and STAT5-mediated ROS production in the resistance of CML to TKIs remains to be elucidated. Thus, in this study, we aimed to examine the role of STAT5-mediated ROS production and aberrant p53 apoptotic signaling in the resistance of CML to imatinib.

Materials and methods

Cell culture

The human K562 CML cell line was purchased from the Shanghai Institutes for Biological Sciences of the Chinese Academy of Sciences (Shanghai, China) and imatinib-resistant K562 cells (K562/G) were obtained from the Department of Pharmacology at the Institute of Hematology, Chinese Academy of Medical Sciences (Tianjin, China). The cells were cultured in Iscove's modified Dulbecco's medium (IMDM) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 1% penicillin and 1% streptomycin, in a 5% CO2 incubator at 37°C. The cells were cultured in the presence or absence of 1 µM imatinib. All cells were passaged every 2–3 days.

Clinical specimens

All patients were enrolled from Anhui Provincial Hospital of Anhui Medical University (Hefei, China) between July, 2015 and September, 2016. The present study was approved by the Ethics Committee of the Affiliated Provincial Hospital of Anhui Medical University. Peripheral blood mononuclear cells (PBMCs) were isolated from patients with CML during routine examinations following the provision of informed consent from all patients, in accordance with the Declaration of Helsinki. Aliquots of the PBMCs were used for subsequent analysis. The molecular responses to imatinib were assessed according to the BCR-ABL/ABL ratio, standardized to the International Scale (13). According to the 2013 European Leukemia Net guidelines (https://www.leukemia-net.org), responses were assessed using standardized reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis at 3 and 6 months (14). BCR-ABL1 transcript levels ≤10% at 3 months and <1% at 6 months were defined as optimal responses, whereas levels >10% at 6 months were defined as failed responses (14). BCR-ABL KD mutations and p53 mutations were assessed as described previously (15). The characteristics of the patients, and the results of the BCR-ABL KD mutations and p53 mutations are presented in Table I.

Table I

Clinical characteristics of patients.

Table I

Clinical characteristics of patients.

CharacteristicChronic myeloid leukemia cases (n=63)Non-IM-resistant (n=31)IM-resistant (n=32)
Age at diagnosis (years)
 Median374036
 Range20.4–83.622.3–79.520.4–83.6
Sex
 Male422121
 Female211011
White blood cells (107/ml)49.16±69.6812.17±6.5584.99±83.52
International standard
 <1%31310
 >1%32032
BCR/ABL mutation rate (%)19.019.6028.13
p53 mutation rate (%)0.000.000.00

[i] Non-IM-resistant patients had a BCR-ABL1/ABL1 ratio of 9.6%. BCR-ABL1 mutations, were D276G, F317L and F359I. IM-resistant patients had a BCR-ABL1/ABL1 ratio of 28.13%. BCR-ABL1 mutations were G250E, M244V, M387L, V299L, Y253H, E255K, E297K, F317L and G250E. IM, imatinib; BCR-ABL1, breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1.

DNA damage signaling pathway PCR array

The K562 and K562/G cells lines (~1×107 cells) were collected, respectively. RNA extraction and first strand cDNA synthesis were performed via a routine protocol (16). A Human DNA Damage Signaling Pathway RT2 Profiler™ PCR array was purchased from SABiosciences (Frederick, MD, USA). Total RNA was extracted using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and RNA samples (1 µg) were reverse transcribed into cDNA using the RT2 PCR Array First Strand Synthesis kit (Qiagen). Subsequently, 91 µl of ddH2O was added to each 20 µl cDNA synthesis reaction and mixed well. The following components were mixed in a 5-ml tube or a multi-channel reservoir: 1,050 µl of 2X SuperArray PCR master mix, 105 µl of the diluted first-strand cDNA synthesis reaction and 945 µl of ddH2O. The cocktails were then added to the PCR array. Real-time PCR detection was performed under the following thermocycling conditions: 95°C for 10 min; 40 cycles of 95°C for 15 sec and 60°C for 1 min. RT-qPCR and data analyses via the 2−ΔΔCq method were performed according to the manufacturer's protocol. The expression data were normalized to that of the housekeeping gene GAPDH. Differences in the levels of gene expression are presented as the fold increase/decrease, relative to the levels of the housekeeping gene (Table II).

Table II

mRNA fold-differences in DNA damage response genes between K562 and K562G cells.

Table II

mRNA fold-differences in DNA damage response genes between K562 and K562G cells.

GeneAbbreviationK562G/K562 (fold-difference)
Bloom syndrome, RecQ helicase-likeBLM−1.53
Calcium and integrin binding 1 (calmyrin)CIB1−2.02
Growth arrest and DNA-damage-inducible, αGADD45A−1.51
Nth endonuclease III-like 1NTHL1−1.89
Cell cycle checkpoint protein RAD17RAD17−1.55
Tumor protein p53TP53−2.56
Tumor protein p53 binding protein 1TP53BP1−1.93
Hypoxanthine phosphoribosyltransferase 1HPRT1−1.73
Cell counting assay

A cell counting kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Shanghai, China) was used to determine the survival rate of the K562 and K562/G cells following incubation with imatinib. The cells were seeded in a 96-well plate at a density of 5×103 cells/well in IMDM containing 10% FBS. Subsequently, various concentrations of imatinib (0.02–4 µM for K562 cells and 0.2–400 µM for K562/G cells) were added. Following incubation of the cells at 37°C in 5% CO2 for 24 h, 10 µl of CCK-8 solution was added to each well and the cells were incubated for another 4 h. The absorbance was measured at 450 nm with a microplate reader. A well containing medium and CCK-8 solution only was used as a blank control. A control group of cells were incubated with cell culture medium and CCK-8 solution. The half maximal inhibitory concentration (IC50) values of imatinib were determined as the mean of three independent experiments.

Cell apoptosis assessment using Annexin V-FITC and propidium iodide (PI) staining

Briefly, the cells were harvested and washed with cooled PBS at 4°C. Cell suspensions (5×104 cells each) were incubated with Annexin V-FITC (2 µg/ml) and PI (0.5 µg/ml) at room temperature for 15 min in the dark, and subsequently analyzed using flow cytometry (17). The cells were considered to be apoptotic if they exhibited Annexin V+/PI (early apoptotic) or Annexin V+/PI+ (late apoptotic) staining. The data are presented as the mean of three separate experiments, each performed in duplicate.

Detection of intracellular ROS

The accumulation of intracellular ROS was detected using 2′,7′-dichlorofluorescindiacetate (DCFH-DA) probes (Molecular Probes®; Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. In brief, 2×105 cells/ml of K562 and K562/G cells were stained with 2.5 µM DCFH-DA at 37°C for 15 min. The samples were then washed and resuspended in phosphate-buffered saline (PBS), and the fluorescence intensity was analyzed using flow cytometry (FACSCanto II) using BD FACSDiva software v6.1.3 (both from BD Biosciences, San Jose, CA, USA). Additionally, K562/G cells were pretreated with 5 mM N-acetylcysteine (NAC; Meilunbio, Dalian, China) or 20 µM SH-4–54 (Selleck Chemicals, Houston, TX, USA) for 24 h in a humidified atmosphere at 37°C and 5% CO2, and then subjected to the same analysis. For each sample, ~104 cells contained in the gated regions were counted. ROS-positive cells were stained with DCFH-DA. The experiments were repeated three times, and all data were analyzed with BD FACSDiva software.

RT-qPCR analysis

Total RNA was extracted from the cultured cell lines and PBMCs of the patients with CML using TRIzol reagent (Thermo Fisher Scientific, Inc.) and quantified with a NanoDrop spectrophotometer (Amoy Diagnostics, Xiamen, China). A total of 0.5 µg of the isolated RNA was used for cDNA synthesis and qPCR analysis was subsequently performed on a 7500 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) with a SYBR-Green reaction kit (Invitrogen; Thermo Fisher Scientific, Inc.). GAPDH was used as a housekeeping gene to normalize the levels of target gene expression. The changes in mRNA expression levels were calculated using the comparative Cq method, as follows: Fold change = 2−ΔΔCq = [(Cq gene of interest-Cq internal control) sample A-(Cq gene of interest-Cq internal control) sample B] (18). The primer sequences used are listed in Table III.

Table III

Primer sequences used in gene expression analysis.

Table III

Primer sequences used in gene expression analysis.

GenePrimer sequence (5′-3′)Product length (bp)
GAPDHF: GGAGCGAGATCCCTCCAAAAT
R: GGCTGTTGTCATACTTCTCATGG
197
STAT5AF: GCAGAGTCCGTGACAGAGG
R: CCACAGGTAGGGACAGAGTCT
106
STAT5BF: CAGAACACGTATGACCGCTG
R: CTGGAGAGCTACCATTGTTGG
106
p53F: CCAGCAGCTCCTACACCGGC
R: GAAACCGTAGCTGCCCTG
99
Bcl-2F: CATGTGTGTGGAGAGCGTCAA
R: GCCGGTTCAGGTACTCAGTCA
147
BaxF: GCGTCCACCAAGAAGCTGAG
R: ACCACCCTGGTCTTGGATCC
113
MDM2F: GAATCATCGGACTCAGGTACATC
R: TCTGTCTCACTAATTGCTCTCCT
167

[i] STAT5, signal transducer and activator of transcription 5; Bcl-2, B-cell lymphoma-2; Bax, Bcl-2-associated X protein; MDM2, mouse double minute 2 homolog; F, forward; R, reverse.

Immunofluorescence analysis of the expression of phosphorylated H2AX (γ-H2AX)

γ-H2AX forms microscopically visible foci, and the number of γ-H2AX foci has been found to correlate well with the number of DNA double-strand breaks (DSBs) (19). The K562 and K562/G cells (5×104) were plated on glass slides via a cell concentrator and fixed with 4% paraformaldehyde for 30 min at room temperature. Following three washes with PBS containing 0.2% Tween-20, the cells were permeabilized with PBS containing 0.3% Triton X-100 at room temperature for 30 min, and then blocked with blocking buffer containing 5% goat serum (SL038; Solarbio, Beijing, China) and 0.3% Triton X-100 in PBS for 1 h at room temperature. Incubation with primary γ-H2AX antibody (1:200; monoclonal rabbit anti-H2AX; cat. no. 9718; Cell Signaling Technology, Inc., Danvers, MA, USA) was performed in blocking solution overnight at 4°C. The cells were then washed three times and incubated with anti-rabbit IgG antibodies (1:600; cat. no. A-1101; AlexaFluor V® 488 goat anti-mouse; Molecular Probes®) at room temperature for 1 h. The nuclei were stained with 4′,6-diamidino-2-phenylindole and images were captured with a laser scanning confocal microscope (DMI6000B TCS SP5; Leica Microsystems GmbH, Mannheim, Germany), using microscope imaging software (LAS AF6500; Leica Microsystems GmbH, Wetzlar, Germany).

Western blot analysis

The K562 and K562/G cells were collected and lysed with RIPA buffer containing a protease inhibitor cocktail. The lysates were centrifuged at 12,000 × g for 10 min, 4°C and the supernatant was collected. The total protein concentration in the supernatant was determined using a BCA protein assay. Equal quantities of protein (30 µg) were subjected to 10–12% SDS-polyacrylamide gel electrophoresis at a constant voltage of 80 V for 30 min and 120 V for another 1.5 h. The resolved proteins were electrophoretically transferred onto PVDF membranes (EMD Millipore, Billerica, MA, USA), and the membranes were blocked in 5% skimmed milk for 1 h. Subsequently, the membranes were incubated overnight at 4°C with primary monoclonal antibodies at 1:1,000 dilutions. The following day, the membranes were exposed to horseradish peroxidase-conjugated anti-mouse or anti-rabbit secondary antibodies (1:1,000 dilution; A21010, A21020-1; Abbkine, Wuhan, China) for 2 h at room temperature. To visualize the protein bands, the membranes were treated with an enhanced chemiluminescence kit solution (GE Healthcare Life Sciences; Chalfont, UK) and digitalized by scanning (Fusion Solo3 v 16.12; Fusion FX Vilber Lourmat, France). The primary antibodies against STAT5 (94205S), phosphorylated (p-)STAT5 (9359S), p53 (2524S), B-cell lymphoma-2 (Bcl-2) (2870S), mouse double minute 2 homolog (MDM2; 86934) and Bcl-2-associated X protein (Bax) (5023T) were obtained from Cell Signaling Technology, Inc., and anti-β-actin was purchased from Santa Cruz Biotechnology, Inc. (sc-47778; Dallas, TX, USA)

Statistical analysis

All data in the present study are presented as the mean ± standard deviation of three independent experiments unless stated otherwise. The results were analyzed using one-way analysis of variance and unpaired Student's t-tests to evaluate the significance of differences between groups. P<0.05 was considered to indicate a statistically significant difference. Data analyses were performed using Prism software version 5.0 (GraphPad Software, Inc., La Jolla, CA, USA).

Results

Elevated levels of STAT5 correlate with BCR-ABL1 mutation and imatinib sensitivity in vivo and in vitro

Several previous studies have reported that the activation of STAT5 contributes to imatinib resistance in BCR/ABL1 CML. In a previous study, Warsch et al revealed that imatinib-resistant patients had upregulated levels of STAT5 in leukemic cells (10). In the present study, it was found that patients with CML and imatinib resistance exhibited higher mRNA expression levels of STAT5A and STAT5B (P=0.0093 and P=0.0091) (Fig. 1A). Of note, the expression levels of STAT5 were significantly higher in patients with BCR-ABL1 mutations, compared with those without BCR-ABL1 mutations (sensitive, P=0.02 and P=0.009; resistant P=0.01 and P=0.009), and this effect was independent of the state of imatinib resistance (Fig. 1B). Additionally, the K562/G cells exhibited elevated mRNA levels of STAT5A and STAT5B, compared with the imatinib-sensitive K562 cells (P<0.001 and P<0.001) (Fig. 1C). Consistently, the results of the western blot analysis demonstrated that the protein expression levels of STAT5 and p-STAT5 were markedly increased in the K562/G cells, compared with those in K562 cells (Fig. 1D).

ROS accumulation contributes to enhanced levels of DNA DSBs in K562/G cells

Significantly higher levels of ROS were detected in the imatinib-resistant K562/G cells, compared with those in the imatinib-sensitive K562 cells (P<0.001). Following treatment with 5 mM NAC, a scavenger of ROS, the levels of ROS in the K562/G cells were significantly decreased (P=0.323) (Fig. 2A). Additionally, the present study investigated whether STAT5 contributed to the accumulation of ROS by using the STAT5 inhibitor, SH-4-54. As shown in Fig. 2B, exposure to 20 µM SH-4-54 markedly reduced the levels of ROS in the K562/G cells (P=0.872 vs. K562; P=0.01 vs. K562/G). It is well established that ROS can induce DNA damage, including DNA DSBs, and is generally recognized as an inducer of resistance mutation; γ-H2AX is frequently observed in regions of histone lesions resulting from ROS damage and may be used to quantify DSBs (20). The present study showed that higher levels of γ-H2AX were detected in the K562/G cells than in the K562 cells, which suggested that higher levels of ROS and secondary DNA damage were present in the K562/G cells. To confirm the link between STAT5-mediated ROS production and DSB generation, the K562/G cells were pretreated with NAC. As expected, NAC pretreatment decreased the level of γ-H2AX staining (Fig. 2C).

Involvement of the p53 apoptotic pathway in the reduced apoptotic rate and resistance of K562/G cells to imatinib

Imatinib, as a first generation TKI, is used as a principle treatment for patients with CML (4). A significant difference in the rate of ABL mutation was observed between the imatinib-resistant group and the non-resistant group (28.13 vs. 9.60%) (Table I). The imatinib-resistant K562/G CML cell line was used in the present study, and the resistance of the K562/G cells was first characterized and compared with that of normal K562 cells. The K562 and K562/G cells were treated with imatinib, and a CCK-8 assay was used to assess the cytotoxic effects of various concentrations of imatinib on the cell lines, for which the IC50 values were determined. The results showed that the ratio of IC50 values between the K562/G and K562 cells was 87:1 (14.78±0.43 vs. 0.17±0.07) (Fig. 3). Therefore, the K562/G cell line was resistant to imatinib and maintained a high level of drug resistance, which confirmed the efficacy of the imatinib-resistant cell model. The apoptotic rates of the K562 and K562/G cells in response to different concentrations of imatinib (0.1, 1, 5, 10 and 15 µM) were measured using flow cytometric analysis. The results showed that the rate of apoptosis in the K562/G cells was significantly lower, compared with that in the K562 cells at all concentrations of imatinib (P<0.05). This effect was evident in early and late apoptosis (Fig. 4A–C). Previous evidence indicates that ROS-induced apoptosis is characterized by the upregulation of tumor suppressor protein p53 (21), delocalization of cytochrome c, caspase activation, DNA fragmentation, and suppressed expression of histone deacetylasein CML cells (22). In the present study, using a human DNA damage signaling pathway array, the downregulated transcription levels of DDR genes were found in K562/G cells. Among these downregulated genes, tumor protein p53 and tumor protein p53-binding protein 1 were involved in the apoptotic pathway (Table II). Although the K562 and K562/G cells exhibited low protein expression levels of p53, the mRNA level of p53 was significantly lower in the K562/G cells, compared with that in the K562 cells (P<0.01) (Fig. 4D and E). Similarly, the expression levels of Bax were significantly decreased in the K562/G cells (P<0.01), whereas those of Bcl-2 and MDM2 were significantly increased (P<0.01) (Fig. 4D). The p53 mutation was not found in the CML patients (Table I).

Discussion

CML is commonly considered to be a typical model for understanding the molecular pathogenesis of malignancy. Although the treatment and prognosis of CML have markedly improved since the development of TKIs, including imatinib, which is now used as a standard first-line therapeutic agent, not all patients with CML respond well. Even following long-term imatinib treatment, stem cells in the majority of patients with CML continue to express BCR-ABL mRNA. Additionally, following long-term therapy, it has been reported that 60% of patients experience relapse at the molecular level, whereas 15% of patients developed drug resistance and subsequently discontinued imatinib following a sustained complete molecular response (23). The most frequently reported cause for TKI resistance is a gene mutation in the KD of BCR-ABL1, which typically occurs at a frequency of 40–90% (24). Other mechanisms may include the upregulation of BCR-ABL1, increased expression of drug transporter ABCB1, elevated levels of granulocyte-macrophage colony-stimulating factor, and inactivation of TP53 (25,26). In the present study, 63 blood samples from imatinib-resistant (n=32) and non-resistant (n=31) patients were analyzed by DNA sequencing, and 28% of the imatinib-resistant cases were found to harbor mutations within the KD of BCR-ABL1 (Table I).

It is generally accepted that STAT5 exerts key effects in various hematological malignancies, including anti-apoptotic effects and growth-stimulatory functions (27). To date, STAT5 has been confirmed to support BCR-ABL1-triggered CML via four key mechanisms: Cell cycle control, cell viability maintenance, ROS generation and TKI resistance (28). The importance of STAT5 in the pathogenesis of CML is further highlighted by findings that STAT5 is upregulated during disease progression and that high STAT5 levels can significantly decrease imatinib sensitivity. In the present study, significantly increased expression levels of STAT5 were consistently found in the blood samples of imatinib-resistant patients and in imatinib-resistant K562/G cells. This was in agreement with a previous study that identified significantly increased mRNA levels of STAT5A in patients presenting with secondary imatinib resistance without BCR-ABL1 mutations, compared with newly diagnosed imatinib responders (29). The present study investigated the potential connection between the transcription factor STAT5 and the occurrence of BCR-ABL1 mutations. The data showed that significantly higher levels of STAT5 were expressed in the BCR-ABL1 mutation group, compared with those in the non-BCR-ABL1 mutation group, regardless of TKI resistance state.

ROS are considered to be potent signaling mediators, and may interfere with gene regulatory pathways, including the mitogen-activated protein kinase and hypoxia-responsive element/hypoxia-inducible factor pathways, which are also known to be regulated by STAT5 transcription factors. In the present study, as expected, the STAT5 inhibitor SH-4-54 caused a marked reduction in the levels of ROS in K562/G cells. However, under environmental stress, ROS levels increase markedly, which can cause significant damage to cell structures, and ultimately mutation. In BCR-ABL1+ cells, high levels of STAT5 have been associated with elevated levels of endogenous ROS (30). In the present study, the levels of ROS in K562/G cells were elevated, compared with those in K562 cells. Previous studies have shown that certain chemotherapeutic drugs can induce cell death by increasing the generation of intracellular ROS (31,32). The production of ROS can lead to DNA damage (33). DNA DSBs are considered to be the most destructive form of DNA damage, and have a high probability of resulting in cumulative mutations. In the present study, it was observed that ROS increased the levels of γ-H2AX, which was alleviated by NAC.

The function of p53 as a tumor suppressor has been attributed to its ability to promote cell death or permanently inhibit cell proliferation. p53 is subject to a wide range of post-translational modifications, including phosphorylation, acetylation, methylation and ubiquitination. Regulation of the gene expression of p53 usually occurs mainly at the protein level (34). Somatic TP53 mutations have been identified in several types of cancer, at various frequencies depending on the cancer type. Overall, TP53 mutations are found in 5–10% of de novo cases of myelodysplastic syndrome MDS and acute myeloid leukemia cases, and have been associated with complex karyotypes and reduced survival rates (35,36). p53 mutations are considered to be high-risk factors for the development of leukemia, and indicators of a poor response to chemotherapy and poor prognosis (37). In the patients with CML enrolled in the present study, no p53 mutations were detected. However, whether the contributions of p53-associated TKI resistance occur mainly via mutations or epigenetic modifications remains to be elucidated. In a previous study, the deletion of p53 was associated with the progression of CML, and the wild-type p53 protein was present in the KBM5 chronic phase cell line, whereas the low expression or absence of p53 was observed in K562 cells (an advanced stage CML cell line) (38). In the present study, the results of the Human DNA Damage Signaling Pathway Array revealed downregulated transcription levels of DDR genes, including p53 and p53-binding protein 1, in the K562/G cells. The expression levels of p53, Bcl-2 and Bax, proteins of the p53 apoptotic pathway, were assessed using western blot analysis, and the downregulation of Bax was observed in K562/G cells. These experimental results are consistent with the findings of a previous study involving the deletion of p53 in K562 and K562/G cells (38).

In conclusion, the present study identified elevated levels of STAT5 in patients with imatinib-resistant CML and K562/G cells. In addition, the high levels of STAT5 and subsequent accumulation of ROS were associated with increased chronic oxidative damage to DNA, as evidenced by increased DSBs within the DNA of K562/G cells, which may be a cause of gene mutations associated with resistance. It was also found that aberrant regulation and expression of the p53 pathway was involved in imatinib resistance. The expression of p53 can be modulated by changes in transcriptional and translational events, and the activity and protein levels of p53 are negatively regulated by the E3 ubiquitin ligase MDM2 (39). The present study detected lower mRNA and protein expression levels of MDM2 in K562 cells, compared with those in the K562/G cells. Therefore, a time profile of the induced expression of p53 and its epigenetic modification (methylation status, post transcriptional regulation by microRNAs) during the establishment of resistance to imatinib require further examination in future investigations.

Acknowledgments

This study was supported by the Anhui Provincial Natural Science Foundation (grant no. 1708085MH223)

References

1 

Hochhaus A, Saglio G, Hughes TP, Larson RA, Kim DW, Issaragrisil S, le Coutre PD, Etienne G, Dorlhiac-Llacer PE, Clark RE, et al: Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 30:1044–1054. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Hochhaus A, Rosti G, Cross NC, Steegmann JL, le Coutre P, Ossenkoppele G, Petrov L, Masszi T, Hellmann A, Griskevicius L, et al: Frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: Results from the European ENEST1st study. Leukemia. 30:57–64. 2016. View Article : Google Scholar :

3 

Breccia M, Stagno F, Luciano L, Abruzzese E, Annunziata M, D'Adda M, Maggi A, Sgherza N, Russo-Rossi A, Pregno P, et al: Dasatinib first-line: Multicentric Italian experience outside clinical trials. Leuk Res. 40:24–29. 2016. View Article : Google Scholar

4 

Hughes TP and Ross DM: Moving treatment-free remission into mainstream clinical practice in CML. Blood. 128:17–23. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Vaidya S, Vundinti BR, Shanmukhaiah C, Chakrabarti P and Ghosh K: Evolution of BCR/ABL gene mutation in CML is time dependent and dependent on the pressure exerted by tyrosine kinase inhibitor. PLoS One. 10:e01148282015. View Article : Google Scholar : PubMed/NCBI

6 

Parker WT, Yeung DT, Yeoman AL, Altamura HK, Jamison BA, Field CR, Hodgson JG, Lustgarten S, Rivera VM, Hughes TP, et al: The impact of multiple low-level BCR-ABL1 mutations on response to ponatinib. Blood. 127:1870–1880. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Soverini S, Hochhaus A, Nicolini FE, Gruber F, Lange T, Saglio G, Pane F, Müller MC, Ernst T, Rosti G, et al: BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: Recommendations from an expert panel on behalf of European LeukemiaNet. Blood. 118:1208–1215. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Turhan AG: STAT5 as a CML target: STATinib therapies? Blood. 117:3252–3253. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Warsch W, Grundschober E and Sexl V: Adding a new facet to STAT5 in CML: Multitasking for leukemic cells. Cell Cycle. 12:1813–1814. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Warsch W, Kollmann K, Eckelhart E, Fajmann S, Cerny-Reiterer S, Hölbl A, Gleixner KV, Dworzak M, Mayerhofer M, Hoermann G, et al: High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia. Blood. 117:3409–3420. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Walz C, Ahmed W, Lazarides K, Betancur M, Patel N, Hennighausen L, Zaleskas VM and Van Etten RA: Essential role for Stat5a/b in myeloproliferative neoplasms induced by BCR-ABL1 and JAK2(V617F) in mice. Blood. 119:3550–3560. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Carter BZ, Mak PY, Mak DH, Ruvolo VR, Schober W, McQueen T, Cortes J, Kantarjian HM, Champlin RE, Konopleva M, et al: Synergistic effects of p53 activation via MDM2 inhibition in combination with inhibition of Bcl-2 or Bcr-Abl in CD34+ proliferating and quiescent chronic myeloid leukemia blast crisis cells. Oncotarget. 6:30487–30499. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Hughes T, Deininger M, Hochhaus A, Branford S, Radich J, Kaeda J, Baccarani M, Cortes J, Cross NC, Druker BJ, et al: Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: Review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 108:28–37. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Baccarani M, Deininger MW, Rosti G, Hochhaus A, Soverini S, Apperley JF, Cervantes F, Clark RE, Cortes JE, Guilhot F, et al: European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 122:872–884. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Dong HJ, Zhou LT, Zhu DX, Wang DM, Fang C, Zhu HY, Zhuang Y, Miao KR, Xu W and Li JY: The prognostic significance of TP53 mutations in Chinese patients with chronic lymphocytic leukemia is independent of del(17p13). Ann Hematol. 90:709–717. 2011. View Article : Google Scholar

16 

Lan X, Zhao C, Chen X, Zhang P, Zang D, Wu J, Chen J, Long H, Yang L, Huang H, et al: Nickel pyrithione induces apoptosis in chronic myeloid leukemia cells resistant to imatinib via both Bcr/Abl-dependent and Bcr/Abl-independent mechanisms. J Hematol Oncol. 9:1292016. View Article : Google Scholar : PubMed/NCBI

17 

Shi X, Chen X, Li X, Lan X, Zhao C, Liu S, Huang H, Liu N, Liao S, Song W, et al: Gambogic acid induces apoptosis in imatinib-resistant chronic myeloid leukemia cells via inducing proteasome inhibition and caspase-dependent Bcr-Abl downregulation. Clin Cancer Res. 20:151–163. 2014. View Article : Google Scholar :

18 

Wu L, Chen X, Huang L, Tian J, Ke F, Xu J, Chen Y and Zheng M: A Novobiocin derivative, XN4, inhibits the proliferation of chronic myeloid leukemia cells by inducing oxidative DNA damage. PLoS One. 10:e01233142015. View Article : Google Scholar : PubMed/NCBI

19 

Ismail IH, Wadhra TI and Hammarsten O: An optimized method for detecting gamma-H2AX in blood cells reveals a significant interindividual variation in the gamma-H2AX response among humans. Nucleic Acids Res. 35:e362007. View Article : Google Scholar : PubMed/NCBI

20 

Ren G, Luo W, Sun W, Niu Y, Ma DL, Leung CH, Wang Y, Lu JJ and Chen X: Psoralidin induced reactive oxygen species (ROS)-dependent DNA damage and protective autophagy mediated by NOX4 in breast cancer cells. Phytomedicine. 23:939–947. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Maillet A and Pervaiz S: Redox regulation of p53, redox effectors regulated by p53: a subtle balance. Antioxid Redox Signal. 16:1285–1294. 2012. View Article : Google Scholar

22 

Banerjee K, Ganguly A, Chakraborty P, Sarkar A, Singh S, Chatterjee M, Bhattacharya S and Choudhuri SK: ROS and RNS induced apoptosis through p53 and iNOS mediated pathway by a dibasic hydroxamic acid molecule in leukemia cells. Eur J Pharm Sci. 52:146–164. 2014. View Article : Google Scholar

23 

Chomel JC, Bonnet ML, Sorel N, Bertrand A, Meunier MC, Fichelson S, Melkus M, Bennaceur-Griscelli A, Guilhot F and Turhan AG: Leukemic stem cell persistence in chronic myeloid leukemia patients with sustained undetectable molecular residual disease. Blood. 118:3657–3660. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Branford S, Rudzki Z, Walsh S, Parkinson I, Grigg A, Szer J, Taylor K, Herrmann R, Seymour JF, Arthur C, et al: Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood. 102:276–283. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Raja T: Optimizing second-line therapy for chronic myeloid leukemia. Indian J Cancer. 49:46–56. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Wang Y, Cai D, Brendel C, Barett C, Erben P, Manley PW, Hochhaus A, Neubauer A and Burchert A: Adaptive secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF) mediates imatinib and nilotinib resistance in BCR/ABL+ progenitors via JAK-2/STAT-5 pathway activation. Blood. 109:2147–2155. 2007. View Article : Google Scholar

27 

Hoelbl A, Schuster C, Kovacic B, Zhu B, Wickre M, Hoelzl MA, Fajmann S, Grebien F, Warsch W, Stengl G, et al: Stat5 is indispensable for the maintenance of bcr/abl-positive leukaemia. EMBO Mol Med. 2:98–110. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Hoelbl A, Kovacic B, Kerenyi MA, Simma O, Warsch W, Cui Y, Beug H, Hennighausen L, Moriggl R and Sexl V: Clarifying the role of Stat5 in lymphoid development and Abelson-induced transformation. Blood. 107:4898–4906. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Zhang WW, Cortes JE, Yao H, Zhang L, Reddy NG, Jabbour E, Kantarjian HM and Jones D: Predictors of primary imatinib resistance in chronic myelogenous leukemia are distinct from those in secondary imatinib resistance. J Clin Oncol. 27:3642–3649. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Mallette FA, Moiseeva O, Calabrese V, Mao B, Gaumont-Leclerc MF and Ferbeyre G: Transcriptome analysis and tumor suppressor requirements of STAT5-induced senescence. Ann NY Acad Sci. 1197:142–151. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Gorrini C, Harris IS and Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 12:931–947. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Reczek CR and Chandel NS: ROS-dependent signal transduction. Curr Opin Cell Biol. 33:8–13. 2015. View Article : Google Scholar :

33 

Brem R, Li F, Montaner B, Reelfs O and Karran P: DNA breakage and cell cycle checkpoint abrogation induced by a therapeutic thiopurine and UVA radiation. Oncogene. 29:3953–3963. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Gu B and Zhu WG: Surf the post-translational modification network of p53 regulation. Int J Biol Sci. 8:672–684. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, Kantarjian H, Raza A, Levine RL, Neuberg D, et al: Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 364:2496–2506. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Kulasekararaj AG, Smith AE, Mian SA, Mohamedali AM, Krishnamurthy P, Lea NC, Gäken J, Pennaneach C, Ireland R, Czepulkowski B, et al: TP53 mutations in myelodysplastic syndrome are strongly correlated with aberrations of chromosome 5, and correlate with adverse prognosis. Br J Haematol. 160:660–672. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Perrotti D, Jamieson C, Goldman J and Skorski T: Chronic myeloid leukemia: Mechanisms of blastic transformation. J Clin Invest. 120:2254–2264. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Woo SM, Choi YK, Kim AJ, Cho SG and Ko SG: p53 causes butein mediated apoptosis of chronic myeloid leukemia cells. Mol Med Rep. 13:1091–1096. 2016. View Article : Google Scholar

39 

Haupt Y, Maya R, Kazaz A and Oren M: Mdm2 promotes the rapid degradation of p53. Nature. 387:296–299. 1997. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 41 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng Y, Hao Y, Zhang A, Hu C, Jiang X, Wu Q and Xu X: Persistent STAT5-mediated ROS production and involvement of aberrant p53 apoptotic signaling in the resistance of chronic myeloid leukemia to imatinib. Int J Mol Med 41: 455-463, 2018
APA
Cheng, Y., Hao, Y., Zhang, A., Hu, C., Jiang, X., Wu, Q., & Xu, X. (2018). Persistent STAT5-mediated ROS production and involvement of aberrant p53 apoptotic signaling in the resistance of chronic myeloid leukemia to imatinib. International Journal of Molecular Medicine, 41, 455-463. https://doi.org/10.3892/ijmm.2017.3205
MLA
Cheng, Y., Hao, Y., Zhang, A., Hu, C., Jiang, X., Wu, Q., Xu, X."Persistent STAT5-mediated ROS production and involvement of aberrant p53 apoptotic signaling in the resistance of chronic myeloid leukemia to imatinib". International Journal of Molecular Medicine 41.1 (2018): 455-463.
Chicago
Cheng, Y., Hao, Y., Zhang, A., Hu, C., Jiang, X., Wu, Q., Xu, X."Persistent STAT5-mediated ROS production and involvement of aberrant p53 apoptotic signaling in the resistance of chronic myeloid leukemia to imatinib". International Journal of Molecular Medicine 41, no. 1 (2018): 455-463. https://doi.org/10.3892/ijmm.2017.3205