Skip to main content
Erschienen in: BMC Cancer 1/2012

Open Access 01.12.2012 | Research article

Wnt/β-catenin pathway regulates ABCB1 transcription in chronic myeloid leukemia

verfasst von: Stephany Corrêa, Renata Binato, Bárbara Du Rocher, Morgana TL Castelo-Branco, Luciana Pizzatti, Eliana Abdelhay

Erschienen in: BMC Cancer | Ausgabe 1/2012

Abstract

Background

The advanced phases of chronic myeloid leukemia (CML) are known to be more resistant to therapy. This resistance has been associated with the overexpression of ABCB1, which gives rise to the multidrug resistance (MDR) phenomenon. MDR is characterized by resistance to nonrelated drugs, and P-glycoprotein (encoded by ABCB1) has been implicated as the major cause of its emergence. Wnt signaling has been demonstrated to be important in several aspects of CML. Recently, Wnt signaling was linked to ABCB1 regulation through its canonical pathway, which is mediated by β-catenin, in other types of cancer. In this study, we investigated the involvement of the Wnt/β-catenin pathway in the regulation of ABCB1 transcription in CML, as the basal promoter of ABCB1 has several β-catenin binding sites. β-catenin is the mediator of canonical Wnt signaling, which is important for CML progression.

Methods

In this work we used the K562 cell line and its derived MDR-resistant cell line Lucena (K562/VCR) as CML study models. Real time PCR (RT-qPCR), electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP), flow cytometry (FACS), western blot, immunofluorescence, RNA knockdown (siRNA) and Luciferase reporter approaches were used.

Results

β-catenin was present in the protein complex on the basal promoter of ABCB1 in both cell lines in vitro, but its binding was more pronounced in the resistant cell line in vivo. Lucena cells also exhibited higher β-catenin levels compared to its parental cell line. Wnt1 and β-catenin depletion and overexpression of nuclear β-catenin, together with TCF binding sites activation demonstrated that ABCB1 is positively regulated by the canonical pathway of Wnt signaling.

Conclusions

These results suggest, for the first time, that the Wnt/β-catenin pathway regulates ABCB1 in CML.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-12-303) contains supplementary material, which is available to authorized users.
Luciana Pizzatti and Eliana Abdelhay contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SC performed the experiments, statistical analysis and drafted the manuscript. RB participated and assisted the electrophoretic mobility shift assays and Luciferase experiments. BR participated in flow cytometry experiments. MTLCB performed immunofluorescence assays. LP and EA made substantial contributions to the study conception and design and critically revised the manuscript for intellectual content. All authors read and approved the final manuscript.

Background

Chronic myeloid leukemia (CML) is a myeloproliferative disease characterized by the BCR-ABL constitutive tyrosine kinase (TK) oncoprotein, the result of the balanced reciprocal translocation of chromosomes 9 and 22 (t(9;22)(q34;q11)) [1]. BCR-ABL signaling is responsible for the pathogenesis of CML and is the primary molecular target for disease therapy with imatinib mesylate (Glivec, Gleevec, IM), a TK inhibitor. CML progresses in three phases: an initial phase known as the chronic phase (CP), the accelerated phase (AP) and the blastic crisis (BC) [2]. CML progression to BC has been associated, among others, with the canonical pathway of Wnt signaling. Activation of this pathway leads to nuclear accumulation of β-catenin, which activates the TCF/LEF1 family of transcriptional factors. The canonical pathway plays an important role in CML progression by activating several targets, such as c-MYC, ROK13A, cadherin, MDI1, prickle 1, and FZD2 [3]. Recently, this pathway was demonstrated to be crucial in disease maintenance through the sustenance of CML stem cells [46]. Hu and colleagues indicated that β-catenin is essential for the survival and self-renewal of CML stem cells even in mice subjected to kinase inhibition therapy [7].
Mechanisms surrounding the response to IM therapy in CML have been mostly associated with BCR-ABL oncoprotein mutations and BCR-ABL amplification. Nevertheless, some patients do not present either mechanism or respond to therapy, suggesting other mechanisms, the so-called BCR-ABL-independent mechanisms. Among them is the multidrug resistance (MDR) phenotype that is dependent on the expression of proteins that function as extrusive pumps [810]. In leukemia, the product of ABCB1, P-glycoprotein (Pgp), is most commonly implicated in the development of drug resistance. It is known that ABCB1 can be regulated by several pathways in different conditions and that there is redundancy in this regulation [11]. However, despite the complex pattern of the ABCB1 promoter region, the existence of seven TCF/LEF1 consensus binding sites on the basal promoter of this gene [12] indicates the possibility of ABCB1 regulation by the canonical Wnt pathway. Indeed, Yamada and colleagues [13], Flahaut and colleagues [14] and Bourguignon and colleagues [15] revealed the involvement of the Wnt/β-catenin pathway in ABCB1 regulation in early colorectal cancer, neuroblastoma and breast cancer, respectively. Regarding CML, studies with patients have shown that polymorphisms of the ABCB1 gene can alter the response to therapy [1621]. A few works have confirmed that ABCB1 can be overexpressed in the AP of disease [2226], and IM-resistant cell lines also overexpress ABCB1[2729]. Our previously work have demonstrated that MDR cell line Lucena, which over-expresses ABCB1 (800-fold increase) and Pgp (45-fold increase) is cross-resistant to IM and IM-resistant patients present ABCB1 over-expressed, despite of disease phase [30]. Therefore, the aim of this work was to investigate the involvement of the WNT/β-catenin pathway in the regulation of ABCB1 transcription in CML. Our results provide unprecedented information regarding ABCB1 regulation in CML.

Methods

Culture conditions

Lucena (K562 multidrug-resistant cell line induced by vincristine (VCR)) cells overexpressing ABCB1 were kindly provided by Dra. Vivian Rumjanek (Departamento de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Brazil) [31]. The human myelogenous leukemia cell line (K562) and its vincristine-resistant derivative, the Lucena cell line, were grown in RPMI 1640 medium (Invitrogen) supplemented with 10% FBS (Invitrogen), 50 units/mL penicillin G (Invitrogen), 50 μg/L streptomycin (Invitrogen) and 2 mM l-glutamine (Invitrogen) at 37 °C in a humidified atmosphere containing 5% CO2. Lucena medium was supplemented with 60 nM VCR (Sigma).

Electrophoretic mobility shift assays (EMSAs)

Syntheses of double-stranded oligonucleotides for the seven TCF sites in the ABCB1 sequence from the upstream promoter followed the protocols of Labialle and colleagues see reference [12]. They were named S1 to S7, with S7 being the shortest distance from the coding sequence. TCF sites are distributed throughout the ABCB1 promoter, with S1 and S2 being distal sites.
S1: 5′-CAACTCGTCAAAGGAATTAT-3′
S2: 5′-GGTGTTGATCAAAGGTACAA-3′
S3: 5′-GCAGAACTCAAAGAAACAGA-3′
S4: 5′-ATGTCAAAACAAAGGAGATT-3′
S5: 5′-AAACAAAGTTTGCTCCTCTT-3′
S6: 5′-GTAGGAAATACAAAGAATACT-3′
S7: 5′-GCCTAAGAACAAAGAGAGAG-3′
Dephosphorylated oligonucleotides (25 pmol) were end-labeled with [γ-32P] ATP and T4 polynucleotide kinase. For binding reactions, 7 μg of nuclear protein extracts, prepared as previously described [32], was incubated with 80.000 cpm of labeled probe in 2 μL of 1× binding buffer (50 mM HEPES - pH 7.4, 300 mM KCl, 5 mM EDTA, 5 mM DTT, 11.5% Ficoll) and 1 μg of poly(dI-dC)(dI-dC) (GE) in a total volume of 20 μL for 40 min at room temperature (25 °C). Reactions were resolved by 4.5% polyacrylamide gel electrophoresis in 0.5× TBE for 90 min at 4 °C. In all EMSA experiments, the concentration chosen for competition experiments was a 200-fold molar excess. For competition reactions, a 200-fold molar excess of nonlabeled competitor DNA was added 20 min before the addition of the probe. The Opt oligonucleotide, described by Pizzatti and colleagues, was also used as a competitor because it only possesses the TCF consensus binding site in its sequence [33]. Opt 5′-GGTAAGATCAAAGGG-3′
For supershift analysis of the ABCB1 promoter, protein extracts were incubated for 2 h with 1 μg of the anti-β-catenin (Sigma) and anti-Smad8 (Santa Cruz Technologies) antibodies at 4 °C before the addition of the probe. Anti-Smad8 was used as a negative control.

Chromatin immunoprecipitation (ChIP) assays on native chromatin

Chromatin from K562 and Lucena cells was fractionated by incubation of purified nuclei with micrococcal nuclease and its immunoprecipitation with anti-β-catenin antibody was performed as described previously [34]. DNA extractions from bound fractions were performed following the Abcam protocol. The immunoprecipitated DNA was amplified for sequences containing binding sites by using the following ABCB1 promoter sequence primers: ABCB1p (F) 5′-CAACTCGTCAAAGGAATTAT-3′ and ABCB1p (R) 5′-TTGTACCTTTGATCAACACC-3′.
Quantification was evaluated by RT-qPCR analysis. Immunoprecipitation of Protein A (Santa Cruz Technologies) was used for non-specific binding, and Smad8 (Santa Cruz Technologies) was used as a positive control.

Real-time quantitative PCR (RT-qPCR)

Analysis of ABCB1 WNT1 β-catenin and β-ACTIN mRNA levels was performed by RT-qPCR. Two micrograms of Trizol (Invitrogen) extracted RNA from cell lines was treat with DNAse Amplification Grade I (Invitrogen) and reverse-transcribed with Superscript II Reverse Transcriptase® (Invitrogen). cDNAs dilutions (1:100) were mixed with SYBR Green PCR Master Mix® (Applied Biosystems) and the following primers: ABCB1: (F) 5′-CCC ATC ATT GCA ATA GCA GG-3′ and (R) 5′-GTT CAA ACT TCT GCT CCT GA-3′; WNT1: (F) 5′-TGG TTT GCA AAG ACC ACC TCC A-3′, and (R) 5′-TGA TTC CAG GAG GCA AAC GCA T-3′; β-CATENIN: (F) 5´-AAG ACA TCA CTG AGC CTG CCAT-3´ and (R) 5´-CGA TTT GCG GGA CAA AGG GCA A-3´; β-ACTIN: (F) 5′-ACC TGA GAA CTC CAC TAC CCT-3′ and (R) 5′-GGT CCC ACC CAT GTT CCA G-3′. RT-qPCR was performed in a Rotor Gene 6000 thermocycler (Corbett) with 50 cycles of 20 s at 95 °C, 30 s at 60 °C and 30 s at 72 °C. For each sample, the expression of target genes was normalized to β-actin mRNA levels. Changes in the mRNA levels of genes were evaluated [35].

WNT/β-catenin activation by LiCl treatment

Cell cultures were exposed to 10 mM LiCl [36]. Treatment was performed in 12-well culture plates for 24 and 48 h at a cellular density of 2.0 × 105 cells/mL. The toxicity of the assay was evaluated by FACS analysis, and ABCB1 mRNA levels were analyzed by RT-qPCR.

Flow cytometry (FACS) analysis

Viability was evaluated via the analysis of propidium iodide (PI) staining (Sigma-Aldrich). Briefly, K562 and Lucena cells (approximately 3.0 x 105 cells) treated with 10 mM LiCl (24 h and 48 h) were harvested and washed in 500 μL of PBS. PI (1.5 μg/mL) was added to the incubated tubes prior to FACS analysis. PI(−) cells were considered viable. All procedures were performed according to the manufacturer’s protocol. For β-catenin expression detection, cells were harvested (approximately 2.5 x 105 cells) and fixed with PBS/1% Formol. As follow, they were permeabilized with 0.5% Tween 20 to allow intracellular staining and labeled with anti-β-catenin polyclonal antibody (Sigma). Results are expressed as mean relative fluorescence intensity (MRFI), which was calculated by subtracting the mean fluorescence intensity (MFI) for specific antibody by the MFI of the respective secondary antibody (which served as negative isotype control). Ten thousand events were analyzed for each sample in a FACSCalibur Flow Cytometer (Becton Dickinson). The data were analyzed using CellQuest v.3.1 software (Becton Dickinson). All experiments were performed in triplicate.

RNAi knockdown (siRNA) and transfection

All RNA oligonucleotides described in this study were synthesized and purified using high-performance liquid chromatography at Integrated DNA Technologies (Coralville), and the duplex sequences are available upon request. siRNA and transfections were performed following the manufacturer’s protocols of the TriFECTa Dicer-Substrate RNAi kit (Integrated DNA Technologies) and the Trifectin reagent (Integrated DNA Technologies). K562 and Lucena cells (5.5 × 104 cells per well) were split in 24-well plates at 60% confluence in RPMI medium 1 day prior to transfection. The TriFECTa kit contains control sequences for RNAi experiments, including a fluorescently labeled transfection control duplex and a scrambled universal negative control RNA duplex that is absent in human, mouse and rat genomes. Fluorescence microscopy was used to monitor the transfection efficiency according to the manufacturer’s recommendations. Only experiments in which transfection efficiencies were ≥ 80% were evaluated. mRNA levels were measured 48 h after transfection. Duplexes were evaluated at 10 nM. All transfections were minimally performed in duplicate, and the data were averaged. Wnt1 and β-Catenin depletion and RT-qPCR analyses were performed as described above.

Western blot analysis

Cell lysates from K562 and Lucena cells (control - untreated - and treated with LiCl 10 mM) were run on 15% sodium dodecyl sulfate-polyacrylamide gels (SDS-PAGE), transferred to nitrocellulose membranes (Bioead) and incubated with β-catenin (Santa Cruz Technologies), P-GSK3α/β (Cell signaling) and α-tubulin (Sigma) antibodies. Antibody binding was detected using enhanced chemiluminescence ECL Plus Western Blotting detection Reagents (GE).

Immunofluorescence staining and confocal laser microscopy

Cytospin preparations of K562 and Lucena cells were fixed with methanol, permeabilized with 0.2% triton X-100 PBS, and incubated for 1 h at room temperature with 1% bovine serum albumin (BSA) and 2.0% FBS blocking buffer under shaking. Slides were rinsed once with PBS and 0.05% Tween 20 and then incubated with appropriately diluted primary antibodies in PBS. Cells were incubated with anti-β-catenin polyclonal antibody (Sigma) overnight at 4 °C. After incubation, the slides were rinsed three times and incubated with Alexa® 546 conjugated anti-rabbit IgG (Molecular Probes) for 1 h at room temperature. Sections from each sample were incubated with secondary antibody and served as negative isotype control. The slides were rinsed three times, air-dried and mounted in an antifading medium containing 4',6-diamidino-2-phenylindole (DAPI) (Vector Labs). Expression and localization of the proteins were observed with a Leica TCS-SP5 AOBS confocal laser scanning microscope (Leica), for capturing representative images of each sample.

Reporter vectors design

The ABCB1 reporter constructs were synthesized by Gene Art (Germany) and cloned into the firefly pGL3-Basic vector (Promega) upstream of the Luciferase reporter gene. The constructs named pGL3α, containing just the basal promoter (−1019/+1); pGL3β, containing the basal promoter and one TCF binding site (−1067/+1) and pGL3γ, containing the basal promoter and three TCF binding sites (−3187/+1) were inserted into KpnI and BgLII restriction sites of pGL3-basic.

Transient transfection and luciferase reporter assay

For the transient assays, 1.0 x 105 cells from both cell lines (with or without LiCl 10 mM treatment) were co-transfected using Lipofectamine LTX 2000 (Invitrogen) with 1 μg of each Luciferase construct and 100 ng of pRL-SV40 vector (Promega), according to the manufacturer’s instructions. Firefly and Renilla Luciferase activities were measured in cell lysates 48 hours after transfection using the DualGlo Luciferase Assay System (Promega) on a Veritas TM Microplate Luminometer (Turner Biosystems), following the manufacturer’s protocol. All experiments were performed in triplicate. Ratios of Renilla luciferase readings to firefly luciferase readings were taken for each experiment and triplicates were averaged. The average values of the tested constructs were normalized to the activity of the empty pGL3-basic vector, which was arbitrarily set at value 1.

Statistical analysis

Comparison between K562 and Lucena results from different assays was performed by an unpaired t-test. P- Values less than 0.05 were considered as statistically significant (*p < 0.05,**p < 0.01, and ***p < 0.001). Statistical analyses and graphical representations were performed using GraphPad Prism™ software (GraphPad).

Results

β-catenin binds to the ABCB1promoter at the TCF-binding site

Protein binding to the seven oligonucleotides containing TCF consensus binding sites (S1, S2, S3, S4, S5, S6 and S7) was determined by EMSA (see additional file 1 for all EMSAs for TCF binding sites). We also performed competition assays to verify whether protein complex binding was specific for the TCF consensus binding site. Competition analyses using a 200-fold excess of unlabeled S4, S5 and Opt oligonucleotides demonstrated that the specific binding was reduced (Figure1). In addition, we investigated the presence of β-catenin in the protein complexes formed at the TCF consensus binding site in both S4 and S5 oligonucleotides as these TCF binding sites showed the clearer and stronger signal in EMSA analysis. Supershift assays were performed using human β-catenin antibody, and these assays demonstrated that β-catenin was present in the protein complexes (Figure1). These results suggest that the TCF consensus binding site is important for the formation of a protein complex. The appearance of a shifted band in the supershift assay with both the S4 and S5 oligonucleotides ensures the presence of β-catenin among protein complexes binding at the ABCB1 promoter.
To confirm the EMSA results, we performed ChIP assays. The ChIP assay allows in vivo analysis of nuclear protein-DNA interactions. Chromatin fractions bound to the β-catenin antibody in K562 and Lucena cells were quantified by RT-qPCR using primers to amplify the promoter region that contains TCF binding sites. A 2-fold increase in β-catenin binding was verified in Lucena cells compared to that in K562 cells after normalization with unspecific binding of protein A (Figure2A). Qualitative analysis of ABCB1 promoter amplification is shown in Figure2B.
In order to investigate if the more pronounced bind observed in ChIP assay was due to different β-catenin expression between cell lines, we evaluated its expression by RT-qPCR, western blot, flow cytometry and immunofluorescence assays (Figure3 and Figure4). The results show a higher expression of β-catenin in Lucena cell line.

The Wnt/β-catenin signaling pathway regulates ABCB1expression

To verify whether the binding of β-catenin to the ABCB1 promoter could lead to ABCB1 transcriptional activation, we activated the canonical WNT pathway in K562 and Lucena cells by LiCl 10 mM treatment, as described by Stambolic and colleagues see reference [36]. LiCl inhibits GSK3-β, resulting in β-catenin stabilization and its consequent nuclear translocation. This treatment is currently used for this purpose in the literature [37]. The nuclear translocation of β-catenin was more pronounced in K562 cell line than in Lucena cell line (Figure4). Moreover after LiCl treatment we demonstrated that phosphorylated GSK3-β was abolished in both cell lines (Figure5) indicating that degradation of β-catenin was prevented, enhancing its nuclear translocation (Figure5).
We examined cell viability by FACS analysis prior to RNA extraction (data not shown). As LiCl treatment did not alter cell viability, we evaluated ABCB1 mRNA levels in K562 and Lucena cells treated with LiCl 10 mM by RT-qPCR analysis at 24 h. Untreated cells were used as a control. The increase in ABCB1 mRNA levels was more significant in K562 cells than in Lucena cells (Figure6).
To strengthen our observations, we performed functional analyses of WNT1 and β-catenin depletion in K562 and Lucena cell lines using RT-qPCR. Using a siRNA approach after 48 h of transfection, a reduction in WNT1 expression of more than 85% and β-catenin of more than70% were achieved in both cell lines when compared to scrambled control sequence-treated cells (Figure7A and 8A). Thus, we used these samples to evaluate ABCB1 mRNA levels to verify how the Wnt pathway could be involved in ABCB1 regulation. WNT1 depletion in Lucena cells resulted in an 80% reduction in ABCB1 expression (Figure7B). Despite the 85% reduction of WNT1 expression in siRNA-treated K562 cells, we did not find evidence of significant reductions in ABCB1 mRNA levels compared to the levels in scramble control-treated cells, suggesting that probably other Wnt ligands could play a role in this activation (Figure7B).
However β-catenin reduction in both cell lines confirmed that indeed the canonical WNT pathway regulates ABCB1 expression as shown by 60% and 71% reduction of ABCB1 mRNA levels in K562 and Lucena cells respectively (Figure8B).

ABCB1promoter TCF binding sites role in transcriptional activity

To further evaluate the relative contribution of TCF transcription factor to the regulation of ABCB1 promoter activity, we performed transient transfection assays using K562 and Lucena cells with constructions containing TCF binding sites (Figure9A). These constructions were transfected with or without LiCl 10 mM and Luciferase activity was measured using Luciferase assay approach.
These results showed that Luciferase activity increases in the presence of TCF binding site in both K562 and Lucena cell lines (Figure9B, 9C). Even with only one TCF binding site-pGL3β construct transfection, we observed a higher Luciferase activity compared with basal promoter without TCF binding sites. The Luciferase activity increases with constructions with more than one TCF binding sites (Figure9B, 9C).
In K562 and Lucena cells treated with LiCl we observed a reduction in the Luciferase activity when compared with the untreated cells. As LiCl treatment results in the translocation of β-catenin to the nucleus this reduction reflects the lack of cytoplasmatic β-catenin necessary to activate TCF binding sites in the constructs that are in the cytoplasm.

Discussion

Resistance to chemotherapy is a recurrent issue in all cancer types. Because CML has the propensity to evolve from the CP to the AP and BC, with different responses to targeted therapy such as TK inhibitors, the molecular understanding of the mechanism of resistance in this neoplasia is advancing rapidly.
IM was the first molecularly targeted therapy rationally designed to specifically inhibit BCR-ABL TK activity [38]. However, despite the effectiveness and good tolerability of IM, drug resistance does emerge. Although a hematological response is observed in over 95% of CP patients, primary resistant can occur [39]. Otherwise, AP patients initially respond to IM but inevitably relapse with treatment-refractory disease because they acquire other mutations in addition to BCR-ABL amplification or kinase domain mutations [4042].
Since the demonstration that IM could be extruded from CML cells through Pgp action [43, 44], ABCB1 has become an interesting subject in IM resistance studies. Our previous results indicated that ABCB1 is overexpressed in CML patients with intrinsic and acquired resistance to IM therapy compared to its expression in IM-responsive patients and healthy bone marrow donors see reference [30]. This finding contrasts the idea that only individuals in the BC stage can exhibit ABCB1 overexpression, as suggested in the literature. Even though we have analyzed a small cohort of patients, our results corroborate those of Vasconcelos and colleagues see reference [24], ratifying the importance of ABCB1/Pgp in CML.
Interestingly, in CML, Jamieson and colleagues demonstrated that the granulocyte-macrophage progenitor pools from patients in BC and IM-resistant patients exhibited elevated levels of nuclear β-catenin compared with those in granulocyte-macrophage progenitors from healthy donors. Moreover, these progenitors acquired self-renewal ability [45]. These data indicated the important role of the Wnt/β-catenin pathway in the self-renewal of CML progenitors and the acquisition of resistance. Wnt signaling involvement in TK inhibitor resistance was also demonstrated through its noncanonical pathway by Gregory and colleagues [46]. Their results indicated that Wnt/Ca2+/NFAT signaling maintains the survival of Ph+ leukemic cells under BCR-ABL inhibition. Altogether, we can speculate that the deregulation of Wnt signaling leads to key modifications in the biology of cells, allowing them to become intrinsically more resistant to drug therapy. However, a link between TK inhibitor resistance, Wnt signaling and drug efflux mechanisms such as MDR has never been considered. Despite some recent findings demonstrating that TCF consensus sites for β-catenin were functional in the ABCB1 promoter in other types of cancer, in CML, this regulation has not yet been investigated.
In this work, using MDR (overexpressing ABCB1) and non-MDR cell lines (Lucena and K562, respectively) as models of CML, we demonstrated through EMSA and ChIP analyses that β-catenin binds to the TCF/LEF consensus binding site in the ABCB1 promoter. RT-qPCR analyses indicated that this binding occurred at 2-fold higher levels in Lucena cells than in K562 cells. As it has been demonstrated that the BCR-ABL protein can establish β-catenin expression in CML via TK-mediated phosphorylation [47], this finding suggests that in drug resistance in CML, the canonical Wnt pathway could be more strongly activated to positively regulate ABCB1 transcription, as previously evidenced in other types of cancer. Interestingly we could demonstrate by RT-qPCR, western blot and FACS, β-catenin higher expression in the MDR cell line. Furthermore, it is known that BCR is a negative regulator of the Wnt/β-catenin pathway. The fusion gene BCR-ABL formed in CML decreases BCR transcription and BCR translation, and thus, there is no more BCR available to complex with β-catenin, leading to the translocation of β-catenin to the nucleus and Wnt/β-catenin pathway activation [48].
To verify whether Wnt/β-catenin could regulate ABCB1 mRNA levels, we modulated the canonical pathway in CML cell lines. We demonstrated that activation of β-catenin signaling by LiCl treatment resulted in increased ABCB1 mRNA levels in both cell lines, with higher levels observed in K562 cells. As discussed previously, K562 cells exhibited less β-catenin binding to the ABCB1 promoter than Lucena cells. ABCB1 mRNA levels were not altered significantly in Lucena cells, suggesting that these cells exhibit saturation of the Wnt pathway. By silencing the pathway using a siRNA approach with WNT 1 and β-catenin knockdown, we verified the opposite—a significant decrease in ABCB1 mRNA levels in Lucena cells—indicating that when Wnt/β-catenin signaling is downregulated, ABCB1 transcription is also downregulated.
ABCB1 gene promoter presents seven TCF binding sites that have been until now poorly investigated in the regulation of gene transcriptional activity in cancer cells. However the more proximal TCF binding site have already been demonstrated to be functional in some cancer types see references [1315]. In this study we could demonstrate that all the seven TCF binding sites are potentially functional (supplementary material) and moreover we showed that two of them are functional in vivo. Transfection experiments suggest that TCF binding sites can function in combination to enhance transcriptional activity.
These findings suggest that the canonical pathway of Wnt signaling regulates ABCB1 in CML. Several studies have demonstrated that quiescent CML stem cells do not undergo apoptosis even in the presence of high-dose or more potent TK inhibitors. Moreover, seminal studies demonstrated that a quiescent population of CML stem cells with BCR-ABL kinase domain mutation that is detectable before the initiation of IM therapy gives rise to leukemic cells that persist after treatment see references [17, 4952]. These findings suggest that CML stem cells contribute to CML persistence and disease progression. A question to be further addressed is whether ABCB1 is also regulated by the canonical Wnt pathway in CML stem cells, as this pathway has also been correlated with self-renewal.
CML stem cells and normal hematopoietic stem cells (HSC) share several characteristics despite exhibiting remarkable differences. Self-renewal is an essential stem cell property, but self-renewal pathway activation has also been increasingly recognized as a hallmark of cancer. Interestingly, HSC and CML stem cells also exhibit increased levels of drug efflux-related molecules such as the product of ABCB1, Pgp and decreased levels of OCT1, a transporter involved in the uptake of IM, rendering them more resistant to drugs [53, 54].

Conclusion

By the data presented in this work, we provided evidence that the canonical pathway of Wnt signalling is involved in ABCB1 transcriptional activation in CML.

Acknowledgements

We thank Dra. Vivian Rumjanek (Departamento de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Brazil) for providing the Lucena cell line. This work was supported by FINEP, FAPERJ, CNPQ and INCT of Cancer Control.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SC performed the experiments, statistical analysis and drafted the manuscript. RB participated and assisted the electrophoretic mobility shift assays and Luciferase experiments. BR participated in flow cytometry experiments. MTLCB performed immunofluorescence assays. LP and EA made substantial contributions to the study conception and design and critically revised the manuscript for intellectual content. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves A, Eaves C: Chronic myeloid leukemia stem cells possess multiple unique features of resistance to BCR-ABL targeted therapies. Leukemia. 2007, 21: 926-935.PubMed Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves A, Eaves C: Chronic myeloid leukemia stem cells possess multiple unique features of resistance to BCR-ABL targeted therapies. Leukemia. 2007, 21: 926-935.PubMed
2.
Zurück zum Zitat Jørgensen HG, Holyoake TL: Characterization of cancer stem cells in chronic myeloid leukaemia. Biochem Soc Trans. 2007, 35: 1347-1351. 10.1042/BST0351347.CrossRefPubMed Jørgensen HG, Holyoake TL: Characterization of cancer stem cells in chronic myeloid leukaemia. Biochem Soc Trans. 2007, 35: 1347-1351. 10.1042/BST0351347.CrossRefPubMed
3.
Zurück zum Zitat Radich JP, Dai H, Mao M, Oehler V, Schelter J, Druker B, Sawyers C, Shah N, Stock W, Willman CL, Friend S, Linsley PS: Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci U S A. 2006, 103: 2794-2799. 10.1073/pnas.0510423103.CrossRefPubMedPubMedCentral Radich JP, Dai H, Mao M, Oehler V, Schelter J, Druker B, Sawyers C, Shah N, Stock W, Willman CL, Friend S, Linsley PS: Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci U S A. 2006, 103: 2794-2799. 10.1073/pnas.0510423103.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, Lagoo A, Reya T: Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007, 12: 528-541. 10.1016/j.ccr.2007.11.003.CrossRefPubMedPubMedCentral Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, Lagoo A, Reya T: Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007, 12: 528-541. 10.1016/j.ccr.2007.11.003.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Chen Y, Peng C, Sullivan C, Li D, Li S: Critical molecular pathways in cancer stem cells of chronic myeloid leukemia. Leukemia. 2010, 24: 1545-1554. 10.1038/leu.2010.143.CrossRefPubMedPubMedCentral Chen Y, Peng C, Sullivan C, Li D, Li S: Critical molecular pathways in cancer stem cells of chronic myeloid leukemia. Leukemia. 2010, 24: 1545-1554. 10.1038/leu.2010.143.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Deshpande AJ, Buske C: Knocking the Wnt out of the sails of leukemia stem cell development. Cell Stem Cell. 2007, 1: 597-598. 10.1016/j.stem.2007.11.006.CrossRefPubMed Deshpande AJ, Buske C: Knocking the Wnt out of the sails of leukemia stem cell development. Cell Stem Cell. 2007, 1: 597-598. 10.1016/j.stem.2007.11.006.CrossRefPubMed
7.
Zurück zum Zitat Hu Y, Chen Y, Douglas L, Li S: beta-Catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia. 2009, 23: 109-116. 10.1038/leu.2008.262.CrossRefPubMed Hu Y, Chen Y, Douglas L, Li S: beta-Catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia. 2009, 23: 109-116. 10.1038/leu.2008.262.CrossRefPubMed
8.
Zurück zum Zitat Hochhaus A: Chronic myelogenous leukemia (CML): resistance to tyrosine kinase inhibitors. Ann Oncol. 2006, 17: 274-279. 10.1093/annonc/mdl273.CrossRef Hochhaus A: Chronic myelogenous leukemia (CML): resistance to tyrosine kinase inhibitors. Ann Oncol. 2006, 17: 274-279. 10.1093/annonc/mdl273.CrossRef
9.
Zurück zum Zitat Mughal TI, Goldman JM: Emerging strategies for the treatment of mutant Bcr-Abl T315I myeloid leukemia. Clin Lymphoma Myeloma. 2007, 7: S81-S84.CrossRefPubMed Mughal TI, Goldman JM: Emerging strategies for the treatment of mutant Bcr-Abl T315I myeloid leukemia. Clin Lymphoma Myeloma. 2007, 7: S81-S84.CrossRefPubMed
10.
Zurück zum Zitat Quintás-Cardama A, Kantarjian HM, Cortes JE: Mechanisms of Primary and Secondary Resistance to Imatinib in Chronic Myeloid Leukemia. Cancer Control. 2009, 16: 122-131.PubMed Quintás-Cardama A, Kantarjian HM, Cortes JE: Mechanisms of Primary and Secondary Resistance to Imatinib in Chronic Myeloid Leukemia. Cancer Control. 2009, 16: 122-131.PubMed
11.
Zurück zum Zitat Shtil AA, Azare J: Redundancy of biological regulation as the basis of emergence of multidrug resistance. Int Rev Cytol. 2005, 246: 1-29.CrossRefPubMed Shtil AA, Azare J: Redundancy of biological regulation as the basis of emergence of multidrug resistance. Int Rev Cytol. 2005, 246: 1-29.CrossRefPubMed
12.
Zurück zum Zitat Labialle S, Gayet L, Marthinet E, Rigal D, Baggetto LG: Transcriptional regulators of the human multidrug resistance 1 gene: recent views. Biochem Pharmacol. 2002, 64: 943-948. 10.1016/S0006-2952(02)01156-5.CrossRefPubMed Labialle S, Gayet L, Marthinet E, Rigal D, Baggetto LG: Transcriptional regulators of the human multidrug resistance 1 gene: recent views. Biochem Pharmacol. 2002, 64: 943-948. 10.1016/S0006-2952(02)01156-5.CrossRefPubMed
13.
Zurück zum Zitat Yamada T, Takaoka AS, Naishiro Y, Hayashi R, Maruyama K, Maesawa C, Ochiai A, Hirohashi S: Transactivation of the multidrug resistance 1 gene by T-cell factor 4/beta-catenin complex in early colorectal carcinogenesis. Cancer Res. 2000, 60: 4761-4766.PubMed Yamada T, Takaoka AS, Naishiro Y, Hayashi R, Maruyama K, Maesawa C, Ochiai A, Hirohashi S: Transactivation of the multidrug resistance 1 gene by T-cell factor 4/beta-catenin complex in early colorectal carcinogenesis. Cancer Res. 2000, 60: 4761-4766.PubMed
14.
Zurück zum Zitat Flahaut M, Meier R, Coulon A, Nardou KA, Niggli FK, Martinet D, Beckmann JS, Joseph JM, Mühlethaler-Mottet A, Gross N: The Wnt receptor FZD1 mediates chemoresistance in neuroblastoma through activation of the Wnt/beta-catenin pathway. Oncogene. 2009, 28: 2245-2256. 10.1038/onc.2009.80.CrossRefPubMed Flahaut M, Meier R, Coulon A, Nardou KA, Niggli FK, Martinet D, Beckmann JS, Joseph JM, Mühlethaler-Mottet A, Gross N: The Wnt receptor FZD1 mediates chemoresistance in neuroblastoma through activation of the Wnt/beta-catenin pathway. Oncogene. 2009, 28: 2245-2256. 10.1038/onc.2009.80.CrossRefPubMed
15.
Zurück zum Zitat Bourguignon LYW, Xia W, Wong G: Hyaluronan-mediated CD44 interaction with p300 and SIRT1 regulates beta-catenin signaling and NFkappaB-specific transcription activity leading to MDR1 and Bcl-xL gene expression and chemoresistance in breast tumor cells. J Biol Chem. 2009, 284: 2657-2671.CrossRefPubMedPubMedCentral Bourguignon LYW, Xia W, Wong G: Hyaluronan-mediated CD44 interaction with p300 and SIRT1 regulates beta-catenin signaling and NFkappaB-specific transcription activity leading to MDR1 and Bcl-xL gene expression and chemoresistance in breast tumor cells. J Biol Chem. 2009, 284: 2657-2671.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Gurney H, Wong M, Balleine RL, Rivory LP, McLachlan AJ, Hoskins JM, Wilcken N: Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype. Clin Pharmacol Ther. 2007, 82: 33-40. 10.1038/sj.clpt.6100201.CrossRefPubMed Gurney H, Wong M, Balleine RL, Rivory LP, McLachlan AJ, Hoskins JM, Wilcken N: Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype. Clin Pharmacol Ther. 2007, 82: 33-40. 10.1038/sj.clpt.6100201.CrossRefPubMed
17.
Zurück zum Zitat Dulucq S, Bouchet S, Turcq B, Lippert E, Etienne G, Reiffers J, Molimard M, Krajinovic M, Mahon FX: Multidrug resistance gene (MDR1) polymorphisms are associated with major molecular responses to standard-dose imatinib in chronic myeloid leukemia. Blood. 2008, 112: 2024-2027. 10.1182/blood-2008-03-147744.CrossRefPubMed Dulucq S, Bouchet S, Turcq B, Lippert E, Etienne G, Reiffers J, Molimard M, Krajinovic M, Mahon FX: Multidrug resistance gene (MDR1) polymorphisms are associated with major molecular responses to standard-dose imatinib in chronic myeloid leukemia. Blood. 2008, 112: 2024-2027. 10.1182/blood-2008-03-147744.CrossRefPubMed
18.
Zurück zum Zitat Kim DH, Sriharsha L, Xu W, Kamel-Reid S, Liu X, Siminovitch K, Messner HA, Lipton JH: Clinical relevance of a pharmacogenetic approach using multiple candidate genes to predict response and resistance to imatinib therapy in chronic myeloid leukemia. Clin Cancer Res. 2009, 15: 4750-4758. 10.1158/1078-0432.CCR-09-0145.CrossRefPubMed Kim DH, Sriharsha L, Xu W, Kamel-Reid S, Liu X, Siminovitch K, Messner HA, Lipton JH: Clinical relevance of a pharmacogenetic approach using multiple candidate genes to predict response and resistance to imatinib therapy in chronic myeloid leukemia. Clin Cancer Res. 2009, 15: 4750-4758. 10.1158/1078-0432.CCR-09-0145.CrossRefPubMed
19.
Zurück zum Zitat Ni LN, Li JY, Miao KR, Qiao C, Zhang SJ, Qiu HR, Qian SX: Multidrug resistance gene (MDR1) polymorphisms correlate with imatinib response in chronic myeloid leukemia. Med Oncol. 2011, 28: 265-269. 10.1007/s12032-010-9456-9.CrossRefPubMed Ni LN, Li JY, Miao KR, Qiao C, Zhang SJ, Qiu HR, Qian SX: Multidrug resistance gene (MDR1) polymorphisms correlate with imatinib response in chronic myeloid leukemia. Med Oncol. 2011, 28: 265-269. 10.1007/s12032-010-9456-9.CrossRefPubMed
20.
Zurück zum Zitat Maffiolia M, Camósa M, Gayaa A, Hernández-Boludab JC, Álvarez-Larránc A, Domingoa A, Granella M, Guillemb V, Vallansota R, Costad D, Bellosillo B, Colomerd D, Cervantesa F: Correlation between genetic polymorphisms of the hOCT1 and MDR1 genes and the response to imatinib in patients newly diagnosed with chronic-phase chronic myeloid leukemia. Leuk Res. 2011, 35: 1014-1019. 10.1016/j.leukres.2010.12.004.CrossRef Maffiolia M, Camósa M, Gayaa A, Hernández-Boludab JC, Álvarez-Larránc A, Domingoa A, Granella M, Guillemb V, Vallansota R, Costad D, Bellosillo B, Colomerd D, Cervantesa F: Correlation between genetic polymorphisms of the hOCT1 and MDR1 genes and the response to imatinib in patients newly diagnosed with chronic-phase chronic myeloid leukemia. Leuk Res. 2011, 35: 1014-1019. 10.1016/j.leukres.2010.12.004.CrossRef
21.
Zurück zum Zitat Yamakawa Y, Hamada A, Nakashima R, Yuki M, Hirayama C, Kawaguchi T, Saito H: Association of Genetic Polymorphisms in the Influx Transporter SLCO1B3 and the Efflux Transporter ABCB1 With Imatinib Pharmacokinetics in Patients With Chronic Myeloid Leukemia. Ther Drug Monit. 2011, 33: 244-250.PubMed Yamakawa Y, Hamada A, Nakashima R, Yuki M, Hirayama C, Kawaguchi T, Saito H: Association of Genetic Polymorphisms in the Influx Transporter SLCO1B3 and the Efflux Transporter ABCB1 With Imatinib Pharmacokinetics in Patients With Chronic Myeloid Leukemia. Ther Drug Monit. 2011, 33: 244-250.PubMed
22.
Zurück zum Zitat List AF, Kopecky KJ, Willman CL, Head DR, Slovak ML, Douer D, Dakhil SR, Appelbaum FR: Cyclosporine inhibition of P-glycoprotein in chronic myeloid leukemia blast phase. Blood. 2002, 100: 1910-1912.PubMed List AF, Kopecky KJ, Willman CL, Head DR, Slovak ML, Douer D, Dakhil SR, Appelbaum FR: Cyclosporine inhibition of P-glycoprotein in chronic myeloid leukemia blast phase. Blood. 2002, 100: 1910-1912.PubMed
23.
Zurück zum Zitat Stromskaya TP, Rybalkina EY, Kruglov SS, Zabotina TN, Mechetner EB, Turkina AG, Stavrovskaya AA: Role of P-glycoprotein in evolution of populations of chronic myeloid leukemia cells treated with imatinib. Biochemistry (Mosc). 2008, 73: 29-37. 10.1134/S0006297908010045.CrossRef Stromskaya TP, Rybalkina EY, Kruglov SS, Zabotina TN, Mechetner EB, Turkina AG, Stavrovskaya AA: Role of P-glycoprotein in evolution of populations of chronic myeloid leukemia cells treated with imatinib. Biochemistry (Mosc). 2008, 73: 29-37. 10.1134/S0006297908010045.CrossRef
24.
Zurück zum Zitat Vasconcelos FC, Silva KL, Souza PS, Silva LF, Moellmann-Coelho A, Klumb CE, Maia RC: Variation of MDR proteins expression and activity levels according to clinical status and evolution of CML patients. Cytometry B Clin Cytom. 2011, 80: 158-166.CrossRefPubMed Vasconcelos FC, Silva KL, Souza PS, Silva LF, Moellmann-Coelho A, Klumb CE, Maia RC: Variation of MDR proteins expression and activity levels according to clinical status and evolution of CML patients. Cytometry B Clin Cytom. 2011, 80: 158-166.CrossRefPubMed
25.
Zurück zum Zitat Racil Z, Razga F, Polakova KM, Buresova L, Polivkova V, Dvorakova D, Zackova D, Klamova H, Cetkovsky P, Mayer J: Assessment of adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) mRNA expression in patients with de novo chronic myelogenous leukemia: the role of different cell types. Leuk Lymphoma. 2011, 52: 331-334. 10.3109/10428194.2010.533220.CrossRefPubMed Racil Z, Razga F, Polakova KM, Buresova L, Polivkova V, Dvorakova D, Zackova D, Klamova H, Cetkovsky P, Mayer J: Assessment of adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) mRNA expression in patients with de novo chronic myelogenous leukemia: the role of different cell types. Leuk Lymphoma. 2011, 52: 331-334. 10.3109/10428194.2010.533220.CrossRefPubMed
26.
Zurück zum Zitat Reis FR, Vasconcelos FC, Pereira DL, Moellman-Coelho A, Silva KL, Maia RC: Survivin and P-glycoprotein are associated and highly expressed in late phase chronic myeloid leukemia. Oncol Rep. 2011, 26: 471-478.PubMed Reis FR, Vasconcelos FC, Pereira DL, Moellman-Coelho A, Silva KL, Maia RC: Survivin and P-glycoprotein are associated and highly expressed in late phase chronic myeloid leukemia. Oncol Rep. 2011, 26: 471-478.PubMed
27.
Zurück zum Zitat Mahon FX, Deininger M, Schultheis B, Chabrol J, Reiffers J, Goldman JM, Melo JV: Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the signal transduction inhibitor STI571: diverse mechanisms of resistance. Blood. 2000, 96: 1070-1079.PubMed Mahon FX, Deininger M, Schultheis B, Chabrol J, Reiffers J, Goldman JM, Melo JV: Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the signal transduction inhibitor STI571: diverse mechanisms of resistance. Blood. 2000, 96: 1070-1079.PubMed
28.
Zurück zum Zitat Tang C, Schafranek L, Watkins DB, Parker WT, Moore S, Prime JA, White DL, Hughes TP: Tyrosine kinase inhibitor resistance in chronic myeloid leukemia cell lines: investigating resistance pathways. Leuk Lymphoma. 2011, 52: 2139-2147. 10.3109/10428194.2011.591013.CrossRefPubMed Tang C, Schafranek L, Watkins DB, Parker WT, Moore S, Prime JA, White DL, Hughes TP: Tyrosine kinase inhibitor resistance in chronic myeloid leukemia cell lines: investigating resistance pathways. Leuk Lymphoma. 2011, 52: 2139-2147. 10.3109/10428194.2011.591013.CrossRefPubMed
29.
Zurück zum Zitat Yamada O, Ozaki K, Furukawa T, Machida M, Wang YH, Motoji T, Mitsuishi T, Akiyama M, Yamada H, Kawauchi K, Matsuoka R: Activation of STAT5 confers imatinib resistance on leukemic cells through the transcription of TERT and MDR1. Cell Signal. 2011, 23: 1119-1127. 10.1016/j.cellsig.2011.02.005.CrossRefPubMed Yamada O, Ozaki K, Furukawa T, Machida M, Wang YH, Motoji T, Mitsuishi T, Akiyama M, Yamada H, Kawauchi K, Matsuoka R: Activation of STAT5 confers imatinib resistance on leukemic cells through the transcription of TERT and MDR1. Cell Signal. 2011, 23: 1119-1127. 10.1016/j.cellsig.2011.02.005.CrossRefPubMed
30.
Zurück zum Zitat Corrêa S, Pizzatti L, Du Rocher B, Mencalha A, Pinto D, Abdelhay E: A comparative proteomic study identified LRPPRC and MCM7 as putative actors in imatinib mesylate cross-resistance in Lucena cell line. Proteome Sci. 2012, 10: 23-10.1186/1477-5956-10-23. Epub ahead of printCrossRefPubMedPubMedCentral Corrêa S, Pizzatti L, Du Rocher B, Mencalha A, Pinto D, Abdelhay E: A comparative proteomic study identified LRPPRC and MCM7 as putative actors in imatinib mesylate cross-resistance in Lucena cell line. Proteome Sci. 2012, 10: 23-10.1186/1477-5956-10-23. Epub ahead of printCrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Rumjanek VM, Trindade GS, Wagner-Souza K, de-Oliveira MC, Marques-Santos LF, Maia RC, Capella MA: Multidrug resistance in tumour cells: characterization of the multidrug resistant cell line K562-Lucena 1. An Acad Bras Cienc. 2001, 73: 57-69. 10.1590/S0001-37652001000100007.CrossRefPubMed Rumjanek VM, Trindade GS, Wagner-Souza K, de-Oliveira MC, Marques-Santos LF, Maia RC, Capella MA: Multidrug resistance in tumour cells: characterization of the multidrug resistant cell line K562-Lucena 1. An Acad Bras Cienc. 2001, 73: 57-69. 10.1590/S0001-37652001000100007.CrossRefPubMed
32.
Zurück zum Zitat Binato R, Alvarez Martinez CE, Pizzatti L, Robert B, Abdelhay E: SMAD 8 binding to mice Msx1 basal promoter is required for transcriptional activation. Biochem J. 2006, 393: 141-150. 10.1042/BJ20050327.CrossRefPubMed Binato R, Alvarez Martinez CE, Pizzatti L, Robert B, Abdelhay E: SMAD 8 binding to mice Msx1 basal promoter is required for transcriptional activation. Biochem J. 2006, 393: 141-150. 10.1042/BJ20050327.CrossRefPubMed
33.
Zurück zum Zitat Pizzatti L, Binato R, Cofre J, Gomes BE, Dobbin J, Haussmann ME, D'Azambuja D, Bouzas LF, Abdelhay E: SUZ12 is a candidate target of the non-canonical WNT pathway in the progression of chronic myeloid leukemia. Genes Chromosomes Cancer. 2010, 49: 107-118.PubMed Pizzatti L, Binato R, Cofre J, Gomes BE, Dobbin J, Haussmann ME, D'Azambuja D, Bouzas LF, Abdelhay E: SUZ12 is a candidate target of the non-canonical WNT pathway in the progression of chronic myeloid leukemia. Genes Chromosomes Cancer. 2010, 49: 107-118.PubMed
34.
Zurück zum Zitat Wagschal A, Delaval K, Pannetier M, Arnaud P, Feil R: Chromatin Immunoprecipitation (ChIP) on Unfixed Chromatin from Cells and Tissues to Analyze Histone Modifications. Cold Spring Harb. Protoc. 2007 Wagschal A, Delaval K, Pannetier M, Arnaud P, Feil R: Chromatin Immunoprecipitation (ChIP) on Unfixed Chromatin from Cells and Tissues to Analyze Histone Modifications. Cold Spring Harb. Protoc. 2007
35.
Zurück zum Zitat Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C (T)) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C (T)) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed
36.
Zurück zum Zitat Stambolic V, Ruel L, Woodgett JR: Lithium inhibits glycogen synthase kinase-3 activity and mimics wingless signalling in intact cells. Curr Biol. 1996, 6: 1664-1668. 10.1016/S0960-9822(02)70790-2.CrossRefPubMed Stambolic V, Ruel L, Woodgett JR: Lithium inhibits glycogen synthase kinase-3 activity and mimics wingless signalling in intact cells. Curr Biol. 1996, 6: 1664-1668. 10.1016/S0960-9822(02)70790-2.CrossRefPubMed
37.
Zurück zum Zitat Lim JC, Kania KD, Wijesuriya H, Chawla S, Sethi JK, Pulaski L, Romero IA, Couraud PO, Weksler BB, Hladky SB, Barrand MA: Activation of beta-catenin signalling by GSK-3 inhibition increases p-glycoprotein expression in brain endothelial cells. J Neurochem. 2008, 106: 1855-1865.PubMedPubMedCentral Lim JC, Kania KD, Wijesuriya H, Chawla S, Sethi JK, Pulaski L, Romero IA, Couraud PO, Weksler BB, Hladky SB, Barrand MA: Activation of beta-catenin signalling by GSK-3 inhibition increases p-glycoprotein expression in brain endothelial cells. J Neurochem. 2008, 106: 1855-1865.PubMedPubMedCentral
38.
Zurück zum Zitat Melo JV, Barnes DJ: Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nat Rev Cancer. 2007, 7: 441-453. 10.1038/nrc2147.CrossRefPubMed Melo JV, Barnes DJ: Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nat Rev Cancer. 2007, 7: 441-453. 10.1038/nrc2147.CrossRefPubMed
39.
Zurück zum Zitat Perrotti D, Jamieson C, Goldman J, Skorski T: Chronic myeloid leukemia: mechanisms of blastic transformation. J Clin Invest. 2010, 120: 2254-2264. 10.1172/JCI41246.CrossRefPubMedPubMedCentral Perrotti D, Jamieson C, Goldman J, Skorski T: Chronic myeloid leukemia: mechanisms of blastic transformation. J Clin Invest. 2010, 120: 2254-2264. 10.1172/JCI41246.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Jabbour E, Fava C, Kantarjian H: Advances in the biology and therapy of patients with chronic myeloid leukaemia. Best Prac Res Clin Haematol. 2009, 22: 395-407. 10.1016/j.beha.2009.09.002.CrossRef Jabbour E, Fava C, Kantarjian H: Advances in the biology and therapy of patients with chronic myeloid leukaemia. Best Prac Res Clin Haematol. 2009, 22: 395-407. 10.1016/j.beha.2009.09.002.CrossRef
41.
Zurück zum Zitat Swords R, Alvarado Y, Giles F: Novel Abl kinase inhibitors in chronic myeloid leukemia in blastic phase and Philadelphia chromosome-positive acute lymphoblastic leukemia. Clin Lymphoma Myeloma. 2007, 7: S113-S119.CrossRefPubMed Swords R, Alvarado Y, Giles F: Novel Abl kinase inhibitors in chronic myeloid leukemia in blastic phase and Philadelphia chromosome-positive acute lymphoblastic leukemia. Clin Lymphoma Myeloma. 2007, 7: S113-S119.CrossRefPubMed
42.
Zurück zum Zitat Roychowdhury S, Talpaz M: Managing resistance in chronic myeloid leukemia. Blood Reviews. 2011, 25: 279-290. 10.1016/j.blre.2011.09.001.CrossRefPubMed Roychowdhury S, Talpaz M: Managing resistance in chronic myeloid leukemia. Blood Reviews. 2011, 25: 279-290. 10.1016/j.blre.2011.09.001.CrossRefPubMed
43.
Zurück zum Zitat Hegedus T, Orfi L, Seprodi A, Váradi A, Sarkadi B, Kéri G: Interaction of tyrosine kinase inhibitors with the human multidrug transporter proteins, MDR1 and MRP1. Biochim Biophys Acta. 2002, 1587: 318-325. 10.1016/S0925-4439(02)00095-9.CrossRefPubMed Hegedus T, Orfi L, Seprodi A, Váradi A, Sarkadi B, Kéri G: Interaction of tyrosine kinase inhibitors with the human multidrug transporter proteins, MDR1 and MRP1. Biochim Biophys Acta. 2002, 1587: 318-325. 10.1016/S0925-4439(02)00095-9.CrossRefPubMed
44.
Zurück zum Zitat Hamada A, Miyano H, Watanabe H, Saito H: Interaction of Imatinib Mesilate with Human P-Glycoprotein. J Pharmacol Exp Ther. 2003, 307: 824-828. 10.1124/jpet.103.055574.CrossRefPubMed Hamada A, Miyano H, Watanabe H, Saito H: Interaction of Imatinib Mesilate with Human P-Glycoprotein. J Pharmacol Exp Ther. 2003, 307: 824-828. 10.1124/jpet.103.055574.CrossRefPubMed
45.
Zurück zum Zitat Mahon FX, Belloc F, Lagarde V, Chollet C, Moreau-Gaudry F, Reiffers J, Goldman JM, Melo JV: MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models. Blood. 2003, 101: 2368-2373. 10.1182/blood.V101.6.2368.CrossRefPubMed Mahon FX, Belloc F, Lagarde V, Chollet C, Moreau-Gaudry F, Reiffers J, Goldman JM, Melo JV: MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models. Blood. 2003, 101: 2368-2373. 10.1182/blood.V101.6.2368.CrossRefPubMed
46.
Zurück zum Zitat Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, Gotlib J, Li K, Manz MG, Keating A, Sawyers CL, Weissman IL: Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004, 351: 657-667. 10.1056/NEJMoa040258.CrossRefPubMed Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, Gotlib J, Li K, Manz MG, Keating A, Sawyers CL, Weissman IL: Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004, 351: 657-667. 10.1056/NEJMoa040258.CrossRefPubMed
47.
Zurück zum Zitat Gregory MA, Phang TL, Neviani P, Alvarez-Calderon F, Eide CA, O'Hare T, Zaberezhnyy V, Williams RT, Druker BJ, Perrotti D, Degregori J: Wnt/Ca2+/NFAT signaling maintains survival of Ph + leukemia cells upon inhibition of Bcr-Abl. Cancer Cell. 2010, 18: 74-87. 10.1016/j.ccr.2010.04.025.CrossRefPubMedPubMedCentral Gregory MA, Phang TL, Neviani P, Alvarez-Calderon F, Eide CA, O'Hare T, Zaberezhnyy V, Williams RT, Druker BJ, Perrotti D, Degregori J: Wnt/Ca2+/NFAT signaling maintains survival of Ph + leukemia cells upon inhibition of Bcr-Abl. Cancer Cell. 2010, 18: 74-87. 10.1016/j.ccr.2010.04.025.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Coluccia AM, Vacca A, Duñach M, Mologni L, Redaelli S, Bustos VH, Benati D, Pinna LA, Gambacorti-Passerini C: Bcr-Abl stabilizes beta-catenin in chronic myeloid leukemia through its tyrosine phosphorylation. EMBO J. 2007, 26: 1456-1466. 10.1038/sj.emboj.7601485.CrossRefPubMedPubMedCentral Coluccia AM, Vacca A, Duñach M, Mologni L, Redaelli S, Bustos VH, Benati D, Pinna LA, Gambacorti-Passerini C: Bcr-Abl stabilizes beta-catenin in chronic myeloid leukemia through its tyrosine phosphorylation. EMBO J. 2007, 26: 1456-1466. 10.1038/sj.emboj.7601485.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Ress A, Moelling K: Bcr is a negative regulator of the Wnt signalling pathway. EMBO Reports. 2005, 11: 1095-1100.CrossRef Ress A, Moelling K: Bcr is a negative regulator of the Wnt signalling pathway. EMBO Reports. 2005, 11: 1095-1100.CrossRef
50.
51.
Zurück zum Zitat Chomel JC, Turhan AG: Chronic myeloid leukemia stem cells in the era of targeted therapies: resistance, persistence and long-term dormancy. Oncotarget. 2011, 2: 713-727.CrossRefPubMedPubMedCentral Chomel JC, Turhan AG: Chronic myeloid leukemia stem cells in the era of targeted therapies: resistance, persistence and long-term dormancy. Oncotarget. 2011, 2: 713-727.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Leber B: CML biology for the clinician in 2011: six impossible things to believe before breakfast on the way to cure. Curr Oncol. 2011, 18: 185-190.CrossRef Leber B: CML biology for the clinician in 2011: six impossible things to believe before breakfast on the way to cure. Curr Oncol. 2011, 18: 185-190.CrossRef
53.
Zurück zum Zitat Chomel JC, Bonnet ML, Sorel N, Bertrand A, Meunier MC, Fichelson S, Melkus M, Bennaceur-Griscelli A, Guilhot F, Turhan AG: Leukemic stem cell persistence in chronic myeloid leukemia patients with sustained undetectable molecular residual disease. Blood. 2011, 118: 3657-3660. 10.1182/blood-2011-02-335497.CrossRefPubMed Chomel JC, Bonnet ML, Sorel N, Bertrand A, Meunier MC, Fichelson S, Melkus M, Bennaceur-Griscelli A, Guilhot F, Turhan AG: Leukemic stem cell persistence in chronic myeloid leukemia patients with sustained undetectable molecular residual disease. Blood. 2011, 118: 3657-3660. 10.1182/blood-2011-02-335497.CrossRefPubMed
54.
Zurück zum Zitat Helgason GV, Young GA, Holyoake TL: Targeting chronic myeloid leukemia stem cells. Curr Hematol Malig Rep. 2010, 5: 81-87. 10.1007/s11899-010-0043-0.CrossRefPubMed Helgason GV, Young GA, Holyoake TL: Targeting chronic myeloid leukemia stem cells. Curr Hematol Malig Rep. 2010, 5: 81-87. 10.1007/s11899-010-0043-0.CrossRefPubMed
Metadaten
Titel
Wnt/β-catenin pathway regulates ABCB1 transcription in chronic myeloid leukemia
verfasst von
Stephany Corrêa
Renata Binato
Bárbara Du Rocher
Morgana TL Castelo-Branco
Luciana Pizzatti
Eliana Abdelhay
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2012
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-12-303

Weitere Artikel der Ausgabe 1/2012

BMC Cancer 1/2012 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.