Skip to main content

Characterization of SN38-resistant T47D breast cancer cell sublines overexpressing BCRP, MRP1, MRP2, MRP3, and MRP4

Abstract

Background

Although several novel resistant breast cancer cell lines have been established, only a few resistant breast cancer cell lines overexpress breast cancer resistance proteins (BCRP). The aim of this study was to establish new resistant breast cancer cell lines overexpressing BCRP using SN38 (7-ethyl-10-hydroxycamptothecin), an active metabolite of irinotecan and was to discover genes and mechanisms associated with multidrug resistance.

Methods

SN38-resistant T47D breast cancer cell sublines were selected from the wild-type T47D cells by gradually increasing SN38 concentration. The sensitivity of the cells to anti-cancer drugs was assessed by 3-(4,5-methylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. Expression profiles of the resistance-related transporters were examined using RT-qPCR, and western blot analysis. Intracellular fluorescent dye accumulation in the resistant cells was determined using flow cytometry.

Results

The SN38-resistant T47D breast cancer cell sublines T47D/SN120 and T47D/SN150 were established after long-term exposure (more than 16 months) of wild-type T47D cells to 120 nM and 150 nM SN38, respectively. T47D/SN120 and T47D/SN150 cells were more resistant to SN38 (14.5 and 59.1 times, respectively), irinotecan (1.5 and 3.7 times, respectively), and topotecan (4.9 and 12 times, respectively), than the wild-type parental cells. Both T47D/SN120 and T47D/SN150 sublines were cross-resistant to various anti-cancer drugs. These resistant sublines overexpressed mRNAs of MRP1, MRP2, MRP3, MRP4, and BCRP. The DNA methylase inhibitor 5-aza-2′-deoxycytidine and the histone deacetylase inhibitor trichostatin A increased the expression levels of BCRP, MRP1, MRP2, MRP3, and MRP4 transcripts in T47D/WT cells. Fluorescent dye accumulation was found to be lower in T47D/SN120 and T47D/SN150 cells, compared to that in T47D/WT cells. However, treatment with known chemosensitizers increased the intracellular fluorescent dye accumulation and sensitivity of anti-tumor agents.

Conclusion

T47D/SN120 and T47D/SN150 cells overexpressed MRP1, MRP2, MRP3, MRP4, and BCRP, which might be due to the suppression of epigenetic gene silencing via DNA hypermethylation and histone deacetylation. Although these resistant cells present a higher resistance to various anti-cancer drugs than their parental wild-type cells, multidrug resistance was overcome by treatment with chemosensitizers. These SN38 resistant T47D breast cancer cell sublines expressing resistance proteins can be useful for the development of new chemosensitizers.

Peer Review reports

Background

Breast cancer is not only a frequently diagnosed cancer worldwide, it is also a leading cause of cancer-related death in women [1]. The treatment of breast cancer is largely classified into two categories: local treatment including surgery, radiation, or both modalities; and systemic treatment including cytotoxic chemotherapy, endocrine therapy, biological therapy, or a combination of these. Systemic treatment, which is used in adjuvant, neoadjuvant, and palliative settings, plays an important role in the treatment of breast cancer at various stages. Indeed, systemic agents are effective at the beginning of therapy in 69–95% of primary breast cancers [2,3,4,5,6] and 50% of metastatic cancers [7,8,9]. However, cancer progression often occurs after a variable duration of chemotherapy, owing to the development of chemotherapy resistance, which is divided into primary resistance and acquired resistance. Primary resistance includes cases that are unresponsive despite the use of appropriate initial chemotherapy, with continued tumor growth during the treatment [10]. Acquired resistance involves cases where the tumor cells initially seem to respond well to the chemotherapy but acquire resistance to the anti-cancer drugs due to repeated exposure [10]; in this case, cells are resistant to some agents of the same class but sensitive to drugs of different classes. However, eventual cross-resistance to multiple anti-cancer drugs of apparently different structures and functions is observed; this phenomenon is known as multidrug resistance (MDR) [11].

The mechanisms and pathways of MDR are complicated and multi-factorial. One of the mechanisms of MDR is associated with the alteration of anti-cancer drug transporter. One of the classic MDR mechanisms involves decreased drug accumulation due to increased expression of drug efflux pump in the tumor cell membrane [12]. This MDR phenotype is mediated by ATP (adenosine triphosphate)-binding cassette (ABC) transporters [12], including P-glycoprotein (Pgp) [13,14,15] and members of the multidrug resistance protein (MRP) family [16,17,18]. Another novel transporter, breast cancer resistance protein (BCRP), has been identified as an ABC half-transporter and is distributed in the placenta and various cancer types [19,20,21,22]. Meanwhile, it was reported that a redistribution of the anti-cancer drug from the nucleus to the cytoplasm is related to non-ABC transporters-mediated MDR [23]. Lung resistance protein (LRP) is involved in the nucleo-cytoplasmic transport and cytoplasmic sequestration of anti-cancer drugs [24,25,26]. Another mechanism of MDR is associated with alterations in topoisomerase [27,28,29,30,31].

Based on comprehensive knowledge about the mechanisms of drug resistance, recent studies on the mechanism of resistance to anticancer drugs in breast cancer have been reported [32]. Moreover, exploring the mechanism of MDR and finding molecular targets for drug resistance is important for the development of new therapeutic agents to treat cancer. Thus, further studies are vital for a better understanding of MDR mechanisms in breast cancer and development of various types of resistant cancer cell lines.

Recently, several novel resistant breast cancer cell lines have been established. Notably, although BCRP expression is observed in primary breast cancer as well as in normal breast tissue [33,34,35], only a few resistant breast cancer cell lines overexpress BCRP [36,37,38,39]. Tumor cell lines resistant to the camptothecin-derived topoisomerase I inhibitor topotecan have been shown to overexpress BCRP and display a significant cross-resistance to CPT11, SN38 (7-ethyl-10-hydroxycamptothecin), and 9-aminocamptothecin [28, 40,41,42]. SN38, an active metabolite of irinotecan, possesses a much stronger cytotoxicity against tumor cells than irinotecan [43, 44] through the inhibition of DNA topoisomerase I [45]. Among breast cancer cell lines, MDA-MB 231 is estrogen receptor (ER)-negative, while T47D and MCF-7 are ER-positive [46]. Recently, SN-38-resistant MCF-7 and MDA-MB-231 sublines have been generated [47]. However, no SN38-resistant breast cancer cell T47D sublines have been developed so far. Therefore, the specific aim of this study was to establish SN38-resistant breast cancer cell sublines, anticipating the overexpression of transporters, such as BCRP and to investigate MDR mechanisms. Herein, we describe two SN38-resistant breast cancer cell lines, T47D/SN120 and T47D/SN150, characterized by MRP1, MRP2, MRP3, MRP4, and BCRP overexpression. These resistant cancer cell sublines will be used to develop chemosensitizers that can reverse the resistance.

Methods

Cell culture

The human breast cancer cell line T47D was purchased from the Cancer Research Center in Seoul National University (Seoul, South Korea). The cells were cultured in RPMI-1640 (Gibco BRL Grand Island, NY, USA) supplemented with 10% fetal bovine serum (Sigma-Aldrich, St. Louis, MO, USA). The cells were allowed to adhere to the culture dish and form a monolayer. The cells were sub-cultured once they reached confluence.

Establishment of the SN38-resistant breast cancer cell sublines

The SN38-resistant breast cancer cell sublines T47D/SN120 and T47D/SN150 were established by gradually increasing the concentration of SN38 from 15 nM (IC50 value) to final concentrations of 120 nM (IC50 × 8) and 150 nM (IC50 × 10), respectively in the parental T47D cells. Stable T47D/SN120 and T47D/SN150 cell lines were obtained in a timeframe of 16 months.

Calculation of cellular population doubling time

The cells were seeded into 24-well culture plates at a density of 5 × 104 cells per well and incubated at 37 °C for 24 h. The cell numbers were counted every 24 h for 4 days. The cell doubling time (Td) was calculated using the formula: Td = T x log2/(log Nt- log N0), where N0 and Nt represent the number of cells at the beginning and end of the culture during time T, respectively.

Chemosensitivity test using MTT assay

The 3-(4,5-methylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT; Sigma-Aldrich) assay was performed to assess the sensitivity of T47D/SN120 and T47D/SN150 cells to anti-cancer drugs. 50% inhibitory concentration (IC50) was defined as the drug concentration that causes a 50% reduction in the number of cells compared to the untreated control. The IC50 values were determined directly from the dose-response curves. Resistance factor (RF) was calculated from the ratios of the IC50 values of T47D/SN120 and T47D/SN150 to T47D/WT cells.

RNA extraction and reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay

Total RNA was extracted from the cells using the RNeasy mini kit (Qiagen, Hilden, Germany). mRNA expression was determined by RT-qPCR and normalized to β-actin mRNA expression level. Genes as well as the conventional and real-time primer pairs are listed in Table 1.

Table 1 Primer sequences of PCR

RNA was reverse transcribed using Moloney murine leukemia virus reverse transcriptase (Bethesda Research Laboratories, USA) and an oligo (dT) primer for 1 h at 37 °C. The synthesized cDNA was diluted 1:5 with water and amplified using 2.5 units of Taq DNA polymerase (TaKaRa, Tokyo, Japan) and 10 pmol of each primer, under defined PCR conditions, using a GeneAmp PCR system 9600 (Perkin-Elmer-Cetus, Waltham, MA, USA). After the final cycle, all the PCR products were subjected to a final extension at 72 °C for 5 min. The PCR products were electrophoresed on agarose gel. qPCR was conducted by LightCycler® 2.0 (Roche, USA) using TB Green® Premix Ex Taq (Tli RNaseH Plus) (TaKaRa, Tokyo, Japan). The endpoint used in PCR quantification (Ct) was defined as the PCR cycle number that crosses an arbitrarily placed signal threshold.

Western blot analysis

Cells were washed with phosphate-buffered saline (PBS) and lysed in 50 mM Tris-HCl (pH 7.4), 250 mM NaCl, 0.5% Triton X-100, 10% glycerol, 1 mM DTT, 1 mM phenylmethylsulfonyl fluoride, and protease inhibitor cocktail (Pierce Biotechnology, Rockford, IL, USA). The cell lysates were centrifuged and then resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis. Western blotting was performed using a slight modification of the method described previously [48]. The membrane was incubated with primary rabbit polyclonal antibodies against MRP1 (1:1000; Invitrogen, Carlsbad, CA, USA), MRP2 (1:5000; Sigma-Aldrich), MRP3 (1:50; Abcam, Cambridge, England), MRP4 (1:50; Abcam), BCRP (1:500; Santa Cruz Biotechnology, Santa Cruz, CA, USA), and GAPDH (1:6000; Santa Cruz Biotechnology, Texas, USA). The membrane was then washed and incubated with horseradish peroxidase-conjugated secondary antibodies (1:2500 for BCRP and GAPDH) for 1 h. The signal was then detected using the ECL detection kit (Amersham, Piscataway, NJ, USA). Densities of blots were determined by densitometric analysis using a Kodak Image Station 4000MM (Eastman Kodak, Rochester, NY, USA).

Fluorescent dye accumulation assay

Cell suspensions (5 × 105 cells) in PBS were exposed to 1 μM rhodamine 123, 50 nM calcein-AM, and 20 μM mitoxantrone at 37 °C for 1 h. Additionally, cells were incubated in the presence of each fluorescent substrate with 10 nM PSC833, 1 mM probenecid, 5 mM probenecid, 200 μM genistein, and 2 mM cyanide in PBS at 37 °C for 1 h. After incubation, cellular fluorescent dye accumulation was determined using a flow cytometer (FACSCalibur, Becton Dickinson, MA, USA), which detected drug fluorescence. A focused argon laser beam (488 nm) excited the cells in a laminar sheath flow, following which the fluorescence emissions at 530 nm (for rhodamine 123 and calcein-AM) and 670 nm (for mitoxantrone) were detected to generate the histogram.

Screening of chemosensitizers

IC50 values of SN38 were obtained in the presence and absence of chemosensitizers for SN38-resistant T47D sublines and the ratios were defined as chemosensitizing index.

Determination of involvement of epigenetic gene silencing of transporters in the T47D/WT cells

We tested if hypermethylation and deacetylation are involved in epigenetic gene silencing by treating the cells with the DNA methyltransferase inhibitor (2.5 uM 5-aza-2′-deoxycytidine) for 96 h and the histone deacetylase (HDAC) inhibitor (100 ng /ml trichostatin A) for 48 h, respectively.

Statistical analysis

All experiments were repeated more than three times. Statistical significance of the data was determined using student’s t-test. P values less than 0.05 were considered significant.

Results

Establishment and characterization of resistant cell lines

SN38-resistant T47D/SN120 and T47D/SN150 cell sublines were established from the wild-type T47D cells following long-term exposure (of more than 16 months) to 120 nM and 150 nM SN38, respectively. Microscopic observation showed some distinct features in the SN38-resistant T47D sublines compared to their parental cell line. In monolayer, T47D/WT cells were relatively consistent in size and shape, while the resistant cells presented a spindle-shaped morphology and were smaller in size (Supplementary Fig. 1). Interestingly, the doubling times of T47D/SN120 and T47D/SN150 cell lines were shorter as 38.0 ± 1.33 h and 46.7 ± 2.64 h, respectively, than that of the parental cell lines (70.5 ± 3.84 h) (Supplementary Fig. 2).

Cross-resistance to other anti-cancer drugs of resistant cell lines

MTT assay showed that T47D/SN120 and T47D/SN150 cells were more resistant to SN38 (14.5 and 59.1 times, respectively), irinotecan (1.5 and 3.7 times, respectively), and topotecan (4.9 and 12 times, respectively) as compared to the wild-type drug-sensitive parental cells. In addition to topoisomerase I inhibitors, both T47D/SN120 and T47D/SN150 sublines were cross-resistant to various anti-cancer drugs that are used in breast cancer treatment, including microtubule inhibitors (paclitaxel and vinblastine), anti-metabolites (5-fluorouracil), topoisomerase II inhibitors (doxorubicin and mitoxantrone), estrogen receptor blockers (endoxifen), and a tyrosine kinase inhibitor (gefitinib) (Table 2).

Table 2 The sensitivity of the T47D/WT, T47D/SN120 and T47D/SN150 cells to SN38 and other anticancer drugs. Drugs are sorted according to the order of relative resistance of T47D/SN120 cells to T47D/WT cells

As shown in Table 2, both resistant sublines were highly resistant to paclitaxel and vinblastine. Additionally, T47D/SN150 cells showed a high resistance to SN38, mitoxantrone and doxorubicin and moderate resistance to topotecan. However, the sensitivity to gefinitib, endoxifen, 5-FU and methotrexate appeared similar between both resistant sublines.

Expression profiles of transporters in the T47D/SN sublines

RT-qPCR confirmed that T47D/SN120 and T47D/SN150 cells overexpressed MRP1 (7-fold and 11-fold, respectively), MRP2 (795-fold and 1061-fold, respectively), MRP3 (57-fold and 96-fold, respectively), MRP4 (204-fold both), and BCRP (536-fold and 3083-fold, respectively), compared to T47D/WT cells (Fig. 1).

Fig. 1
figure 1

Reverse transcription quantitative PCR assay for MRP1, MRP2, MRP3, MRP4 and BCRP mRNA. RT-qPCR confirmed that T47D/SN120 and T47D/SN150 cells overexpressed MRP1 (7-fold and 11-fold, respectively), MRP2 (795-fold and 1061-fold, respectively), MRP3 (57-fold and 96-fold, respectively), MRP4 (204-fold both), and BCRP (536-fold and 3083-fold, respectively), compared to T47D/WT cells. Numbers above column refer relative fold increase as compared with WT cells. *, P < 0.05 vs WT (n = 3)

Expression levels of MRP1, MRP2, MRP3, MRP4, and BCRP were determined by western blot analysis. MRP1, MRP2, MRP3, and MRP4 expression levels in T47D/SN150 cells were compared with those in T47D/SN120 cells. Expression levels of MRP4 and BCRP but not MRP1, MRP2 and MRP3 were significantly different between both resistant sublines (Fig. 2, Supplementary Fig. 3).

Fig. 2
figure 2

Western blot analysis for MRP1, MRP2, MRP3, MRP4 and BCRP protein. MRP1, MRP2, MRP3, and MRP4 expression levels in T47D/SN150 cells were compared with those in T47D/SN120 cells. T47D/SN150 cells overexpressed MRP4 (2.2-fold) and BCRP (4-fold) protein. Compared to T47D/WT cells, which expressed a trace amount of BCRP, T47D/SN120 and T47D/SN150 cells displayed 85.3-fold and 327.5-fold higher BCRP protein expression, respectively. (n = 3)

Compared to T47D/WT cells, which expressed a trace amount of BCRP, T47D/SN120 and T47D/SN150 cells displayed 85.3-fold and 327.5-fold higher BCRP protein expression, respectively. T47D/SN150 cells displayed 4-fold higher expression of BCRP, as compared to T47D/SN120 cells (Fig. 2).

Sensitivity of SN38-resistant T47D sublines to SN38 in the presence of various chemosensitizers

The effects of several chemosensitizers, including Pgp [verapamil, PSC833, and 7,3′,4-trimethoxyflavone (TMF)], MRP (probenecid), and BCRP (genistein) inhibitors, were assessed on SN38-resistant T47D sublines [49]. Among the Pgp inhibitors, only TMF showed chemosensitizing effects on SN38 in a concentration-dependent manner. Probenecid and genistein also sensitized SN38-resistant T47D sublines to SN38 in a concentration-dependent manner (Fig. 3). The chemosensitizing effects of probenecid were similar between T47D/SN120 and T47D/SN150 cells whereas those of TMF and genistein in the T47D/SN150 cells was more sensitive as compared to the T47D/SN120 cells. As shown in Table 3, the chemosensitizing index was calculated by dividing the IC50 value in the absence of the chemosensitizer with that in its presence. Chemosensitizing indices of probenecid (5 and 50 μM) and genistein (1 and 10 μM) were 1.3–2.0 and 1.2–10.8, respectively, in both the cell lines, whereas those for verapamil (1 and 10 μM) and PSC833 (5 and 50 nM) were less than 1.0, with the exception of PSC833 in the T47D/SN120 cell line, which displayed a chemosensitizing index of 1.3.

Fig. 3
figure 3

Sensitivity of SN38-resistant T47D sublines to SN38 in the presence of various chemosensitizers. The effects of several chemosensitizers, including Pgp [verapamil, PSC833, and 7,3′,4-trimethoxyflavone (TMF)], MRP (probenecid), and BCRP (genistein) inhibitors, were assessed on SN38-resistant T47D sublines. Among the Pgp inhibitors, only TMF showed chemosensitizing effects on SN38 in a concentration-dependent manner. Probenecid and genistein also sensitized SN38-resistant T47D sublines to SN38 in a concentration-dependent manner. (n = 3). Vp, verapamil; TMF, 7,3′,4-trimethoxyflavone; PB, probenecid; GS, genistein

Table 3 Effect of chemosensitizers on SN38-resistant T47D sublines

Involvement of epigenetic gene silencing of MRP1, MRP2, MRP3, MRP4, and BCRP in T47D/WT cells

Next, we investigated whether MRP1, MRP2, MRP3, MRP4, and BCRP could be epigenetically induced in T47D/WT cells following treatment with 2.5 μM 5-aza-2′-deoxycytidine for 96 h or 100 ng/mL trichostatin A (TSA) for 48 h. RT-qPCR indicated that 5-aza-2′-deoxycytidine induced the mRNA expression of MRP2, MRP3, MRP4, and BCRP, while TSA induced the mRNA expression of MRP1, MRP2, MRP4, and BCRP in T47D/WT cells (Fig. 4).

Fig. 4
figure 4

Effects of 5-aza-2′-deoxcytidine (AdC) and trichostatin A (TSA) on expression of MRP1, MRP2, MRP3, MRP4 and BCRP in T47D/WT cells. RT-qPCR indicated that 5-aza-2′-deoxycytidine induced the mRNA expression of MRP2, MRP3, MRP4, and BCRP, while TSA induced the mRNA expression of MRP1, MRP2, MRP4, and BCRP in T47D/WT cells. *, P < 0.05 vs CTL (n = 4)

Decreased fluorescent dye accumulation in T47D/SN120 and T47D/SN150 cells

Fluorescent dye accumulation was assayed to estimate the functional activity of transporters in T47D/SN120 and T47D/SN150 cells. Calcein AM and mitoxantrone were used as fluorescent substrates for MRP and BCRP, respectively, and detected using flow cytometry. Accumulation of both the substrates decreased in T47D/SN120 and T47D/SN150 cells, compared to that in T47D/WT cells. However, upon treatment with the inhibitors probenecid and genistein, there was an increase in the accumulation of both the substrates (Figs. 5-6).

Fig. 5
figure 5

Intracelluar calcein-AM accumulation of the T47D/WT, T47D/SN120 and T47D/SN150 cells. Accumulation of calcein AM decreased in T47D/SN120 and T47D/SN150 cells, compared to that in T47D/WT cells. However, calcein AM accumulation was increased after treatment with MRP inhibitor (probenecid) and ATP deplete cyanide. *, P < 0.05 vs WT (n = 3)

Fig. 6
figure 6

Intracelluar mitoxantrone accumulation of the T47D/WT, T47D/SN120 and T47D/SN150 cells. Mitoxantrone accumulation decreased in T47D/SN120 and T47D/SN150 cells, compared to that in T47D/WT cells. However, mitoxantrone accumulation was increased after treatment with BCRP inhibitor (genistein) and ATP deplete cyanide. *, P < 0.05 vs WT (n = 3)

Meanwhile, intracellular levels of rhodamine 123, a fluorescent substrate of Pgp, were not affected by treatment with a Pgp inhibitor PCS833 in T47D/SN120 and T47D/SN150 cells (Supplementary Fig. 4).

Discussion

Although there is continuous development of new strategies for breast cancer prognosis and treatment, chemotherapy resistance is frequently encountered and remains a major obstacle in the management of breast cancer. Eventually, MDR develops in most systemic recurrent or initially metastatic breast cancer patients. Overcoming MDR could significantly improve the efficacy of chemotherapy.

The aim of this study was to establish new resistant breast cancer cell lines that could aid the development of novel chemosensitizers to eventually overcome MDR and to investigate MDR mechanisms.

We established two new resistant breast cancer cell sublines, T47D/SN120 and T47D/SN150. Both sublines showed a wide variety of resistance to anticancer drugs tested in this study from huge resistance (paclitaxel and vinblastine) to low or no resistance (5-FU and methotrexate).

Generally, the levels of resistance are classified as high (RF >20x), moderate (RF 5-15x), and low or no resistance (RF <5x) [50]. Considering that common substrates for both MRPs and BCRP are SN38, irinotecan and methotrexate, and substrates for MRP only are paclitaxel, vinblastine and cisplatin and those for BCRP only are doxorubicin, topotecan, gefitinib, mitoxantrone [51,52,53], the resistance profiles of both resistant sublines to anticancer drugs seems to be closely related to the expression levels of transporters although there are some exceptions which remain to be determined.

Compared to their parental cell line, T47D/SN120 and T47D/SN150 cells displayed faster proliferation and a shorter doubling time. These features were not consistent with other resistant cell lines such as MCF-7/TAX [54], BEL-7402/5-FU [55], MCF-7/Doc, and MCF-7/Adr [36]. This may be a result of the short incubation period. Most cells follow an S-shaped pattern of growth; however, we calculated the doubling time at a time point prior to the S-shaped growth curve.

The mRNA expression profiles of the transporter genes in T47D/SN120 and T47D/SN150 cells were compared to those in the T47D/WT cell line. The T47D/WT did not express Pgp and MRP1 mRNAs, which had already been reported in another study [56]. We found that T47D/SN120 and T47D/SN150 cells overexpress MRP1, MRP2, MRP3, MRP4, and BCRP. However, we did not perform RNA sequencing although a number of splicing variants of MPR mRNA had been reported [57]. Moreover, T47D/SN150 cells obtained following exposure to a higher concentration of SN38 expressed a higher level of transporter genes associated with MDR. This suggests that MDR is related to the drug concentration used as well as overexpression of MDR-related genes.

MTT assays were performed to assess the sensitivity of SN38-resistant T47D sublines to SN38 in the presence of inhibitors (Pgp inhibitors: verapamil, PSC833, and TMF; MRP inhibitor: probenecid; BCRP inhibitor: genistein) [49]. TMF, probenecid, and genistein showed a concentration-dependent chemosensitizing effect. The MRP and BCRP expression in T47D/SN150 cells was higher than that in T47D/SN120 cells and the chemosensitizing effects of TMF and genistein were more effective in T47D/SN150 than that in T47D/SN120 cell lines. Interestingly, administration of the well-known Pgp inhibitors, verapamil and PSC833 did not sensitize the cells to SN38; however, the sensitivity was recovered upon administration of TMF. In addition, the sensitivity recovery pattern observed in SN38-resistant breast cancer cells treated with TMF was similar to that observed in SN38-resistant breast cancer cells treated with genistein. This suggests that TMF not only functions as a Pgp inhibitor, but also affects other MDR mechanisms. The inhibitory effects of TMF on non-Pgp transporters remain to be investigated.

Next, experiments were designed to identify the relationship between the expression of MRP1, MRP2, MRP3, MRP4, and BCRP and epigenetic gene silencing. Generally, alteration of gene function in cancer is attributed to either genetic alterations (such as mutations or deletions) or epigenetic alterations, which alter the gene expression status [58, 59]. Epigenetic alterations include promoter methylation and chromatin remodeling, such as histone modification without an alteration in the DNA sequence [60, 61]. Changes in gene expression caused by epigenetic alterations in cancer can be divided into three categories, transcriptional regression by methylation of the promoter CpG islands (lesions in the genome rich in sequences consisting of a cytosine preceding a guanine), increased gene expression by hypomethylation, and decreased gene expression associated with histone deacetylase (HDAC) [62,63,64]. Aberrant DNA methylation in normally unmethylated gene promoter CpG islands results in a decrease in gene expression. Hypermethylation of promoter CpG islands in tumor suppressor genes is a hallmark of all human cancers [65,66,67]. Moreover, global hypomethylation of the DNA contributes to carcinogenesis by inducing genomic instability [68,69,70]. DNA hypomethylation is associated with activation of proto-oncogenes [71]. Lastly, activated HDAC induces promoter DNA methylation and represses gene expression [58, 64, 72]. Consequently, epigenetic gene silencing occurs by DNA hypermethylation and histone deacetylation. Therefore, DNA methyltransferase inhibitors and HDAC inhibitors, which inhibit DNA methylation and histone deacetylation, induce the expression of genes that were abnormally suppressed. Thus, they have been used as therapeutic agents to treat cancer caused by epigenetic gene silencing. In this study, expression of MRP2, MRP3, MRP4, and BCRP were found to be significantly increased in T47D/WT cancer cells treated with the DNA methyltransferase inhibitor, 5-aza-2′-deoxycytidine. Similarly, treatment of T47D/WT cancer cells with the HDAC inhibitor, trichostatin A significantly increased the expression of MRP1, MRP2, MRP4, and BCRP. In other words, contrary to what occurs in cancer induced by epigenetic gene silencing, overexpression of MRP1, MRP2, MRP3, MRP4, and BCRP might be caused by DNA demethylation and histone acetylation. These results suggest that the expression of MRP1, MRP2, MRP3, MRP4, and BCRP could be induced by the suppression of epigenetic gene silencing. These findings are consistent with reports indicating that epigenetic gene silencing is involved in irinotecan sensitivity in colorectal cancer cells [73,74,75]. Intracellular irinotecan is inactivated by UGT1A1 or pumped out by transporters such as ABCB1 (Pgp), ABCC1 (MRP1), and ABCC2 (MRP2) [76, 77]. Since silencing of UGT1A1 and transporters occurs via DNA methylation, DNA methyltransferase inhibitors can restore gene expression and thereby, enhance SN38 inactivation [74]. ABC transporter gene silencing occurs through histone deacetylation [73, 75] . In this study, surprisingly, the expression of MRP3 decreased after treatment with TSA; however, the underlying mechanism remains to be elucidated and is difficult to explain because studies on the epigenetic profile of the ABC loci are still incomplete. Also, other possible mechanisms of overexpression of ABC transporters includes gene amplification and post-transcriptional regulations by miRNAs [78,79,80].

BCRP, MRP1, MRP2, MRP3, and MRP4 decreased the intracellular fluorescent dye accumulation. When the BCRP and MRP inhibitors genistein and probenecid were added as chemosensitizers, the intracellular fluorescent dye concentrations were found to be increased. However, addition of the Pgp inhibitor PSC833 did not increase the intracellular fluorescent dye accumulation, suggesting that the resistance of T47D/SN120 and T47D/SN150 cells to anti-cancer drugs is not associated with Pgp.

Conclusions

Two SN38-resistant breast cancer cell sublines, T47D/SN120 and T47D/SN150, were established by gradual exposure to SN38. The resistant cells overexpressed MRP1, MRP2, MRP3, MRP4, and BCRP, which might be due to the suppression of epigenetic gene silencing via DNA hypermethylation and histone deacetylation. These resistant cells presented classical MDR phenotype, characterized by cross-resistance to various other anti-cancer drugs. This MDR phenotype decreased intracellular drug accumulation, which was reversed by known MRP and BCRP chemosensitizers, probenecid and genistein, respectively. Thus, the both SN-38-resistant sublines overexpressing both MRP and BCRP are valuable for screening chemosensitizers inhibiting both transporters.

Availability of data and materials

The datasets used during the current study are available from the corresponding author on reasonable request.

Abbreviations

BCRP:

Breast cancer resistance protein

SN38:

7-ethyl-10-hydroxycamptothecin

MTT:

3-(4,5-methylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide

MDR:

Multidrug resistance

ABC:

ATP (adenosine triphosphate)-binding cassette

MRP1:

Multidrug resistance protein 1

MRP2:

Multidrug resistance protein 2

MRP3:

Multidrug resistance protein 3

MRP4:

Multidrug resistance protein 4

LRP:

Lung resistance protein

RF:

Resistance factor

RT-qPCR:

Reverse transcription-quantitative polymerase chain reaction

PBS:

Phosphate-buffered saline

TMF:

7,3′,4-trimethoxyflavone

Pgp:

P-glycoprotein

TSA:

Trichostatin A

HDAC:

Histone deacetylase

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  PubMed  Google Scholar 

  2. Fisher B, Bryant J, Wolmark N, Mamounas E, Brown A, Fisher ER, et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol. 1998;16(8):2672–85.

    Article  CAS  PubMed  Google Scholar 

  3. Bear HD, Anderson S, Brown A, Smith R, Mamounas EP, Fisher B, et al. The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: preliminary results from National Surgical Adjuvant Breast and bowel project protocol B-27. J Clin Oncol. 2003;21(22):4165–74.

    Article  CAS  PubMed  Google Scholar 

  4. Early breast Cancer Trialists’ collaborative G. Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018;19(1):27–39.

    Article  Google Scholar 

  5. Heys SD, Sarkar T, Hutcheon AW. Primary docetaxel chemotherapy in patients with breast cancer: impact on response and survival. Breast Cancer Res Treat. 2005;90(2):169–85.

    Article  CAS  PubMed  Google Scholar 

  6. Hutcheon AW, Heys SD, Sarkar TK, Aberdeen BG. Neoadjuvant docetaxel in locally advanced breast cancer. Breast Cancer Res Treat. 2003;79(Suppl 1):S19–24.

    Article  CAS  PubMed  Google Scholar 

  7. Blohmer JU, Schmid P, Hilfrich J, Friese K, Kleine-Tebbe A, Koelbl H, et al. Epirubicin and cyclophosphamide versus epirubicin and docetaxel as first-line therapy for women with metastatic breast cancer: final results of a randomised phase III trial. Ann Oncol. 2010;21(7):1430–5.

    Article  PubMed  Google Scholar 

  8. Nabholtz J-M, Falkson C, Campos D, Szanto J, Martin M, Chan S, et al. Docetaxel and doxorubicin compared with doxorubicin and cyclophosphamide as first-line chemotherapy for metastatic breast Cancer: results of a randomized, multicenter, phase III trial. J Clin Oncol. 2003;21(6):968–75.

    Article  CAS  PubMed  Google Scholar 

  9. Zielinski C, Beslija S, Mrsic-Krmpotic Z, Welnicka-Jaskiewicz M, Wiltschke C, Kahan Z, et al. Gemcitabine, Epirubicin, and paclitaxel versus fluorouracil, Epirubicin, and cyclophosphamide as first-line chemotherapy in metastatic breast Cancer: a central European cooperative oncology group international, multicenter, prospective, randomized phase III trial. J Clin Oncol. 2005;23(7):1401–8.

    Article  CAS  PubMed  Google Scholar 

  10. Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Snyder N, et al. Drug resistance in cancer: an overview. Cancers (Basel). 2014;6(3):1769–92.

    Article  Google Scholar 

  11. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5(3):219–34.

    Article  CAS  PubMed  Google Scholar 

  12. Dallavalle S, Dobricic V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, et al. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat. 2020;50:100682.

    Article  PubMed  Google Scholar 

  13. Rosenberg MF, Callaghan R, Ford RC, Higgins CF. Structure of the multidrug resistance P-glycoprotein to 2.5 nm resolution determined by electron microscopy and image analysis. J Biol Chem. 1997;272(16):10685–94.

    Article  CAS  PubMed  Google Scholar 

  14. Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem. 1993;62:385–427.

    Article  CAS  PubMed  Google Scholar 

  15. Ferry DR, Russell MA, Cullen MH. P-glycoprotein possesses a 1,4-dihydropyridine-selective drug acceptor site which is alloserically coupled to a vinca-alkaloid-selective binding site. Biochem Biophys Res Commun. 1992;188(1):440–5.

    Article  CAS  PubMed  Google Scholar 

  16. Riordan JR, Ling V. Genetic and biochemical characterization of multidrug resistance. Pharmacol Ther. 1985;28(1):51–75.

    Article  CAS  PubMed  Google Scholar 

  17. Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92(16):1295–302.

    Article  CAS  PubMed  Google Scholar 

  18. Muller M, de Vries EG, Jansen PL. Role of multidrug resistance protein (MRP) in glutathione S-conjugate transport in mammalian cells. J Hepatol. 1996;24(Suppl 1):100–8.

    CAS  PubMed  Google Scholar 

  19. Allikmets R, Schriml LM, Hutchinson A, Romano-Spica V, Dean M. A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res. 1998;58(23):5337–9.

    CAS  PubMed  Google Scholar 

  20. Natarajan K, Xie Y, Baer MR, Ross DD. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol. 2012;83(8):1084–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li W, Zhang H, Assaraf YG, Zhao K, Xu X, Xie J, et al. Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies. Drug Resist Updat. 2016;27:14–29.

    Article  CAS  PubMed  Google Scholar 

  22. Lage H, Dietel M. Effect of the breast-cancer resistance protein on atypical multidrug resistance. Lancet Oncol. 2000;1:169–75.

    Article  CAS  PubMed  Google Scholar 

  23. Larsen AK, Escargueil AE, Skladanowski A. Resistance mechanisms associated with altered intracellular distribution of anticancer agents. Pharmacol Ther. 2000;85(3):217–29.

    Article  CAS  PubMed  Google Scholar 

  24. Dalton WS, Scheper RJ. Lung resistance-related protein: determining its role in multidrug resistance. J Natl Cancer Inst. 1999;91(19):1604–5.

    Article  CAS  PubMed  Google Scholar 

  25. Scheffer GL, Schroeijers AB, Izquierdo MA, Wiemer EA, Scheper RJ. Lung resistance-related protein/major vault protein and vaults in multidrug-resistant cancer. Curr Opin Oncol. 2000;12(6):550–6.

    Article  CAS  PubMed  Google Scholar 

  26. Chen YL, Yang TY, Wu CL, Chen KC, Hsu SL, Hsueh CM. Mechanisms underlying lung resistance-related protein (LRP)-mediated doxorubicin resistance of non-small cell lung cancer cells. Chin J Physiol. 2016;59(6):331–47.

    Article  CAS  PubMed  Google Scholar 

  27. Wtorek K, Dlugosz A, Janecka A. Drug resistance in topoisomerase-targeting therapy. Postep Hig Med Dosw. 2018;72:1073–83.

    Article  Google Scholar 

  28. Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006;6(10):789–802.

    Article  CAS  PubMed  Google Scholar 

  29. Chrencik JE, Staker BL, Burgin AB, Pourquier P, Pommier Y, Stewart L, et al. Mechanisms of camptothecin resistance by human topoisomerase I mutations. J Mol Biol. 2004;339(4):773–84.

    Article  CAS  PubMed  Google Scholar 

  30. Gongora C, Vezzio-Vie N, Tuduri S, Denis V, Causse A, Auzanneau C, et al. New topoisomerase I mutations are associated with resistance to camptothecin. Mol Cancer. 2011;10:64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ganapathi RN, Ganapathi MK. Mechanisms regulating resistance to inhibitors of topoisomerase II. Front Pharmacol. 2013;4:89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Ji X, Lu Y, Tian H, Meng X, Wei M, Cho WC. Chemoresistance mechanisms of breast cancer and their countermeasures. Biomed Pharmacother. 2019;114:108800.

    Article  CAS  PubMed  Google Scholar 

  33. Burger H, Foekens JA, Look MP, Meijer-van Gelder ME, Klijn JG, Wiemer EA, et al. RNA expression of breast cancer resistance protein, lung resistance-related protein, multidrug resistance-associated proteins 1 and 2, and multidrug resistance gene 1 in breast cancer: correlation with chemotherapeutic response. Clin Cancer Res. 2003;9(2):827–36.

    CAS  PubMed  Google Scholar 

  34. Wang M, Wang X, Yuan J, Guo L. Expression of the breast cancer resistance protein and 5fluorouracil resistance in clinical breast cancer tissue specimens. Mol Clin Oncol. 2013;1(5):853–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Maliepaard M, Scheffer GL, Faneyte IF, van Gastelen MA, Pijnenborg AC, Schinkel AH, et al. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res. 2001;61(8):3458–64.

    CAS  PubMed  Google Scholar 

  36. Li WJ, Zhong SL, Wu YJ, Xu WD, Xu JJ, Tang JH, et al. Systematic expression analysis of genes related to multidrug-resistance in isogenic docetaxel- and adriamycin-resistant breast cancer cell lines. Mol Biol Rep. 2013;40(11):6143–50.

    Article  CAS  PubMed  Google Scholar 

  37. Doyle LA, Yang. W, Abruzzo LV, Krogmann T, Gao Y, rishi AK, et al. a multidrug resistance transporter from human MCF-7 breast cancer cells. [erratum appears in proc Natl Acad Sci U S a 1999 mar 2;96(5):2569]. Proc Natl Acad Sci U S A. 1998;95(26):15665–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu F, Fan D, Qi J, Zhu H, Zhou Y, Yang C, et al. Co-expression of cytokeratin 8 and breast cancer resistant protein indicates a multifactorial drug-resistant phenotype in human breast cancer cell line. Life Sci. 2008;83(13):496–501.

    Article  CAS  PubMed  Google Scholar 

  39. Robey RW, Medina-Perez WY, Nishiyama K, Lahusen T, Miyake K, Litman T, et al. Overexpression of the ATP-binding cassette half-transporter, ABCG2 (Mxr/BCrp/ABCP1), in flavopiridol-resistant human breast cancer cells. Clin Cancer Res. 2001;7(1):145–52.

    CAS  PubMed  Google Scholar 

  40. Schellens JH, Maliepaard M, Scheper RJ, Scheffer GL, Jonker JW, Smit JW, et al. Transport of topoisomerase I inhibitors by the breast cancer resistance protein. Potential clinical implications. Ann N Y Acad Sci. 2000;922:188–94.

    Article  CAS  PubMed  Google Scholar 

  41. Beretta GL, Perego P, Zunino F. Mechanisms of cellular resistance to camptothecins. Curr Med Chem. 2006;13(27):3291–305.

    Article  CAS  PubMed  Google Scholar 

  42. Zander SA, Sol W, Greenberger L, Zhang Y, van Tellingen O, Jonkers J, et al. EZN-2208 (PEG-SN38) overcomes ABCG2-mediated topotecan resistance in BRCA1-deficient mouse mammary tumors. Plos One. 2012;7(9):e45248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kaneda N, Nagata H, Furuta T, Yokokura T. Metabolism and pharmacokinetics of the camptothecin analogue CPT-11 in the mouse. Cancer Res. 1990;50(6):1715–20.

    CAS  PubMed  Google Scholar 

  44. Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K. Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res. 1991;51(16):4187–91.

    CAS  PubMed  Google Scholar 

  45. Hsiang YH, Hertzberg R, Hecht S, Liu LF. Camptothecin induces protein-linked DNA breaks via mammalian DNA topoisomerase I. J Biol Chem. 1985;260(27):14873–8.

    Article  CAS  PubMed  Google Scholar 

  46. Engel LW, Young NA. Human breast carcinoma cells in continuous culture: a review. Cancer Res. 1978;38(11 Pt 2):4327–39.

    CAS  PubMed  Google Scholar 

  47. Jandu H, Aluzaite K, Fogh L, Thrane SW, Noer JB, Proszek J, et al. Molecular characterization of irinotecan (SN-38) resistant human breast cancer cell lines. BMC Cancer. 2016;16:34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. 1979. Biotechnology. 1992;24:145–9.

    CAS  PubMed  Google Scholar 

  49. Jeong JM, Choi CH, Kang SK, Lee IH, Lee JY, Jung H. Antioxidant and chemosensitizing effects of flavonoids with hydroxy and/or methoxy groups and structure-activity relationship. J Pharm Pharm Sci. 2007;10(4):537–46.

    Article  CAS  PubMed  Google Scholar 

  50. Snow K, Judd W. Characterisation of adriamycin- and amsacrine-resistant human leukaemic T cell lines. Br J Cancer. 1991;63(1):17–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport--an update. AAPS J. 2015;17(1):65–82.

    Article  CAS  PubMed  Google Scholar 

  52. Zhang YK, Wang YJ, Gupta P, Chen ZS. Multidrug resistance proteins (MRPs) and Cancer therapy. AAPS J. 2015;17(4):802–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang JQ, Yang Y, Cai CY, Teng QX, Cui Q, Lin J, et al. Multidrug resistance proteins (MRPs): structure, function and the overcoming of cancer multidrug resistance. Drug Resist Updat. 2021;54:100743.

    Article  PubMed  Google Scholar 

  54. Chen SY, Hu SS, Dong Q, Cai JX, Zhang WP, Sun JY, et al. Establishment of paclitaxel-resistant breast cancer cell line and nude mice models, and underlying multidrug resistance mechanisms in vitro and in vivo. Asian Pac J Cancer Prev. 2013;14(10):6135–40.

    Article  PubMed  Google Scholar 

  55. Gu W, Fang FF, Li B, Cheng BB, Ling CQ. Characterization and resistance mechanisms of a 5-fluorouracil- resistant hepatocellular carcinoma cell line. Asian Pac J Cancer Prev. 2012;13(9):4807–14.

    Article  PubMed  Google Scholar 

  56. Maas RA, Bruning PF, Breedijk AJ, Top B, Peterse HL. Immunomagnetic purification of human breast carcinoma cells allows tumor-specific detection of multidrug resistance gene 1-mRNA by reverse transcriptase polymerase chain reaction in fine-needle aspirates. Lab Investig. 1995;72(6):760–4.

    CAS  PubMed  Google Scholar 

  57. Grant CE, Kurz EU, Cole SP, Deeley RG. Analysis of the intron-exon organization of the human multidrug-resistance protein gene (MRP) and alternative splicing of its mRNA. Genomics. 1997;45(2):368–78.

    Article  CAS  PubMed  Google Scholar 

  58. Baylin SB, Esteller M, Rountree MR, Bachman KE, Schuebel K, Herman JG. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Hum Mol Genet. 2001;10(7):687–92.

    Article  CAS  PubMed  Google Scholar 

  59. Baylin SB, Jones PA. Epigenetic determinants of Cancer. Cold Spring Harb Perspect Biol. 2016;8:a019505.

  60. Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity (Edinb). 2010;105(1):4–13.

    Article  CAS  Google Scholar 

  61. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27–36.

    Article  CAS  PubMed  Google Scholar 

  62. Park JY. Promoter hypermethylation in prostate cancer. Cancer Control. 2010;17(4):245–55.

    Article  PubMed  Google Scholar 

  63. Yelina N, Diaz P, Lambing C, Henderson IR. Epigenetic control of meiotic recombination in plants. Sci China Life Sci. 2015;58(3):223–31.

    Article  CAS  PubMed  Google Scholar 

  64. Hernandez-Rosas F, Hernandez-Oliveras A, Flores-Peredo L, Rodriguez G, Zarain-Herzberg A, Caba M, et al. Histone deacetylase inhibitors induce the expression of tumor suppressor genes Per1 and Per2 in human gastric cancer cells. Oncol Lett. 2018;16(2):1981–90.

    PubMed  PubMed Central  Google Scholar 

  65. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–28.

    Article  CAS  PubMed  Google Scholar 

  66. Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP. Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res. 1998;72:141–96.

    Article  CAS  PubMed  Google Scholar 

  67. Bilgrami SM, Qureshi SA, Pervez S, Abbas F. Promoter hypermethylation of tumor suppressor genes correlates with tumor grade and invasiveness in patients with urothelial bladder cancer. Springerplus. 2014;3:178.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301(5895):89–92.

    Article  CAS  PubMed  Google Scholar 

  69. Roberts LR, Gores GJ. Hepatocellular carcinoma: molecular pathways and new therapeutic targets. Semin Liver Dis. 2005;25(2):212–25.

    Article  CAS  PubMed  Google Scholar 

  70. Lee HS, Kim BH, Cho NY, Yoo EJ, Choi M, Shin SH, et al. Prognostic implications of and relationship between CpG island hypermethylation and repetitive DNA hypomethylation in hepatocellular carcinoma. Clin Cancer Res. 2009;15(3):812–20.

    Article  CAS  PubMed  Google Scholar 

  71. Calvisi DF, Ladu S, Gorden A, Farina M, Lee JS, Conner EA, et al. Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma. J Clin Invest. 2007;117(9):2713–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Rountree MR, Bachman KE, Herman JG, Baylin SB. DNA methylation, chromatin inheritance, and cancer. Oncogene. 2001;20(24):3156–65.

    Article  CAS  PubMed  Google Scholar 

  73. Huo H, Magro PG, Pietsch EC, Patel BB, Scotto KW. Histone methyltransferase MLL1 regulates MDR1 transcription and chemoresistance. Cancer Res. 2010;70(21):8726–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Gagnon JF, Bernard O, Villeneuve L, Tetu B, Guillemette C. Irinotecan inactivation is modulated by epigenetic silencing of UGT1A1 in colon cancer. Clin Cancer Res. 2006;12(6):1850–8.

    Article  CAS  PubMed  Google Scholar 

  75. Hauswald S, Duque-Afonso J, Wagner MM, Schertl FM, Lubbert M, Peschel C, et al. Histone deacetylase inhibitors induce a very broad, pleiotropic anticancer drug resistance phenotype in acute myeloid leukemia cells by modulation of multiple ABC transporter genes. Clin Cancer Res. 2009;15(11):3705–15.

    Article  CAS  PubMed  Google Scholar 

  76. Iyer L, King CD, Whitington PF, Green MD, Roy SK, Tephly TR, et al. Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J Clin Invest. 1998;101(4):847–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Innocenti F, Kroetz DL, Schuetz E, Dolan ME, Ramirez J, Relling M, et al. Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J Clin Oncol. 2009;27(16):2604–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Bakos E, Homolya L. Portrait of multifaceted transporter, the multidrug resistance-associated protein 1 (MRP1/ABCC1). Pflugers Arch. 2007;453(5):621–41.

    Article  CAS  PubMed  Google Scholar 

  79. Gomes BC, Honrado M, Armada A, Viveiros M, Rueff J, Rodrigues AS. ABC efflux transporters and the circuitry of miRNAs: kinetics of expression in Cancer drug resistance. Int J Mol Sci. 2020;21(8):2985.

  80. Reustle A, Fisel P, Renner O, Buttner F, Winter S, Rausch S, et al. Characterization of the breast cancer resistance protein (BCRP/ABCG2) in clear cell renal cell carcinoma. Int J Cancer. 2018;143(12):3181–93.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supportive by research fund from Chosun University 2017.

Author information

Authors and Affiliations

Authors

Contributions

HJL and CHC conceived and designed the analysis, collected the data and analyzed the data. All authors were involved in the drafting, critical review, and approval of the final manuscript and the decision to submit for publication.

Corresponding author

Correspondence to Cheol-Hee Choi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, HJ., Choi, CH. Characterization of SN38-resistant T47D breast cancer cell sublines overexpressing BCRP, MRP1, MRP2, MRP3, and MRP4. BMC Cancer 22, 446 (2022). https://doi.org/10.1186/s12885-022-09446-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-022-09446-y

Keywords