Abstract
Gemcitabine is commonly used to treat various cancer types, including human non-small cell lung cancer (NSCLC). However, even cases that initially respond rapidly commonly develop acquired resistance, limiting our ability to effectively treat advanced NSCLC. To gain insight for developing a strategy to overcome gemcitabine resistance, the present study investigated the mechanism of gemcitabine resistance in NSCLC according to the involvement of ATP-binding cassette subfamily B member 6 (ABCB6) and heme biosynthesis. First, an analysis of ABCB6 expression in human NSCLCs was found to be associated with poor prognosis and gemcitabine resistance in a hypoxia-inducible factor (HIF)-1-dependent manner. Further experiments showed that activation of HIF-1α/ABCB6 signaling led to intracellular heme metabolic reprogramming and a corresponding increase in heme biosynthesis to enhance the activation and accumulation of catalase. Increased catalase levels diminished the effective levels of reactive oxygen species, thereby promoting gemcitabine-based resistance. In a mouse NSCLC model, inhibition of HIF-1α or ABCB6, in combination with gemcitabine, strongly restrained tumor proliferation, increased tumor cell apoptosis, and prolonged animal survival. These results suggest that, in combination with gemcitabine-based chemotherapy, targeting HIF-1α/ABCB6 signaling could result in enhanced tumor chemosensitivity and, thus, may improve outcomes in NSCLC patients.
Supplementary Information
The online version contains supplementary material available at 10.1007/s00018-022-04360-9.
Keywords: ABCB6, Hypoxia-inducible factor-1, Transcriptional activation, Non-small cell lung cancer, Gemcitabine resistance
Background
Lung cancer is one of the most common forms of malignancy and the leading cause of cancer-related deaths in both men and women worldwide [1]. An estimated 1.8 million people are diagnosed with lung cancer annually, and an estimated 1.6 million people die from this disease every year [2]. In China, approximately 733,300 new cases were diagnosed and 610,200 patients died of lung cancer in 2015 [3]. Non-small cell lung cancer (NSCLC), which is divided into three subtypes (large-cell carcinoma, squamous cell carcinoma, and adenocarcinoma), accounts for more than 85% of all lung cancer cases [4]. Since most cases are diagnosed at advanced stage with advanced local invasion or distant metastasis, the 5-year survival rate for lung cancer patients is only 4–17% [5]. Despite advancements in surgical treatments and targeted therapies, systemic chemotherapy remains the mainstay of treatment for advanced lung cancer. Gemcitabine, a deoxycytidine analog, exerts an anticancer effect by catalyzing the complex intracellular conversion to gemcitabine diphosphate and triphosphate, compounds that are important for terminating DNA synthesis [6]. Clinically, gemcitabine has been used in one of the standard regimens for NSCLC [7]. However, gemcitabine-based chemotherapy resistance has continued to limit our ability to effectively treat advanced NSCLC. In addition to inherent resistance to gemcitabine, even cases that initially respond to gemcitabine treatment frequently develop acquired resistance. Hence, it is critically important to determine the mechanism of gemcitabine-based resistance in NSCLC and to urgently discover an effective approach to overcome chemotherapy resistance.
The adenosine triphosphate (ATP)-binding cassette (ABC) transporter family consists of 11 members that facilitate translocation of heterogeneous substrates, including saccharides, amino acids, peptides and proteins, lipids and sterols, metabolic products, inorganic and organic ions, metals and drugs, across the cell membrane [8]. Some of the ABC transporters have been shown to play crucial roles in the development of drug resistance via the efflux of anticancer agents outside of cancer cells [9]. However, this activity cannot fully explain the underlying mechanism of acquired chemotherapy resistance. ABC transporter subfamily B member 6 (ABCB6) encodes an 842-amino-acid protein [10], and as an energy-dependent transporter, ABCB6 is capable of binding with heme and is involved in heme biosynthesis [11]. To investigate the contribution of ABCB6 to gemcitabine resistance in NSCLC cells, in the present study, we established a gemcitabine-resistant (GR) human lung squamous cancer (LUSC) call line and a GR lung adenocarcinoma (LUAD) cell line and then examined and analyzed ABCB6 expression as well as heme synthesis in these cells. We found that gemcitabine treatment alters intracellular heme biosynthesis by enhancing the levels of ABCB6, whereas inhibition of ABCB6 restrains intracellular heme accumulation. Interestingly, we also found this interaction is regulated by gemcitabine-induced hypoxia-inducible factor-1α (HIF-1α). HIF-1α, which is stably expressed in response to low oxygen conditions (hypoxia), is a transcriptional activator that regulates the expression of thousands of target genes [12]. In addition to hypoxia, stimuli such as chemotherapeutic agents and radiation increase the stabilization and accumulation of HIF-1α [13, 14]. We observed stable expression of HIF-1α in the GR lung cancer cell lines even under normoxic conditions. In addition, we demonstrate that ABCB6 is a direct HIF-1α target gene, and further identify relevant hypoxia response elements (HREs) in the ABCB6 gene.
Chemotherapeutic drug-induced cell apoptosis is associated with enhanced levels of reactive oxygen species (ROS), primarily superoxide (O2−) and hydrogen peroxide (H2O2), that can cause functional and structural damage to cell components [15]. Cancer cells, thus, protect themselves from chemotherapy by activating a variety of defense mechanisms to detoxify any adverse effects caused by an accumulation of chemotherapy-induced ROS. Catalase, an enzyme that degrades H2O2 by converting it into H2O and O2, is generally recognized as an antioxidant enzyme that confers cellular protection [16]. Heme metabolism is an important cellular mechanism that influences catalase activities. As a heme-containing enzyme, the regulation of H2O2 decomposition and peroxidase activities by catalase are heme-dependent [17]. To determine whether reprogramming of HIF-1α/ABCB6-regulated heme metabolism influences catalase activity, we analyzed catalase protein expression and activity in GR NSCLC cell lines and observed a dramatic induction of both catalase protein expression and activity, which further facilitates ROS scavenging to directly contribute to promoting gemcitabine-based resistance.
Materials and methods
Statistical analysis of microarray data and survival
Gene expression data from the LUSC and LUAD datasets of The Cancer Genome Atlas (TCGA) were obtained from http://ualcan.path.uab.edu/index.html or http://xena.ucsc.edu. Pearson’s correlation analysis was used to determine P values for co-expression. The prognostic significance of ABCB6 expression in lung cancer was analyzed using Kmplot (www.kmplot.com) [18]. Briefly, the gene was entered into the online database to obtain a Kaplan–Meier survival plot. The log-rank P values were calculated and displayed on the webpage. The survival rates were investigated according to a median cutoff (low represents patients with indicated gene mRNA levels less than the median, and high represents patients with indicated gene mRNA levels greater than the median).
Patients and clinical specimens
Patients diagnosed with advanced NSCLC were enrolled in the cancer center of the First Affiliated Hospital (Southwest Hospital) of Army Medical University (Chongqing, China) from January 2010 to December 2014. All patients received routine gemcitabine-platinum chemotherapy (completed 2–4 cycles). Forty-one specimens were obtained from patients with chemotherapy failure (including progressive disease, PD) after gemcitabine-based chemotherapy, and constituted the GR group. Forty specimens were obtained from patients who had a positive response after gemcitabine therapy including a partial response (PR), and these samples constituted the gemcitabine-sensitive group. Adjacent normal specimens were collected from 10 NSCLC cancer patients who underwent surgery in Southwest Hospital. The histomorphology of all specimens had been confirmed by the Department of Pathology. This clinical study complied with the ethical standards codified in the 1964 Declaration of Helsinki. The protocol for immunohistochemistry (IHC) examination of patient tissues was approved by the Ethics Committee of the First Affiliated Hospital (Southwest Hospital), Army Medical University, and all patients or family members involved provided written informed consent. All slides were prepared from stored pretreated, paraffin-embedded tissue blocks from lung cancer patients who underwent surgery at Southwest Hospital. All specimens were confirmed by pathological examination, and tissue microarrays were prepared as described previously [19].
Cell culture and drug treatment
All human NSCLC cell lines were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in Dulbecco’s Modified Eagle Medium (DMEM, A549 cells) or RPMI 1640 medium (NCI-H1703 cells) supplemented with 10% fetal bovine serum (Gibco), penicillin (100 U/ml), and streptomycin (0.1 mg/ml) (Beyotime Institute of Biotechnology, China). Cells were cultured in an incubator at 37 °C in a humidified atmosphere containing 5% CO2. The GR cell lines (A549-GR and H1703-GR) were established as follows: A549 or H1703 parental cells were exposed to gradually increasing concentrations of gemcitabine (Sigma-Aldrich, St. Louis, MO, USA) from 10 nM initially up to 10 μM over a 7-month period. For hypoxia exposure, cells were placed in a modular incubator chamber that was flushed with a 1% O2/5% CO2/94% N2 gas mixture and sealed. Dimethyloxalylglycine (DMOG, Sigma-Aldrich) was initially dissolved in dimethyl sulfoxide (DMSO) and subsequently diluted in the treatment appropriate culture medium to the indicated concentrations. For the vehicle control group, a final concentration of 1% (v/v) aqueous DMSO was used throughout. Tomatidine was purchased from MedChemExpress (purity: > 98.0% Cat No. HY-N2149, Shanghai, China), and fresh solutions were prepared by further dilution of a stock solution (100 mmol/l in DMSO). Tomatidine was added to cell culture wells to reach the final various working concentrations (0, 2, 6, 8, and 10 μmol/l). Cells were incubated for 24 h before subsequent experiments.
Lentivirus transduction
The pLKO.1-puro lentiviral vectors encoding short hairpin (sh)RNA targeting HIF-1α (sh1α-1, Clone ID:NM_001530.x-2671s1c1; sh1α-2, Clone ID:NM_001530.x-1048s1c1), ABCB6 (shABCB6-1, Clone ID:NM_005689.1-1581s1c1; shABCB6-2, Clone ID:NM_005689.1-2814s21c1), and non-targeting control (NTC) were purchased from Sigma-Aldrich. Lentiviruses were packaged, and cells were transduced and subjected to puromycin selection as described previously [20].
Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis
Total cellular RNA was extracted using TRIzol (Invitrogen), precipitated with isopropanol, treated with DNase I (Ambion), and reverse transcribed with the iScript cDNA Synthesis kit (Bio-Rad). qPCR analysis was performed using SYBR Green (Bio-Rad) with the iCycler Real-time PCR Detection System (Bio-Rad). The 2−ΔΔCt method was used to calculate relative gene expression levels as described previously [21]. The primer sequences used in this study are listed in Table S1.
Chromatin immunoprecipitation (ChIP) assay
ChIP assays were performed as previously described [19]. Briefly, A549 and H1703 cells were cross-linked in 3.7% formaldehyde for 10 min and lysed with sodium dodecyl sulfate (SDS) lysis buffer. Chromatin was sheared by sonication, and lysates were pre-cleared with salmon sperm DNA/protein A-agarose slurry (Millipore) and incubated with IgG (Novus Biologicals) or antibodies against the following proteins: anti-HIF-1α (Santa Cruz Biotechnology), anti-HIF-2α (Cell Signaling Technology), anti-HIF-1β (Novus Biologicals), anti-H3K27ac (GeneTex) and anti-H3K4me3 (GeneTex). Salmon sperm DNA/protein A-agarose slurry was added, and the agarose beads were washed sequentially with low- and high-salt immune complex wash buffers; LiCl immune complex wash buffer; and twice with Tris–EDTA (TE) buffer (10 mM Tris–HCl/1 mM EDTA). DNA was eluted in 1% SDS with 0.1 M NaHCO3, and crosslinks were reversed by addition of 0.2 M NaCl. DNA was purified by phenol–chloroform extraction and ethanol precipitation and then suspended in 50-μl TE buffer. A 2-μl aliquot was used for qPCR with the primer sequences listed in Table S1.
Western blot assay
Immunoblot assays were performed as previously described [20]. Aliquots of whole-cell lysates were prepared in radioimmunoprecipitation assay (RIPA) lysis buffer, and proteins were separated by SDS–polyacrylamide gel electrophoresis (PAGE), blotted onto nitrocellulose membranes, and further probed with primary antibodies followed by horseradish peroxidase (HRP)-conjugated secondary antibodies (GE Healthcare). The chemiluminescent signal was developed using ECL Plus (GE Healthcare). Antibodies used in immunoblot assays included: anti-HIF-1α (BD Transduction Laboratory); anti-ABCB6 (Novus Biologicals); anti-catalase (Novus Biologicals); and anti-β-actin (Santa Cruz Biotechnology).
Heme detection
The heme content in whole cells was determined by the fluorometric method [22]. Briefly, cells were washed and homogenized in phosphate-buffered saline (PBS) buffer, and the protein content was determined using the Bio-Rad protein assay system (Bio-Rad, Munich, Germany). For heme detection, 10-μg protein samples were incubated with 500 μl of 2 M oxalic acid (Sigma-Aldrich) at 100 °C for 30 min. Samples were subsequently centrifuged at 18,000g for 10 min. The amount of heme in the supernatant was determined by fluorometric detection using a F4500 fluorescence spectrophotometer (Hitachi) with the excitation and emission wavelengths were set at 405 and 662 nm, respectively. The background was evaluated by measuring fluorescence in non-boiled samples.
Catalase activity
For the detection of catalase activity, cells were seeded into 6-well plates at a density of 105 cells/well and then later harvested and homogenized in 50 ml of 0.1 M PBS buffer. The mixture was centrifuged at 800g for 5 min at 4 °C, and the supernatant was separated and assayed for protein content (Bradford assay, Bio-Rad). Catalase activity was determined calorimetrically in 10 μg protein samples using the Catalase Assay Kit (No. CAT-100, Sigma-Aldrich) according to the manufacturer’s instructions by spectrophotometry. Briefly, all working solutions were prepared from the reagents provided in the kit. The sample (10 µl), mixed with 750 µl of 1X assay buffer and 25 µl colorimetric assay substrate solution, was incubated for 5 min. The reaction was stopped using 900 µl of stop solution, and the tubes were kept inverted. Within 15 min after the enzymatic reaction, 10 µl aliquots of the reaction mixture were transferred to 1 ml of the color reagent. After incubation for 15 min, the absorbance of the reaction mixture was measured at 520 nm. Specific catalase activity is reported as μmol H2O2 consumed min−1 μg protein−1.
Colony formation assay
For detection of the clonogenic ability of the cells after drug treatment, cells were trypsinized and seeded into 6-well plates at a density of 1000 cells per well. The cells were then cultured in medium containing gemcitabine at the indicated concentrations, which was refreshed every 3 days. After 15 days, cells were fixed with 4% paraformaldehyde for 10 min and then stained with 0.1% crystal violet in ddH2O for 10 min.
Cell viability assay
To evaluate cell viability, 1 × 104 cells/well were seeded into 96-well plates and cultured at 37 °C overnight. Cells were then treated with various concentrations (0.001, 0.01, 0.1, 1, 10, or 100 μM) of gemcitabine for 72 h. Cell viability was determined using the CellTiter 96 AQueous One Solution Cell Proliferation Assay (MTS [3-(4,5-dimethylthiazol-2-yl)-5(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium]; Promega, USA) following the manufacturer’s instructions. All experiments were done in triplicate and repeated more than three times.
Soft agar assay
Cells were suspended in 1 ml top agar made of 0.35% agarose in medium supplemented with 10% FBS, and the mixture was seeded in 6-well plates containing a basal layer of 0.6% agarose at 5000 cells/well. After 3 weeks, colonies were stained with 0.05% crystal violet for 1 h, and the colony size and colony number were measured by pictomicrography. Viable colonies larger than 0.1 mm in diameter were counted.
TUNEL assay
To evaluate cell apoptosis in tumor tissues, we performed terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-digoxigenin nick-end labeling (TUNEL) on formalin-fixed tumor samples in paraffin blocks using the one-step TUNEL apoptosis assay kit (Beyotime Institute of Biotechnology). Tissue sections (4–5 μm) mounted on glass slides were deparaffinized, rehydrated with a graded series of alcohol solutions, treated with 20 μg/ml proteinase K (37 °C, 20 min), and then washed in PBS buffer. The TUNEL assay was performed according to the manufacturer’s instructions. Images of stained tissue sections were acquired using a confocal microscope (Leica TSC-SP5, Germany). Ten fields were randomly selected on each tumor slide.
ROS detection
Total intracellular ROS was determined by staining cells with dichlorofluorescin diacetate (DCFH-DA, Beyotime Institute of Biotechnology) following the manufacturer’s instruction. Briefly, cells were washed with PBS and incubated in darkness with 10 μM DCFH-DA at 37 °C for 30 min. As a positive control, cells were treated with Rosup (agent provided in ROS assay kit). Cells were then washed twice with PBS, and ROS levels were determined by fluorospectrophotometry (excitation wavelength 488 nm and emission wavelength 535 nm).
IHC staining
Tumors and adjacent normal tissue were fixed in 10% formalin and paraffin embedded. Tissue microarrays were dewaxed in xylene and hydrated with graded ethanol solutions, followed by antigen retrieval using citrate buffer (pH 6.1). The instantaneous SP supersensitive kit (SP-9000, Beijing Zhongshan Jinqiao Biotechnology) was used with antibodies against ABCB6 (Novus Biologicals). Sections were counterstained with Mayer’s hematoxylin (Sigma). For quantitative measurement of ABCB6-positive staining in clinical samples (adjacent normal lung tissue, GR tumors and gemcitabine-sensitive tumors), ICH staining of ABCB6 was quantified using ImageJ software (NIH) as described previously [23]. Specifically, five random fields on each sample (magnification, 200 ×) were selected for scoring, and a mean score for each slide was calculated in the final analysis. The percentage of stained cells was scored as follows: 0 (no positive cells), 1 (< 10% positive cells), 2 (10–40% positive cells), 3 (40–70% positive cells), and 4 (> 70% positive cells). The intensity of positive staining was scored as follows: 0 (negative staining), 1 (weak staining exhibited as light yellow), 2 (moderate staining exhibited as yellow brown), and 3 (strong staining exhibited as brown). The proportion and intensity scores were added to obtain a total score (range 0, 2–7). Samples were divided into two categories depending on the IHC score: low if < 4 and high if ≥ 4. Slides were examined and scored independently using the Gynecologic Oncology Group (GOG) criteria [24] by two histopathologists (F Wu and Y Ling) who were blinded to other pathological information.
Animal studies
The animal experiments were approved by the Institutional Animal Care and Use Committee of the Army Medical University (Chongqing, China) and were performed in accordance with the guidelines for animal experiments of the university, which meet the ethical guidelines for experimental animals in China. Six-to-eight-week-old male severe combined immunodeficiency (SCID) mice were purchased from the Institute of Experimental Animal of the Army Medical University. For transfer into the mice, A549 and H1703 GR and gemcitabine-sensitive subclones were harvested by trypsinization and resuspended at 107 cells/ml in a 50:50 mix of PBS:Matrigel (BD Biosciences) for subcutaneous injection of 2 × 106 cells into the inguinal area of mice (16 mice per group). The following groups were prepared: NTC-transfected A549 or H1703 wild-type (WT) group (treated with saline), NTC-transfected A549 or H1703 GR group (treated with saline), NTC-transfected A549 or H1703 GR group (treated with gemcitabine), shHIF-1α expression vector-transfected A549 or H1703 GR group (treated with gemcitabine), shABCB6 expression vector-transfected A549 or H1703 GR group (treated with gemcitabine), and shHIF-1α/ABCB6 expression vector-transfected A549 or H1703 GR group (treated with gemcitabine). Tumor volume (mm3) was calculated as 0.52 × L × W × T (L: Length, W: width, T: thickness). Tumor volumes and body weights were monitored twice weekly. After palpable tumors had formed (8 days after tumor implantation), mice received intraperitoneal injections of saline (250 µl) or gemcitabine (20 mg/kg) twice per week. After 42 days, tumors were excised and weighed (n = 6). Xenografts were harvested for analysis by TUNEL assay. To assess survival, the remaining animals (n = 10) were monitored for 100 days until being euthanized. The distribution of survival percentages over time was estimated using the Kaplan–Meier method. The log-rank test was used to determine P values. For the WT A549 animal experiment, 2 × 106 cells of A549 subclones (NTC, shHIF-1α, or shABCB6) were injected subcutaneously into the inguinal area of mice (n = 5 mice per group). Tumor volumes were monitored twice weekly. After palpable tumors had formed (26 days after tumor implantation), mice received intraperitoneal injection of gemcitabine (20 mg/kg) or saline (250 µl) twice per week until day 53.
Statistical analysis
All data are expressed as mean ± standard error of the mean (SEM). Differences between two groups were analyzed using an unpaired two-tailed Student’s t test, and differences among multiple groups were detected by analysis of variance (ANOVA) followed by Bonferroni post-test. Survival data were analyzed using Kaplan–Meier survival plots, and P values were calculated using the Wilcoxon rank sum test. A P value of < 0.05 was considered statistically significant.
Results
ABCB6 expression is increased after chemotherapy in GR lung cancer tissues and is associated with poor prognosis in human NSCLC
To investigate whether ABCB6 expression has clinical significance in human NSCLCs, we compared ABCB6 gene expression in LUSC and LUAD tissues to that in normal lung tissue using data from TCGA database. This analysis revealed that ABCB6 mRNA levels were significantly greater in human NSCLC tissues (LUSC and LUAD) than in the normal lung tissues (Fig. 1A, B). Moreover, analysis of ABCB6 mRNA expression in 1926 human lung cancer specimens [19, 25] revealed that levels above the median were associated with a significant decrease in patient survival (Fig. 1C). To evaluate whether the expression of ABCB6 in human NSCLC tissue differed according to varying gemcitabine-based chemotherapy sensitivity, we performed ICH staining to examine the expression of ABCB6 in the GR tumor tissues of 41 patients, gemcitabine-sensitive tumor tissues of 40 patients, and adjacent normal lung tissues of 6 individuals. Representative images of ABCB6 expression in GR tumor tissues, gemcitabine-sensitive tumor tissues, and adjacent normal lung tissues are shown in Fig. 1D. Quantification of the staining density of ABCB6 in multiple samples indicated that ABCB6 expression was increased by more than twofold in GR tumors compared with gemcitabine-sensitive tumors (Fig. 1E). However, no positive or weak staining was seen in the adjacent normal lung tissues (Fig. 1D, E). The demographic and clinical characteristics of the patients are summarized in Table 1. We found no correlations between ABCB6 expression and patient’s age, gender, histology, differentiation, or clinical stage (all P > 0.05; Table 1). Interestingly, a high level of ABCB6 staining was significantly correlated with a negative response to gemcitabine treatment, including progressive disease (PD; P < 0.001) as well as with distant metastasis (P < 0.001; Table 1). Moreover, the level of ABCB6 staining was positively correlated with gemcitabine resistance in NSCLCs (P < 0.001; Table 2).
Table 1.
Variables | Total | Number of patients | Chi-square test | |
---|---|---|---|---|
Low ABCB6 expression | High ABCB6 expression | P value | ||
Age (year) | ||||
< 60 | 43 | 26 | 17 | n.s |
≥ 60 | 38 | 14 | 24 | |
Gender | ||||
Male | 59 | 30 | 29 | n.s |
Female | 22 | 10 | 12 | |
Grade | ||||
Well and moderately differentiated | 49 | 22 | 27 | n.s |
Poorly and non-differentiated | 32 | 18 | 14 | |
Cell type | ||||
Squamous | 66 | 34 | 32 | n.s |
Adenocarcinoma | 15 | 6 | 9 | |
Clinical stage | ||||
Early stage (II) | 33 | 22 | 11 | n.s |
Advanced stage (III–IV) | 48 | 18 | 30 | |
cT stage | ||||
cT1–2 | 40 | 24 | 16 | n.s |
cT3–4 | 41 | 16 | 25 | |
cN stage | ||||
cN0 | 31 | 18 | 13 | n.s |
cN + | 50 | 22 | 28 | |
cM stage | ||||
cM0 | 45 | 31 | 14 | *** |
cM1 | 36 | 9 | 27 | |
Gemcitabine response | ||||
PR | 34 | 25 | 9 | *** |
SD | 6 | 4 | 2 | |
PD | 41 | 11 | 30 |
Median values were used as cutoff points for definition of age subgroups
PD progressive disease, SD stable disease, PR partial response, n.s not significant
a, Pearson Chi-square test; b, Fisher’s exact test
Pearson Chi-square test was used to calculate P values, ***P < 0.001
Table 2.
ABCB6 staining | Total | P value | ||||
---|---|---|---|---|---|---|
0–3 | 4–6 | 7–9 | 10–12 | (n = 81) | ||
Gemcitabine-sensitive | 11 | 18 | 7 | 4 | 40 | *** |
Gemcitabine-resistant | 2 | 9 | 20 | 10 | 41 |
Two-sided ANOVA with Bonferroni post hoc t test correction was used to calculate the P value, ***P < 0.001
Establishment of GR NSCLC cell lines
To address the mechanism of gemcitabine resistance, we generated lung cancer cell line (A549 and NCI-H1703) models with acquired gemcitabine resistance. For this, the WT A549 and NCI-H1703 lung cancer cell lines were cultured with increasing concentrations of gemcitabine over a period of approximately 7 months. To determine the differences in resistant phenotypes between gemcitabine-treated cells and WT cells, we first measured the viability of each line after exposure to different concentrations of gemcitabine (0.001–100 μM). Results from the MTS colorimetric assay demonstrated that A549-GR cells were more resistant than A549-WT cells to increasing gemcitabine concentrations (Fig. S1A). Similar results for H1703 cells were obtained from cell viability assays (Fig. S1B). We also observed an increase in the clonogenic ability of the A549-GR cells (Fig. S1C–S1D) and H1703-GR cells (Fig. S1E–S1F) under control and gemcitabine treatment as well as increased colony number and size in soft agar assays. These data indicate that A549-GR and H1703-GR cells had an increased capability for chemoresistance to gemcitabine treatment.
ABCB6 expression is induced in GR lung cancer cells in a HIF-1-dependent manner
We next investigated ABCB6 gene and protein expression in GR lung cancer cell lines by performing RT-qPCR and immunoblot analyses, respectively. We observed that A549-GR and H1703-GR cells both expressed significantly elevated levels of ABCB6 mRNA (Fig. 2A, B) and protein (Fig. 2C) compared with the respective WT cells, indicating that ABCB6 may participate in gemcitabine-induced chemotherapy resistance in lung cancers. We also performed qPCR assays to detect altered expression of other genes in the ABC family in the GR cells. As shown in Supplemental Fig. 1, the mRNA expression levels of ABCA1, ABCB1, and ABCG2 also were increased in GR cells as compared to gemcitabine-sensitive cells (Fig. S2A, S2C and S2I). Interestingly, ABCA1, ABCB1, and ABCG2 are all regulated by hypoxia and HIFs [26–29]. It is possible that ABCB6 is also regulated by HIF-1α. We tested this hypothesis by qPCR analysis and found that similar to ABCA1, ABCB1, and ABCG2 expression, ABCB6 mRNA expression was increased remarkably in hypoxic cells compared to normoxic cells (Fig. S2A, S2C, S2F, S2I). HIF-1α is a transcription factor that serves as a master regulator of cellular responses to hypoxia and is always associated with aggressive phenotypes, including resistance to radiation and chemotherapy, metastasis, and poor patient prognosis [30]. Interestingly, chemotherapy induces HIF-1α expression even under normoxic conditions [31]. We observed that HIF-1α expression was induced in A549-GR and H1703-GR cells under normoxia (Fig. 2C), indicating that HIF-1α is participate in acquired gemcitabine resistance.
We next hypothesized that gemcitabine-induced ABCB6 expression is regulated by the transcription factor HIF-1α in GR lung cancer cells. To test this hypothesis, the gene expression data from 1267 human LUSC specimens and 706 LUAD specimens in TCGA database were analyzed to compare the expression levels of ABCB6 mRNA with a HIF metagene signature based on the expression of LDHA, PLOD1, PLOD2, P4HA1, P4HA2, SLC16A3, SLC2A1, ANGPTL4, PDK1, L1CAM, CA9, BNIP3, IGFBP1, PKM2, and MET mRNA, which are all HIF-regulated genes. Statistical analysis of the expression levels of individual genes in the HIF signature revealed that ABCB6 expression was significantly correlated with the expression levels of 12 of the 15 HIF target genes in the LUSC and LUAD databases, respectively (Tables 3, 4). These findings indicate that ABCB6 mRNA expression may be HIF-regulated in human NSCLCs. To confirm this possibility, we measured ABCB6 expression in A549 and H1703 (including WT and GR cells) subclones, which were stably transfected with an expression vector encoding shRNA targeting HIF-1α (sh1α-1 and sh1α-2) or a NTC shRNA. We observed that the induction of ABCB6 mRNA expression was significantly decreased in A549 (Fig. 2D) and H1703 subclones (Fig. 2E) when HIF-1α was silenced. Induction of ABCB6 protein expression also was abrogated in A549 (Fig. 2F) and H1703 subclones (Fig. 2G) transfected with two different shRNAs (sh1α-1 and sh1α-2) targeting HIF-1α. It is well accepted that HIF-1α accumulates and is activated under hypoxia. In addition, DMOG, a small molecule inhibitor of the HIF prolyl hydroxylases, can also be used to mimic hypoxia in vitro. We next evaluated HIF-1α and ABCB6 protein expression in WT A549 or H1703 cells exposed to hypoxic conditions (1% O2 for 24 h or 48 h) or treated with varying concentrations (0 µM, DMSO vehicle, 100 µM, 500 µM, 1 mM) of DMOG under normoxia for 24 h. As shown in Fig. 3H, I hypoxia or DMOG treatment stabilized HIF-1α expression in a dose- and time-dependent manner. The ABCB6 protein levels in A549 (Fig. 2H) and H1703 (Fig. 2I) cells were upregulated with the increase in HIF-1α expression under hypoxia and DMOG treatment. Taken together, the data presented in Figs. 1 and 2 indicate that ABCB6 expression is induced in GR lung cancer cells in a HIF-1α-dependent manner.
Table 3.
Category | GENE | ABCB6 | CA9 | LDHA | PDK1 |
---|---|---|---|---|---|
HIF targets | P4HA1 | *** | *** | *** | *** |
LDHA | *** | *** | – | *** | |
PLOD2 | *** | *** | *** | *** | |
SLC16A3 | *** | *** | *** | n.s | |
SLC2A1 | *** | *** | *** | *** | |
ANGPTL4 | n.s | *** | *** | n.s | |
PDK1 | *** | *** | *** | – | |
L1CAM | n.s | ** | *** | n.s | |
CA9 | *** | – | *** | *** | |
BNIP3 | *** | *** | *** | *** | |
IGFBP1 | *** | n.s | *** | n.s | |
PKM2 | *** | *** | *** | *** | |
MET | * | * | ** | n.s | |
PLOD1 | n.s | *** | *** | *** | |
P4HA2 | *** | ** | *** | n.s | |
ABCB6 | – | *** | *** | *** |
Positive correlation *P < 0.05, **P < 0.01, ***P < 0.001, n.s, not significant
Table 4.
Category | GENE | ABCB6 | CA9 | LDHA | PDK1 |
---|---|---|---|---|---|
HIF targets | P4HA1 | *** | *** | *** | *** |
LDHA | *** | *** | – | *** | |
PLOD2 | *** | *** | *** | *** | |
SLC16A3 | * | *** | *** | *** | |
SLC2A1 | *** | *** | *** | *** | |
ANGPTL4 | n.s | *** | *** | *** | |
PDK1 | *** | *** | *** | – | |
L1CAM | * | *** | n.s | *** | |
CA9 | ** | – | *** | *** | |
BNIP3 | *** | n.s | *** | *** | |
IGFBP1 | *** | n.s | *** | *** | |
PKM2 | *** | *** | *** | *** | |
MET | ### | n.s | ** | n.s | |
PLOD1 | * | *** | *** | *** | |
P4HA2 | ## | *** | *** | *** | |
ABCB6 | – | ** | *** | *** |
Positive correlation *P < 0.05, **P < 0.01, ***P < 0.001, negative correlation ##P < 0.01, ###P < 0.001, n.s, not significant
Identification of HREs in the human ABCB6 gene
HIF proteins bind with co-factors to HREs in the promoter regions of their target genes to coordinate a wide-ranging transcriptional program in response to hypoxia [32, 33] as well as to other stimuli such as radiation and chemotherapy [34]. We hypothesized that ABCB6 is a direct HIF target gene. To test this hypothesis, we analyzed the ABCB6 locus for the presence of matches to the consensus HIF-binding site sequence (5′-RCGTG-3′) located in the DNase I-hypersensitive chromatin domains using the UCSC Genome Bioinformatics database. This analysis identified two candidate sites that met these criteria (Fig. S3). For validation, we performed ChIP-qPCR assays in A549-WT and A549-GR cells, which demonstrated binding of HIF-1α and HIF-1β, but not HIF-2α, to the two candidate sites (− 155 bp site and + 87 bp site) in A549-GR cells (Fig. 3A, H), which was in contrast to the findings in A549-WT cells (Fig. 3B, I). Notably, enrichment of IgG at these HREs was not significantly induced, verifying the specificity of the assay (Fig. 3B, I). Since HREs are primarily confirmed under hypoxic conditions, which promote HIF-1α protein stability, we confirmed our results by performing ChIP-qPCR assays in A549-WT cells exposed to normoxia (20% O2) or hypoxia (1% O2) for 16 h. The candidate binding sites (site 1 and site 2) showed significant hypoxia-induced binding of HIF-1α and HIF-1β but not HIF-2α (Fig. 3C, J).
In general, the transcription start sites of actively transcribed genes are marked by trimethylated acetylated H3K27 (H3K27ac) and H3K4 (H3K4me3) [35]. Thus, we next investigated the enrichment of these histone modifications on specific HREs. We performed ChIP assays with antibodies against H3K27ac and H3K4me3, followed by qPCR with primers flanking the HIF-1-binding sites (site 1 and site 2) of ABCB6 in A549-WT and A549-GR cells. Obvious enrichment of these histone marks of transcriptional activation (H3K27ac and H3K4me3) at HRE site 1 (Fig. 3D, E) and site 2 (Fig. 3K, L) was observed in GR cells compared to WT cells. We further exposed A549-WT cells to normoxia (20% O2) or hypoxia (1% O2) for 16 h and performed ChIP with antibodies against H3K27ac and H3K4me3. Again, hypoxic treatment significantly increased H3K27ac (Fig. 3F, M) and H3K4me3 (Fig. 3G, N) marks at the HIF-1-binding sites of ABCB6. Similar results were obtained by performing ChIP-qPCR assays in H1703-GR cells as compared to H1703-WT cells (Fig. S4C and S4E) and further confirmed in H1703 cells exposed to hypoxia compared to normoxia (Fig. S4D and S4F). Taken together, the results of these experiments demonstrate that HIF-1, but not HIF-2, binds directly to HRE sites adjacent to the transcription start sites of the ABCB6 gene in GR lung cancer cells as well as in lung cancer cells exposed to hypoxia. Induction of H3K27ac and H3K4me3 modifications at specific HREs of ABCB6 promotes chromatin accessibility and HIF-1 binding to these regions in response to chemotherapy or hypoxia.
HIF-1α/ABCB6 axis regulates heme synthesis to confer chemoresistance to gemcitabine treatments
ABCB6 is an ATP-binding cassette half-transporter and a specific heme-binding protein [36]. Heme plays a vital role in many biological processes such as oxidative metabolism and ATP generation, detoxification, oxygen transport, circadian rhythm, and regulation of transcription and translation [37, 38]. To investigate the mechanism by which the HIF-1α/ABCB6 axis regulates acquired gemcitabine resistance in NSCLC cells, we first explored the function of ABCB6 in heme synthesis. First, A549 and H1703 subclones were stably transfected with expression vectors encoding a shRNA targeting ABCB6 (shABCB6) or shRNAs targeting both HIF-1α and ABCB6 (shHIF-1α/ABCB6). Since HIF-1α protein is mostly degraded in WT cells under normoxia, we verified the efficiency of HIF-1α and ABCB6 double knockdown in A549-GR and H1703-GR subclones (Fig. S4A and S4B) and then detected heme biosynthesis in each of the subclones. Heme biosynthesis was markedly increased in A549-GR and H1703-GR transfected with the NCT compared to the WT groups (Fig. 4A, B). Knockdown of ABCB6 led to significantly decreased heme accumulation in both the GR and WT subclones (Fig. 4A, B). Gemcitabine-induced heme accumulation was also decreased clearly in HIF-1α knockdown cells and to a greater extent in HIF-1α/ABCB6 double-knockdown cells (Fig. 4A, B). These data suggest that gemcitabine-induced HIF-1α/ABCB6 activity promotes intracellular heme synthesis.
As a co-factor, heme has been demonstrated to be critically important in regulating the stability and activity of catalase, a key cellular and tissue antioxidant enzyme [11, 39]. We further confirmed that the protein expression and activity of catalase were highly induced in A549-GR and H1703-GR cells compared to WT cells (Fig. 4C–E). Again, catalase protein expression (Fig. 4C) and activity (Fig. 4D, E) were largely decreased in ABCB6 or HIF-1α knockdown subclones, particularly in the HIF-1α/ABCB6 double-knockdown groups, which indicates the vital role of the HIF-α/ABCB6 axis in regulating the heme-dependent antioxidant defense enzyme catalase.
We then analyzed intracellular ROS-mediated oxidative stress and observed significantly decreased levels of intracellular ROS in A549-GR (Fig. 4F) and H1703-GR (Fig. 4G) cells but not in WT cells. Knockdown of ABCB6, HIF-1α or HIF-1α/ABCB6 all led to increased intracellular ROS accumulation (Fig. 4F, G). We further observed an increased sensitivity to gemcitabine treatment in A549-GR (Fig. 4H) and H1703-GR (Fig. 4I) cells transfected with shHIF-1α or shABCB6 expression vectors. Notably, significant inhibition of cell survival was observed in A549-GR and H1703-GR cells transfected with both shHIF-1α and shABCB6 expression vectors upon treatment with gemcitabine (Fig. 4H, I).
To further verify the effects of ABCB6 inhibition on reversing gemcitabine resistance in cancer cells, tomatidine, an ATP synthase inhibitor, was used for inhibition of ABCB6 protein expression. Tomatidine, a steroidal alkaloid derived from the Solanaceae family, is known for its confirmed pharmacological safety and many biologic activities, including cardioprotective, anti-inflammatory, and immunostimulatory effects, as well as its ability to inhibit ATP synthase [40–42]. It has been demonstrated that tomatidine acts as an inhibitor of ABCB1-mediated drug transport [43]. Immunoblot analysis showed that ABCB6 protein expressions in both A549 (Fig. 5A) and H1703 (Fig. 5B) cells was gradually inhibited with increasing tomatidine concentration. Inhibition of ABCB6 by tomatidine largely reduced heme accumulation and catalase activity in both A549-WT (Fig. 5C, E) and H1703-WT (Fig. 5D, F) cells, and to a greater extent in GR cells. Finally, we observed an expected inhibition of cell survival in A549-GR and H1703-GR cells treated with both gemcitabine and tomatidine (Fig. 5G, H). Taken together, these results demonstrate that GR cells have increased HIF-1α/ABCB6 axis-induced heme biosynthesis and catalase activity, and further indicate that their poor responsiveness to gemcitabine can be reversed by targeting the HIF-1α/ABCB6 axis.
Inhibition of the HIF-α/ABCB6 axis sensitizes NSCLCs to gemcitabine therapy in animal models
Mouse xenograft models were established using A549 or H1703 subclones treated with or without gemcitabine to determine whether knockdown of ABCB6 and/or HIF-1α confers a sensitive phenotype to lung cancers previously resistant to gemcitabine therapy in vivo. The grouping and treatments of animals are outlined in Fig. 6. All animals received intraperitoneal injection of gemcitabine (20 mg/kg) or saline (250 µl) after palpable tumors had formed. Tumor growth was monitored twice per week. We observed similar results in both A549 (Fig. 6A) and H1703 (Fig. 6C) groups. The growth of GR tumors in mice transfected with shHIF-1α or shABCB6 was significantly decreased compared to that of GR tumors in mice transfected with NTC upon treatment with gemcitabine (Fig. 6A, C). The GR tumors in mice transfected with shHIF-1α/ABCB6 demonstrated a further decrease in the tumor volume compared with those in the NTC group after gemcitabine administration (Fig. 6A, C). The tumor images on day 42 are shown in Fig. S5A and S5B. Kaplan–Meier curves were plotted to evaluate the survival of mice in each group (Fig. 6B, D), and the log-rank test demonstrated a statistical difference in survival between mice in the GR NTC A549 tumors with gemcitabine treatment group and mice in the A549-GR shABCB6 plus gemcitabine group or the A549-GR shHIF-1α plus gemcitabine group (P = 0.0046 and 0.0004, respectively; Fig. 6B). The combination of HIF-1α and ABCB6 inhibition (shHIF-1α/ABCB6) with gemcitabine treatment also significantly increased survival compared with that of the gemcitabine-only treated groups (P = 0.0004; Fig. 6B). Again, similar results were obtained from the H1703 tumor-bearing mice groups as shown in Fig. 6D (GR NTC + Gem vs. GR shABCB6 + Gem, P = 0.0008; GR NTC + Gem vs. GR shHIF-1α + Gem, P = 0.0002; GR NTC + Gem vs. GR shHIF-1α/ABCB6 + Gem, P = 0.0003). However, the A549-GR and H1703-GR shHIF-1α/ABCB6 plus gemcitabine groups did not exhibit prolonged survival compared with the GR shABCB6 plus gemcitabine groups or the GR shHIF-1α plus gemcitabine groups (Fig. 6B, D).
At the end of the experiment, the animals were killed, and the xenografts were harvested. TUNEL staining was performed to evaluate apoptosis within the tumors. As expected, gemcitabine treatment caused marked apoptosis of A549-GR tumor cells in the shHIF-1α, the shABCB6, and particularly the shHIF-1α/ABCB6 transfected groups (Fig. 6E, G). These findings were further confirmed in the H1703 groups, for which we observed similar results (Fig. 6F, H).
We also studied the tumor growth rate in A549-WT tumor cells with HIF-1α or ABCB6 depletion combined with or without gemcitabine treatment. Animals were treated with saline (250 µl, intraperitoneal [i.p.] administration, twice per week) or gemcitabine (20 mg/kg, i.p. administration, twice per week) from day 26 and were monitored to day 53. As shown in Fig. S6A and S6B, the growth of A549 tumors in mice transfected with shABCB6 was obviously decreased compared to that of A549 tumors in mice transfected with NTC when treated with saline. The A549 tumors in mice transfected with shHIF-1α demonstrated a further decrease in the tumor volume compared with the NTC group. ABCB6 or HIF-1α depletion inhibited growth of A549 xenograft tumors even further when combined with gemcitabine treatment (Fig. S6A and S6B). Taken together, these data presented demonstrate that knockdown of HIF-α and ABCB6 dramatically delayed tumor growth, promoted apoptosis, and increased survival of mice with GR tumors under chemotherapy, indicating that inhibition of the HIF-α/ABCB6 axis may sensitize NSCLCs to gemcitabine therapy.
Discussion
Although gemcitabine is approved as the first-line chemotherapeutic drug for pancreatic adenocarcinoma treatment, it is also critically and widely used to treat breast, bladder, and non-small cell lung cancers [44]. Unfortunately, patients commonly develop acquired resistance shortly after beginning gemcitabine treatment, which ultimately leads to treatment failure [45]. Cancer cells are known to employ a variety of mechanisms to acquire resistance against gemcitabine-based treatment. Most previous studies have focused on acquired resistance due to changes in the rate of drug influx or efflux. For instance, ABC transporters such as ABCB1, ABCC1 and ABCG2 have crucial roles in developing resistance via the efflux of anticancer drugs outside of cancer cells [46]. In general, ABC transporters use ATP to facilitate not only the transmembrane flux of drugs, but also drug metabolites including saccharides, amino acids, peptides and proteins, and lipids and sterols, and they also participate in a variety of biological processes, such as antigen presentation, insulin secretion, and intracellular metabolic reprogramming [11]. However, the critical functions of ABC transporters as regulators or co-factors to activate intracellular signal pathways or metabolic reprogramming remain relatively unexplored. The present study demonstrates a mechanism of gemcitabine chemoresistance by which cancer cells increase intracellular ABCB6 expression in a HIF-1α-dependent manner that can, in turn, render antioxidant enzyme catalase effective by promoting heme biosynthesis. The results of the present study provide strong evidence for the inhibition of the HIF-α/ABCB6 axis in targeting lung cancer cell heme metabolism to disturb mitochondrial redox homeostasis, increase oxidant levels, and induce cell apoptosis, thereby abrogating gemcitabine resistance in NSCLCs (Fig. 7).
Previous studies have established that intratumoral hypoxia increases the stabilization and accumulation of HIF-1α, a critical transcriptional factor that responds to low oxygen condition, and that HIF-1α expression in associated with the development of resistance to chemotherapy and radiotherapy in multiple human cancer types [47]. However, further research has revealed that in addition to hypoxia, chemotherapeutic agents and radiation also can increase the levels of HIF-1α, which regulates multiple downstream target genes associated with many key steps of cancer progression, particularly acquired chemotherapy resistance [48]. HIFs are heterodimers composed of HIF-1α or HIF-2α subunits and a constitutively expressed HIF-1β subunit. Under hypoxic conditions, the O2-regulated prolyl hydroxylation-induced proteasomal degradation of HIF-1α is inhibited, leading to rapid accumulation of HIF-1α, dimerization with HIF-1β, binding to the consensus DNA sequence 5′-RCGTG-3′ within HREs located in target genes, and transcriptional activation [19]. Conversely, in normoxic conditions, HIF-1α is subject to ubiquitination and degradation. Intriguingly, we observed obvious stabilization and accumulation of HIF-1α even under normoxic conditions in GR lung cancer cell lines that had been exposed to gemcitabine for months. Our results demonstrate that gemcitabine-induced HIF-1α increased ABCB6 transcriptional activities by binding with two HREs (− 155 bp site and + 87 bp site) in the ABCB6 gene sequence. Moreover, we observed induction of H3K27ac and H3K4me3 modifications at specific HREs of the ABCB6 gene that promotes chromatin accessibility and HIF-1 binding to these regions in response to chemotherapy or hypoxia. However, the activation mechanism of HIF-1α expression under normoxia in GR cells remains unclear. It is possible that another protein functions as a modulator or coregulator in coordination with HIF-1α to facilitate its stability under normoxia. A study by Chaika et al. demonstrated that type I transmembrane protein physically interacts with HIF-1α and p300 and stabilizes HIF-1α at the protein level [49]. Shukla et al. also reported upregulation of HIF-1α protein in normoxic GR pancreatic cancer cells [31].
Multiple molecular mechanisms contributed to the acquired resistance of cancer cells to chemotherapeutics, and metabolic rewiring of tumor cells has been identified as a critical step in acquired drug resistance [50]. Cancer cells alter their metabolism by monitoring nutrient uptake (such as glucose and glutamine) and metabolite secretion, which leads to the modulation of certain metabolic pathways in response. As a critical transcriptional factor, HIF-1α has been shown to modulate multiple genes related to cancer metabolic reprogramming, including glycolysis [51], branched-chain amino acid metabolism [52], nucleotide synthesis [53], and lipid synthesis [54]. In addition, our previous studies and results from other groups consistently showed the involvement of HIF-1α in the regulation of other important metabolic pathways of cancer cells, such as glutamine metabolism and glutathione synthesis [13, 19, 55]. In the present study, we focused on heme metabolism. Heme is a central metabolic and signaling molecule that regulates diverse molecular and cellular processes related to oxidative metabolism and ATP generation. A recent study by Sohoni et al. showed that heme synthesis and uptake are enhanced in NSCLC cells [38]. Here, we show that the levels of heme biosynthesis are upregulated in GR NSCLC cells relative to gemcitabine-sensitive NSCLC cells. Our results further demonstrated that this increase is regulated by the HIF-α/ABCB6 axis. Heme binds to and directly regulates the activities of many proteins controlling processes ranging from tyrosine kinase signaling to redox homeostasis [11, 56, 57]. The present study suggests that increased heme biosynthesis in GR cells causes the elevation of ROS scavenger catalase activity, which in turn leads to redox homeostasis destruction and tumor survival.
Gemcitabine combined with platinum is a standard therapy for NSCLC. However, platinum-based chemotherapy has been shown to produce limited clinical benefits for cancer patients due to various adverse effects, and acquired tumor resistance remains a common problem [58, 59]. Accordingly, increasing evidence has suggested that the combination of chemotherapy with another form of anticancer therapy such as a tyrosine kinase inhibitor (TKI) [60], angiogenesis inhibitor [61], or immune checkpoint inhibitor [62] can produce better therapeutic outcomes. Given that hypoxia and HIF-1α activation are prominent features of various solid tumors that are, at least in part, responsible for chemotherapy resistance in these tumors, reversal of hypoxia or inhibition of HIF-1α signaling can improve the effectiveness of cancer therapies [63–65]. Our results suggest that a combination therapy consisting of a HIF-1α inhibitor and gemcitabine offered improved anti-tumor efficacy in the NSCLC tumor model. This combination therapy inhibited tumor growth and prolonged animal survival through increased chemotherapy sensitivity, leading to suppression of tumor cell proliferation as well as increased tumor cell apoptosis. Therefore, gemcitabine/HIF-1α-trap combination therapy might represent a more effective alternative for human lung cancer. The results of the present study provide rationale for human clinical studies to investigate the benefits of such combination therapy in lung cancer.
Supplementary Information
Below is the link to the electronic supplementary material.
Acknowledgements
We gratefully acknowledge Dr. Feng Wu and Dr. Yong Lin from the Department of Pathophysiology, Third Military Medical University for their kind help in assessing IHC results.
Author contributions
LX, BS and GX designed the study and wrote the paper. LX, GX, JL and FN performed the experiments. LX, SW, YG and HD analyzed the data. BS and GX advised on experimental procedures and revision of the paper. YW revised the paper. All the authors contributed to this manuscript. All the authors read and approved the final manuscript.
Funding
This work was supported by the Sichuan Science and Technology Program (2020YJ0046, 2020YFS0272 and 2019YJ0041), the Natural Science Foundation of Chongqing (cstc2018jcyjAX0512 and cstc2019jcyj-msxmX0791), and the National Science and Technology Major Project (2017ZX09304023).
Data availability
All data generated or analyzed during this study are included in this article.
Declarations
Conflict of interest
The authors declare that they have no competing interests.
Ethics approval and consent to participate
The experimental protocol was established according to the ethical guidelines of the 1964 Declaration of Helsinki. The protocol for IHC staining of patient tissues was approved by the Ethics Committee of the First Affiliated Hospital (Southwest Hospital), the Third Military Medical University (Army Medical University), and all patients or family members involved provided written informed consent.
Consent for publication
Written informed consent for publication was obtained from all participants.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Bin Shao, Email: sklbshaobin@scu.edu.cn.
Ganfeng Xie, Email: xiegf@aliyun.com.
References
- 1.Torre LA, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
- 2.Ferlay J, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–E386. doi: 10.1002/ijc.29210. [DOI] [PubMed] [Google Scholar]
- 3.Gao P, et al. miR-3607-3p suppresses non-small cell lung cancer (NSCLC) by targeting TGFBR1 and CCNE2. PLoS Genet. 2018;14(12):e1007790. doi: 10.1371/journal.pgen.1007790. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 4.Jemal A, et al. Cancer statistics, 2010. CA Cancer J Clin. 2010;60(5):277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
- 5.Hirsch FR, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311. doi: 10.1016/S0140-6736(16)30958-8. [DOI] [PubMed] [Google Scholar]
- 6.Mini E, et al. Cellular pharmacology of gemcitabine. Ann Oncol. 2006;17(Suppl 5):v7–12. doi: 10.1093/annonc/mdj941. [DOI] [PubMed] [Google Scholar]
- 7.Toschi L, et al. Role of gemcitabine in cancer therapy. Future Oncol. 2005;1(1):7–17. doi: 10.1517/14796694.1.1.7. [DOI] [PubMed] [Google Scholar]
- 8.Hlavata I, et al. The role of ABC transporters in progression and clinical outcome of colorectal cancer. Mutagenesis. 2012;27(2):187–196. doi: 10.1093/mutage/ger075. [DOI] [PubMed] [Google Scholar]
- 9.Cole SP, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science. 1992;258(5088):1650–1654. doi: 10.1126/science.1360704. [DOI] [PubMed] [Google Scholar]
- 10.Krishnamurthy PC, et al. Identification of a mammalian mitochondrial porphyrin transporter. Nature. 2006;443(7111):586–589. doi: 10.1038/nature05125. [DOI] [PubMed] [Google Scholar]
- 11.Boswell-Casteel RC, Fukuda Y, Schuetz JD. ABCB6, an ABC transporter impacting drug response and disease. Aaps J. 2017;20(1):8. doi: 10.1208/s12248-017-0165-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lee JW, et al. Hypoxia-inducible factor (HIF-1)alpha: its protein stability and biological functions. Exp Mol Med. 2004;36(1):1–12. doi: 10.1038/emm.2004.1. [DOI] [PubMed] [Google Scholar]
- 13.Lu H, et al. Chemotherapy triggers HIF-1-dependent glutathione synthesis and copper chelation that induces the breast cancer stem cell phenotype. Proc Natl Acad Sci U S A. 2015;112(33):E4600–E4609. doi: 10.1073/pnas.1513433112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kim WY, et al. Targeting heat shock protein 90 overrides the resistance of lung cancer cells by blocking radiation-induced stabilization of hypoxia-inducible factor-1alpha. Cancer Res. 2009;69(4):1624–1632. doi: 10.1158/0008-5472.CAN-08-0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Nantapong N, et al. The effect of reactive oxygen species (ROS) and ROS-scavenging enzymes, superoxide dismutase and catalase, on the thermotolerant ability of Corynebacterium glutamicum. Appl Microbiol Biotechnol. 2019;103(13):5355–5366. doi: 10.1007/s00253-019-09848-2. [DOI] [PubMed] [Google Scholar]
- 16.Goyal MM, Basak A. Human catalase: looking for complete identity. Protein Cell. 2010;1(10):888–897. doi: 10.1007/s13238-010-0113-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sichak SP, Dounce AL. Analysis of the peroxidatic mode of action of catalase. Arch Biochem Biophys. 1986;249(2):286–295. doi: 10.1016/0003-9861(86)90004-4. [DOI] [PubMed] [Google Scholar]
- 18.Gyorffy B, et al. Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS ONE. 2013;8(12):e82241. doi: 10.1371/journal.pone.0082241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Xiang L, et al. Glutaminase 1 expression in colorectal cancer cells is induced by hypoxia and required for tumor growth, invasion, and metastatic colonization. Cell Death Dis. 2019;10(2):40. doi: 10.1038/s41419-018-1291-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Xiang L, et al. Hypoxia-inducible factor 1 mediates TAZ expression and nuclear localization to induce the breast cancer stem cell phenotype. Oncotarget. 2014;5(24):12509–12527. doi: 10.18632/oncotarget.2997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhang H, et al. HIF-1-dependent expression of angiopoietin-like 4 and L1CAM mediates vascular metastasis of hypoxic breast cancer cells to the lungs. Oncogene. 2012;31(14):1757–1770. doi: 10.1038/onc.2011.365. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 22.Sassa S, Kappas A. Induction of aminolevulinate synthase and porphyrins in cultured liver cells maintained in chemically defined medium. Permissive effects of hormones on induction process. J Biol Chem. 1977;252(7):2428–2436. doi: 10.1016/S0021-9258(17)40571-0. [DOI] [PubMed] [Google Scholar]
- 23.Xiang L, et al. Ganetespib blocks HIF-1 activity and inhibits tumor growth, vascularization, stem cell maintenance, invasion, and metastasis in orthotopic mouse models of triple-negative breast cancer. J Mol Med (Berl) 2014;92(2):151–164. doi: 10.1007/s00109-013-1102-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Benda JA, Zaino R. GOG pathology manual: gynecologic oncology group. NY: Buffalo; 1994. [Google Scholar]
- 25.Nagy A, et al. Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep. 2018;8(1):9227. doi: 10.1038/s41598-018-27521-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bogomolova AM, et al. Hypoxia as a factor involved in the regulation of the apoA-1, ABCA1, and complement C3 gene expression in human macrophages. Biochemistry (Mosc) 2019;84(5):529–539. doi: 10.1134/S0006297919050079. [DOI] [PubMed] [Google Scholar]
- 27.Plosch T, et al. The liver X receptor (LXR) and its target gene ABCA1 are regulated upon low oxygen in human trophoblast cells: a reason for alterations in preeclampsia? Placenta. 2010;31(10):910–918. doi: 10.1016/j.placenta.2010.07.009. [DOI] [PubMed] [Google Scholar]
- 28.Chou CW, et al. Tumor cycling hypoxia induces chemoresistance in glioblastoma multiforme by upregulating the expression and function of ABCB1. Neuro Oncol. 2012;14(10):1227–1238. doi: 10.1093/neuonc/nos195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.He X, et al. Hypoxia regulates ABCG2 activity through the activation of ERK1/2/HIF-1alpha and contributes to chemoresistance in pancreatic cancer cells. Cancer Biol Ther. 2016;17(2):188–198. doi: 10.1080/15384047.2016.1139228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Albadari N, Deng S, Li W. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy. Expert Opin Drug Discov. 2019;14(7):667–682. doi: 10.1080/17460441.2019.1613370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Shukla SK, et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell. 2017;32(1):71–87.e7. doi: 10.1016/j.ccell.2017.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Coulet F, et al. Identification of hypoxia-response element in the human endothelial nitric-oxide synthase gene promoter. J Biol Chem. 2003;278(47):46230–46240. doi: 10.1074/jbc.M305420200. [DOI] [PubMed] [Google Scholar]
- 33.Elks PM, et al. Exploring the HIFs, buts and maybes of hypoxia signalling in disease: lessons from zebrafish models. Dis Model Mech. 2015;8(11):1349–1360. doi: 10.1242/dmm.021865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bhattarai D, Xu X, Lee K. Hypoxia-inducible factor-1 (HIF-1) inhibitors from the last decade (2007 to 2016): a "structure-activity relationship" perspective. Med Res Rev. 2018;38(4):1404–1442. doi: 10.1002/med.21477. [DOI] [PubMed] [Google Scholar]
- 35.Kimura H. Histone modifications for human epigenome analysis. J Hum Genet. 2013;58(7):439–445. doi: 10.1038/jhg.2013.66. [DOI] [PubMed] [Google Scholar]
- 36.Krishnamurthy P, Xie T, Schuetz JD. The role of transporters in cellular heme and porphyrin homeostasis. Pharmacol Ther. 2007;114(3):345–358. doi: 10.1016/j.pharmthera.2007.02.001. [DOI] [PubMed] [Google Scholar]
- 37.Chiabrando D, et al. Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes. Front Pharmacol. 2014;5:61. doi: 10.3389/fphar.2014.00061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sohoni S, et al. Elevated heme synthesis and uptake underpin intensified oxidative metabolism and tumorigenic functions in non-small cell lung cancer cells. Cancer Res. 2019;79(10):2511–2525. doi: 10.1158/0008-5472.CAN-18-2156. [DOI] [PubMed] [Google Scholar]
- 39.Kumar J, et al. Nutraceutical profile and evidence of alleviation of oxidative stress by Spirogyra porticalis (Muell.) Cleve inhabiting the high altitude Trans-Himalayan Region. Sci Rep. 2019;9(1):4091. doi: 10.1038/s41598-018-35595-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dyle MC, et al. Systems-based discovery of tomatidine as a natural small molecule inhibitor of skeletal muscle atrophy. J Biol Chem. 2014;289(21):14913–14924. doi: 10.1074/jbc.M114.556241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chiu FL, Lin JK. Tomatidine inhibits iNOS and COX-2 through suppression of NF-kappaB and JNK pathways in LPS-stimulated mouse macrophages. FEBS Lett. 2008;582(16):2407–2412. doi: 10.1016/j.febslet.2008.05.049. [DOI] [PubMed] [Google Scholar]
- 42.Vestergaard M, Roshanak S, Ingmer H. Targeting the ATP synthase in Staphylococcus aureus small colony variants, Streptococcus pyogenes and pathogenic fungi. Antibiotics (Basel) 2021;10(4):376. doi: 10.3390/antibiotics10040376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lavie Y, et al. Inhibitory effect of steroidal alkaloids on drug transport and multidrug resistance in human cancer cells. Anticancer Res. 2001;21(2a):1189–1194. [PubMed] [Google Scholar]
- 44.de Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16. doi: 10.1016/j.ejphar.2014.07.041. [DOI] [PubMed] [Google Scholar]
- 45.Gebregiworgis T, et al. Insights into gemcitabine resistance and the potential for therapeutic monitoring. Metabolomics. 2018;14(12):156. doi: 10.1007/s11306-018-1452-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hu J, et al. Effect of ceritinib (LDK378) on enhancement of chemotherapeutic agents in ABCB1 and ABCG2 overexpressing cells in vitro and in vivo. Oncotarget. 2015;6(42):44643–44659. doi: 10.18632/oncotarget.5989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Manoochehri Khoshinani H, Afshar S, Najafi R. Hypoxia: a double-edged sword in cancer therapy. Cancer Invest. 2016;34(10):536–545. doi: 10.1080/07357907.2016.1245317. [DOI] [PubMed] [Google Scholar]
- 48.O'Donnell JL, et al. Oncological implications of hypoxia inducible factor-1alpha (HIF-1alpha) expression. Cancer Treat Rev. 2006;32(6):407–416. doi: 10.1016/j.ctrv.2006.05.003. [DOI] [PubMed] [Google Scholar]
- 49.Chaika NV, et al. MUC1 mucin stabilizes and activates hypoxia-inducible factor 1 alpha to regulate metabolism in pancreatic cancer. Proc Natl Acad Sci USA. 2012;109(34):13787–13792. doi: 10.1073/pnas.1203339109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bosc C, Selak MA, Sarry JE. Resistance is futile: targeting mitochondrial energetics and metabolism to overcome drug resistance in cancer treatment. Cell Metab. 2017;26(5):705–707. doi: 10.1016/j.cmet.2017.10.013. [DOI] [PubMed] [Google Scholar]
- 51.Nagao A, et al. HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 2019;20(2):238. doi: 10.3390/ijms20020238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhang B, et al. Regulation of branched-chain amino acid metabolism by hypoxia-inducible factor in glioblastoma. Cell Mol Life Sci. 2020;78:195–206. doi: 10.1007/s00018-020-03483-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wu J, et al. HIF-1α in the heart: remodeling nucleotide metabolism. J Mol Cell Cardiol. 2015;82:194–200. doi: 10.1016/j.yjmcc.2015.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mylonis I, Simos G, Paraskeva E. Hypoxia-inducible factors and the regulation of lipid metabolism. Cells. 2019;8(3):214. doi: 10.3390/cells8030214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Xiang L, et al. Knock-down of glutaminase 2 expression decreases glutathione, NADH, and sensitizes cervical cancer to ionizing radiation. Biochim Biophys Acta. 2013;1833(12):2996–3005. doi: 10.1016/j.bbamcr.2013.08.003. [DOI] [PubMed] [Google Scholar]
- 56.Yao X, et al. Heme controls the regulation of protein tyrosine kinases Jak2 and Src. Biochem Biophys Res Commun. 2010;403(1):30–35. doi: 10.1016/j.bbrc.2010.10.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Quick-Cleveland J, et al. The DGCR8 RNA-binding heme domain recognizes primary microRNAs by clamping the hairpin. Cell Rep. 2014;7(6):1994–2005. doi: 10.1016/j.celrep.2014.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Paz-Ares L, et al. Phase III study of gemcitabine and cisplatin with or without aprinocarsen, a protein kinase C-alpha antisense oligonucleotide, in patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2006;24(9):1428–1434. doi: 10.1200/JCO.2005.04.3299. [DOI] [PubMed] [Google Scholar]
- 59.Heinemann V, et al. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol. 2006;24(24):3946–3952. doi: 10.1200/JCO.2005.05.1490. [DOI] [PubMed] [Google Scholar]
- 60.Zhou C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12(8):735–742. doi: 10.1016/S1470-2045(11)70184-X. [DOI] [PubMed] [Google Scholar]
- 61.Bello L, et al. Low-dose chemotherapy combined with an antiangiogenic drug reduces human glioma growth in vivo. Cancer Res. 2001;61(20):7501–7506. [PubMed] [Google Scholar]
- 62.Tallón de Lara P, et al. Gemcitabine synergizes with immune checkpoint inhibitors and overcomes resistance in a preclinical model and mesothelioma patients. Clin Cancer Res. 2018;24(24):6345–6354. doi: 10.1158/1078-0432.CCR-18-1231. [DOI] [PubMed] [Google Scholar]
- 63.Raykov Z, et al. Myo-inositol trispyrophosphate-mediated hypoxia reversion controls pancreatic cancer in rodents and enhances gemcitabine efficacy. Int J Cancer. 2014;134(11):2572–2582. doi: 10.1002/ijc.28597. [DOI] [PubMed] [Google Scholar]
- 64.Gstalder C, Ader I, Cuvillier O. FTY720 (Fingolimod) inhibits HIF1 and HIF2 signaling, promotes vascular remodeling, and chemosensitizes in renal cell carcinoma animal model. Mol Cancer Ther. 2016;15(10):2465–2474. doi: 10.1158/1535-7163.MCT-16-0167. [DOI] [PubMed] [Google Scholar]
- 65.Zhao T, et al. Inhibition of HIF-1α by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget. 2015;6(4):2250–2262. doi: 10.18632/oncotarget.2948. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data generated or analyzed during this study are included in this article.