1. Introduction
Lung cancer is the leading cause of cancer-related death worldwide [
1]. Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases, and includes lung adenocarcinoma (ADC), squamous cell lung carcinoma (SCC), and large cell lung carcinoma (LCC) [
2]. Although multiple therapeutic options, including targeted therapies and immunotherapy, are available for the treatment of lung cancer, and despite advances in cancer treatment including immunotherapy, platinum-based chemotherapy is still a commonly used treatment option [
3,
4,
5]. Unfortunately, the overall survival (OS) of NSCLC patients remains unsatisfactory, with a 5-year OS rate of approximately 19% [
6]. Certain subtypes of NSCLC, particularly lung ADC, are often resistant to conventional chemotherapies [
7]. Therefore, understanding the drug-resistant mechanisms and development of new effective treatments are important tasks required to improve the outcomes among patients with lung cancer.
Although the presence of reactive oxygen species (ROS) can facilitate tumor growth/progression and promote certain drug-resistant phenotypes, high levels of ROS are detrimental to cells [
8]. It is known that antioxidant capacity is elevated in some cancer cells and that it confers drug resistance [
8,
9]. Indeed, reprogramming of energy metabolism is now recognized as a hallmark of cancer [
10]. In particular, cancer cells can increase the uptake of glucose and glutamine for nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH) production to balance intracellular ROS [
11,
12]. Therefore, disabling NADPH and GSH production has been considered a potential strategy to sensitize tumor cells to chemotherapy and other antitumor treatments [
13,
14].
The wild-type isocitrate dehydrogenase 2 (IDH2) is a mitochondrial enzyme that catalyzes the interconverts between isocitrate and α-ketoglutaric acid (α-KG) in the TCA cycle, using NADP+ and NADPH for the redox reaction. Thus, abrogation of wild-type IDH2 could cause significant alterations in mitochondrial metabolism and redox status [
15,
16]. Furthermore, gain-of-function mutations in IDH1 and IDH2 have been discovered in several cancers, and IDH inhibitors, including ivosidenib (AG-120) and enasidenib (AG-221), are now clinically available for treatment of cancers with IDH mutations [
17]. In addition to the mutated versions, wild-type IDH2 has been shown to play a role in antioxidant defense [
18,
19] and to promote cell growth and survival of cancer cells [
20,
21,
22]. In a previous work, we found that IDH2 was upregulated in lung cancer and promoted lung cancer cell proliferation and tumor growth [
23]. However, the mechanism by which IDH2 promotes lung cancer and its therapeutic potential have yet to be further investigated. In this study, we examined the association between IDH2 and survival among lung cancer patients undergoing cisplatin chemotherapy and radiotherapy. We also examined the effects of combining IDH2 inhibition with cisplatin or radiation for the treatment of lung cancer and explored the mechanisms underlying the combination effect, with a focus on the impact of IDH2 abrogation on mitochondrial metabolism and ROS generation.
2. Materials and Methods
The ReagentsAnnexin V-FITC and propidium iodide (PI) were obtained from BD Biosciences (San Jose, CA, USA). The IDH2 inhibitor AGI-6780 was from Selleckchem (Houston, TX, USA). The antibodies used to detect IDH2 were from Abcam (Cambridge, UK, ab55271), and the antibody used to detect β-actin was from Cell Signaling Technology (Danvers, MA, USA, cst4970).
Cell culture
A549 and HCC827 cell lines were cultured in DMEM with 10% FBS; A549-CR cell line was purchased from Cell Resource Center, Peking Union Medical College (Beijing, China), and cultured in McCoy’s 5A with 10% FBS, as recommended by the vendor. All cell lines were incubated in an incubator at 37 °C with 5% CO2. For comparison of CDDP cytotoxicity, both cell lines were first cultured inDMEM+10% FBS for several days before being subjected to drug exposure and cell viability assay.
Generation of IDH2-overexpressing and IDH2-knockdown cells
The shRNA plasmids (GV-248-sh-IDH2#1 and GV-248-sh-IDH2#2) and the non-target control vector GV-248 were purchased from Genechem (Shanghai, China). For lentivirus production, each plasmid was co-transfected with the packaging (psPAX2) and envelope (pMD2.G) vectors into HEK293T cells. Lentivirus was harvested at 48 h post-transfection from the supernatants. A549 and HCC827 cells were infected with the lentivirus and selected in 1 μg/mL puromycin (Selleckchem, China) for three days.
Assays of cell proliferation and cell viability
For cell proliferation analysis, cells were seeded into a 6-well plate (8 × 104 cells/well), and cell numbers were counted using the trypan blue exclusion method in an auto-counting chamber. Apoptosis was stained with an annexin V-FITC/PI kit and analyzed using a Beckman cytoFLEX flow cytometer (Beckman Coulter Life Sciences, Indianapolis, IN, USA). Cell viability was determined in 96-well plates (2 × 103 cells/well) using the MTS reagent from Promega (Madison, WI, USA). The MTS regent was added into culture medium with a ratio of 20:100 and incubated at 37 °C with 5% CO2 for 3 h. The culture medium was used as the background control. The absorbance value at 490 nm was measured using a MultiSkan plate reader (Thermo, Helsinki, Finland). For colony formation assay, cells were seeded into 6-well plates (2 × 103 cells/well) and cultured with CDDP (dissolved in warm water) for two weeks. The culture medium was changed at day 7; cell colonies were stained with crystal violet and counted. We also determined the impact of an IDH2 inhibitor (AGI-6780) and cisplatin by incubating lung cancer cells with various concentrations of the compounds and performing the above assays.
Radiation assay
To determine the effect of AGI-6780 and radiation on colony formation, cells were seeded into 6-well plates (1 × 103 cells/well) and cultured for 24 h, treated with IDH2 inhibitor (AGI-6780) for four hours, and then exposed to the indictated doses of radiation. Cell colonies were counted after two weeks.
RT-qPCR
Total RNA was isolated using the Trizol reagent (Thermo Fisher Scientific, Waltham, MA, USA) and then converted to cDNA using a reverse transcription kit (Thermo Fisher Scientific, Waltham, MA, USA). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed using the ABI7500 qPCR system (Applied Biosystems, Waltham, MA, USA). The re-actions contained the indicated cDNA, primers, and SYBR®®Green Real-Time PCR Master Mixes (Thermo Fisher Scientific). The sequences of primers were as follows: beta-actin forward AGAGCTACGAGCTGCCTGAC, reverse AGCACTGTGTTGGCGTACAG; IDH2 forward CCTGCTCGTTCGCTCTCCA, reverse ACGGGTCATCTCATCACCATC.
Western blot analysis
Cells were harvested and washed twice with cold PBS, lysed in protein-IP lysis buffer with protein inhibitors, and centrifuged at 12,000× g for 20 min. The supernatant containing protein lysates (20 μg, as quantified by the BCA Protein Assay Kit, Thermo Fisher Scientific) was subjected to SDS–PAGE and transferred to a PVDF membrane (Millipore, Bedford, MA, USA). Membranes were blocked with 5% non-fat milk for 1 h at room temperature and then incubated with the specific primary antibody, washed with TBST, and then incubated with the corresponding HRP-conjugated secondary antibody. Protein bands were visualized using the Western lightening plus-ECL kit (Thermo Fisher Scientific).
ROS assay
Cells (5 × 104) were seeded into a 6-well plate and incubated with AGI-6780 and or cisplatin for 24 h, followed by staining with Dihydroethidium (1 μM). Cells were then harvested, washed twice with PBS, and analyzed using the Beckman cytoFLEX flow cytometer.
Analysis of cellular and mitochondrial metabolism
Cells (5 × 104) were seeded into a 24-well plate and incubated with AGI-6780 and or cisplatin for 24 h. The oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured using a Seahorse XF24 metabolic analyzer according to the proto-cols recommended by the manufacturer (Agilent Technologies, Inc., Santa Clara, CA, USA). Oligomycin (1 μM), FCCP (1 μM) and antimycin A + rotenone (0.5 μM) was used in OCR analysis as indicated. Exogenous glucose (10 mM), oligomycin (1 μM), and 2-DG (50 mM) were used in the ECAR measurement, as indicated in the respective figure legends. Mitochondrial metabolic parameters, including basal respiration, mitochondrial ATP production, maximal respiration capacity, spared respiratory capacity, and proton leak were calculated from the OCR curves according to the methods provided by the manufacturer of the Seahorse metabolic analyzer (Agilent Technologies, Inc.).
Animal study
Animal experiments were conducted according to protocols approved by the Sun Yat-sen University Animal Care Committee. Nude mice (6 weeks old) were injected in their flanks with either A549 cells with IDH2-knockdown cells (2 × 106) or the corresponding control cells. Tumor growth was assessed by measuring the length and width of the tumors. Tumor size was calculated using the following formula: length × width2/2. For drug treatment, cisplatin was injected intraperitoneally at a dose of 3 mg/kg/week.
Analysis of databases
Publicly available datasets were from Kaplan–Meier Plotter database (
http://kmplot.com/analysis/index.php?p=service&cancer=lung, accessed on 1 November 2020) and GEO databases, as indicated in the respective figure legends. Kmplot was used to compare the survival pro-files of cancer patients with high or low IDH2 expression [
24]. The GEO (NCBI) dataset (GDS3101) was utilized to analyze the mRNA levels in the A549 cell line and the corresponding cisplatin-resistant line.
Statistical analysis
Student’s t-test was used to test the statistical difference between two groups of samples, such as cancer tissues compared to normal tissues, or control cells compared to the drug-treated cells. Two-tailed unpaired t-tests were performed using GraphPad Prism 7. A p value of less than 0.05 was considered statistically significant.
4. Discussion
Mitochondrial isocitrate dehydrogenase catalyzes the metabolic conversion between isocitrate and α-ketoglutarate, using NAD(P)+/NAD(P)H for the redox reaction in the TCA cycle. As such, IDH2 plays a key role in mitochondrial energy metabolism and redox regulation, which could significantly affect cell survival and proliferation. Our recent study showed that IDH2 enhanced the survival of colon cancer cells by promoting redox homeostasis [
15] and leukemia cells by promoting a reductive TCA cycle [
16]. In this study, we showed that IDH2 was upregulated in drug-resistant lung cancer cells and enhanced cancer cell survival in the presence of cisplatin treatment. The ability of IDH2 to promote resistance to cisplatin seems to provide an explanation for the poorer clinical outcome in lung cancer patients with high IDH2 expression compared to those with low IDH2 expression among the patients in the chemotherapy group.
Cisplatin exerts its cytotoxic effect through covalent binding with the purine bases in DNA, leading to DNA damage response and activation of signaling pathways that trigger apoptosis [
24]. Cancer cells could become resistant to cisplatin through multiple mechanisms, which include an increase in the drug export, upregulation of DNA repair capacity, inactivation of the p53-mediated apoptotic response to DNA damage, and aberrant cell cycle regulation [
25]. Based on these mechanisms, various strategies have been developed to overcome cisplatin resistance or sensitize cancer cells to drug treatment. Such strategies include inhibition of glutathione synthesis, suppression of glutathione S-transferases, abrogation of DNA repair, and activation of apoptotic response to DNA damage [
24,
26,
27]. Although these approaches to overcoming cisplatin resistance have met with some success in experimental models and in clinical studies, the overall results are not very satisfactory. Since cisplatin remains as a major front-line chemotherapeutic agent for lung cancer treatment, more effective strategies to overcome cisplatin resistance and enhance its therapeutic activity are needed. Our study showed that IDH2 might play an important role in cisplatin resistance, and that abrogation of IDH2 could increase the sensitivity of lung cancer cells to cisplatin. We used multiple assays to evaluate the effect of the drug on cell viability, cell proliferation, and metabolic activity, with consistent results. However, the effect of IDH2 knockdown on cellular sensitivity to CDDP varied to some degree in multiple assays, although the overall impacts were in the same vein (an increase in drug sensitivity). These data suggest that each assay has its own limitation, and thus the results from multiple assays should be considered together when evaluating the overall impact of IDH2 on cell viability and drug sensitivity.
Interestingly, a recent study showed that treatment of cisplatin-resistant lung cancer cells (H69 and H460) with Riluzole, a drug used in clinical treatment of amyotrophic lateral sclerosis, resulted in an inhibition of lactate dehydrogenase A (LDHA) and xCT antiporter (SLC7A11), leading to a blockage of glutamate transport, an elevation of cellular ROS, and cytotoxicity to lung cancer cells [
28]. This study suggests that modulation of cellular redox metabolism and elevated ROS generation could be a potentially effective strategy to overcome cisplatin resistance. Our study, however, showed that LDHA and SLC7A11 were not upregulated in cisplatin-resistant A549 cells (
Figure S2C,G). Instead, we found that IDH2 expression was elevated and promoted cancer cell resistance to cisplatin. Importantly, abrogation of IDH2 by shRNA-mediated knockdown or by pharmacological inhibition could significantly sensitize the drug-resistant cells to cisplatin. Induction of ROS stress seems to be a key mechanism for this sensitization effect, since the drug-induced ROS alterations were positively correlated with the cytotoxic effect and antioxidant NAC was able to reverse this sensitization. Although our Seahorse analysis showed no significant changes in OCR when A549 cells were treated with 4–8 μg/mL cisplatin in the presence or absence of AGI-6780, these results do not necessarily suggest that the observed increase in ROS was outside of the mitochondria for the following reasons. The majority of ROS generated in the mitochondria are mainly due to the capture of electrons that “leak” from the respiratory chain by molecular oxygen to form superoxide, which is then converted to H
2O
2 by superoxide dismutase (SOD). With this mechanism of ROS generation, it only takes 1–2% of the total oxygen consumption to generate a massive amount of ROS. As such, an alteration in electron transport efficiency due to cisplatin and/or AGI-6780 treatment could cause only a subtle change in OCR, but a significant increase in ROS. Additionally, a decrease in NADPH generation due to IDH2 inhibition by AGI-6780 could also contribute to a ROS increase in the drug combination scenario.
The upregulation of antioxidant capacity often confers drug resistance in cancer cells [
8,
9]. Indeed, inhibition of antioxidant defense enzymes, and the subsequent disruption of NADPH and GSH production, can sensitize tumors to chemotherapy [
13,
14]. Our study has identified the mitochondrial metabolic enzyme IDH2 as an important redox regulator in lung cancer cells, in which it helps to maintain redox homeostasis. Inhibition of IDH2 leads to an abnormal accumulation of ROS, likely due to suppression of the IDH2-mediated NADPH generation, and renders the cancer cells more vulnerable to ROS stress induced by cisplatin. Furthermore, since the upregulation of antioxidant capacity is an important mechanism for radiation tolerance and targeting mitochondrial metabolism is considered as a potential strategy to reverse radio resistance [
29], the unique roles of IDH2 in both redox regulation and mitochondrial metabolism seem to make this enzyme a potentially important target for sensitization to radiotherapy. Our study showed that inhibition of IDH2 indeed increased the sensitivity of lung cancer cells to radiation in colony formation assay. Further in vitro and in vivo studies are needed to test if IDH2 could be targeted to enhance the efficacy of radiotherapy.
It is of particular interest to note that the pro-tumor role of wild-type IDH2 and its underlying mechanisms are different from the oncogenic function of IDH2 mutations, which are most often observed in certain gliomas and leukemia, such as acute myeloid leukemia (AML) [
30,
31]. Mutation in IDH2 leads to increased generation of oncogenic metabolite 2-hydroxyglutarate (2-HG), which promotes carcinogenesis by causing alterations in epigenetics, whereas wild-type IDH2 promotes cancer cell survival and proliferation through enhancing the reductive TCA cycle for the utilization of glutamine for lipid synthesis and increasing the antioxidant capacity of the cancer cells to counteract oxidative stress [
15]. It is important to note that the clinical drug Enasidenib (AG-221) used in clinical treatment of AML is a specific inhibitor of mutant IDH2 [
17], and thus is unlikely to inhibit wild-type IDH2 effectively. Our study showed that a specific knockdown of wild-type IDH2 in lung cancer cells increased their sensitivity to cisplatin in a mouse xenograft model. It would be important to test if the IDH2 inhibitor AGI-6780 could also potentiate antitumor activity of cisplatin in animals. To enhance the therapeutic effect of cisplatin or radiation via inhibition of IDH2, new inhibitors of wild-type IDH2 still need to be developed and evaluated in experimental models and in clinical settings.