Curcumin, a plant polyphenol, is a widely studied chemopreventive agent with demonstrated antitumor activities in preclinical studies and low toxicity profiles in multiple clinical trials against human malignancies. We previously showed that curcumin radiosensitizes cervical tumor cells without increasing the cytotoxic effects of radiation on normal human fibroblasts. Here we report that an inhibitory activity of curcumin on the antioxidant enzyme thioredoxin reductase-1 (TxnRd1) is required for curcumin-mediated radiosensitization of squamous carcinoma cells. Stable knockdown of TxnRd1 in both HeLa and FaDu cells nearly abolished curcumin-mediated radiosensitization. TxnRd1 knockdown cells showed decreased radiation-induced reactive oxygen species and sustained extracellular signal-regulated kinase 1/2 activation, which we previously showed was required for curcumin-mediated radiosensitization. Conversely, overexpressing catalytically active TxnRd1 in HEK293 cells, with low basal levels of TxnRd1, increased their sensitivity to curcumin alone and to the combination of curcumin and ionizing radiation. These results show the critical role of TxnRd1 in curcumin-mediated radiosensitization and suggest that TxnRd1 levels in tumors could have clinical value as a predictor of response to curcumin and radiotherapy. Cancer Res; 70(5); 1941–50

The success of radiotherapy depends on the ability to target malignant cells with radiation while simultaneously protecting surrounding normal tissues from its cytotoxic effects. One pharmacologic approach for increasing the clinical response to radiotherapy is the use of radiation response modifiers that either preferentially sensitize tumor cells or protect normal tissues from radiation-induced damage. Curcumin, a naturally occurring plant polyphenol, has an extensive history of use in diet and traditional medicine. Several preclinical studies carried out over the last 20 years have shown direct antitumorigenic activity in mouse tumor models (1). Curcumin's antitumor properties occur through pleiotropic mechanisms, including inhibition of proliferation, induction of apoptosis, inhibition of angiogenesis, immune modulation, and suppression of inflammation (reviewed in ref. 2). Curcumin has a low toxicity profile and has been well tolerated in multiple phase I and II clinical trials for malignant and nonmalignant diseases (2).

Several groups, including our own, have shown that curcumin selectively radiosensitizes tumor cells (35) without enhancing the effects of radiation on nontransformed cells or normal tissue (6, 7). We previously showed that curcumin sensitized cervical tumor cells, but not normal fibroblasts to ionizing radiation (IR). Mechanistically, we found that pretreatment of tumor cells with curcumin and IR resulted in elevated levels of reactive oxygen species (ROS) leading to sustained activation extracellular signal-regulated kinase 1/2 (ERK1/2)–mitogen-activated protein kinase (MAPK) and that both were required for curcumin-mediated radiosensitization (3). However, the molecular mechanism accounting for the differential effects of curcumin in fully transformed and normal cells remained unclear.

Thioredoxin reductase 1 (TxnRd1) is one of several identified molecular targets of curcumin (8). TxnRds are essential mammalian selenocysteine containing flavoenzymes that act in homodimeric form to catalyze NADPH-dependent reduction of thioredoxin and small molecular weight oxidants including ROS (9, 10). TxnRd1 and TxnRd2 (the cytosolic/nuclear and mitochondrial form, respectively) are two ubiquitously expressed isoforms of this enzyme family. They play a key role in maintaining redox-regulated cellular functions, including transcription, DNA damage recognition and repair, proliferation, and apoptosis (9). In response to oxidative stress, TxnRds sustain signaling pathways that regulate transcription of genes to protect the cell from oxidative damage (1113). Cytosolic TxnRd1 expression is often upregulated in human cancers, wherein it is associated with aggressive tumor growth and poor prognosis (14). TxnRd1 has been shown to confer protection against the lethal effects of IR in tumor cells (11). Moreover, agents that selectively target TxnRd1 have shown promising results as anticancer drugs in preclinical and clinical studies (15, 16) when used alone or when combined with IR (11). Fang and colleagues recently (8) showed that curcumin covalently binds to the selenocysteine residue of TxnRd1, thereby inhibiting its catalytic activity.

Based on these findings and our previous studies (3), we hypothesized that curcumin-mediated inhibition of TxnRd1 activity could enhance radiation sensitivity by decreasing the capacity of TxnRd1 to scavenge IR-induced ROS and/or decrease the activity of prosurvival signaling cascades activated by IR. We report that knockdown of TxnRd1 in squamous carcinoma cell lines significantly decreases curcumin-mediated radiosensitization. Conversely, overexpression in cells with low basal levels of TxnRd1 increased their sensitivity to curcumin alone and to the combination of curcumin and IR. These results show that TxnRd1 is required for curcumin-mediated radiosensitization.

Cell culture

HeLa, SiHa, FaDu (American Type Culture Collection), SCC-1 (a gift from Dr. Bert O'Malley, Jr., University of Pennsylvania), HEK293-pIRES, and HEK293-TxnRd1 (a gift from Dr. Giannis Spyrou, Academy of Athens, Greece) cell lines were grown in DMEM supplemented with penicillin/streptomycin and 10% fetal bovine serum. MSK-Leuk1 (a gift from Dr. Peter Sacks and Memorial Sloane Kettering Hospital) and keratinocytes (Invitrogen) were maintained in defined keratinocyte serum-free media with growth factor supplements.

Reagents

All chemical reagents were obtained from Sigma-Aldrich. Curcumin and tetrahydrocurcumin were dissolved in DMSO (10 mmol/L stock solution). Puromycin and G418 were dissolved in water (1 and 200 mg/mL stock solutions, respectively). 2′,7′-Dichlorofluorescein diacetate (DCF-DA) was dissolved in DMSO (20 mmol/L stock solution).

Generation of stable TxnRd1 knockdown cells

A set of nontargeting shRNA (shNT) and four shRNA plasmids targeting different coding regions of the TxnRd1 gene (shTR1) were purchased (SA Biosciences, KH02104P). HeLa and FaDu cells were transfected using Lipofectamine 2000 reagent (Invitrogen). At 48 h posttransfection, equal numbers of cells were plated from mock and plasmid-transfected plates. At 24 h postplating, cells were treated with 2 μg/mL puromycin for 48 h, and clones were allowed to grow. Selected clones were maintained in 0.5 μg/mL puromycin.

Clonogenic survival assays (low density)

HeLa cells were plated in 60-mm dishes at low densities and allowed to attach overnight. Cells were irradiated in the presence of curcumin or DMSO, using a 137Cs source (dose rate of 1.3 Gy/min). Colony formation, fixation, and counting were performed as described (3). Survival curves were fitted using a second-order polynomial function. The average normalized surviving fraction from three independent experiments was reported (±SE). Dose enhancement ratio (DER) values were calculated at 0.37 survival fraction.

Clonogenic survival assays (high density)

FaDu cells were plated in 60-mm dishes at high densities and allowed to attach overnight. Cells were pretreated for 8 h with curcumin and then irradiated using a 137Cs source (dose rate of 1.3 Gy/min). At 1 h after irradiation, cells were trypsinized and subcultured at low densities. Colony formation, fixation, and counting were performed as described (3). Survival curves were fitted using a second-order polynomial function. The average normalized surviving fraction from three independent experiments is reported (±SE). DER values were calculated at 0.37 survival fraction.

Cell growth assay (MTT assay)

MTT assays were performed using the cell proliferation kit (Roche Applied Sciences). Cells were plated in 24-well plates and treated with different curcumin doses for 8 h. Cell viability was assessed according to the manufacturer's protocol when the untreated controls reached confluence. Aliquots (200 μL) from each well were transferred into a 96-well plate, and absorbance was measured at 570 nm with an automated plate reader (Thermo Scientific).

Analysis of ROS levels

HeLa clones (shNT and shTR1) were treated with DMSO or 10 μmol/L curcumin for 8 h. Cells were incubated with 20 μmol/L DCF-DA for 30 min at 37°C. The probe was washed off with PBS, and the cells were either mock irradiated or exposed to a 4-Gy dose of IR. At 30 min later, the cells were trypsinized, resuspended in PBS, and analyzed for DCF fluorescence by flow cytometry (FACSCalibur, CellQuest Pro; BD Biosciences).

Immunoblotting

Whole-cell lysates were obtained as described previously (17). For detection of TxnRd1, TxnRd2, pERK1/2, ERK1/2, and β-actin, 20 to 50 μg of total protein was resolved on 10% SDS-PAGE gels, transferred to polyvinylidene difluoride membrane, blocked for 30 min in TBS-Tween solution containing 5% milk, and incubated for 1 to 2 h with the following antibodies in TBS-Tween solution containing 1% milk: anti-TxnRd1 antibody (Abcam), anti-TxnRd2 antibody (Abcam), anti–β-actin (Sigma), anti-pERK1/2, and anti-ERK1/2 (Cell Signaling). All membranes were incubated with horseradish peroxidase–conjugated antimouse or antirabbit secondary antibodies (Santa Cruz Biotechnology), and immunoreactive bands were detected using ECL Plus chemiluminescence (GE Healthcare).

Real-time reverse transcription–PCR

RNA was isolated from cells following TRI reagent protocol (Invitrogen). Reverse transcription was performed using AMV Reverse Transcriptase (Promega). Real-time PCR was done on Applied Biosystems 7300 Real-Time PCR System using Power SYBR Green PCR Master Mix. The primer sequences for detecting human TxnRd1 cDNA were ccactggtgaaagaccacgtt (forward) and aggagaaaagatcatcactgctgat (reverse). Primer sequences for 18S rRNA were aaacggctaccacatccaag (forward) and caattacagggcctcgaaag (reverse).

TxnRd activity assay

Cells were plated to 90% confluence, allowed to attach overnight, and treated with 10 μmol/L curcumin or tetrahydrocurcumin for 8 h. Following treatment, media were aspirated and replaced with 0.5 mL of media with Na-R-Lipoate (Na-RALA, GeroNova Research, Inc.) at a final concentration of 1 mmol/L. After a 2-h incubation, an aliquout was removed from the media overlying the cell monolayer and spun at 1,000 × g for 5 min to get rid of cellular debris. Fifty microliters of cleared media were then added to 100 μL of 5,5′-dithiobis(2-nitrobenzoic acid) solution in triplicate wells of a 96-well plate. The plates were incubated for 10 min in the dark. Absorbance was measured at 450 nm using a plate reader (Thermo Scientific). TxnRd activity was defined by the concentration of sodium-lipoate (disulfide) reduced to dehydrolipoate (dithiol) per milligram protein.

Statistical analysis

For all the clonogenic survival assays, mean and SEM of survival fraction from three independent experiments at 4 and 6 Gy for each treatment group were calculated. ANOVA was used to determine whether there were significant differences among three or more independent experimental conditions; specific pairwise comparisons were evaluated by planned contrasts. Student's t test was used to test for differences between two independent experimental conditions. Statistical significance was set at 0.05. All tests were two-sided. Statistical analyses were performed with SPSS Version 17 software (SPSS, Inc.).

Tumor cells express higher levels of TxnRd1 compared with normal or minimally transformed cells

TxnRd1 expression is shown to be elevated in several primary human malignancies and in a number of human cancer cell lines (14, 18). A subset of aggressive head and neck cancers also exhibit elevated expression of TxnRd1 mRNA and protein when compared with normal mucosa (14). We analyzed the expression of TxnRd1 protein levels and activity in various squamous carcinoma cell lines (FaDu, SCC-1, and HeLa) and compared it with that in primary keratinocytes and in MSK-Leuk1, a premalignant cell line derived from a human oral leukoplakia. Based on immunoblot analysis, TxnRd1 expression was elevated by >50-fold in FaDu and HeLa cells and by >20-fold in SCC-1 compared with either MSK-Leuk1 or keratinocytes (Fig. 1A). Total TxnRd activity was determined using an assay based on work by Arner and colleagues (19) and Biaglow and colleagues (20). These investigators showed that mammalian TxnRds reduce lipoate to dehydrolipoate more efficiently than lipoate dehydrogenase (19) and that the reduction of lipoate in intact tumor cells was largely dependent on TxnRd activity (20). As shown in Fig. 1B, basal TxnRd activity correlated with levels of TxnRd1 protein; TxnRd activity was ∼20-fold higher in FaDu and HeLa cells and ∼7-fold higher in SCC-1 compared with that in keratinocytes or MSK-Leuk1 cells.

Figure 1.

TxnRd1 protein and activity levels in cells with different transformation status correlate with response to curcumin. A, MSK-Leuk1, human keratinocytes, and three squamous carcinoma cell lines [HeLa (cervical), FaDu, and SCC-1 (head and neck)] were analyzed for TxnRd1 protein levels by immunoblot analysis. β-Actin was used as a loading control. B, basal levels of whole-cell TxnRd activity. Activity was measured as nanomoles dihydrolipoate formed per milligram protein and was normalized to that in MSK-Leuk1 cells. Columns, average of three independent experiments; bars, SE. C, the effect of curcumin on TxnRd activity. FaDu and HeLa cells were treated with 0, 5, 10, 20, and 50 μmol/L curcumin or tetrahydrocurcumin for 8 h and assayed for TxnRd activity. Activity was measured as nanomoles dihydrolipoate formed per milligram protein and normalized to own cells untreated control. D, keratinocytes, MSK-Leuk1, FaDu, and HeLa cells were treated with DMSO at 10, 20, or 50 μmol/L curcumin for 8 h. Whole-cell lysates were analyzed for curcumin-induced apoptosis by immunoblot assay using antibodies against cleaved PARP and β-actin.

Figure 1.

TxnRd1 protein and activity levels in cells with different transformation status correlate with response to curcumin. A, MSK-Leuk1, human keratinocytes, and three squamous carcinoma cell lines [HeLa (cervical), FaDu, and SCC-1 (head and neck)] were analyzed for TxnRd1 protein levels by immunoblot analysis. β-Actin was used as a loading control. B, basal levels of whole-cell TxnRd activity. Activity was measured as nanomoles dihydrolipoate formed per milligram protein and was normalized to that in MSK-Leuk1 cells. Columns, average of three independent experiments; bars, SE. C, the effect of curcumin on TxnRd activity. FaDu and HeLa cells were treated with 0, 5, 10, 20, and 50 μmol/L curcumin or tetrahydrocurcumin for 8 h and assayed for TxnRd activity. Activity was measured as nanomoles dihydrolipoate formed per milligram protein and normalized to own cells untreated control. D, keratinocytes, MSK-Leuk1, FaDu, and HeLa cells were treated with DMSO at 10, 20, or 50 μmol/L curcumin for 8 h. Whole-cell lysates were analyzed for curcumin-induced apoptosis by immunoblot assay using antibodies against cleaved PARP and β-actin.

Close modal

Curcumin inhibits TxnRd activity and enhances apoptosis in a manner-dependent on transformation status

We subsequently examined the ability of curcumin to inhibit TxnRd activity in HeLa and FaDu cells. Curcumin treatment resulted in a dose-dependent decrease in TxnRd activity with an IC50 of ∼10 μmol/L in both cell lines (Fig. 1C). These results are in agreement with those of Fang and colleagues, wherein TxnRd1 activity was measured by assaying the reduction of insulin in curumin-treated and untreated HeLa cell lysates (8). In that study, the Michael acceptor function of curcumin was proposed to mediate the inhibition of TxnRd activity. To test this possibility more stringently, we used tetrahydrocurcumin, a curcumin metabolite lacking the α,β-unsaturated carbon double bond that is presumably required for curcumin to covalently inhibit TxnRd1. As predicted, tetrahydrocurcumin had no significant effect on TxnRd activity at doses of up to 50 μmol/L (Fig. 1C).

Curcumin has been shown to selectively induce apoptosis in transformed cells (21). Therefore, we determined the effects of curcumin on levels of cleaved poly(ADP-ribose) polymerase (PARP), a marker of apoptosis, in keratinocytes (normal untransformed cells), MSK-Leuk1 (minimally transformed), FaDu, and HeLa cells (fully transformed) at 24 hours after curcumin treatment. Levels of cleaved PARP did not appreciably increase in keratinocytes or MSK-Leuk1 cells treated with curcumin doses of up to 20 μmol/L. However, there was a modest increase in cleaved PARP observed with 50 μmol/L curcumin. In contrast, curcumin induced a robust increase in cleaved PARP in both FaDu and HeLa cells. This increase exhibited a dose dependence, which was evident at doses as low as 10 μmol/L in FaDu (Fig. 1D). In accordance with reports that tetrahydrocurcumin retains the antioxidant effects, but not the proapoptotic activity, of the parental drug (22), we did not observe an increase in cleaved PARP in any of the cells lines treated with tetrahydrocurcumin doses up to 50 μmol/L (data not shown). These results show that curcumin enhances apoptosis preferentially in fully transformed squamous carcinoma cells and that this activity parallels its inhibitory activity on TxnRd1.

Efficient knockdown of TxnRd1 in squamous carcinoma cell lines

To test whether TxnRd1 is required for curcumin-mediated radiosensitization, we generated stable clones from both the HeLa and FaDu cell lines expressing an shRNA targeting a unique sequence in TxnRd1 gene (shTR1) or a nontargeting shRNA (shNT). For HeLa cells, two stable clones (shTR1-3.2 and shTR1-3.3) were selected using shRNA-3, whereas another two stable clones (shTR1-1.2 and shTR1-1.5) were selected using shRNA-1. For FaDu cells, one stable clone (shTR1-1.1) was selected using shRNA-1 and one stable clone using (shTR1-2.1) was selected using shRNA-2. All six knockdown clones exhibited >70% reduction in TxnRd1 mRNA levels compared with corresponding shNT clones transfected with the shNT plasmid (Fig. 2A). Consistent with mRNA levels, the HeLa-shTR1 clones showed over a 90% decrease (Fig. 2B, left) and FaDu-shTR1 clones showed an 80% decrease (Fig. 2B, right) in TxnRd1 protein levels as determined by immunoblot analysis. Moreover, there was a 50% reduction of TxnRd activity observed in HeLa-shTR1 clones (Fig. 2C, left) and 40% reduction in FaDu-shTR1 clones (Fig. 2C, right) compared with the respective shNT clones. The residual activity most likely reflects activity due to the mitochondrial TxnRd2 isoform, whose levels are not substantially affected by these shRNA sequences (Fig. 2B).

Figure 2.

Stable knockdown of TxnRd1 in HeLa and FaDu squamous carcinoma cell lines. Stable clones were generated from HeLa (left) and FaDu (right) cells by expressing a nontargeting shRNA (shNT) or an shRNA targeting human TxnRd1 (shTR1). A, untransfected cells, shNT, and shTR1 clones were analyzed for TxnRd1 mRNA levels by real-time reverse transcription–PCR analysis. TxnRd1 mRNA levels were normalized to 18s rRNA control and normalized to mRNA levels in untransfected HeLa or FaDu cells. PCR reactions were done in triplicate (±SEM); *, P < 0.05. B, TxnRd1 protein levels assayed by immunoblot analysis. β-Actin was used as a loading control. C, TxnRd enzymatic activity. Results represent the average of three independent experiments (±SEM); *, P < 0.05.

Figure 2.

Stable knockdown of TxnRd1 in HeLa and FaDu squamous carcinoma cell lines. Stable clones were generated from HeLa (left) and FaDu (right) cells by expressing a nontargeting shRNA (shNT) or an shRNA targeting human TxnRd1 (shTR1). A, untransfected cells, shNT, and shTR1 clones were analyzed for TxnRd1 mRNA levels by real-time reverse transcription–PCR analysis. TxnRd1 mRNA levels were normalized to 18s rRNA control and normalized to mRNA levels in untransfected HeLa or FaDu cells. PCR reactions were done in triplicate (±SEM); *, P < 0.05. B, TxnRd1 protein levels assayed by immunoblot analysis. β-Actin was used as a loading control. C, TxnRd enzymatic activity. Results represent the average of three independent experiments (±SEM); *, P < 0.05.

Close modal

Stable TxnRd1 knockdown in squamous carcinoma cells inhibits curcumin-mediated radiosensitization

To determine whether TxnRd1 is involved in the enhancement of the cytotoxic response of curcumin in combination with IR, we performed clonogenic survival assays on shNT and shTR1 clones from HeLa, SiHa (another cervical squamous carcinoma cell line), and FaDu cells. As predicted from our previous report (3), HeLa, SiHa, and FaDu cells stably expressing control shRNA (shNT clones) were significantly sensitized to IR by pretreatment with curcumin. The DERs at a surviving fraction of 0.37 ranged from 1.32 to 1.92 (Fig. 3A–D; Table 1). Knocking down TxnRd1 in HeLa and FaDu cells resulted in enhanced sensitivity to IR, in agreement with previous reports (ref. 12; Fig. 3A–D). Notably, curcumin failed to increase the radiosensitivity in the HeLa and FaDu shTR1 clones (Fig. 3A–D). Interestingly, in one of the HeLa clones (shTR1-3.2), curcumin induced a statistically significant radioprotective effect (Fig. 3A). Taken together, these results indicate that TxnRd1 is required for curcumin-mediated radiosensitization of squamous carcinoma cell lines.

Figure 3.

Squamous carcinoma cells with TxnRd1 knockdown are less sensitive to curcumin-mediated radiosensitization. HeLa (A, B) and FaDu (C, D) shNT control and shTR1 clones were treated with DMSO or 10 μmol/L curcumin for 8 h followed by 0, 2, 4, or 6 Gy doses of IR. Survival was assessed by clonogenic assays. Points, averages of three independent experiments; bars, SEM. Data were analyzed by pairwise comparisons. Treatment groups with statistically significant differences are indicated by brackets; *, P < 0.05. Differences in survival in response to IR between untreated (no curcumin) shNT and shTR1 cells were analyzed by ANOVA and were found to be statistically significant at 6 Gy (P < 0.05; see Results for details).

Figure 3.

Squamous carcinoma cells with TxnRd1 knockdown are less sensitive to curcumin-mediated radiosensitization. HeLa (A, B) and FaDu (C, D) shNT control and shTR1 clones were treated with DMSO or 10 μmol/L curcumin for 8 h followed by 0, 2, 4, or 6 Gy doses of IR. Survival was assessed by clonogenic assays. Points, averages of three independent experiments; bars, SEM. Data were analyzed by pairwise comparisons. Treatment groups with statistically significant differences are indicated by brackets; *, P < 0.05. Differences in survival in response to IR between untreated (no curcumin) shNT and shTR1 cells were analyzed by ANOVA and were found to be statistically significant at 6 Gy (P < 0.05; see Results for details).

Close modal
Table 1.

Summary of DERs in TxnRd1 knockdown and overexpressing HEK293 cells

Cell linePlasmid (clone)nDER (at 0.37 surviving fraction)
HeLa shNT-5 1.32 ± 0.06 
shTR1-3.2 0.78 ± 0.18 
shTR1-3.3 0.97 
shTR1-1.2 1.04 ± 0.07 
shTR1-1.5 0.95 
SiHa shNT-2 1.92 
shTR1-1.1 1.50 
FaDu shNT-1 1.75 ± 0.28 
shTR1-1.1 1.10 ± 0.27 
shTR1-2.1 1.04 ± 0.14 
HEK293 pIRES 1.06 ± 0.08 
TxnRd1 1.52 ± 0.19 
Cell linePlasmid (clone)nDER (at 0.37 surviving fraction)
HeLa shNT-5 1.32 ± 0.06 
shTR1-3.2 0.78 ± 0.18 
shTR1-3.3 0.97 
shTR1-1.2 1.04 ± 0.07 
shTR1-1.5 0.95 
SiHa shNT-2 1.92 
shTR1-1.1 1.50 
FaDu shNT-1 1.75 ± 0.28 
shTR1-1.1 1.10 ± 0.27 
shTR1-2.1 1.04 ± 0.14 
HEK293 pIRES 1.06 ± 0.08 
TxnRd1 1.52 ± 0.19 

NOTE: DERs (expressed as the ratio of radiation dose required to produce 0.37% survival with no curcumin present to the dose required to produce the same survival) in presence of 10 μmol/L curcumin. n represents the number of times the experiment was replicated.

Abbreviations: shNT, the control, nontargeting vector; shTR1, TxnRd1 knockdown clones; pIRES, empty expression vector; TxnRd1, TxnRd1 expressing vector.

TxnRd1 knockdown leads to reduced ROS generation and ERK1/2 activation following curcumin and IR treatments

In our previous study (3), we showed that pretreatment of tumor cells with curcumin led to enhanced production of IR-induced ROS resulting in sustained ERK1/2-MAPK activation. To test the effect of TxnRd1 knockdown on ROS generation following combined treatment with curcumin and IR, we used DCF-DA, a probe whose fluorescence is directly proportional to intracellular ROS levels (23). Treatment of HeLa shNT (control) and shTR1-3.2 (TxnRd1 knockdown) clones with curcumin did not significantly increase ROS levels (Fig. 4A). A single dose of 4 Gy induced an increase in ROS levels that was significantly higher in the shTR1 clones, which is consistent with the role of TxnRd1 as a ROS scavenger. Combined curcumin and IR treatment caused a substantial increase in ROS in the shNT clone, but not in the shTR1-3.2 clone. Quantitation of three independent experiments showed a similar trend (Fig. 4B). Similar results in terms of curcumin-mediated ROS changes were observed in a second HeLa clone, shTR1-1.2 (Supplementary Fig. S1). Interestingly, clone shTR1-3.2, which exhibited a larger increase in ROS induction upon IR treatment compared with shTR1-1.2, also showed a larger effect in terms of radiosensitization upon knockdown of TxnRd1. These results indicate that higher levels of TxnRd1 result in increased levels of ROS in response to the combined treatment of curcumin and IR.

Figure 4.

Effect of TxnRd1 knockdown on ROS production and ERK1/2 activation. A, increase in ROS in HeLa-shNT or shTR1-3.2 clones was analyzed by flow cytometry using DCF-DA. At least 30,000 events were collected for each treatment condition. Results are representative of three independent experiments. B, results from A were quantified and represented as percentage increase in fluorescence normalized to shNT unirradiated control (±SEM); *, P < 0.05. C, ERK phosphorylation in response to curcumin treatment. shNT and shTR1-3.2 cells were serum-starved for 24 h. The cells were pretreated with DMSO or 10 μmol/L curcumin for 8 h, followed by mock irradiation or irradiation with 6 Gy. Whole-cell lysates were analyzed by immunoblot using antibodies against p-ERK1/2 and total ERK1/2. The results are representative of three independent experiments.

Figure 4.

Effect of TxnRd1 knockdown on ROS production and ERK1/2 activation. A, increase in ROS in HeLa-shNT or shTR1-3.2 clones was analyzed by flow cytometry using DCF-DA. At least 30,000 events were collected for each treatment condition. Results are representative of three independent experiments. B, results from A were quantified and represented as percentage increase in fluorescence normalized to shNT unirradiated control (±SEM); *, P < 0.05. C, ERK phosphorylation in response to curcumin treatment. shNT and shTR1-3.2 cells were serum-starved for 24 h. The cells were pretreated with DMSO or 10 μmol/L curcumin for 8 h, followed by mock irradiation or irradiation with 6 Gy. Whole-cell lysates were analyzed by immunoblot using antibodies against p-ERK1/2 and total ERK1/2. The results are representative of three independent experiments.

Close modal

Our previous study (3) indicated that increased ROS production following treatment with curcumin and IR led to sustained ERK1/2 activation. To determine the effects of TxnRd1 knockdown on ERK1/2 activation, we performed immunoblot analysis on irradiated HeLa-shNT and HeLa-shTR1 cells that were pretreated with either curcumin or DMSO. As shown in Fig. 4C, irradiated cells showed a transient increase in phosphorylated ERK1/2 levels that decreased to baseline within an hour after irradiation. Pretreatment with curcumin prolonged IR-induced ERK1/2 activation in the HeLa-shNT cells for up to 6 hours after IR. This sustained ERK1/2 activation was not observed in the HeLa-shTR1 cells after curcumin treatment. These results show that the increased production of ROS and the sustained ERK1/2 activation (which we have previously shown is required for curcumin-induced radiosensitization) are dependent on the levels of TxnRd1 protein.

Overexpression of TxnRd1 increases radiosensitivity to curcumin

To determine if overexpression of TxnRd1 is sufficient to cause curcumin-mediated radiosensitization, we attempted to overexpress TxnRd1 in keratinocytes and the minimally transformed MSK-Leuk1 cells. However, the intricate mammalian selenoprotein synthesis machinery imposes a major obstacle in constructing stable selenoprotein-expressing mammalian cell lines. Specifically, previous studies showed that TxnRd1 expression is tightly regulated by multiple AU-rich sequences in the 3′-untranslated region (UTR; refs. 24, 25). Indeed, our attempts to overexpress TxnRd1 in either keratinocytes or MSK-Leuk1 cells were unsuccessful.

To overcome this problem, we searched for cell lines with low basal TxnRd1 levels, which could allow for TxnRd1 overexpression. One such line reported in the literature is the HEK293 cell line (24). We obtained the HEK293 cells stably expressing the full-length TxnRd1 gene (HEK293-TxnRd1) and also HEK293 cells expressing the empty pIRES vector (HEK293-pIRES). Immunoblot analysis confirmed the overexpression of TxnRd1 protein in HEK293-TxnRd1 cells (Fig. 5A). Moreover, TxnRd1 activity was 6-fold higher in HEK293-TxnRd1 cells compared with HEK293-pIRES cells, indicating that the expressed protein was enzymatically active (Fig. 5B). Also as shown in Fig. 5C, HEK293-TxnRd1 cells exhibited significantly reduced survival in response to curcumin compared with the HEK293-pIRES cells, whereas tetrahydrocurcumin did not result in significant cytotoxicity in either of the HEK293 cell lines, confirming the role of TxnRd1 in determining curcumin mediated cytotoxicity.

Figure 5.

TxnRd1 overexpression enhances curcumin-induced toxicity and radiosensitivity in HEK293 cells. A, immunoblot analysis of HEK293 cells stably expressing an empty pIRES-neo vector (HEK293-pIRES) or the pIRES vector containing the full-length TxnRd1 gene (HEK293-TxnRd1). β-Actin was used as a loading control. B, basal TxnRd activity in HEK293-pIRES and HEK293-TxnRd1 cells. Columns, average of three independent experiments; bars, SEM. C, cytotoxic effect of curcumin and tetrahydrocurcumin on HEK293-pIRES and HEK293-TxnRd1 cells. Cells were treated with increasing concentrations of curcumin or tetrahydrocurcumin for 8 h. When the untreated cells were confluent, MTT assays were performed. Points, averages of four independent experiments; bars, SEM. D, effect of curcumin on radiosensitivity of HEK293-pIRES and HEK293-TxnRd1 cells. Cells were treated with DMSO or 10 μmol/L curcumin for 8 h followed by 0, 2, 4, or 6 Gy doses of IR. Long-term survival was assessed by high-density clonogenic survival assay. Points, averages of three independent experiments; bars, SEM. Data were analyzed by pairwise comparisons. Treatment groups with statistically significant differences are indicated by brackets; *, P < 0.05.

Figure 5.

TxnRd1 overexpression enhances curcumin-induced toxicity and radiosensitivity in HEK293 cells. A, immunoblot analysis of HEK293 cells stably expressing an empty pIRES-neo vector (HEK293-pIRES) or the pIRES vector containing the full-length TxnRd1 gene (HEK293-TxnRd1). β-Actin was used as a loading control. B, basal TxnRd activity in HEK293-pIRES and HEK293-TxnRd1 cells. Columns, average of three independent experiments; bars, SEM. C, cytotoxic effect of curcumin and tetrahydrocurcumin on HEK293-pIRES and HEK293-TxnRd1 cells. Cells were treated with increasing concentrations of curcumin or tetrahydrocurcumin for 8 h. When the untreated cells were confluent, MTT assays were performed. Points, averages of four independent experiments; bars, SEM. D, effect of curcumin on radiosensitivity of HEK293-pIRES and HEK293-TxnRd1 cells. Cells were treated with DMSO or 10 μmol/L curcumin for 8 h followed by 0, 2, 4, or 6 Gy doses of IR. Long-term survival was assessed by high-density clonogenic survival assay. Points, averages of three independent experiments; bars, SEM. Data were analyzed by pairwise comparisons. Treatment groups with statistically significant differences are indicated by brackets; *, P < 0.05.

Close modal

Using a dose of 10 μmol/L curcumin, we performed clonogenic survival assays on irradiated HEK293-pIRES and HEK293-TxnRd1 cells. As predicted from the work of Smart and colleagues (12), HEK293-TxnRd1 cells were more radioresistant than HEK 293-pIRES cells. Pretreating HEK293-pIRES cells with curcumin did not induce significant radiosensitization. In contrast, the HEK293-TxnRd1 cells were significantly radiosensitized by 10 μmol/L curcumin, with a DER of 1.52 at 0.37 survival fraction (Fig. 5D). Taken together, these results indicate that TxnRd1 confers increased radioresistance and that TxnRd1-overexpressing cells exhibit enhanced sensitivity to combined treatment with radiation and curcumin, supporting the role for TxnRd1 as a crucial target mediating curcumin-induced radiosensitization.

Several reports have identified curcumin as a potent protector of normal tissue against radiation-induced damage. Administration of curcumin significantly reduced various normal tissue toxicities in rodent models treated with whole-body radiation (6, 26, 27). Intriguingly, curcumin has also been shown to radiosensitize various tumor cell lines in vitro (35) and induce a pronounced tumor growth delay following irradiation in mouse tumor models (28, 29). These findings indicate that curcumin has the ability to preferentially radiosensitize tumor, but not normal tissue, a remarkable property for a radiation response modifier that could translate into substantial clinical benefit. However, the mechanism behind this selective property of curcumin has remained elusive, and potential targets, including NF-κB, Akt, etc. (4, 30, 31), have not been rigorously tested as causative factors in this effect.

Growing evidence suggests that cancer cells produce higher basal levels of ROS (32) than normal cells. Under this persistent intrinsic oxidative stress, cancer cells develop an enhanced endogenous antioxidant capacity, which makes them more resistant to exogenous oxidants (33, 34). The upregulation of the antioxidant enzyme TxnRd1 is observed in multiple primary human malignancies, and its loss has been associated with a reversal of tumor phenotype and a decrease in tumorigenicity (35). These observations support the speculation that malignant cells could be sensitized to oxidants, including IR, by inhibition of this key antioxidant protein (12, 36).

It has been hypothesized that TxnRd1 may be necessary to counteract IR-induced changes in intracellular protein thiol oxidation/reduction status and to directly scavenge cytotoxic free radicals formed during exposure to IR. This hypothesis was tested by Smart and colleagues (12) using cell lines overexpressing wild-type or dominant-negative form of TxnRd1 (cysteine mutant). This study showed that HeLa cells overexpressing the wild-type but not the dominant-negative form of TxnRd1 were more resistant to the lethal effects of IR, suggesting that TxnRd1 is a clinically relevant target for novel radiosensitizing agents. Moreover, in a separate study, motexafin gadolinium, a potent inhibitor of TxnRd (37), has been shown to enhance tumor cell response to IR (38) and currently is in phase I clinical trials for patients with brain metastases from lung and breast cancers (15). Our results showing a significant increase in the radiosensitivity of HeLa and FaDu cells with knockdown TxnRd1 levels are in agreement with these data and further support the role of TxnRd1 as a major determinant of intrinsic tumor cell radiosensitivity (39).

Our attempts to overexpress TxnRd1 in nontransformed primary keratinocytes that normally express low levels of the protein were not successful, even when using a plasmid that had been used to express TxnRd1 in HEK293 cells (24). However, this was not unexpected, because previous reports showed that major obstacles exist to overexpressing TxnRd1 in mammalian cells (24, 25). TxnRd1 expression is regulated by sequences in the 3′ UTR including AU-rich elements (ARE) that exert stringent regulatory control over translation of TxnRd1 mRNA. ARE-binding proteins and microRNAs that selectively recognize AU-rich sequences can affect the stability of TxnRd1 mRNA levels and protein expression (40). One or more of these factors may contribute to the differential expression of TxnRd1 in tumor and normal cells. This is an area of ongoing investigation in our laboratory.

Based on the findings presented here, we propose that the preferential radiosensitization of tumor cells by curcumin is, in part, due to the elevated expression of the TxnRd1. The involvement of TxnRd1 in curcumin-mediated radiosensitization seems to be 2-fold. First, curcumin covalently binds to TxnRd1, irreversibly inhibiting its ability to reduce thioredoxin, which in turn is required for many of the pleiotropic antioxidant effects associated with TxnRd1 (9). Second, modification of the selenocysteine residue of TxnRd1 converts the protein from essential antioxidant enzyme to a protein with NADPH oxidase activity, thereby elevating oxidative stress. Both effects would enhance the oxidative burden on the cell, leading to greater clonogenic cell death (a finding supported by our previous work showing ablation of curcumin-mediated radiosensitization by the antioxidant NAC; ref. 3). Normal cells that express very low levels of TxnRd1 are not radiosensitized by curcumin. Whereas this phenomenon could be explained by low levels of TxnRd1, we have not excluded other possible mechanisms of radioprotection. For example, at least in vivo, it has been postulated that the radioprotective effects of curcumin could be due to its inhibitory effects on the expression of inflammatory and fibrogenic cytokines (6).

Our findings further suggest that TxnRd1 may mediate radiosensitization by additional natural and chemical compounds. Curcumin reacts as a potent Michael acceptor due to the presence of α,β-unsaturated ketone moieties coupled with electron withdrawing phenolic hydroxyl groups. Curcumin-induced inhibition of TxnRd1 is dependent on the Michael acceptor function by which curcumin binds covalently to the highly nucleophilic selenocysteine residue in the COOH terminus of TxnRd1 (8, 41). Tetrahydrocurcumin, which lacks the α,β-unsaturated ketone moieties, does not inhibit TxnRd1 activity and does not radiosensitize tumor cells. These observations suggest that compounds with Michael acceptor functionality could represent a novel family of radiosensitizers that are selective for transformed cells. Moreover, direct inhibition of TxnRd1 activity could be a useful means for screening natural and synthetic compounds for Michael acceptor functionality. Notably, caffeic acid phenethyl ester and oleocanthal acid, two additional natural compounds with Michael acceptor functionality, are also inhibitors of TxnRd1 and potent tumor radiosensitizers.4

4S.W. Tuttle and colleagues, unpublished observations

In conclusion, our results support a critical role of TxnRd1 in curcumin-mediated radiosensitization. Analysis of TxnRd1 levels in the tumors of patients enrolled in curcumin clinical trials could provide a valuable tool in identifying patients who would respond favorably to combined use of curcumin and radiotherapy.

No potential conflicts of interest were disclosed.

We thank Dr. Peter Sacks (Memorial Sloan-Kettering Hospital) for his gift of MSK-Leuk1 cells, Dr. Giannis Spyrou for his gift of HEK293-pIRES and HEK293-TxnRd1 cells and pIRES TxnRd1 plasmid, and Dr. Bert W. O'Malley, Jr., for his gift of SCC-1 cells.

Grant Support: National Cancer Institute grants CA104922 (C. Koumenis) and CA137398 (S.W. Tuttle).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Howells
LM
,
Moiseeva
EP
,
Neal
CP
, et al
. 
Predicting the physiological relevance of in vitro cancer preventive activities of phytochemicals
.
Acta Pharmacol Sin
2007
;
28
:
1274
304
.
2
Goel
A
,
Kunnumakkara
AB
,
Aggarwal
BB
. 
Curcumin as “curecumin”: from kitchen to clinic
.
Biochem Pharmacol
2008
;
75
:
787
809
.
3
Javvadi
P
,
Segan
AT
,
Tuttle
SW
,
Koumenis
C
. 
The chemopreventive agent curcumin is a potent radiosensitizer of human cervical tumor cells via increased reactive oxygen species production and overactivation of the mitogen-activated protein kinase pathway
.
Mol Pharmacol
2008
;
73
:
1491
501
.
4
Chendil
D
,
Ranga
RS
,
Meigooni
D
,
Sathishkumar
S
,
Ahmed
MM
. 
Curcumin confers radiosensitizing effect in prostate cancer cell line PC-3
.
Oncogene
2004
;
23
:
1599
607
.
5
Khafif
A
,
Hurst
R
,
Kyker
K
,
Fliss
DM
,
Gil
Z
,
Medina
JE
. 
Curcumin: a new radio-sensitizer of squamous cell carcinoma cells
.
Otolaryngol Head Neck Surg
2005
;
132
:
317
21
.
6
Okunieff
P
,
Xu
J
,
Hu
D
, et al
. 
Curcumin protects against radiation-induced acute and chronic cutaneous toxicity in mice and decreases mRNA expression of inflammatory and fibrogenic cytokines
.
Int J Radiat Oncol Biol Phys
2006
;
65
:
890
8
.
7
Choudhary
D
,
Chandra
D
,
Kale
RK
. 
Modulation of radioresponse of glyoxalase system by curcumin
.
J Ethnopharmacol
1999
;
64
:
1
7
.
8
Fang
J
,
Lu
J
,
Holmgren
A
. 
Thioredoxin reductase is irreversibly modified by curcumin: a novel molecular mechanism for its anticancer activity
.
J Biol Chem
2005
;
280
:
25284
90
.
9
Mustacich
DP
,
Powis
G
. 
G. Thioredoxin reductase
.
Biochem J
2000
;
346
:
1
8
.
10
Becker
K
,
Gromer
S
,
Schirmer
RH
,
Müller
S
. 
Thioredoxin reductase as a pathophysiological factor and drug target
.
Eur J Biochem
2000
;
267
:
6118
25
.
11
Karimpour
S
,
Lou
J
,
Lin
LL
, et al
. 
Thioredoxin reductase regulates AP-1 activity as well as thioredoxin nuclear localization via active cysteines in response to ionizing radiation
.
Oncogene
2002
;
21
:
6317
27
.
12
Smart
DK
,
Ortiz
KL
,
Mattson
D
, et al
. 
Thioredoxin reductase as a potential molecular target for anticancer agents that induce oxidative stress
.
Cancer Res
2004
;
64
:
6716
24
.
13
Choi
JH
,
Kim
TN
,
Kim
S
, et al
. 
Overexpression of mitochondrial thioredoxin reductase and peroxiredoxin III in hepatocellular carcinomas
.
Anticancer Res
2002
;
22
:
3331
5
.
14
Lincoln
DT
,
Ali Emadi
EM
,
Tonissen
KF
,
Clarke
FM
. 
The thioredoxin-thioredoxin reductase system: over-expression in human cancer
.
Anticancer Res
2003
;
23
:
2425
33
.
15
Carde
P
,
Timmerman
R
,
Mehta
MP
, et al
. 
Multicenter phase Ib/II trial of the radiation enhancer motexafin gadolinium in patients with brain metastases
.
J Clin Oncol
2001
;
19
:
2074
83
.
16
Urig
SBK
. 
On the potential of thioredoxin reductase inhibitors for cancer therapy
.
Semin Cancer Biol
2006
;
16
:
452
65
.
17
Koumenis
C
,
Naczk
IC
,
Koritzinsky
M
, et al
. 
Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2a
.
Mol Cell Biol
2002
;
22
:
7405
16
.
18
Tamura
T
,
Stadtman
TC
. 
A new selenoprotein from human lung adenocarcinoma cells: purification, properties, and thioredoxin reductase activity
.
Proc Natl Acad Sci U S A
1996
;
93
:
1006
11
.
19
Arnér
ESJ
,
Nordberg
J
,
Holmgren
A
. 
Efficient reduction of lipoamide and lipoic acid by mammalian thioredoxin reductase
.
Biochem Biophys Res Commun
1996
;
225
:
268
74
.
20
Biaglow
JE
,
Donahue
J
,
Tuttle
S
,
Held
K
,
Chrestensen
C
,
Mieyal
J
. 
A method for measuring disulfide reduction by cultured mammalian cells: relative contributions of glutathione-dependent and glutathione-independent mechanisms
.
Anal Biochem
2000
;
281
:
77
86
.
21
Syng-ai
C
,
Kumari
AL
,
Khar
A
. 
Effect of curcumin on normal and tumor cells: role of glutathione and bcl-2
.
Mol Cancer Ther
2004
;
3
:
1101
8
.
22
Somparn
P
,
Phisalaphong
C
,
Nakornchai
S
,
Unchern
S
,
Morales
NP
. 
Comparative antioxidant activities of curcumin and its demethoxy and hydrogenated derivatives
.
Biol Pharm Bull
2007
;
30
:
74
8
.
23
Brandt
R
,
Keston
AS
. 
Synthesis of dichlorodihydrofluorescin: a stable reagent for fluorometric analysis
.
Anal Biochem
1965
;
11
:
6
9
.
24
Nalvarte
I
,
Damdimopoulos
AE
,
Nystom
C
, et al
. 
Overexpression of enzymatically active human cytosolic and mitochondrial thioredoxin reductase in HEK-293 cells: effect on cell growth and differentiation
.
J Biol Chem
2004
;
279
:
54510
7
.
25
Gasdaska
JR
,
Harney
JW
,
Gasdaska
PY
,
Powis
G
,
Berry
MJ
. 
Regulation of human thioredoxin reductase expression and activity by 3′-untranslated region selenocysteine insertion sequence and mRNA instability elements
.
J Biol Chem
1999
;
274
:
25379
85
.
26
Inano
H
,
Onoda
M
. 
Radioprotective action of curcumin extracted from curcuma longa LINN: inhibitory effect on formation of urinary 8-hydroxy-2-deoxyguanosine, tumorigenesis, but not mortality, induced by γ-ray irradiation
.
Int J Radiat Oncol Biol Phys
2002
;
53
:
735
43
.
27
Thresiamma
KC
,
George
J
,
Kuttan
R
. 
Protective effect of curcumin, ellagic acid and bixin on radiation induced toxicity
.
Indian J Exp Biol
1996
;
34
:
845
7
.
28
Kunnumakkara
AB
,
Diagaradjane
P
,
Guha
S
, et al
. 
Curcumin sensitizes human colorectal cancer xenografts in nude mice to {γ}-radiation by targeting nuclear factor-{κ}B-regulated gene products
.
Clin Cancer Res
2008
;
14
:
2128
36
.
29
Li
M
,
Zhang
Z
,
Hill
DL
,
Wang
H
,
Zhang
R
. 
Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway
.
Cancer Res
2007
;
67
:
1988
96
.
30
Kunnumakkara
AB
,
Diagaradjane
P
,
Guha
S
, et al
. 
Curcumin sensitizes human colorectal cancer xenografts in nude mice to {γ}-radiation by targeting nuclear factor-{κ} B-regulated gene products
.
Clin Cancer Res
2008
;
14
:
2128
2136
.
31
Dhandapani
KM
,
Mahesh
VB
,
Brann
DW
. 
Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFκB transcription factors
.
J Neurochem
2007
;
102
:
522
38
.
32
Toyokuni
S
,
Okamoto
K
,
Yodoi
J
,
Hiai
H
. 
Persistent oxidative stress in cancer
.
FEBS Lett
1995
;
358
:
1
3
.
33
Pervaiz
S
,
Clement
MV
. 
Tumor intracellular redox status and drug resistance - serendipity or a causal relationship?
Curr Pharm Des
2004
;
10
:
1969
77
.
34
Tiligada
E
. 
Chemotherapy: induction of stress responses
.
Endocr Relat Cancer
2006
;
13
:
S115
24
.
35
Yoo
M
,
Xu
X
,
Carlson
BA
,
Gladyshev
VN
,
Hatfield
DL
. 
Thioredoxin reductase 1 deficiency reverses tumor phenotype and tumorigenicity of lung carcinoma cells
.
J Biol Chem
2006
;
281
:
13005
8
.
36
Kirkpatrick
DL
,
Kuperus
M
,
Dowdeswell
M
, et al
. 
Mechanisms of inhibition of the thioredoxin growth factor system by antitumor 2-imidazolyl disulfides
.
Biochem Pharmacol
1998
;
55
:
987
94
.
37
Magda
D
,
Lepp
C
,
Gerasimchuk
N
, et al
. 
Redox cycling by motexafin gadolinium enhances cellular response to ionizing radiation by forming reactive oxygen species
.
Int J Radiat Oncol Biol Phys
2001
;
51
:
1025
36
.
38
Rockwell
S
,
Donnelly
ET
,
Liu
Y
,
Tang
L
. 
Preliminary studies of the effects of gadolinium texaphyrin on the growth and radiosensitivity of EMT6 cells in vitro
.
Int J Radiat Oncol Biol Phys
2002
;
54
:
536
41
.
39
Gius
D
. 
Redox-sensitive signaling factors and antioxidants: how tumor cells respond to ionizing radiation
.
J Nutr
2004
;
134
:
3213S
4S
.
40
Von Roretz
C
,
Gallouzi
I
. 
Decoding ARE-mediated decay: is microRNA part of the equation?
J Cell Biol
2008
;
181
:
189
94
.
41
Moos
PJ
,
Edes
K
,
Mullally
JE
,
Fitzpatrick
FA
. 
Curcumin impairs tumor suppressor p53 function in colon cancer cells
.
Carcinogenesis
2004
;
25
:
1611
7
.

Supplementary data