Open Access

Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling

  • Authors:
    • Justin Lucas
    • Tze-Chen Hsieh
    • H. Dorota Halicka
    • Zbigniew Darzynkiewicz
    • Joseph M. Wu
  • View Affiliations

  • Published online on: August 1, 2018     https://doi.org/10.3892/ijo.2018.4512
  • Pages: 1469-1480
  • Copyright: © Lucas et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Programmed cell death ligand 1 (PD‑L1) expressed in cancer cells interacting with its receptor programmed cell death 1 (PD‑1) expressed in immune cells represents a regulatory axis linked to the suppression and evasion of host immune functions. The blockade of PD‑1/PD‑L1 interaction using monoclonal antibodies has emerged as an effective therapy for several solid tumors; however, durable response has been observed in a subset of patients with PD‑L1-positive tumors. Thus, the understanding of the mechanisms responsible for the expression of PD‑L1 in tumor cells may help to improve the response to PD‑L1 blockade therapies. In this study, we investigated whether resveratrol, a grape-derived stilbenoid with immunoregulatory activity, modulates the expression of PD‑L1 in breast and colorectal cancer cells. The surface expression of PD‑L1 was determined by flow cytometry in cancer cells treated with resveratrol and/or piceatannol. Each stilbenoid alone induced PD‑L1 and when used in combination, elicited a synergistic upregulation of PD‑L1 in some cell lines. The induction of PD‑L1 by the combined use of stilbenoids was most pronounced in the Cal51 triple-negative breast cancer (TNBC) and SW620 colon cancer cells. The observed induction of PD‑L1 was transcriptionally mediated by nuclear factor (NF)-κB, as shown by NF‑κB reporter assays, the nuclear accumulation of the p65 subunit of NF‑κB, inhibition by the IKK inhibitor, BMS‑345541, and histone the modification inhibitors, resminostat, entinostat or anacardic acid. Combined treatment with resveratrol and piceatannol also decreased tumor cell survival as indicated by the upregulation of the DNA damaging marker, γH2AX, the cleavage of caspase 3, the downregulation of the survival markers, p38-MAPK/c‑Myc, and G1-to-S cell cycle arrest.

Introduction

Programmed cell death ligand 1 (PD-L1) is a functional ligand of programmed cell death 1 (PD-1) (1). The binding of tumor cell PD-L1 to immune T-cell PD-1 inhibits T-cell activation and attenuates T-cell-mediated immunosuppression (24). This results in the evasion of host antitumor immunity, potentially reducing the efficacy of anticancer therapies and resulting in a poor clinical outcome (2,5,6). To counter the escape from the host immune surveillance system by tumor cells, blockade strategies using monoclonal antibodies (mAbs) to prevent the binding of PD-L1 to PD-1 have been developed (7-12). The clinical efficacy of this approach has been demonstrated in certain cancer types, including melanoma (13-16), non-small-cell lung cancer (17-19) and renal carcinoma (20). Blockade therapy differs from tumoricidal chemotherapy in that its antitumorigenic effects involve boosting host immunity concomitant with the modulation of the expression/activity of the repertoire of cytotoxic T-cell and T-regulatory cells (2124). Although success in anti-PD-1/PD-L1 therapy has made it possible to achieve tumor eradication and disease remission/cure, outstanding challenges remain. Only 31% of patients with advanced melanoma treated intravenously with anti-PD-1 drugs (nivolumab) have exhibited objective tumor regression (14). Likewise, the same therapeutic regimen has shown response rates ranging from 25% in renal cancer (20), to 19% in non-squamous non-small-cell lung cancer (17) and to 20% in squamous non-small-cell lung cancer (18), respectively. Moreover, a positive therapeutic response typically occurs in patients with PD-L1-positive tumors (13,25).

Since the expression of PD-L1 in cancer cells may affect the patient response to immune blockade therapy, it is of interest to identify agents capable of modulating the expression of PD-L1. In this study, we focused on resveratrol, a stilbenoid present abundantly in red wine, red grape skin and peanuts (26,27). Interest in resveratrol stems largely from the report in 1997 by Jang et al, showing that the molecule prevents the development of pre-neoplastic lesions in carcinogen-exposed mammary glands, and the inhibition of initiation and promotion of skin cancer in a mouse model (28). Since then, numerous studies have demonstrated its broad-spectrum beneficial health effects, including, anti-inflammatory (29,30) and anticancer activities (26). Several mechanisms (31,32) and target proteins for the biological and pharmacological activities of resveratrol have been identified and characterized (3342). Resveratrol has also been reported to exert immunomodulatory effects, as illustrated by the induction of interferon (IFN)-γ expression in CD8+ T-cells both ex vivo and in vivo (43), and the inhibition of the proliferation of CD4+ T-cells (43,44). Craveiro et al (45) recently demonstrated that low-dose resveratrol (20 μM) activates human CD4+ cells and induces DNA damage response, while high-dose resveratrol (100 μM) induces G1 phase cell cycle arrest, suggesting that resveratrol may act on host immune cell types in a dose-dependent manner. The chemopreventive activity of resveratrol was first demonstrated using skin and breast cancer models (28), and recent clinical trials support the use of resveratrol in colorectal cancer (46,47). Thus, in this study, we selected breast and colorectal cancer cell lines to examine the regulatory effects of resveratrol and its biotransformed product, piceatannol, on the expression of PD-L1. The results revealed that both dietary stilbenoids, alone or in combination, copiously increased the expression level of PD-L1 in some breast and colorectal cancer cells via HDAC3/p300-mediated nuclear factor (NF)-κB signaling. In addition, both stilbenoids exerted cytotoxic effects on the tumor cells.

Materials and methods

Reagents

Resveratrol, piceatannol, resminostat, entinostat, mocetinostat, vorinostat, curcumin, garcinol, anacardic acid and Tip60i were purchased from Selleckchem (Houston, TX, USA). MB-3 and BMS-345541 were purchased from MilliporeSigma (Burlington, MA, USA). Pterostilbene and myricetin were from LKT Laboratories (St. Paul, MN, USA) and trimethoxy-resveratrol (trans-3,5,4′-trimethoxystilbene) was from Cayman Chemical Co. (Ann Arbor, MI, USA). Stock solutions of the chemicals were prepared based on the information provided by the manufacturer and maintained at −20°C. The antibodies for human PD-L1 (E1L3N, 13684), p38 MAPK (D13E1, 8690), NF-κB p65 (D14E12, 8242), γH2AX (20E3, 9718), cleaved caspase 3 (D3E9, 9579), IRF-1 (D5E4, 8478) and rabbit IgG isotype monoclonal antibody (DA1E, 5742) conjugated to PE were obtained from Cell Signaling Technology (Beverly, MA, USA). c-Myc antibody (9E10, sc-40) was obtained from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). Fetal bovine serum, RPMI-1640, DMEM, streptomycin and penicillin were from Gibco/Thermo Fisher Scientific (Waltham, MA, USA). All other chemicals and solvents were of analytical grade.

Cell culture and treatment

Human BT549 (breast cancer), BT474 (invasive ductal carcinoma), SKBR3 (breast cancer), HCT116 (colon cancer), SW480 (colon cancer), HT29 (recto-sigmoid adenocarcinoma) and SW620 (colon cancer) cells were obtained from the American Type Culture Collection (ATCC, Rockville, MD, USA). Human Cal51 (breast cancer) cells were from the Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures (Braunschweig, Germany). The cells were maintained in RPMI-1640 or DMEM culture media supplemented with penicillin, streptomycin and 10% heat-inactivated fetal bovine serum according to the manufacturer's instructions. The cells were split once a week and the media were changed every 3-4 days.

In all experiments described in this study, parent/parental or DMSO treated cells all refer to untreated, control cells. For treated cells, the conditions (dose and treatment duration) and whether any reagents were used together at specific doses were as indicated in the figure legends.

Immunohistochemistry

Paraffin-embedded SW620 colon cancer cells were immunohistochemically stained to evaluate the protein expression of PD-L1, c-Myc, p38 MAPK, γH2AX and cleaved caspase 3. Following deparaffinization and rehydration, sections of SW620 cells were prepared. The slides were heated in the Retriever 2000 pressure cooker (Electron Microscopy Sciences, Hatfield, PA, USA) in Borg buffer pH 9.5 (Biocare Medical, Concord, CA, USA) and cooled to room temperature. Endogenous peroxidase activity was inactivated with Peroxidazed 1 (Biocare Medical) for 10 min. Non-specific protein interactions were blocked for 10 min with Background Punisher (Biocare Medical). The sections were incubated with the primary antibodies, indicated above, at a dilution of 1/200 for 1 h, washed in TBS and incubated with SignalStain Boost IHC Detection Reagent (Cell Signaling Technology) for 30 min. Following washes in TBS, immunoreactivity was visualized by development with 3,3′-diaminobenzidine (DAB+, Dako, Carpinteria, CA, USA) for 5 min. Immunostained sections were briefly counterstained with CAT hematoxylin, washed in tap water, dehydrated in a graded alcohol series, cleared in xylene and coverslipped with Permount mounting medium (Fisher Scientific Co. L.L.C., Pittsburgh, PA, USA).

Flow cytometric analysis for the surface expression of PD-L1

The cells were harvested and fixed in 4% parafor-maldehyde for 10 min. The cells were then rinsed twice with phosphate-buffered saline (PBS) before a 30-min staining was performed to prepare the samples for flow cytometry using a rabbit anti-human PD-L1 monoclonal antibody (E1L3N) conjugated to phycoerythrin (PE). As controls, cell samples were also stained for 30 min using rabbit IgG isotype monoclonal antibody (DA1E) conjugated to PE. Following labeling with antibody, the cells were rinsed twice with PBS and re-suspended with PBS. The data shown as the geometric means from n=3-4 independent experiments were acquired on a DB LSR Fortessa X-20, and analyzed with FlowJo version 10 software (BD Biosciences, San Jose, CA, USA).

PD-L1 mRNA analyses using the Cancer Cell Line Encyclopedia (CCLE) database

The basal PD-L1 mRNA expression levels shown in Fig. 2B, presented as transcript per million (TPM) were obtained from a public Cancer Cell Line Encyclopedia (CCLE, https://portals.broadinstitute.org/ccle/home) database for cancer cell lines tested.

Figure 2

Effects of resveratrol and piceatannol on programmed cell death ligand 1 (PD-L1) expression in human breast and colon cancer cells. (A) A panel of 4 breast (Cal51, BT549, BT474 and SKBR3) and colorectal (HCT116, SW480, HT29 and SW620) cancer cell lines were incubated with increasing concentrations of resveratrol or piceatannol, as single agents or in combination for 48 h. Following treatment, the cells were harvested and stained and quantified for PD-L1-PE surface expression by flow cytometry. The induction of PD-L1 expression was based on comparison to a DMSO-treated sample (control used throughout all experiments), and to isotype control staining. The geometric mean of MFI of PE area was used as the PD-L1 readout. The levels of PD-L1 from n=3 experiments were converted to a bar graph to represent the respective changes in PD-L1 expression following treatment. The parental condition represents the untreated control. The statistical difference reflects comparison of treated samples to parental condition. (B) RNA-seq analysis of constitutive mRNA expression of PD-L1 in panel of breast and colon cancer cells. Basal PD-L1 mRNA levels, shown as transcript per million (TPM) were obtained from a public Cancer Cell Line Encyclopedia (CCLE) database and ranked from low to high according to the relative TPM expression by most of the breast and colon cancer cell lines tested in (A) https://portals.broadinstitute.org/ccle/home. The asterisks apply to comparisons made among treated samples: *P<0.05, **P<0.01, ***P<0.001. In addition, the 'x' symbol refers to comparisons made between the untreated, control sample with the treated sample: xP<0.05 (treated vs. control).

NF-κB reporter assay

The A549-Dual cells purchased from InvivoGen (San Diego, CA, USA) were used. These are derivatives of the A549 human lung carcinoma cells containing the stable integration of two inducible reporter constructs. The constructs allow for the co-expression of a secreted embryonic alkaline phosphatase (SEAP) reporter gene under the control of the IFN-β minimal promoter fused 5 five NF-κB binding sites, and, a Lucia luciferase gene encoding a secreted luciferase whose transcription is driven by an ISG54 promoter fused to 5 IFN-stimulated response elements. The cells were treated for the specified amount of time (8, 24 and 48 h) with resveratrol or piceatannol, each at 100 μM or combined, each at 50 μM (referred to as combo-100) to yield a concentration of 100 μM and the secreted alkaline phosphatase and Lucia luciferase in the supernatant of the control and treated cells were detected using the Quanti-Blue reagent from InvivoGen. The results were scored by the fluorescence intensity on a Perkin Elmer EnSpire set at a wavelength of 650 nm (PerkinElmer Inc., Shelton, CT, USA). To determine the role of NF-κB in mediating the induction of PD-L1, the IKK inhibitor, BMS-345541, was administered for 24 h in vitro prior to exposure to the combination of piceatannol and resveratrol, each at 50 μM to yield a concentration of 100 μM, for a further 48 h. BMS-345541 was added at increasing concentrations (1, 4 and 8 μM), while the dose of the combination was kept constant.

Cell cycle/apoptosis analysis

The cells were harvested and washed with PBS then re-suspended in cold 1% formaldehyde in PBS solution for 15 min at 4°C. The cells were washed twice in PBS and re-suspended in ice-cold 70% ethanol and stored at −20°C for 2 h prior to analysis. Prior to fluorescence measurement the cells were stained with 4′,6-diamidine-2′-phenylindole dihydrochloride (DAPI). The intensity of blue fluorescence emission of DAPI stained DNA, excited with the UV laser (355 nm) was measured, recorded and analyzed on a MoFlo flow cytometer (Beckman Coulter Life Sciences, Indianapolis IN, USA) using Kaluza fluorescence intensity analysis software (48). Experiments were repeated and representative data are presented.

Statistical analysis

All data are presented as the mean ± the standard error of the mean. A Student's t-test or two-way ANOVA with Bonferroni correction were performed to determine statistical significance between frequencies or mean fluorescence intensities of assessed cell populations using GraphPad Prism 6 (GraphPad Software). Statistical significance of results was as indicated in each figure.

Results

PD-L1 expression is increased by dietary stilbenoids, resveratrol and/or piceatannol in breast and colorectal cancer cells

We first assessed any alterations in PD-L1 levels using the Cal51 breast cancer and HCT116 colon cancer cells treated with stilbenoids, specifically, resveratrol, piceatannol, pterostilbene and 3,5,4′-trimethoxystilbene, for comparison with the flavonoid, myricetin (the chemical structures of the compounds are shown in Fig. 1A). The surface expression of PD-L1 in the control (DMSO-treated, also referred to as parent/parental cells) and treated Cal51 and HCT116 cells was assayed by flow cytometry. Resveratrol significantly increased the expression of PD-L1 in the Cal51 cells, while treatment with piceatannol resulted in a marked increase in the PD-L1 level in HCT116 cells (Fig. 1B).

To determine whether the upregulation of PD-L1 by resveratrol and piceatannol was broadly or uniquely observed in specific breast or colon cancer cell lines, we assayed any alterations in PD-L1 expression using a panel of breast (Cal51, BT549, BT474 and SKBR3) and colorectal (HCT116, SW480, HT29 and SW620) cancer cell lines. In addition, we also determined whether the synergistic upregulation of PD-L1 may result from treatment with the two stilbenoids. The differential increase in PD-L1 expression induced by resveratrol or piceatannol was observed in 2/4 breast and 3/4 colorectal cancer cell lines treated with either of the stilbenoids as a single agent (Fig. 2A). The combination of resveratrol and piceatannol acted synergistically; 50 μM each of resveratrol and piceatannol combined and referred to as 'combo-100' resulted in significantly greater induction of PD-L1 expression; specifically, ≥4.5-fold in the Cal51 and ≥3.5-fold in the SW620 cells than 50 μM of either stilbenoid added alone (Fig. 2A). Gene expression analyses frequently reveal that the relative abundance of mRNA is only weakly or even inversely associated with the level of protein expression (4951). Thus, in this study, to determine whether the differential expression level of endogenous PD-L1 mRNA might contribute to the observed induction of PD-L1 by resveratrol and piceatannol in these two cell lines, relative to the panel of the other studied cell lines with the same cancer type grouping, the basal PD-L1 mRNA expression levels, shown as TPM were obtained from a public Cancer Cell Line Encyclopedia (CCLE, https://portals.broadinstitute.org/ccle/home) database for cancer cell lines tested. In the breast cancer cell lines, the endogenous level of PD-L1 mRNA ranked as follows: Cal51 ≤ BT474 < SKBR3 ≤ BT549 (Fig. 2B); however, the induction of PD-L1 by co-treatment with resveratrol and piceatannol yielded the opposite result: Cal51 (~7-fold) ≥ BT474 (~ 4-fold) > SKBR3 (~2-fold) ≥ BT549 (~2-fold) (Fig. 2A). A similar trend was also observed in the colorectal cancer cells; namely, the endogenous PD-L1 mRNA ranking was as follows: SW620 ≤ HCT116 < HT29 ≤ SW480 (Fig. 2B), whereas the relative induction of PD-L1 decreased from high to low as follows: SW620 (~4-fold) > HCT116 (~3-fold) > SW480 (~3-fold) > HT29 (~1.5-fold) (Fig. 2A). These results suggest that tumors with a low endogenous mRNA level of PD-L1 are more likely to be affected by resveratrol and/or piceatannol, alone or in combination.

NF-κB mediates the upregulation of PD-L1 induced by resveratrol and/or piceatannol

IFN-γ is known to induce PD-L1 expression by upregulating its transcription through the activation of interferon regulatory transcription factor (IRF-1) (52,53). Thus, in this study, we investigated whether the same mechanism may contribute to the resveratrol-and/or piceatannol-mediated induction of PD-L1 expression. The results of immunohistochemistry revealed no change in IRF-1 expression in the SW620 cells treated for 48 h with 'combo-100' compared to the control (data not shown). Since the NF-κB consensus sequence is also present in the PD-L1 gene promoter (52,53), and NF-κB plays a major role in the transcription of PD-L1 by IFN-γ (54-56), in this study, we examined whether the induction of PD-L1 by resveratrol and/or piceatannol was due to the activation of NF-κB. The A549 cells co-expressing the SEAP reporter gene and Lucia luciferase gene were used to investigate the association between NF-κB activity and the PD-L1 expression levels following treatment with the two stilbenoids, alone or combination. In this dual reporter assay, the secreted SEAP and Lucia luciferase in the culture supernatant were separately measured to provide a quantitative readout of the transcriptional impact of NF-κB and the IFN signaling pathways. The time-dependent (≥24 h) and synergistic induction of NF-κB expression induced by piceatannol alone and by combined treatment with resveratrol was observed (Fig. 3A). In response to stimuli, the inhibitory protein IκB is degraded, which leads to the release/translocation of heterodimer p65/p50 from the cytoplasm to the nucleus (57,58). Therefore, the activation of NF-κB by the combined stilbenoids was examined in the SW620 cells by immunohistochemistry to analyze changes in the localization of p65; 48 h of exposure to 'combo-100' resulted in an increase in the translocation and nuclear accumulation of p65, as shown in Fig. 3B (inset).

Figure 3

Control of nuclear factor (NF)-κB-mediated programmed cell death ligand 1 (PD-L1) expression by resveratrol and piceatannol. (A) A549 dual reporter cells were used to assay the activation of NF-κB following treatment with resveratrol or piceatannol, each at 100 μM or combined, each at 50 μM (referred to as combo-100) to yield a concentration of 100 μM. Resveratrol as a single agent resulted in an optimal early time activation of NF-κB, while piceatannol alone decreased NF-κB signaling at 8 h and followed by a progressive increases at 24 and 48 h, respectively. When used in combination, the two stilbenoids led to a marked increase in NF-κB signaling over the tested duration of 48 h. The parental condition represents the untreated control. The data shown are from 1 experiment. (B) Immunohistochemistry of the subcellular localization of NF-κB p65 subunit in SW620 colon cancer cells treated with a combination of piceatannol and resveratrol, each at 50 μM to yield a concentration of 100 μM vs. the untreated cells. The analysis showed that p65 translocated from the cytoplasm to the nucleus by the combined use of the stilbenoids, indicating the activation of NF-κB signaling. Images were captured at ×20 magnification and cropped to show the field of cells representative of the effect of treatment. The results are representative of 5 sections from 1 experiment. We did not quantify the percentage of cells in which translocation occurred in these samples. The parental condition represents the control. The image in the inset represents a high magnification image of the cells in the field of view. (C) BMS-345541, an inhibitor of IKK, was administered for 24 h in vitro prior to exposure to the combination of piceatannol and resveratrol, each at 50 μM to yield a concentration of 100 μM, for a further 48 h. BMS-345541 was added at increasing concentrations (1, 4 and 8 μM) while the dose of the combination was kept constant. Following treatment, the cells were harvested and stained for PD-L1 expression by flow cytometry. The geometric mean of the mean fluorescent intensity (MFI) of the phytoerythrin (PE) area was used as the readout for PD-L1 expression. The parental condition represents the untreated control. The results are representative of 3 experiments.

The small molecule, BMS-345541, is an IKK kinase inhibitor (59) that prevents IκB phosphorylation, to effectively suppress the translocation of NF-κB into the nucleus for participation in transcriptional activation of NF-κB-responsive genes (57,58). Thus, in this study, we then examined whether the stilbenoid-induced PD-L1 expression can be blocked by BMS-345541. In SW620 cells, the induction of PD-L1 by 'combo-100' was inhibited by 48% with 1 μM BMS-345541 (Fig. 3C). Furthermore, a dose-dependent inhibition was observed with the 1, 4 and 8 μM of IKK inhibitor concentration range (Fig. 3C). These results, showing that the inhibition of IKK significantly decreased PD-L1 expression in SW620 cells suggest that NF-κB activation is involved in the induction of PD-L1 expression by resveratrol or piceatannol either alone, or in combination.

Histone deacetylase inhibitors (HDACis) and histone acetyltransferase inhibitors (HATis) modulate the induction of PD-L1 expression induced by the combination of resveratrol and piceatannol

The expression of PD-L1 can be regulated via histone acetylation/deacetylation (60,61) and resveratrol is an activator of HDAC (62). To investigate whether the induction of PD-L1 by the combination of resveratrol and piceatannol is blocked by inhibitors of HDAC or HAT, HDACis (vorinostat, mocetinostat, resminostat and entinostat) and HATis (curcumin, garcinol, anacardic acid, MB-3 and Tip60i) were used to assess their effects on the modulation of PD-L1 by the combined use of the stilbenoids. The cells were pre-treated for 48 h with individual HDACis/HATis alone or in combination with 60 μM of either of the stilbenoids, followed by the flow cytometric analysis of PD-L1 expression. The addition of HDACis or HATis alone did not affect PD-L1 expression compared to the untreated controls (Fig. 4). When the SW620 cells were pre-treated with histone modification inhibitors, the ability of the stilbenoids to induce PD-L1 was markedly reduced by two HDACis (5 μM of resminostat and entinostat) and also by the HATi, anacardic acid (1 μM) (Fig. 5). These results demonstrated that histone modification inhibitors can suppress the induction of PD-L1 expression by stilbenoids. The data are consistent with the interpretation that the upregulation of PD-L1 by stilbenoids involves transcriptional control.

Induction of apoptotic and cell cycle changes by the combined use of resveratrol and piceatannol

The upregulation of PD-L1 may allow cancers to evade the host immune system and acquire resistance to anticancer drugs. Having demonstrated that the upregulation of PD-L1 expression by stilbenoids in the SW620 colon cancer cells, we then investigated whether stilbenoids affect the survival status of cells by analyzing two biomarkers related to apoptosis, namely, the expression of the DNA damage indicator γH2AX, and that of cleaved caspase 3. In addition, markers associated with cell survival, p38-MAPK and c-Myc, were also assessed using immunohistochemistry. Treatment of the SW620 cells for 48 h with 'combo-100' resveratrol and piceatannol increased the expression of γH2AX and that of cleaved caspase 3, and downregulated the p38-MAPK and c-Myc levels (Fig. 6A). The induction of γH2AX is characteristic of DNA fragmentation and damage during apoptosis, and thus supports the interpretation that exposure to resveratrol and/or piceatannol causes DNA damage and apoptosis via the activation of caspase 3. We then assessed whether treatment with the stilbenoids altered cell cycle distribution by flow cytometric analysis. An increase in the percentage of cells in the S phase of the cell cycle, from 19 to ~30%, a distinct reduction in the proportion of G1 phase cells (from 66 to ~30%), and a marked decrease in the percentage of G2M phase cells, from 12 to ~3% were observed. There was also an increase in the percentage of cells with fractional DNA content ('sub-G1 cells'), an indication of apoptosis from 2.85% in the control cells to 31.84, 35.08 and 36.55% in the cells treated with 50 μM resveratrol, 50 μM piceatannol or 50 μM of these two compounds for 48 h, respectively (Fig. 6B). We also examined and determined that low-dose (10 μM) resveratrol and/or piceatannol did not induce PD-L1 expression, whereas the analysis of cell cycle phase distribution changes using Cal51 and SW620 cell cultures treated for 48 h revealed an apparent increase in the proportion of S-phase cells concomitant with the reduction of G2M-phase cells in cultures treated with resveratrol alone, and to an even greater extent following treatment with both stilbenoids (Fig. 7). In addition, the combination of resveratrol and piceatannol induced apoptosis in both cell types to a much greater degree than each of them alone, as demonstrated by an increase in the percentage of cells with fractional DNA content ('sub-G1 cells'), an indication of apoptosis (compare Fig. 7A with Fig. 7B–D) (48,63,64). Taken together, our findings indicate that stilbenoids not only increase PD-L1 expression, but may also induce DNA damage, leading to increased cell death in tumor cells, such as SW620 colon cancer cells.

Discussion

An association has been observed between a decrease in T-cell proliferation and an increase in apoptosis and tumor immune evasion, with an increase in the expression of T-cell inhibitory protein PD-L1 on cancer cells (2,3), providing the impetus for understanding the control of PD-L1 expression using cancer cells as a model. In this study, we examined the hypothesis that dietary stilbenoids may act as modulators of PD-L1 expression in tumor cells. We focused on the effects of resveratrol, a grape-derived polyphenol that has shown chemopreventive effects in various cancer types (3342), and its hydroxylated derivative, piceatannol. Questions raised and addressed in this study included whether: i) Structurally-modified stilbenoids have the same ability to modulate PD-L1 expression; ii) the effects of stilbenoids on PD-L1 expression can apply to different cancer types; iii) signaling pathways that control the stilbenoid-induced PD-L1 expression are effected; and iv) histone modification inhibitors (HDACis and HATis) block stilbenoid-induced PD-L1 expression.

Stilbenoids in general exert beneficial effects on human health (65). It has been reported that the 4′-hydroxy group of resveratrol is essential for its bioactivity (66). In this study, we used breast and colorectal cancer cells to compare the modulatory effects of stilbenoids on PD-L1 expression, namely, piceatannol (3′,4′,3,5-trans-trihydroxystilbene), 3,5,4′-trimethoxystilbene, and pterostilbene (3,5-dimethoxy-4′-hydroxy-E-stilbene), with resveratrol (3,5,4′-trans-trihydroxystilbene) and myricetin, a naturally occurring flavonoid present in abundance in edible foods (67). The results presented in Figs. 1 and 2 demonstrate that the hydroxyl, but not methoxy groups on stilbenoids are important for the induction of PD-L1. However, no increase in PD-L1 expression was evident in the cells treated with myricetin (3,5,7-trihydroxy-2-(3,4,5-trihydroxyphenyl)-4-chromenone), a flavonoid that contains 6 hydroxyl groups (Fig. 1). These results suggest that the 3,5-dihydroxy-trans-stilbene structure is required for the induction of PD-L1 in the cells used.

The expression level PD-L1 of was significantly increased in the breast and colon cancer cells treated with resveratrol and piceatannol, both as a single agent and when used in combination (Figs. 1 and 2). Moreover, PD-L1 expression was synergistically upregulated 4.5-fold in the Cal51 breast cancer and ≥3.5-fold in the SW620 colon cancer cells by the combined use of resveratrol and piceatannol (Fig. 2A). Of note, both cancer cells express low basal levels of PD-L1 mRNA expression (Fig. 2B). Since, as noted, a positive response and improved clinical outcome to anti-PD-L1 blockade therapy are best observed in patients with PD-L1-positive tumors (presumably reflecting a high expression of PD-L1), we surmise that agents capable of upregulating PD-L1 expression in tumor cells expressing low PD-L1 can sensitize such cancer cells for an improved response to PD-L1 blockade. As a corollary, we also hypothesized that the combined use of resveratrol and piceatannol co-administered with anti-PD-L1 immunotherapy may exhibit clinical benefits in cancer patients with no-or- low-PD-L1 tumors. Whether the efficacy to PD-L1 blockade may be enhanced by the combined intake of resveratrol and/or piceatannol, concomitantly or sequentially, remains to be verified.

This study also provides evidence showing that the resvera-trol- and piceatannol-mediated upregulation of PD-L1 requires the activation of NF-κB (Fig. 3). In the SW620 cells, we showed that the induction of PD-L1 expression induced by resveratrol/piceatannol was attenuated by the IKK inhibitor, BMS 345541 (Fig. 3C). It has been previously demonstrated that the duration and function of nuclear NF-κB is regulated by reversible acetylation/deacetylation (68), and that NF-κB transcriptional response is controlled by the HDAC3-mediated deacetylation of RelA acting as an intranuclear molecular switch for turning 'on-off' the NF-κB response (68); we therefore examined whether histone modification inhibitors affect the resveratrol/ piceatannol-mediated transcriptional control of PD-L1. Our findings revealed that two HDACis (e.g., resminostat and entinostat) and one HATi, anacardic acid, effectively blocked the induction of PD-L1 expression by resveratrol/piceatannol (Fig. 5). Both resminostat and entinostat are HDAC3 inhibitors, thus lending support that the HDAC3-mediated NF-κB response plays a role in resveratrol/piceatannol-induced PD-L1 expression. The suppression of PD-L1 induction by resveratrol using anacardic acid is in accordance with the described effect of anacardic acid as a HATi for p300 and p300/CBP and data reporting that resveratrol is a p300 activator (69,70). These results indicate that the expression of PD-L1 is regulated by the mechanism of histone acetylation/deacetylation and that resveratrol/piceatannol induces PD-L1 expression through HDAC3/p300-mediated NF-κB control.

It should be mentioned that the upregulation of PD-L1 by resveratrol or piceatannol occur at doses not achievable physiologically and may exceed pharmacologically relevant concentrations (26,71). Conceivably, the effective dose could also be modulated by factors present locally at the site of responsive cells/tumors (e.g., different hormones, cytokines, products of cell metabolism or variable oxygen tension) and thus may additionally affect sensitivity of PD-1/PD-L1 checkpoint to these compounds, perhaps amplifying their potential anticancer effect. It should also be noted that the doses used in the present experiments were based on titration studies (data not shown), and that the effectiveness of single or combined agents on the induction of PD-L1 in each cell line is above IC50. It would be of interest to determine what might account for the variations observed in dosage dependence in different cell lines. Since the induction of PD-L1 expression by resveratrol and piceatannol are mediated through the NF-κB signaling pathway; the different dose-dependent responses and the upregulation of PD-L1 by resveratrol and piceatannol may be due to the variation in the endogenous level of NF-κB components, vis-à-vis, NF-κB1 (p105), NF-κB2 (p65), CHUK (IKK-α), IKBKB (IKK-β) and IKBKG (IKK-γ), in each of the cell lines tested. In RNA-seq analyses, we found that Cal51 and SW620, both with a low endogenous level of PD-L1 expression, expressed high levels NF-κB2 (p65) and CHUK (IKK-α) compared to cancer cell lines from same anatomical origin showing high PD-L1 expression (data not shown). Thus, it is tempting to propose that response in the induction of PD-L1 by stilbenoids in different cell lines from identical cancer types may be attributed to the level of expression of NF-κB2 (p65) and CHUK (IKK-α). Currently, experiments are underway to further test and confirm our hypothesis.

Another result of note in this study is that the SW620 cells exposed to a high dose of both stilbenoids were partially restricted in cell cycle transition in the G2/M phase and display evidence of apoptosis (Fig. 6). Furthermore, the accumulation of cells in the S phase of the cell cycle may also be associated with an increase in their expression of PD-L1. This suggests that resveratrol and piceatannol affect cancer cells by a dual mechanism: i) The induction of PD-L1 that sensitizes tumor cells for recognition by anti-PD-L1 antibodies; an effect that could diminish cancer cell evasion from immune surveillance; and ii) the direct induction of cell cycle arrest, increase in DNA damage and cancer cell destruction via induction of apoptosis.

Since cancer patients expressing tumors positive for PD-L1, a negative T-cell regulatory molecule, demonstrate efficacy to anti-PD-L1 blockade therapy with an improved clinical outcome, one might surmise that low PD-L1-expressing tumors may be sensitized and may display an improved responsiveness to PD-L1 blockade therapy using dietary agents. The cell culture experiments used in this study may be considered as a model for testing whether the sensitivity and responsiveness of tumor cells to PD-L1 targeted therapy can be augmented by priming with or co-exposure to stilbenoids, such as resveratrol and/or piceatannol. The hypothesis raised is as follows: The upregulation of membrane-associated PD-L1 in low PD-L1-expressing tumor cells is a 'find-me' approach for targeting by immune checkpoint inhibitors to potentially improve the effi-cacy of anti-PD-L1 blockade therapy via stilbenoids. Indeed, we believe that the elevation of PD-L1 expression, as we have demonstrated in this study using pharmacological doses of the stilbenoids, resveratrol and piceatannol, may underlie the unresolved challenge in that the positive response to immune checkpoint blockade therapy in 19-31% of treated individuals is a limited to number of clinical indications, typically in patients whose tumors express elevated-PD-L1, which we stated explicitly in the Introduction. Thus, while on teleological grounds, the upregulation of PD-L1 by polyphenols in cancer could promote disease progression, we offer the consideration that the observed stilbenoid-induced PD-L1 increase be viewed from the hypothesis that agents capable of upregulating PD-L1 expression in tumor cells could sensitize cancer cells for an improved clinical response to PD-L1 immune checkpoint blockade therapy. Testing these aspects would constitute a novel approach to confirm our hypothesis. These possibilities are under further investigative considerations in our laboratory. Studies are also planned to explore whether stilbenoids may impact host immune response, for example, by affecting PD-1 expression in PD-1-expressing Jurkat T-cells.

Funding

This study was supported in part by the Robert A. Welke Cancer Research Foundation (to HDH and ZD).

Availability of data and materials

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

Authors' contributions

JL performed experiments and analyzed the data (cell culture analysis, immunohistochemistry, flow cytometric analysis, NF-κB reporter assay and statistical analysis) and edited the manuscript. TCH interpreted the data and wrote the manuscript. HDH collected and analyzed/interpreted the data (cell cycle and apoptosis analyses). ZD interpreted the data and wrote the section of cell cycle and apoptosis analyses and edited the manuscript. JMW interpreted the data, and wrote and edited the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors acknowledge that the RNA-seq data presented in Fig. 2B were obtained from the Cancer Cell Line Encyclopedia (CCLE) database (https://portals.broadinstitute.org/ccle/home).

References

1 

Okazaki T and Honjo T: PD-1 and PD-1 ligands: From discovery to clinical application. Int Immunol. 19:813–824. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al: Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat Med. 8:793–800. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, Shin T, Tsuchiya H, Pardoll DM, Okumura K, et al: Expression of programmed death 1 ligands by murine T cells and APC. J Immunol. 169:5538–5545. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, Honjo T, Okazaki T and Tokura Y: Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer. 116:1757–1766. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, Saudemont A and Quesnel B: Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 110:296–304. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, Mikse OR, Cherniack AD, Beauchamp EM, Pugh TJ, et al: Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 3:1355–1363. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, Bruno TC, Richmon JD, Wang H, Bishop JA, et al: Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res. 73:1733–1741. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Mittal D, Gubin MM, Schreiber RD and Smyth MJ: New insights into cancer immunoediting and its three component phases–elimination, equilibrium and escape. Curr Opin Immunol. 27:16–25. 2014. View Article : Google Scholar : PubMed/NCBI

10 

He J, Hu Y, Hu M and Li B: Development of PD-1/PD-L1 Pathway in Tumor Immune Microenvironment and Treatment for Non-Small Cell Lung Cancer. Sci Rep. 5:131102015. View Article : Google Scholar : PubMed/NCBI

11 

Noh H, Hu J, Wang X, Xia X, Satelli A and Li S: Immune checkpoint regulator PD-L1 expression on tumor cells by contacting CD11b positive bone marrow derived stromal cells. Cel. Commun Signal. 13:142015. View Article : Google Scholar

12 

Kim JM and Chen DS: Immune escape to PD-L1/PD-1 blockade: Seven steps to success (or failure). Ann Oncol. 27:1492–1504. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, Brahmer JR, Lawrence DP, Atkins MB, Powderly JD, et al: Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 32:1020–1030. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Gadiot J, Hooijkaas AI, Kaiser AD, van Tinteren H, van Boven H and Blank C: Overall survival and PD-L1 expression in metastasized malignant melanoma. Cancer. 117:2192–2201. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al: Nivolumab versus docetaxel in advanced nonsquamous von-small-cell lung cancer. N Engl J Med. 373:1627–1639. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al: Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 373:123–135. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, et al: Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet. 387:1540–1550. 2016. View Article : Google Scholar

20 

Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al: CheckMate 025 investigators: Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 373:1803–1813. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Chen L and Flies DB: Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 13:227–242. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Yang Y: Cancer immunotherapy: Harnessing the immune system to battle cancer. J Clin Invest. 125:3335–3337. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Chen L and Han X: Anti-PD-1/PD-L1 therapy of human cancer: Past, present, and future. J Clin Invest. 125:3384–3391. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zou W, Wolchok JD and Chen L: PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med. 8:328rv42016. View Article : Google Scholar : PubMed/NCBI

25 

Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al: PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 372:311–319. 2015. View Article : Google Scholar :

26 

Hsieh TC and Wu JM: Resveratrol: Biological and pharmaceutical properties as anticancer molecule. Biofactors. 36:360–369. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Saud SM, Li W, Morris NL, Matter MS, Colburn NH, Kim YS and Young MR: Resveratrol prevents tumorigenesis in mouse model of Kras activated sporadic colorectal cancer by suppressing oncogenic Kras expression. Carcinogenesis. 35:2778–2786. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Jang M, Cai L, Udeani GO, Slowing KV, Thomas CF, Beecher CW, Fong HH, Farnsworth NR, Kinghorn AD, Mehta RG, et al: Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science. 275:218–220. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Rahman I, Biswas SK and Kirkham PA: Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol. 72:1439–1452. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Dvorakova M and Landa P: Anti-inflammatory activity of natural stilbenoids: A review. Pharmacol Res. 124:126–145. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Park EJ and Pezzuto JM: The pharmacology of resveratrol in animals and humans. Biochim Biophys Acta. 1852:1071–1113. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Britton RG, Kovoor C and Brown K: Direct molecular targets of resveratrol: Identifying key interactions to unlock complex mechanisms. Ann N Y Acad Sci. 1348:124–133. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Harikumar KB and Aggarwal BB: Resveratrol: A multitargeted agent for age-associated chronic diseases. Cell Cycle. 7:1020–1035. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Tennen RI, Michishita-Kioi E and Chua KF: Finding a target for resveratrol. Cell. 148:387–389. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Zykova TA, Zhu F, Zhai X, Ma WY, Ermakova SP, Lee KW, Bode AM and Dong Z: Resveratrol directly targets COX-2 to inhibit carcinogenesis. Mol Carcinog. 47:797–805. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Ito Y, Mitani T, Harada N, Isayama A, Tanimori S, Takenaka S, Nakano Y, Inui H and Yamaji R: Identification of carbonyl reductase 1 as a resveratrol-binding protein by affinity chromatography using 4′-amino-3,5-dihydroxy-trans-stilbene. J Nutr Sci Vitaminol (Tokyo). 59:358–364. 2013. View Article : Google Scholar

37 

Buryanovskyy L, Fu Y, Boyd M, Ma Y, Hsieh TC, Wu JM and Zhang Z: Crystal structure of quinone reductase 2 in complex with resveratrol. Biochemistry. 43:11417–11426. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Hsieh TC, Wang Z, Deng H and Wu JM: Identification of glutathione sulfotransferase-pi (GSTP1) as a new resveratrol targeting protein (RTP) and studies of resveratrol-responsive protein changes by resveratrol affinity chromatography. Anticancer Res. 28A:29–36. 2008.

39 

Wang Z, Hsieh TC, Zhang Z, Ma Y and Wu JM: Identification and purification of resveratrol targeting proteins using immobilized resveratrol affinity chromatography. Biochem Biophys Res Commun. 323:743–749. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Athar M, Back JH, Kopelovich L, Bickers DR and Kim AL: Multiple molecular targets of resveratrol: Anti-carcinogenic mechanisms. Arch Biochem Biophys. 486:95–102. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Hsieh TC, Lin CY, Bennett DJ, Wu E and Wu JM: Biochemical and cellular evidence demonstrating AKT-1 as a binding partner for resveratrol targeting protein NQO2. PLoS One. 9:e1010702014. View Article : Google Scholar : PubMed/NCBI

42 

Calleri E, Pochetti G, Dossou KSS, Laghezza A, Montanari R, Capelli D, Prada E, Loiodice F, Massolini G, Bernier M, et al: Resveratrol and its metabolites bind to PPARs. ChemBioChem. 15:1154–1160. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Yang Y, Paik JH, Cho D, Cho JA and Kim CW: Resveratrol induces the suppression of tumor-derived CD4+CD25+ regulatory T cells. Int Immunopharmacol. 8:542–547. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Zou T, Yang Y, Xia F, Huang A, Gao X, Fang D, Xiong S and Zhang J: Resveratrol inhibits CD4+ T cell activation by enhancing the expression and activity of Sirt1. PLoS One. 8:e751392013. View Article : Google Scholar

45 

Craveiro M, Cretenet G, Mongellaz C, Matias MI, Caron O, de Lima MCP, Zimmermann VS, Solary E, Dardalhon V, Dulić V, et al: Resveratrol stimulates the metabolic reprogramming of human CD4+ T cells to enhance effector function. Sci Signal. 10:102017. View Article : Google Scholar

46 

Patel KR, Scott E, Brown VA, Gescher AJ, Steward WP and Brown K: Clinical trials of resveratrol. Ann N Y Acad Sci. 1215:161–169. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Patel KR, Brown VA, Jones DJ, Britton RG, Hemingway D, Miller AS, West KP, Booth TD, Perloff M, Crowell JA, et al: Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 70:7392–7399. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Darzynkiewicz Z, Bruno S, Del Bino G, Gorczyca W, Hotz MA, Lassota P and Traganos F: Features of apoptotic cells measured by flow cytometry. Cytometry. 13:795–808. 1992. View Article : Google Scholar : PubMed/NCBI

49 

Maier T, Güell M and Serrano L: Correlation of mRNA and protein in complex biological samples. FEBS Lett. 583:3966–3973. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Vogel C and Marcotte EM: Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 13:227–232. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Liu Y, Beyer A and Aebersold R: On the dependency of cellular protein levels on mRNA abundance. Cell. 165:535–550. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Ritprajak P and Azuma M: Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol. 51:221–228. 2015. View Article : Google Scholar

53 

Lee SJ, Jang BC, Lee SW, Yang YI, Suh SI, Park YM, Oh S, Shin JG, Yao S, Chen L, et al: Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1 (CD274). FEBS Lett. 580:755–762. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Chen J, Feng Y, Lu L, Wang H, Dai L, Li Y and Zhang P: Interferon-γ-induced PD-L1 surface expression on human oral squamous carcinoma via PKD2 signal pathway. Immunobiology. 217:385–393. 2012. View Article : Google Scholar

55 

Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, Yoshioka Y, Baba T, Konishi I and Mandai M: IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br J Cancer. 112:1501–1509. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Mandai M, Hamanishi J, Abiko K, Matsumura N, Baba T and Konishi I: Dual faces of IFNγ in cancer progression: A role of PD-L1 induction in the determination of pro- and antitumor immunity. Clin Cancer Res. 22:2329–2334. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Goto K, Chiba Y and Misawa M: IL-13 induces translocation of NF-kappaB in cultured human bronchial smooth muscle cells. Cytokine. 46:96–99. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Hideshima H, Yoshida Y, Ikeda H, Hide M, Iwasaki A, Anderson KC and Hideshima T: IKKβ inhibitor in combination with bortezomib induces cytotoxicity in breast cancer cells. Int J Oncol. 44:1171–1176. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Burke JR, Pattoli MA, Gregor KR, Brassil PJ, MacMaster JF, McIntyre KW, Yang X, Iotzova VS, Clarke W, Strnad J, et al: BMS-345541 is a highly selective inhibitor of I kappa B kinase that binds at an allosteric site of the enzyme and blocks NF-kappa B-dependent transcription in mice. J Biol Chem. 278:1450–1456. 2003. View Article : Google Scholar

60 

Kroesen M, Gielen P, Brok IC, Armandari I, Hoogerbrugge PM and Adema GJ: HDAC inhibitors and immunotherapy; a double edged sword. Oncotarget. 5:6558–6572. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Woods DM, Sodré AL, Villagra A, Sarnaik A, Sotomayor EM and Weber J: HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res. 3:1375–1385. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, et al: Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 425:191–196. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Kajstura M, Halicka HD, Pryjma J and Darzynkiewicz Z: Discontinuous fragmentation of nuclear DNA during apoptosis revealed by discrete 'sub-G1' peaks on DNA content histograms. Cytometry A. 71:125–131. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Darzynkiewicz Z, Halicka HD and Zhao H: Analysis of cellular DNA content by flow and laser scanning cytometry. Adv Exp Med Biol. 676:137–147. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Akinwumi BC, Bordun KM and Anderson HD: Biological activities of stilbenoids. Int J Mol Sci. 19:192018. View Article : Google Scholar

66 

Matsuoka A, Takeshita K, Furuta A, Ozaki M, Fukuhara K and Miyata N: The 4′-hydroxy group is responsible for the in vitro cytogenetic activity of resveratrol. Mutat Res. 521:29–35. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Semwal DK, Semwal RB, Combrinck S and Viljoen A: Myricetin: A dietary molecule with diverse biological activities. Nutrients. 8:902016. View Article : Google Scholar : PubMed/NCBI

68 

Chen Lf, Fischle W, Verdin E and Greene WC: Duration of nuclear NF-kappaB action regulated by reversible acetylation. Science. 293:1653–1657. 2001. View Article : Google Scholar

69 

Shakibaei M, Buhrmann C and Mobasheri A: Resveratrol-mediated SIRT-1 interactions with p300 modulate receptor activator of NF-kappaB ligand (RANKL) activation of NF-kappaB signaling and inhibit osteoclastogenesis in bone-derived cells. J Biol Chem. 286:11492–11505. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Zeng Z, Cheng S, Chen H, Li Q, Hu Y, Wang Q, Zhu X and Wang J: Activation and overexpression of Sirt1 attenuates lung fibrosis via P300. Biochem Biophys Res Commun. 486:1021–1026. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Gescher AJ and Steward WP: Relationship between mechanisms, bioavailibility, and preclinical chemopreventive efficacy of resveratrol: A conundrum. Cancer Epidemiol Biomarkers Prev. 12:953–957. 2003.PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 53 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lucas J, Hsieh T, Halicka H, Darzynkiewicz Z and Wu JM: Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling. Int J Oncol 53: 1469-1480, 2018
APA
Lucas, J., Hsieh, T., Halicka, H., Darzynkiewicz, Z., & Wu, J.M. (2018). Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling. International Journal of Oncology, 53, 1469-1480. https://doi.org/10.3892/ijo.2018.4512
MLA
Lucas, J., Hsieh, T., Halicka, H., Darzynkiewicz, Z., Wu, J. M."Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling". International Journal of Oncology 53.4 (2018): 1469-1480.
Chicago
Lucas, J., Hsieh, T., Halicka, H., Darzynkiewicz, Z., Wu, J. M."Upregulation of PD‑L1 expression by resveratrol and piceatannol in breast and colorectal cancer cells occurs via HDAC3/p300‑mediated NF‑κB signaling". International Journal of Oncology 53, no. 4 (2018): 1469-1480. https://doi.org/10.3892/ijo.2018.4512