Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity. Thus, DCs have been regarded as a major target of immunosuppressants for the control of harmful immune responses. In this study, we examined the effect of quercetin, a natural flavonoid found in many vegetables and fruits, on the activation and function of mouse DCs. Quercetin effectively inhibited LPS-induced DC activation by reducing the production of proinflammatory cytokines/chemokines and the expression levels of MHC class II and costimulatory molecules. In addition, quercetin uniquely blocked endocytosis by DCs and the LPS-induced DC migration was diminished by quercetin treatment. Furthermore, quercetin abrogated the ability of LPS-stimulated DCs to induce Ag-specific T cell activation, both in vitro and in vivo. Remarkably, coadministration of quercetin with 2,4-dinitro-1-fluorobenzene prevented 2,4-dinitro-1-fluorobenzene–induced contact hypersensitivity, indicating the potential of quercetin for treating delayed-type hypersensitive diseases. Blockage of LPS-induced ERK, JNK, Akt, and NF-κB activation contributed to the inhibitory effect of quercetin on DCs. These results strongly suggest that quercetin may be a potent immunosuppressive agent and could be used in the prevention and therapy of chronic inflammation, autoimmunity, and transplantation via the abolishment of DC activation and function.

Dendritic cells (DCs), which are the most potent APCs, are critical sentinels of the immune system and have the unique ability to integrate a wide array of incoming signals and convey them to lymphocytes, thus directing the appropriate immune responses against microbial pathogens and tumors (1). After encountering pathogen-associated molecular patterns, DCs become mature to enhance the expression of MHC class II and accessory molecules and produce cytokines and chemokines (2). TLRs are the major pattern recognition receptors in DCs and initiate DC function to regulate immune responses via various signaling pathways (3). Thus, the use of DCs to control cancer and infectious diseases has been proposed (4). However, DCs are also involved in the pathogenesis of several diseases caused by immune dysfunction, such as chronic inflammation and autoimmunity (5, 6). Substances downregulating DC activation can potentially be applied to the treatment of these diseases.

Traditionally, natural products represent a source for discovering bioactive drugs, including immunomodulators, in the pharmaceutical industry (7). Flavonoids are natural products from many vegetables, fruits, herbs, flowers, seeds, and beverages that are present in diet (8). They are polyphenolic compounds and display a variety of biological effects, such as antioxidation, anti-inflammation, anticancer, gastroprotection, cardiovascular protection, oral protection, and memory improvement (913). In addition, flavonoid compounds have been shown to regulate immune responses (14). Thus, flavonoids may have the potential to modulate DC function in the immune system.

There are several subclasses in the flavonoid family. Quercetin (3,5,7,3′,4′-pentahydroxyflavone), which is a typical member in the flavonol subclass, is one of the most common flavonoids in the diet. Its wide range of biological activities has been discussed for several decades (15). In addition, quercetin exerts antimicrobial, antihypertensive, neuroprotective, and chemoprotective effects (16). The immunomodulatory activity of quercetin has been investigated in NK cells (17), macrophages (18), mast cells (19), neutrophils (20), B cells (21), and T cells (22). Although a number of studies have reported the activity of flavonoids in the modulation of DC activation (2327), the effect of quercetin on DCs remains unknown. In this study, we examined the immunomodulatory effect of quercetin on DC activation. Our results showed that quercetin inhibited DC maturation and function, suggesting that quercetin may be a potent immunosuppressant and may have therapeutic applications in inflammatory diseases, such as periodontitis.

C57BL/6 mice were obtained from the National Laboratory Animal Center (Taipei, Taiwan). DCs were generated from mouse bone marrow, as described previously (28). OT-II TCR transgenic mice were provided by Dr. C. Lowell (University of California, San Francisco, CA). All animals were kept in a specific pathogen-free facility (National Health Research Institutes, Miaoli, Taiwan) and handled according to protocols approved by the Institutional Animal Care and Use Committee of the National Health Research Institutes.

DCs were treated with quercetin (dissolved in DMSO and made a stock solution at 25 mM; Sigma-Aldrich, St. Louis, MO) at the indicated concentrations in the absence or presence of LPS for 24 h. DMSO was <0.8% (v/v) in all experiments. Cells were then harvested and stained using the Annexin V kit (Invitrogen, Carlsbad, CA), according to the manufacturer’s instructions. Apoptosis was determined by flow cytometry.

Supernatants were collected from DCs (1 × 106/ml) treated with the indicated conditions. TLR ligands, including LPS (100 ng/ml), zymosan (20 μg/ml), lipoteichoic acid (20 μg/ml) (Sigma-Aldrich), polyinosine polycytidylic acid poly(I:C) (250 μg/ml), flagellin (500 ng/ml), synthetic diacylated lipoprotein Pam2Cys-Ser-Lys4 (20 ng/ml), synthetic CpG oligodeoxynucleotides 1826 (200 nM), and peptidoglycan from Staphylococcus aureus (1 μg/ml) (InvivoGen, San Diego, CA) were used to stimulate DCs. The production of cytokines (TNF-α, IL-1α, IL-1β, IL-6, IL-10, and IL-12 p70) and chemokines (MCP-1, MIP-1α, MIP-1β, and RANTES) was measured using ELISA (R&D Systems, Minneapolis, MN; PeproTech, Rocky Hill, NJ; and eBioscience, San Diego, CA).

DC maturation was determined by the upregulation of MHC class II and costimulatory molecule expression, as described previously (29). Cells were treated with LPS (100 ng/ml), quercetin (50 μM), LPS plus DMSO (0.2%), or LPS plus quercetin for 16 h and stained with mAbs specific for mouse CD11c, I-Ab, CD40, CD80, and CD86 (BioLegend, San Diego, CA) and then analyzed by flow cytometry. The endocytosis by DCs was assessed by dextran-FITC uptake, as performed previously (30). Cells were untreated or treated with LPS (for 1 or 16 h) or quercetin (for 1 h) and then incubated with dextran-FITC. The uptake of dextran-FITC by DCs was detected by flow cytometry.

DCs were treated with LPS (100 ng/ml), quercetin (50 μM), LPS plus DMSO (0.2%), or LPS plus quercetin for 16 h. DC chemotaxis was performed in 24-well Transwell chambers (pore size, 5 μm; Costar Corning, Lowell, MA), as described previously (31). DCs were loaded onto the upper chambers and CCL21 (200 ng/ml; PeproTech) was added to the lower chambers. After incubation for 3 h at 37°C, migrated cells were collected from the lower wells, and the number of CD11c+ cells was determined by flow cytometry. For DC migration in vivo, cells (2 × 106) were labeled with CFSE for 10 min and then injected into the footpad. After 2 d, inguinal lymph node (LN) cells were isolated and the number of CFSE+ cells was calculated by hemocytometry and flow cytometry.

Ag presentation by DCs was analyzed using the OVA-specific T cell proliferation in vitro assay, as described previously (30). Purified DCs were pulsed with OVA323–339 peptide (2 μg/ml) and incubated with LPS (100 ng/ml), quercetin (50 μM), LPS plus DMSO (0.2%), or LPS plus quercetin for 16 h. Cells were then washed with media to remove quercetin. OT-II T cells were added to DC cultures at various DC:T cell ratios, as indicated, and T cell proliferation was determined by [3H]thymidine incorporation. For recall assay in vivo, C57BL/6 mice were immunized with OVA (10 μg; Sigma-Aldrich) and IFA (Sigma-Aldrich) mixed with quercetin (50 μg), LPS (10 μg), LPS plus DMSO, or LPS plus quercetin, via footpad injection. After 10 d, the draining LN cells were cultured with OVA at the indicated concentrations and T cell proliferation was determined by [3H]thymidine incorporation. To measure IFN-γ production, supernatants were collected from DC/OT II T cell and LN cell cultures, and the amount of IFN-γ was determined by ELISA (eBioscience).

2,4-Dinitro-1-fluorobenzene (DNFB; Sigma-Aldrich)-induced contact hypersensitivity (CHS) was described previously (32). The abdomen of mice were painted with vehicle (olive oil/acetone = 1/4), DNFB (0.5% w/v), DNFB plus DMSO, or DNFB plus quercetin (50 μg) for sensitization. In addition to painting, we also used DNFB-pulsed DCs to sensitize mice. Briefly, DCs were treated with vehicle, 0.1% DNFB, DNFB plus DMSO (0.2%), or DNFB plus quercetin (50 μM) for 30 min at 37°C, and 5 × 105 cells were then s.c. injected into mice. After 5 d, all mice were painted on the ears with DNFB (0.2%). CHS response was determined 24 h later by histological analysis using H&E staining. The increase of ear thickness was calculated as follows: 100 × (thickness of the challenged ear − thickness of the unchallenged ear)/thickness of unchallenged ear.

As previously described (30), DCs were treated with DMSO (0.2%) or quercetin (50 μM) and immediately stimulated with LPS (200 ng/ml). Cells were harvested and lysed at the indicated time points, and SDS-PAGE and Western blotting were performed. The phosphorylated (all Abs from Epitomics, Burlingame, CA) and unphosphorylated (all Abs, including anti–I-κB, from Santa Cruz Biotechnology, Santa Cruz, CA) proteins of p38 MAPK, ERK, JNK, and Akt, were determined in cell lysates.

Significance of the inhibition during TLR ligand and quercetin cotreatment in comparison with TLR ligand treatment alone was determined using a Student t test with two-sample equal variance with a two-tailed distribution. A value of p < 0.05 was considered significant.

The anti-inflammatory activity of quercetin in macrophages has been identified; however, the effect of quercetin on DC activation and function is not known. Therefore, we first tested whether quercetin affected the production of TNF-α in LPS-stimulated DCs, which is a hallmark of DC activation. Significantly, TNF-α was downregulated by quercetin in a dose-dependent manner (Fig. 1A), indicating that quercetin may inhibit DC activation. DMSO, the solvent of quercetin, had no effect on LPS-stimulated DCs. Next, we determined whether the suppressive effect of quercetin was caused by its cytotoxicity in DCs. Quercetin induced significant DC apoptosis at a concentration ≥ 100 μM (Fig. 1B), which confirmed the intrinsic inhibitory effect of quercetin at concentrations ≤50 μM. We then examined the efficiency of the inhibitory effect of quercetin. Quercetin blocked the DC activation by LPS at high doses (up to 1 μg/ml) (Fig. 1C). In addition, the production of TNF-α was decreased in all tests, regardless of whether the quercetin treatment was administered to DCs before or after LPS stimulation (Fig. 1D). Furthermore, we stimulated DCs with various TLR ligands, including zymosan, synthetic diacylated lipoprotein Pam2Cys-Ser-Lys4, lipoteichoic acid, peptidoglycan (TLR1, 2, or 6), polyinosine polycytidylic acid (TLR3), flagellin (TLR5), and synthetic CpG oligodeoxynucleotides (TLR9) and found that quercetin significantly reduced TNF-α production in all conditions, although the degree of the effect varied (Fig. 1E). Collectively, we identified the inhibitory effect of quercetin on DC activation and suggested that quercetin may be a very efficient inhibitor of DC activation.

FIGURE 1.

Quercetin efficiently inhibited DC activation. Mouse bone marrow-derived DCs were treated with medium, TLR ligands, TLR ligand + DMSO, or TLR ligands + quercetin (stock solution at 25 mM in DMSO), as indicated. Supernatants were collected 6 h later, and TNF-α production was measured by ELISA. A, Dose-dependent inhibition of the production of TNF-α by quercetin. B, The cytotoxicity of quercetin in DCs without or with LPS was analyzed by Annexin V staining. The percentage of Annexin V+ cells was determined by flow cytometry. C–E, Inhibitory effect of quercetin on DCs treated with the indicated doses of LPS (C), DCs activated with LPS (100 ng/ml) before or after quercetin treatment (D), and DCs stimulated with various TLR ligands, as indicated (E). Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated TLR ligand-activated DCs. All data are representative of three to five independent experiments.

FIGURE 1.

Quercetin efficiently inhibited DC activation. Mouse bone marrow-derived DCs were treated with medium, TLR ligands, TLR ligand + DMSO, or TLR ligands + quercetin (stock solution at 25 mM in DMSO), as indicated. Supernatants were collected 6 h later, and TNF-α production was measured by ELISA. A, Dose-dependent inhibition of the production of TNF-α by quercetin. B, The cytotoxicity of quercetin in DCs without or with LPS was analyzed by Annexin V staining. The percentage of Annexin V+ cells was determined by flow cytometry. C–E, Inhibitory effect of quercetin on DCs treated with the indicated doses of LPS (C), DCs activated with LPS (100 ng/ml) before or after quercetin treatment (D), and DCs stimulated with various TLR ligands, as indicated (E). Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated TLR ligand-activated DCs. All data are representative of three to five independent experiments.

Close modal

In addition to TNF-α, activated DCs secrete a variety of cytokines and chemokines that regulate immune responses. Therefore, we determined whether the inhibitory effect of quercetin on TNF-α production could be extended to other cytokines and chemokines produced by LPS-stimulated DCs. As expected, the generation of cytokines (IL-1α, IL-1β, IL-6, IL-10, and IL-12 p70) (Fig. 2A) and chemokines (MCP-1, MIP-1α, MIP-1β, and RANTES) (Fig. 2B) by activated DCs was impaired by quercetin treatment. Because IL-10 production was also reduced, it is not likely that the suppressive effect of quercetin was mediated through the production of this anti-inflammatory cytokine. Importantly, the expression level of β-actin was not significantly affected by quercetin (Fig. 2C), indicating that quercetin does not have global inhibitory effect on protein expression. These results indicate that quercetin downregulates the cytokines and chemokines secreted by activated DCs, thus disturbing the immunoregulatory function of DCs.

FIGURE 2.

The cytokines and chemokines released from LPS-stimulated DCs were impaired by quercetin. DCs were treated with LPS (100 ng/ml) in the presence of various doses of quercetin or 0.2% DMSO, as indicated. Supernatants were collected after 24 h (6 h for RANTES). The levels of cytokines (A) and chemokines (B) were determined by ELISA. Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. C, The effect of quercetin on expression of β-actin. The expression level of β-actin was determined by Western blot analysis. All data are representative of three independent experiments.

FIGURE 2.

The cytokines and chemokines released from LPS-stimulated DCs were impaired by quercetin. DCs were treated with LPS (100 ng/ml) in the presence of various doses of quercetin or 0.2% DMSO, as indicated. Supernatants were collected after 24 h (6 h for RANTES). The levels of cytokines (A) and chemokines (B) were determined by ELISA. Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. C, The effect of quercetin on expression of β-actin. The expression level of β-actin was determined by Western blot analysis. All data are representative of three independent experiments.

Close modal

Maturation is the key step in the DC-mediated regulation of immune responses. To investigate the effect of quercetin on DC maturation, we examined the expression levels of MHC class II and costimulatory molecules in DCs, which are the major phenotypes of DC maturation, using flow cytometry. LPS stimulation enhanced the expression of I-Ab and the costimulatory molecules CD40, CD80, and CD86 in DCs, but quercetin treatment significantly lowered the expression levels of these molecules (Fig. 3). These data suggest that quercetin attenuates LPS-induced DC maturation and limits the immunostimulatory activity of DCs.

FIGURE 3.

Quercetin attenuated the LPS-induced DC maturation. DCs were treated with medium, LPS (100 ng/ml), LPS + 0.2% DMSO, or LPS + quercetin (50 μM) for 16 h. The expression levels of MHC class II, CD40, CD80, and CD86 were determined by flow cytometry. All data shown were gated on CD11c+ cells. The gray-filled area represents staining with an isotype-matched control Ab. The change of mean fluorescence intensity from LPS to LPS + quercetin was indicated. All data are representative of two to four independent experiments.

FIGURE 3.

Quercetin attenuated the LPS-induced DC maturation. DCs were treated with medium, LPS (100 ng/ml), LPS + 0.2% DMSO, or LPS + quercetin (50 μM) for 16 h. The expression levels of MHC class II, CD40, CD80, and CD86 were determined by flow cytometry. All data shown were gated on CD11c+ cells. The gray-filled area represents staining with an isotype-matched control Ab. The change of mean fluorescence intensity from LPS to LPS + quercetin was indicated. All data are representative of two to four independent experiments.

Close modal

Endocytosis is a major pathway to mediate Ag uptake by DCs. Therefore, we determined the effect of quercetin on endocytosis by DCs by dextran-FITC uptake. Obviously, the ingestion of dextran-FITC was reduced in DCs after quercetin, but not LPS, treatment for 1 h (Fig. 4A). Low uptake of dextran-FITC was observed in mature DC induced by LPS stimulation for 16 h (Fig. 4B). DCs cannot become mature by stimulation for 1 h (data not shown). Thus, the blockage of endocytosis by quercetin is not related to the maturation of DCs. Our data indicate that quercetin can downregulate the endocytosis by DCs, thus impairing the Ag loading in DCs.

FIGURE 4.

Quercetin blocked the endocytosis by DCs. DCs were treated with medium, 0.2% DMSO, quercetin (50 μM), PBS, LPS (100 ng/ml) for 1 h (A), or LPS for 16 h (B) and then incubated with dextran-FITC. The gray-filled area represents the cells incubated with dextran-FITC at 4°C as a control. The change of percentage of dextran-FITC+ cells from untreatment to treatment was indicated. All data are representative of four independent experiments.

FIGURE 4.

Quercetin blocked the endocytosis by DCs. DCs were treated with medium, 0.2% DMSO, quercetin (50 μM), PBS, LPS (100 ng/ml) for 1 h (A), or LPS for 16 h (B) and then incubated with dextran-FITC. The gray-filled area represents the cells incubated with dextran-FITC at 4°C as a control. The change of percentage of dextran-FITC+ cells from untreatment to treatment was indicated. All data are representative of four independent experiments.

Close modal

DC migration is critical for the initiation of adaptive immune responses. As mature DCs are attracted by CCL19 and CCL21, we evaluated DC migration using chemotaxis assay in Transwell chambers. LPS-stimulated DCs efficiently migrated from the upper to the lower wells in response to CCL21; however, this movement was halted by quercetin (Fig. 5A). Furthermore, the migration of LPS-stimulated DCs from the footpad to the inguinal LNs was diminished by quercetin (Fig. 5B). Thus, we concluded that quercetin suppresses DC migration and disconnects the induction of adaptive immune responses.

FIGURE 5.

The migration of LPS-stimulated DCs was diminished by quercetin. DCs were treated with medium, LPS (100 ng/ml), LPS + 0.2% DMSO, or LPS + quercetin (50 μM) for 16 h. A, DCs were transferred into the upper wells of a 24-well Transwell chamber, and CCL21 was included in the lower wells. After 3 h, the number of cells that transferred from the upper to the lower wells was counted by flow cytometry. B, DCs were labeled with CFSE and then injected into mouse footpads. The inguinal LN cells were isolated after 2 d, and CFSE+ cells were determined by hemocytometry and flow cytometry. Data shown were means + SD of samples from three wells (A) or three mice (B). **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. All data are representative of three independent experiments.

FIGURE 5.

The migration of LPS-stimulated DCs was diminished by quercetin. DCs were treated with medium, LPS (100 ng/ml), LPS + 0.2% DMSO, or LPS + quercetin (50 μM) for 16 h. A, DCs were transferred into the upper wells of a 24-well Transwell chamber, and CCL21 was included in the lower wells. After 3 h, the number of cells that transferred from the upper to the lower wells was counted by flow cytometry. B, DCs were labeled with CFSE and then injected into mouse footpads. The inguinal LN cells were isolated after 2 d, and CFSE+ cells were determined by hemocytometry and flow cytometry. Data shown were means + SD of samples from three wells (A) or three mice (B). **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. All data are representative of three independent experiments.

Close modal

The primary function of mature DCs is the induction of Ag-specific T cell activation. Therefore, we determined whether quercetin affected the ability of DCs to activate T cells. DCs were loaded with OVA323–339 peptide and stimulated with LPS in the absence or presence of quercetin for 16 h. DCs were then washed to avoid the effect of quercetin on T cells (22) and cocultured with OT-II T cells. LPS-activated DCs promoted T cell proliferation, which was abrogated by quercetin (Fig. 6A). Next, we performed an in vivo recall assay. C57BL/6 mice were immunized with OVA mixed with IFA plus DMSO, quercetin, LPS, LPS plus DMSO, or LPS plus quercetin. After 10 d, draining LN cells were incubated with OVA for 3 d. Consistent with the in vitro results, quercetin significantly inhibited T cell proliferation in response to OVA (Fig. 6B). Furthermore, quercetin decreased the production of IFN-γ by activated T cells, both in vitro and in vivo (Fig. 6C). These results demonstrate that quercetin abrogates the ability of DCs to induce Ag-specific T cell activation. The abrogation of recall responses by quercetin in vivo is in agreement with the defect of DC maturation, endocytosis, and migration after quercetin treatment (Figs. 35).

FIGURE 6.

Quercetin abrogated the Ag-specific T cell activation induced by LPS-stimulated DCs. A, OT-II CD4+ T cells were cocultured with medium-, quercetin (50 μM)-, LPS (100 ng/ml)-, LPS + 0.2% DMSO-, or LPS + quercetin-treated DCs pulsed with OVA323–339 peptide (2 μg/ml) at the indicated ratios of DC:T cell. T cell proliferation was determined by [3H]thymidine incorporation after 3 d. B, C57BL/6 mice were immunized with OVA (10 μg) mixed with IFA + 0.4% DMSO, IFA + LPS (10 μg), IFA + LPS + DMSO, and IFA + LPS + quercetin (50 μg), via footpad injection. After 10 d, the draining LN cells were incubated with OVA at the indicated concentrations. T cell proliferation was measured by [3H]thymidine incorporation after 3 d. C, Supernatants were collected from cultures in A and B after 4 d. IFN-γ production was assayed by ELISA. Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. All data are representative of two to four independent experiments.

FIGURE 6.

Quercetin abrogated the Ag-specific T cell activation induced by LPS-stimulated DCs. A, OT-II CD4+ T cells were cocultured with medium-, quercetin (50 μM)-, LPS (100 ng/ml)-, LPS + 0.2% DMSO-, or LPS + quercetin-treated DCs pulsed with OVA323–339 peptide (2 μg/ml) at the indicated ratios of DC:T cell. T cell proliferation was determined by [3H]thymidine incorporation after 3 d. B, C57BL/6 mice were immunized with OVA (10 μg) mixed with IFA + 0.4% DMSO, IFA + LPS (10 μg), IFA + LPS + DMSO, and IFA + LPS + quercetin (50 μg), via footpad injection. After 10 d, the draining LN cells were incubated with OVA at the indicated concentrations. T cell proliferation was measured by [3H]thymidine incorporation after 3 d. C, Supernatants were collected from cultures in A and B after 4 d. IFN-γ production was assayed by ELISA. Data shown were means + SD of samples from three wells. NS, p > 0.05; *p < 0.05; **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated LPS-activated DCs. All data are representative of two to four independent experiments.

Close modal

We identified a suppressive effect of quercetin on DCs, which implies that quercetin may prevent DC-mediated diseases. Therefore, we performed DNFB-induced CHS as a model to test this hypothesis. Mice were sensitized with DNFB in the absence or presence of quercetin via injection of DNFB-pulsed DCs (Fig. 7A) or by painting it directly onto the abdomen of animals (Fig. 7B). The CHS response to DNFB was then examined. The ears were obviously swollen in DNFB-sensitized but not in DNFB plus quercetin-sensitized mice, whereas DMSO had no effect on DNFB-sensitized mice (data not shown), indicating that quercetin inhibits the DC-mediated sensitization in CHS. These evidences suggest that quercetin has the potential to prevent delayed-type hypersensitive diseases, such as allergic contact dermatitis.

FIGURE 7.

The CHS response was weakened in mice treated with quercetin. DNFB-induced CHS was described in 1Materials and Methods. Mice were sensitized with vehicle (olive oil/acetone = 1:4), DNFB, or DNFB + quercetin via injection of DCs (A) or by painting the abdomen of the animals (B). After 5 d, the ears of all mice were painted with DNFB and analyzed after 24 h. CHS response was determined by histology using H&E staining, and the thickness of the challenged ear was measured (original magnification ×40). Increases of ear thickness were calculated. Data shown were means + SD of samples from six mice. **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated DNFB-sensitized mice.

FIGURE 7.

The CHS response was weakened in mice treated with quercetin. DNFB-induced CHS was described in 1Materials and Methods. Mice were sensitized with vehicle (olive oil/acetone = 1:4), DNFB, or DNFB + quercetin via injection of DCs (A) or by painting the abdomen of the animals (B). After 5 d, the ears of all mice were painted with DNFB and analyzed after 24 h. CHS response was determined by histology using H&E staining, and the thickness of the challenged ear was measured (original magnification ×40). Increases of ear thickness were calculated. Data shown were means + SD of samples from six mice. **p < 0.01 (Student t test) was for the comparison between quercetin-treated and -untreated DNFB-sensitized mice.

Close modal

TLR ligation induces the activation of the MAPK, Akt, and NF-κB pathways, resulting in DC activation. To explore the molecular mechanism by which quercetin develops its inhibitory effect, we investigated which signal pathway is altered by quercetin in activated DCs. ERK, JNK, p38 MAPK, and Akt were activated in DCs after LPS stimulation; however, quercetin blocked the LPS-induced activation of ERK, JNK, and Akt but not of p38 MAPK, whereas the levels of these proteins were not affected (Fig. 8). In addition, the I-κB degradation was decreased, indicating that the activity of NF-κB was reduced by quercetin (Fig. 8). These results suggest that quercetin suppresses DC activation via disruption of the ERK, JNK, Akt, and NF-κB pathways, which explains the strong inhibitory effect of quercetin on DC activation (Fig. 1).

FIGURE 8.

The activation of ERK, JNK, Akt, and NF-κB in LPS-stimulated DCs was reduced by quercetin. DCs were treated with LPS (200 ng/ml) or quercetin (50 μM) + LPS and then lysed at the indicated time points. Samples were separated on SDS-PAGE, transferred to a polyvinylidene difluoride membrane, and then analyzed by Western blotting. The ERK, JNK, p38 MAPK, and Akt proteins (with and without phosphorylation) were detected by anti–phospho-specific and anti-protein Abs, respectively. The activation of NF-κB was determined by detecting the degradation of I-κB using a specific Ab. All data are representative of three independent experiments.

FIGURE 8.

The activation of ERK, JNK, Akt, and NF-κB in LPS-stimulated DCs was reduced by quercetin. DCs were treated with LPS (200 ng/ml) or quercetin (50 μM) + LPS and then lysed at the indicated time points. Samples were separated on SDS-PAGE, transferred to a polyvinylidene difluoride membrane, and then analyzed by Western blotting. The ERK, JNK, p38 MAPK, and Akt proteins (with and without phosphorylation) were detected by anti–phospho-specific and anti-protein Abs, respectively. The activation of NF-κB was determined by detecting the degradation of I-κB using a specific Ab. All data are representative of three independent experiments.

Close modal

In this study, we reported for the first time that quercetin is an immunosuppressor of DCs and provided strong evidence that quercetin may be a promising agent for the prevention and treatment of inflammatory and autoimmune diseases. Our study also supports the contention that vegetables and fruits containing quercetin can promote health by reducing harmful immunity.

Quercetin is the major flavonoid and forms the backbone of many other flavonoids. The bioactivity of quercetin is probably underestimated, although it has been shown in past decades to have a large spectrum of biological effects (16). Thus, we showed that the suppression of DC function is a new activity of quercetin, and the inhibitory effect of quercetin on DCs exhibits several unique features when compared with other flavonoids. Especially, we found that the ability of DCs to take up dextran-FITC was impaired by quercetin, which represents a maturation-independent reduction of endocytosis (Fig. 4). Therefore, we reported that quercetin is the first flavonoid to inhibit endocytosis by DCs. It could be due to that quercetin binds to actin directly and then block the cytoskeleton rearrangement during endocytosis (33). However, other mechanisms underlying this inhibition of endocytosis by quercetin warrants further investigation.

Another unique activity is the inhibition of DC migration by quercetin. CCR7 expression has been suggested to play a major role in regulating the DC migration (34). However, quercetin had no effect on the expression of CCR7 in DCs after LPS stimulation (Supplemental Fig. 1), indicating the involvement of other mechanisms for the inhibitory effect of quercetin on DC migration. It is possible that the effects of quercetin on cytoskeleton rearrangement, as suggested in the inhibition of endocytosis, may also reduce DC migration (33).

Quercetin targets multiple intracellular signaling pathways in cells, such as NF-κB (35). The reduction of I-κB degradation and blockage of Akt activity by quercetin in LPS-stimulated DCs (Fig. 8) are consistent with what was observed in macrophages (36, 37). However, quercetin inhibited the phosphorylation of ERK and JNK, but not p38 MAPK, in DCs (Fig. 8), which is in contrast with previous studies (38, 39). In addition to the signaling pathways examined in this study, inhibition of STAT1 activation (40), reduction of TBK1 kinase activity in the TIR domain-containing adaptor protein-inducing IFN-β–dependent signaling pathway (41), and disruption of the accumulation of lipid rafts (37) are involved in the inhibition of macrophage activation by quercetin. These pathways are probably related to the suppressive effect of quercetin on DCs.

A possible mechanism by which quercetin develops an immunosuppressive activity is the activation of the aryl hydrocarbon receptor (AhR), which is a cytosolic transcription factor that mediates the immunotoxicity of dioxin (42). AhR has recently been suggested to involve in immunoregulation and Th17 differentiation (43, 44). AhR activation induces regulatory T cell differentiation (45) and forms a complex with Stat1 and NF-κB in macrophages (46) and then modulates inflammatory responses. Recent reports suggest that AhR plays an essential role in the anti-inflammatory effect of other compounds (46, 47). Because quercetin is a ligand of AhR and DCs express high levels of AhR (48), it is likely that AhR may mediate the suppressive effect of quercetin on DCs. In addition, the fact that AhR signaling pathways downmodulate cytokine and growth factor production in other cell types would explain the ability of quercetin to reduce cytokine release in DCs (49). Although AhR is involved in development of immune system (50), DC differentiation was not affected by quercetin treatment (Supplemental Fig. 2). Collectively, quercetin may represent a novel approach to elucidate the role of AhR in DC biology.

Evidence for the possible clinical applications of quercetin is increasing. For instance, quercetin ameliorates EAE by blocking IL-12 signaling and Th1 differentiation (51), it improves the pathogenesis of a rheumatoid arthritis model after the onset of arthritis (52), and it attenuates the lethal systemic inflammation caused by endotoxemia (53). In this paper, we present an additional mechanism by which quercetin suppresses DC activation and function in these disease models. Moreover, AhR activation causes DC-mediated islet allograft-specific tolerance (54) and prevents diabetes in NOD mice (55), which is a pathway that could be presumably induced by quercetin. Currently, we are testing the protective effect of quercetin on chronic marginal periodontitis, as implied by Petti and Scully (11). Because quercetin is a safe agent in vivo (56), additional clinical trials are warranted to support the suitability of quercetin for disease control.

In summary, our study demonstrates that quercetin can suppress DC activation and thus may potentially be used for the prevention and treatment of inflammation, autoimmunity, and transplantation. Our findings provide new insights into the immunopharmacology of quercetin. Recent studies showed that the bioactivity of quercetin can be enhanced by nanotechnical encapsulation (57) and that liposomes can stably and efficiently deliver Ag and quercetin for the Ag-specific suppression of inflammatory arthritis (58). Conceivably, these techniques will hasten the development of the clinical applications of quercetin.

We thank Drs. Clifford Lowell (University of California, San Francisco, CA), Ing-Kang Ho, and Lu-Hai Wang (National Health Research Institutes) for help editing the manuscript. We also thank Drs. Cheng-Jen Chou and Tun-Tschu Chang (Taiwan Forest Research Institute, Tapei, Taiwan) for providing some materials. We sincerely appreciate Kuan-Yin Shen, I-Hua Chen, and Kai-Chieh Chen for excellent technical assistance.

Disclosures The authors have no financial conflicts of interest.

This work was supported by National Health Research Institutes Grant VC096PP04 and National Science Council Grants 962320B400003/972320B400004MY3 (in part) (to C.-L.C.) and 2007CB512403, and the C. Y. Foundation for Advancement of Education, Sciences, and Medicine of Taiwan (to E.F.).

The online version of this article contains supplemental material.

Abbreviations used in this paper:

AhR

aryl hydrocarbon receptor

CHS

contact hypersensitivity

DC

dendritic cell

DNFB

2,4-dinitro-1-fluorobenzene

LN

lymph node.

1
Buckwalter
M. R.
,
Albert
M. L.
.
2009
.
Orchestration of the immune response by dendritic cells.
Curr. Biol.
19
:
R355
R361
.
2
Joffre
O.
,
Nolte
M. A.
,
Spörri
R.
,
Reis e Sousa
C.
.
2009
.
Inflammatory signals in dendritic cell activation and the induction of adaptive immunity.
Immunol. Rev.
227
:
234
247
.
3
Kumar
H.
,
Kawai
T.
,
Akira
S.
.
2009
.
Toll-like receptors and innate immunity.
Biochem. Biophys. Res. Commun.
388
:
621
625
.
4
Steinman
R. M.
,
Banchereau
J.
.
2007
.
Taking dendritic cells into medicine.
Nature
449
:
419
426
.
5
Galkina
E.
,
Ley
K.
.
2009
.
Immune and inflammatory mechanisms of atherosclerosis (*).
Annu. Rev. Immunol.
27
:
165
197
.
6
Oyoshi
M. K.
,
He
R.
,
Kumar
L.
,
Yoon
J.
,
Geha
R. S.
.
2009
.
Cellular and molecular mechanisms in atopic dermatitis.
Adv. Immunol.
102
:
135
226
.
7
Ganesan
A.
2008
.
The impact of natural products upon modern drug discovery.
Curr. Opin. Chem. Biol.
12
:
306
317
.
8
Patil
B. S.
,
Jayaprakasha
G. K.
,
Chidambara Murthy
K. N.
,
Vikram
A.
.
2009
.
Bioactive compounds: historical perspectives, opportunities, and challenges.
J. Agric. Food Chem.
57
:
8142
8160
.
9
García-Lafuente
A.
,
Guillamón
E.
,
Villares
A.
,
Rostagno
M. A.
,
Martínez
J. A.
.
2009
.
Flavonoids as anti-inflammatory agents: implications in cancer and cardiovascular disease.
Inflamm. Res.
58
:
537
552
.
10
Mota
K. S.
,
Dias
G. E.
,
Pinto
M. E.
,
Luiz-Ferreira
A.
,
Souza-Brito
A. R.
,
Hiruma-Lima
C. A.
,
Barbosa-Filho
J. M.
,
Batista
L. M.
.
2009
.
Flavonoids with gastroprotective activity.
Molecules
14
:
979
1012
.
11
Petti
S.
,
Scully
C.
.
2009
.
Polyphenols, oral health and disease: A review.
J. Dent.
37
:
413
423
.
12
Spencer
J. P.
2009
.
The impact of flavonoids on memory: physiological and molecular considerations.
Chem. Soc. Rev.
38
:
1152
1161
.
13
Yang
C. S.
,
Wang
X.
,
Lu
G.
,
Picinich
S. C.
.
2009
.
Cancer prevention by tea: animal studies, molecular mechanisms and human relevance.
Nat. Rev. Cancer
9
:
429
439
.
14
Magrone
T.
,
Candore
G.
,
Caruso
C.
,
Jirillo
E.
,
Covelli
V.
.
2008
.
Polyphenols from red wine modulate immune responsiveness: biological and clinical significance.
Curr. Pharm. Des.
14
:
2733
2748
.
15
Boots
A. W.
,
Haenen
G. R.
,
Bast
A.
.
2008
.
Health effects of quercetin: from antioxidant to nutraceutical.
Eur. J. Pharmacol.
585
:
325
337
.
16
Bischoff
S. C.
2008
.
Quercetin: potentials in the prevention and therapy of disease.
Curr. Opin. Clin. Nutr. Metab. Care
11
:
733
740
.
17
Yu
C. S.
,
Lai
K. C.
,
Yang
J. S.
,
Chiang
J. H.
,
Lu
C. C.
,
Wu
C. L.
,
Lin
J. P.
,
Liao
C. L.
,
Tang
N. Y.
,
Wood
W. G.
,
Chung
J. G.
.
2010
.
Quercetin inhibited murine leukemia WEHI-3 cells in vivo and promoted immune response.
Phytother. Res.
24
:
163
168
.
18
Kim
A. R.
,
Cho
J. Y.
,
Zou
Y.
,
Choi
J. S.
,
Chung
H. Y.
.
2005
.
Flavonoids differentially modulate nitric oxide production pathways in lipopolysaccharide-activated RAW264.7 cells.
Arch. Pharm. Res.
28
:
297
304
.
19
Min
Y. D.
,
Choi
C. H.
,
Bark
H.
,
Son
H. Y.
,
Park
H. H.
,
Lee
S.
,
Park
J. W.
,
Park
E. K.
,
Shin
H. I.
,
Kim
S. H.
.
2007
.
Quercetin inhibits expression of inflammatory cytokines through attenuation of NF-κB and p38 MAPK in HMC-1 human mast cell line.
Inflamm. Res.
56
:
210
215
.
20
Moreira
M. R.
,
Kanashiro
A.
,
Kabeya
L. M.
,
Polizello
A. C.
,
Azzolini
A. E.
,
Curti
C.
,
Oliveira
C. A.
,
T-do Amaral
A.
,
Lucisano-Valim
Y. M.
.
2007
.
Neutrophil effector functions triggered by Fc-γ and/or complement receptors are dependent on B-ring hydroxylation pattern and physicochemical properties of flavonols.
Life Sci.
81
:
317
326
.
21
Gong
J.
,
Chen
S. S.
.
2003
.
Polyphenolic antioxidants inhibit peptide presentation by antigen-presenting cells.
Int. Immunopharmacol.
3
:
1841
1852
.
22
Yu
E. S.
,
Min
H. J.
,
An
S. Y.
,
Won
H. Y.
,
Hong
J. H.
,
Hwang
E. S.
.
2008
.
Regulatory mechanisms of IL-2 and IFNγ suppression by quercetin in T helper cells.
Biochem. Pharmacol.
76
:
70
78
.
23
Kim
J. S.
,
Jobin
C.
.
2005
.
The flavonoid luteolin prevents lipopolysaccharide-induced NF-κB signalling and gene expression by blocking IκB kinase activity in intestinal epithelial cells and bone-marrow derived dendritic cells.
Immunology
115
:
375
387
.
24
Yoon
M. S.
,
Lee
J. S.
,
Choi
B. M.
,
Jeong
Y. I.
,
Lee
C. M.
,
Park
J. H.
,
Moon
Y.
,
Sung
S. C.
,
Lee
S. K.
,
Chang
Y. H.
, et al
.
2006
.
Apigenin inhibits immunostimulatory function of dendritic cells: implication of immunotherapeutic adjuvant.
Mol. Pharmacol.
70
:
1033
1044
.
25
Lee
J. S.
,
Kim
S. G.
,
Kim
H. K.
,
Lee
T. H.
,
Jeong
Y. I.
,
Lee
C. M.
,
Yoon
M. S.
,
Na
Y. J.
,
Suh
D. S.
,
Park
N. C.
, et al
.
2007
.
Silibinin polarizes Th1/Th2 immune responses through the inhibition of immunostimulatory function of dendritic cells.
J. Cell. Physiol.
210
:
385
397
.
26
Kim
Y. J.
,
Choi
S. E.
,
Lee
M. W.
,
Lee
C. S.
.
2008
.
Taxifolin glycoside inhibits dendritic cell responses stimulated by lipopolysaccharide and lipoteichoic acid.
J. Pharm. Pharmacol.
60
:
1465
1472
.
27
Yoneyama
S.
,
Kawai
K.
,
Tsuno
N. H.
,
Okaji
Y.
,
Asakage
M.
,
Tsuchiya
T.
,
Yamada
J.
,
Sunami
E.
,
Osada
T.
,
Kitayama
J.
, et al
.
2008
.
Epigallocatechin gallate affects human dendritic cell differentiation and maturation.
J. Allergy Clin. Immunol.
121
:
209
214
.
28
Chu
C. L.
,
Lowell
C. A.
.
2005
.
The Lyn tyrosine kinase differentially regulates dendritic cell generation and maturation.
J. Immunol.
175
:
2880
2889
.
29
Chu
C. L.
,
Yu
Y. L.
,
Shen
K. Y.
,
Lowell
C. A.
,
Lanier
L. L.
,
Hamerman
J. A.
.
2008
.
Increased TLR responses in dendritic cells lacking the ITAM-containing adapters DAP12 and FcRgamma.
Eur. J. Immunol.
38
:
166
173
.
30
Yu
Y. L.
,
Chen
I. H.
,
Shen
K. Y.
,
Huang
R. Y.
,
Wang
W. R.
,
Chou
C. J.
,
Chang
T. T.
,
Chu
C. L.
.
2009
.
A triterpenoid methyl antcinate K isolated from Antrodia cinnamomea promotes dendritic cell activation and Th2 differentiation.
Eur. J. Immunol.
39
:
2482
2491
.
31
Liu
Q.
,
Chen
T.
,
Chen
G.
,
Shu
X.
,
Sun
A.
,
Ma
P.
,
Lu
L.
,
Cao
X.
.
2007
.
Triptolide impairs dendritic cell migration by inhibiting CCR7 and COX-2 expression through PI3-K/Akt and NF-κB pathways.
Mol. Immunol.
44
:
2686
2696
.
32
Chen
J. P.
,
Liao
N. S.
,
Lai
S. L.
,
Hsu
L.
,
Mao
W. Y.
,
Ku
M. C.
,
Liao
F.
.
2005
.
Reduced 2,4-dinitro-1-fluorobenzene–induced contact hypersensitivity response in IL-15 receptor α-deficient mice correlates with diminished CCL5/RANTES and CXCL10/IP-10 expression.
Eur. J. Immunol.
35
:
690
698
.
33
Böhl
M.
,
Tietze
S.
,
Sokoll
A.
,
Madathil
S.
,
Pfennig
F.
,
Apostolakis
J.
,
Fahmy
K.
,
Gutzeit
H. O.
.
2007
.
Flavonoids affect actin functions in cytoplasm and nucleus.
Biophys. J.
93
:
2767
2780
.
34
Förster
R.
,
Davalos-Misslitz
A. C.
,
Rot
A.
.
2008
.
CCR7 and its ligands: balancing immunity and tolerance.
Nat. Rev. Immunol.
8
:
362
371
.
35
Nam
N. H.
2006
.
Naturally occurring NF-κB inhibitors.
Mini Rev. Med. Chem.
6
:
945
951
.
36
Kim
B. H.
,
Lee
I. J.
,
Lee
H. Y.
,
Han
S. B.
,
Hong
J. T.
,
Ahn
B.
,
Lee
C. K.
,
Kim
Y.
.
2007
.
Quercetin 3-O-β-(2″-galloyl)-glucopyranoside inhibits endotoxin LPS-induced IL-6 expression and NF-κB activation in macrophages.
Cytokine
39
:
207
215
.
37
Kaneko
M.
,
Takimoto
H.
,
Sugiyama
T.
,
Seki
Y.
,
Kawaguchi
K.
,
Kumazawa
Y.
.
2008
.
Suppressive effects of the flavonoids quercetin and luteolin on the accumulation of lipid rafts after signal transduction via receptors.
Immunopharmacol. Immunotoxicol.
30
:
867
882
.
38
Cho
S. Y.
,
Park
S. J.
,
Kwon
M. J.
,
Jeong
T. S.
,
Bok
S. H.
,
Choi
W. Y.
,
Jeong
W. I.
,
Ryu
S. Y.
,
Do
S. H.
,
Lee
C. S.
, et al
.
2003
.
Quercetin suppresses proinflammatory cytokines production through MAP kinases andNF-κB pathway in lipopolysaccharide-stimulated macrophage.
Mol. Cell. Biochem.
243
:
153
160
.
39
Comalada
M.
,
Camuesco
D.
,
Sierra
S.
,
Ballester
I.
,
Xaus
J.
,
Gálvez
J.
,
Zarzuelo
A.
.
2005
.
In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-κB pathway.
Eur. J. Immunol.
35
:
584
592
.
40
Hämäläinen
M.
,
Nieminen
R.
,
Vuorela
P.
,
Heinonen
M.
,
Moilanen
E.
.
2007
.
Anti-inflammatory effects of flavonoids: genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and NF-κB activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only NF-κB activation along with their inhibitory effect on iNOS expression and NO production in activated macrophages.
Mediators Inflamm.
2007
:
45673
.
41
Lee
J. K.
,
Kim
S. Y.
,
Kim
Y. S.
,
Lee
W. H.
,
Hwang
D. H.
,
Lee
J. Y.
.
2009
.
Suppression of the TRIF-dependent signaling pathway of Toll-like receptors by luteolin.
Biochem. Pharmacol.
77
:
1391
1400
.
42
Esser
C.
,
Rannug
A.
,
Stockinger
B.
.
2009
.
The aryl hydrocarbon receptor in immunity.
Trends Immunol.
30
:
447
454
.
43
Kimura
A.
,
Naka
T.
,
Nohara
K.
,
Fujii-Kuriyama
Y.
,
Kishimoto
T.
.
2008
.
Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells.
Proc. Natl. Acad. Sci. USA
105
:
9721
9726
.
44
Veldhoen
M.
,
Hirota
K.
,
Westendorf
A. M.
,
Buer
J.
,
Dumoutier
L.
,
Renauld
J. C.
,
Stockinger
B.
.
2008
.
The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins.
Nature
453
:
106
109
.
45
Quintana
F. J.
,
Basso
A. S.
,
Iglesias
A. H.
,
Korn
T.
,
Farez
M. F.
,
Bettelli
E.
,
Caccamo
M.
,
Oukka
M.
,
Weiner
H. L.
.
2008
.
Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor.
Nature
453
:
65
71
.
46
Kimura
A.
,
Naka
T.
,
Nakahama
T.
,
Chinen
I.
,
Masuda
K.
,
Nohara
K.
,
Fujii-Kuriyama
Y.
,
Kishimoto
T.
.
2009
.
Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses.
J. Exp. Med.
206
:
2027
2035
.
47
Lawrence
B. P.
,
Denison
M. S.
,
Novak
H.
,
Vorderstrasse
B. A.
,
Harrer
N.
,
Neruda
W.
,
Reichel
C.
,
Woisetschläger
M.
.
2008
.
Activation of the aryl hydrocarbon receptor is essential for mediating the anti-inflammatory effects of a novel low-molecular-weight compound.
Blood
112
:
1158
1165
.
48
Jux
B.
,
Kadow
S.
,
Esser
C.
.
2009
.
Langerhans cell maturation and contact hypersensitivity are impaired in aryl hydrocarbon receptor-null mice.
J. Immunol.
182
:
6709
6717
.
49
Haarmann-Stemmann
T.
,
Bothe
H.
,
Abel
J.
.
2009
.
Growth factors, cytokines and their receptors as downstream targets of arylhydrocarbon receptor (AhR) signaling pathways.
Biochem. Pharmacol.
77
:
508
520
.
50
Esser
C.
2009
.
The immune phenotype of AhR null mouse mutants: not a simple mirror of xenobiotic receptor over-activation.
Biochem. Pharmacol.
77
:
597
607
.
51
Muthian
G.
,
Bright
J. J.
.
2004
.
Quercetin, a flavonoid phytoestrogen, ameliorates experimental allergic encephalomyelitis by blocking IL-12 signaling through JAK-STAT pathway in T lymphocyte.
J. Clin. Immunol.
24
:
542
552
.
52
Mamani-Matsuda
M.
,
Kauss
T.
,
Al-Kharrat
A.
,
Rambert
J.
,
Fawaz
F.
,
Thiolat
D.
,
Moynet
D.
,
Coves
S.
,
Malvy
D.
,
Mossalayi
M. D.
.
2006
.
Therapeutic and preventive properties of quercetin in experimental arthritis correlate with decreased macrophage inflammatory mediators.
Biochem. Pharmacol.
72
:
1304
1310
.
53
Tang
D.
,
Kang
R.
,
Xiao
W.
,
Zhang
H.
,
Lotze
M. T.
,
Wang
H.
,
Xiao
X.
.
2009
.
Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function.
Am. J. Respir. Cell Mol. Biol.
41
:
651
660
.
54
Hauben
E.
,
Gregori
S.
,
Draghici
E.
,
Migliavacca
B.
,
Olivieri
S.
,
Woisetschläger
M.
,
Roncarolo
M. G.
.
2008
.
Activation of the aryl hydrocarbon receptor promotes allograft-specific tolerance through direct and dendritic cell-mediated effects on regulatory T cells.
Blood
112
:
1214
1222
.
55
Kerkvliet
N. I.
,
Steppan
L. B.
,
Vorachek
W.
,
Oda
S.
,
Farrer
D.
,
Wong
C. P.
,
Pham
D.
,
Mourich
D. V.
.
2009
.
Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in NOD mice and increases Foxp3 T cells in pancreatic lymph nodes.
Immunotherapy
1
:
539
547
.
56
Harwood
M.
,
Danielewska-Nikiel
B.
,
Borzelleca
J. F.
,
Flamm
G. W.
,
Williams
G. M.
,
Lines
T. C.
.
2007
.
A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties.
Food Chem. Toxicol.
45
:
2179
2205
.
57
Ghosh
A.
,
Mandal
A. K.
,
Sarkar
S.
,
Panda
S.
,
Das
N.
.
2009
.
Nanoencapsulation of quercetin enhances its dietary efficacy in combating arsenic-induced oxidative damage in liver and brain of rats.
Life Sci.
84
:
75
80
.
58
Capini
C.
,
Jaturanpinyo
M.
,
Chang
H. I.
,
Mutalik
S.
,
McNally
A.
,
Street
S.
,
Steptoe
R.
,
O’Sullivan
B.
,
Davies
N.
,
Thomas
R.
.
2009
.
Antigen-specific suppression of inflammatory arthritis using liposomes.
J. Immunol.
182
:
3556
3565
.