As an H+-gated subgroup of the degenerin/epithelial Na+ channel family, acid-sensing ion channels (ASICs) were reported to be involved in various physiological and pathological processes in neurons. However, little is known about the role of ASICs in the function of dendritic cells (DCs). In this study, we investigated the expression of ASICs in mouse bone marrow-derived DCs and their possible role in the function of DCs. We found that ASIC1, ASIC2, and ASIC3 are expressed in DCs at the mRNA and protein levels, and extracellular acid can evoke ASIC-like currents in DCs. We also demonstrated that acidosis upregulated the expression of CD11c, MHC class II, CD80, and CD86 and enhanced the Ag-presenting ability of DCs via ASICs. Moreover, the effect of acidosis on DCs can be abolished by the nonsteroidal anti-inflammatory drugs ibuprofen and diclofenac. These results suggest that ASICs are involved in the acidosis-mediated effect on DC function.

As the most potent APCs of the immune system, dendritic cells (DCs) are specialized to capture, process, and present Ags to T lymphocytes to initiate Ag-specific T cell responses (1, 2). Many reports showed that DCs play an important role in the development of inflammatory diseases, tumors, and autoimmune diseases (36).

pH value is an important physiological indicator of internal environment homeostasis, which usually ranges from pH 7.2–7.4 in the physiological state. However, in inflammatory diseases, pH values in inflamed areas are always lower than those of normal tissues, ranging from pH 5.5–7 (79). Thus, low pH is considered a hallmark of inflammatory reactions. In inflammation, as well as in autoimmune diseases, such as rheumatoid arthritis and asthma, pH values of synovial fluid of affected joints and airway vapor condensate are lower than those in normal tissues (10, 11). Moreover, extracellular pH values range from 5.7–7.8 in solid tumors (1214). However, few studies have focused on the effect of extracellular pH on immune cells and their functions (15), especially DCs. Although it was reported that extracellular acidosis upregulates the expression of cell surface proteins involved in Ag presentation of DCs (16), the molecular mechanism through which DCs are regulated by acidosis remained unknown.

Acid-sensing ion channels (ASICs) represent an H+-gated subgroup of the degenerin/epithelial Na+ channel family of cation channels and are activated by extracellular protons (17). Six ASIC subunit proteins, encoded by four genes, have been identified: ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, and ASIC4. They are widely expressed in peripheral sensory neurons and in the neurons of the CNS. Activation of these channels by protons plays an important role in a variety of physiological and pathological processes, such as nociception, mechanosensation, and acidosis-mediated neuronal injury. Our previous study also demonstrated that ASICs are involved in learning and memory (18). Recent studies reported that ASICs are also expressed in nonneuronal cells and have important effects on their physiological and pathological function (19, 20). Whether ASICs are expressed in DCs and their potential role in DC functions have not been determined.

In the current study, we investigated the expression of ASICs in mouse DCs and the role of ASICs in Ag-presentation–related molecule expression in acidosis. We demonstrated that DCs express ASICs, and the cell-surface molecules involved in Ag presentation are upregulated by acidosis via ASICs.

Female C57BL/6 and BALB/c mice, 6–8 wk old, were obtained from the Institute of Organ Transplantation of Tongji Hospital, Huazhong University of Science and Technology. Neonatal Sprague-Dawley rats (days 0–3) were obtained from the Experimental Animal Center of Tongji Medical College, Huazhong University of Science and Technology. The animals were housed under specific pathogen-free conditions. All studies were performed according to the guidelines of the Animal Care and Use Committee of Tongji Medical College, Huazhong University of Science and Technology.

DCs were generated as previously described (21, 22), with minor modifications. In brief, bone marrow cells were obtained from the femurs and tibias of C57BL/6 mice; suspended at 1 × 106 cells/ml in RPMI 1640 medium (Invitrogen) supplemented with 10% FCS, penicillin (100 U/ml), streptomycin (100 mg/ml), l-glutamine (2 mM), and 2-ME (50 mM); and then seeded in 100-mm dishes at 10 ml/dish. Cultures were supplemented with 10 ng/ml GM-CSF and 5 ng/ml IL-4 (both from PeproTech). The cells were fed on days 3 and 5, by aspirating 75% of the medium and adding back fresh medium with GM-CSF and IL-4 at appropriate concentrations, and cultured for 7 d. By day 7, >85% of the harvested cells expressed CD11c. Immature DCs (iDCs) used in this study were cultured for 7 d. To obtain mature DCs (mDCs), iDCs were activated with 1 μg/ml LPS (Sigma-Aldrich) for 24 h.

Primary rat cortical neurons were isolated as described in our previous study (23, 24), with some modifications. Briefly, the cortex of newborn Sprague-Dawley rats were dissected and rinsed in ice-cold Dulbecco’s PBS. The dissected tissues were treated with 0.125% trypsin in HBSS for 25 min at 37°C and mechanically dissociated using fire-polished Pasteur pipettes. Cells were collected by centrifugation and resuspended in DMEM/F12 (1:1) with 10% FBS. For whole-cell patch-clamp recording, cells (20,000–40,000) were seeded on poly-d-lysine–coated coverslips and kept at 37°C in a 5%-CO2 incubator. After 24 h, the culture medium was changed with DMEM medium supplemented with 2% B27, and the cortical neurons were fed with fresh medium twice a week. Microscopically, glial cells were not apparent by using this protocol. The neurons were maintained for 7–10 d in primary culture until used.

Total RNA was isolated from cells using TRIzol reagent (Invitrogen), according to the manufacturer’s instructions. cDNA synthesis was performed using a PrimeScript 1st Strand cDNA Synthesis Kit (Takara Biotechnology). PCR amplifications of cDNA were performed by standard methods. The following specific primers (forward, reverse) were used: ASIC1, 5′-CAG ATG GCT GAT GAA AAG CA-3′, 5′-AAG TGG CAC GAG AGA AGC AT-3′; ASIC2, 5′-TGA CAT TGG TGG TCA AAT GG-3′, 5′-ATC ATG GCT CCC TTC CTC TT-3′; ASIC3, 5′-AGG GAG AAG TCC CAA AGC AT-3′, 5′-GAC ACT CCA TTC CCA GGA GA-3′ (25); TRPV1, 5′-TCG TCT ACC TCG TGT TCT TGT TTG-3′, 5′-CCA GAT GTT CTT GCT CTC TTG TGC-3′ (26); and GAPDH 5′-TTC ACC ACC ATG GAG AAG GC-3′, 5′-GGC ATG GAC TGT GGT CAT GA-3′ (27). GAPDH was used for normalization. PCR products were analyzed by 2% agarose gel electrophoresis.

Cells were lysed in lysis buffer (50 mM Tris [pH 8], 150 mM NaCl containing 1% Nonidet P-40, 0.02% NaN3, 0.5 mM PMSF, 1 μg/ml aprotinin, and 1 μg/ml leupeptin). Whole-cell lysate was mixed with 5× SDS loading buffer and boiled for 5 min. The proteins were separated by electrophoresis on 10% SDS-polyacrylamide gel and transferred to a polyvinylidene difluoride membrane. The membrane was blocked with 5% (w/v) nonfat milk in TBST for 2 h at 37°C and then incubated with rabbit anti-ASIC1, rabbit anti-ASIC2, or rabbit anti-ASIC3 Abs (all from Alomone Laboratories), as well as mouse anti-actin Ab-5 Ab (BD Biosciences), which served as a loading control, diluted in 5% (w/v) nonfat milk in 0.1% TBST overnight at 4°C. Membranes incubated with Abs that were preincubated with respective antigenic peptides were used to test specificity of Abs. After washing three times (10 min each) in 0.1% TBST, the membranes were incubated with peroxidase-conjugated goat anti-rabbit or goat anti-mouse secondary Ab (both from Pierce) for 2 h at 37°C. The membrane was washed three times in 0.1% TBST, and the proteins were detected using Peroxide Solution (Millipore) with a Kodak Image Station 4000MM.

We used the immunofluorescence double-staining method to determine the localization of ASICs in DCs. First, DCs were plated on polylysine-coated coverslips, cultured for 24 h, washed with PBS once, fixed with 4% paraformaldehyde in PBS for 20 min, washed three times in PBS, permeabilized with 0.3% Triton X-100 in PBS for 20 min, washed three times for 5 min in PBS, and then blocked with 3% BSA for 2 h at 37°C. Cells were incubated with rabbit anti-ASIC1, rabbit anti-ASIC2, or rabbit anti-ASIC3 primary Abs diluted in 3% BSA overnight at 4°C. After washing three times in PBS, cells were incubated with the FITC-conjugated goat anti-rabbit secondary Ab (Pierce) and PE-conjugated anti-CD11c Ab (eBioscience) diluted in 3% BSA for 1 h at room temperature. After extensive washing, coverslips were mounted on glass slides with 50% glycerol. Staining was visualized with an Olympus FV500 confocal microscope.

DCs were fixed with a solution of 4% paraformaldehyde and 1.25% glutaral for 30 min; after washing three times in PBS, cells were permeabilized with 0.5% Triton X-100 and 25 μg/ml proteinase A for 20 min, in sequence. After extensive washing, cells were blocked with normal goat serum for 2 h at room temperature. Cells were incubated with rabbit anti-ASIC1, rabbit anti-ASIC2, or rabbit anti-ASIC3 primary Ab diluted in PBS overnight at 4°C. Then, cells were washed with PBS three times before staining with anti-rabbit IgG (whole molecule)-Gold (Sigma-Aldrich) for 30 min. Cells were postfixed with 1% OsO4 for 1 h and were dehydrated, embedded, made into ultrathin sections, and stained with uranyl acetate, using conventional methods. Images were taken with an FEI-Tecnai G2 12 transmission electron microscope (FEI).

The procedure for whole-cell patch-clamp recording was performed as described in our previous studies (28, 29), with minor modification. The bath solution for recording ASIC currents contained NaCl 150 mM, KCl 5 mM, MgCl2 1 mM, CaCl2 2 mM, HEPES 10 mM, and glucose 10 mM; pH was adjusted to 7.4 with NaOH. Glass pipettes were used with a resistance of ∼2–4 MΩ when filled with the following solution: KCl 140 mM, NaCl 10 mM, MgCl2 1 mM, EGTA 5 mM, Mg-ATP 2 mM, and HEPES 10 mM; pH was adjusted to 7.2 with KOH. After establishing a whole-cell configuration, the adjustment of capacitance compensation and series resistance compensation was done before recording. The current signals were acquired at a sampling rate of 10 kHz and were filtered at 3 kHz. Whole-cell patch-clamp recordings were carried out using an EPC-10 amplifier, driven by Pulse/PulseFit software (both from HEKA Elektronik). Drug actions were measured only after steady-state conditions were reached, which were judged by the amplitudes and time courses of currents remaining constant. All recordings were made at room temperature (20°C–22°C). All experiments were repeated three times using different batches of cells, and at least three or four dishes of cells were used for recording in different batches of cells.

Cells (1 × 106) were stained with PE-conjugated mAbs diluted in PBS containing 1% BSA (FACS buffer). Before staining, cells were incubated with purified anti-mouse CD16/32 (Biolegend) for 10 min on ice to block FcR. The following PE-conjugated Abs (all from eBioscience) were used: anti-CD11, anti-IA/IE (MHC class II), anti-CD80, and anti-CD86. After incubating with Abs for 30 min at 4°C, cells were washed twice with FACS buffer and analyzed by a LSR II flow cytometer (BD Bioscience). The results are shown as mean fluorescence intensity (MFI).

Splenic T cells from normal BALB/c mice were stained with CFSE and used as responders. A total of 2 × 106/ml T cells was cultured with 2 × 105/ml DCs treated with mitomycin C (30 μg/ml) and incubated at 37°C in 5% CO2 for 5 d. After three washes with PBS, the cells were harvested and analyzed for T cell proliferation using a LSR II flow cytometer (BD Bioscience).

Data are presented as mean ± SEM. The Student t test with paired comparisons was used to evaluate differences; p < 0.05 was considered statistically significant.

To determine whether ASIC1, ASIC2, and ASIC3 are expressed in iDCs, we performed RT-PCR to detect the expression of mRNA for ASICs. Cortex neurons served as positive control. The sizes of the expected PCR products for ASIC1, ASIC2, and ASIC3 were 140, 139, and 107 bp, respectively. As a result, mRNAs for all three ASICs were detected in iDCs (Fig. 1A). Then, we confirmed the expression of ASIC proteins by Western blotting. Abs for ASIC1, ASIC2, and ASIC3 were preincubated with the corresponding Ag peptide as specific control (Fig. 1B). Furthermore, we used immunocytochemistry to determine cellular and subcellular distributions of ASICs in iDCs. As shown in Fig. 1C, ASIC1 and ASIC3 proteins were predominantly expressed in cytoplasm. Most ASIC2 is expressed on the plasma membrane. We further observed, by electron microscopy, that ASIC1 was expressed in endoplasmic reticulum and perinuclear regions, whereas ASIC3 was found in mitochondria (Fig. 1D).

FIGURE 1.

Expression and localization of ASIC1, ASIC2, and ASIC3 in iDCs. A, ASIC transcripts were detected in iDCs by RT-PCR. Cortex neurons were used as positive control. B, Western blot analysis of ASIC protein expression in iDCs. Cortex protein was used as positive control. For specificity control, Abs for ASICs were preincubated with corresponding peptide Ags. C, Detection of localization of ASIC proteins in iDCs by double-staining immunofluorescence (original magnification ×400). Nuclei were counterstained with Hoechst33342 (blue). Green, ASICs; red, CD11c. D, Subcellular localization of ASICs was visualized using a transmission electron microscope. ASIC1 and ASIC3 were found in endoplasmic reticulum and mitochondrion, respectively, and ASIC2 was located on the cell membrane. Arrows indicate the localization of ASICs.

FIGURE 1.

Expression and localization of ASIC1, ASIC2, and ASIC3 in iDCs. A, ASIC transcripts were detected in iDCs by RT-PCR. Cortex neurons were used as positive control. B, Western blot analysis of ASIC protein expression in iDCs. Cortex protein was used as positive control. For specificity control, Abs for ASICs were preincubated with corresponding peptide Ags. C, Detection of localization of ASIC proteins in iDCs by double-staining immunofluorescence (original magnification ×400). Nuclei were counterstained with Hoechst33342 (blue). Green, ASICs; red, CD11c. D, Subcellular localization of ASICs was visualized using a transmission electron microscope. ASIC1 and ASIC3 were found in endoplasmic reticulum and mitochondrion, respectively, and ASIC2 was located on the cell membrane. Arrows indicate the localization of ASICs.

Close modal

A whole-cell patch-clamp recording was performed to determine whether the expression of ASIC proteins in iDCs has a function. Cells were held at −60 mV and then a rapid decrease in extracellular pH, from 7.4, was applied to the bath. In 85% (n = 36/42) of the recorded iDCs, the rapid decrease in extracellular pH elicited inward ASIC-like currents, indicating the presence of functional ASIC channels. Usually, this transient inward current contained two phases: early fast and late slow inactivation (Fig. 2A). The mean amplitude of ASIC-like currents induced by pH 6 was 329.63 ± 52.36 pA. Similar to the proton-gated currents in sensory and central neurons, the ASIC-like currents in iDCs were reversibly inhibited by amiloride (100 μM), a blocker of ASICs (n = 6, p < 0.05; Fig. 2A). As shown in Fig. 2B, the response of iDCs to acidosis was pH dependent. The threshold extracellular pH to elicit the inward current was ∼7, and the maximum response appeared at pH 5. The extracellular pH producing 50% effect was 6.08 ± 0.05, and the Hill coefficient was 1.03 ± 0.07 (n = 6) (Fig. 2C).

FIGURE 2.

Electrophysiological characteristics of ASIC-like currents recorded in iDCs by whole-cell patch clamp. A, pH 6 elicited ASIC-like currents in iDCs, and the currents were reversibly inhibited by amiloride (100 μM) (n = 36/42). B and C, ASIC-like currents elicited by acid in iDCs were pH dependent (n = 6 for each pH).

FIGURE 2.

Electrophysiological characteristics of ASIC-like currents recorded in iDCs by whole-cell patch clamp. A, pH 6 elicited ASIC-like currents in iDCs, and the currents were reversibly inhibited by amiloride (100 μM) (n = 36/42). B and C, ASIC-like currents elicited by acid in iDCs were pH dependent (n = 6 for each pH).

Close modal

As a nonselective cation channel, transient receptor potential vanilloid-1 (TRPV1) also can evoke membrane currents at low extracellular pH (<6). Thereby, we examined the effect of capsaicin (Cap), a selective agonist of TRPV1, on iDCs with the whole-cell patch-clamp technique. As shown in Fig. 3A, 10 μM Cap significantly evoked an inward current in cortex neurons. However, 10 or 100 μM Cap failed to induce any inward currents in iDCs, indicating that the inward currents induced by a decrease in extracellular pH may not be mediated by TRPV1 channels in iDCs. To further confirm this ratiocination, RT-PCR was performed to investigate whether TRPV1 is expressed in iDCs, with cortex neurons as positive control. We did not detect TRPV1 mRNA expression in iDCs (Fig. 3B). These results suggested that TRPV1 is not involved in the elicitation of inward currents induced by low extracellular pH in iDCs.

FIGURE 3.

TRPV1 is not functionally expressed in iDCs. A, Cap (10 or 100 μM) did not elicit current in iDCs. Currents evoked by Cap (10 μM) served as positive control. B, No TRPV1 expression at the mRNA level in iDCs was detected by RT-PCR. Cortex neurons were used as positive control.

FIGURE 3.

TRPV1 is not functionally expressed in iDCs. A, Cap (10 or 100 μM) did not elicit current in iDCs. Currents evoked by Cap (10 μM) served as positive control. B, No TRPV1 expression at the mRNA level in iDCs was detected by RT-PCR. Cortex neurons were used as positive control.

Close modal

After demonstrating the characteristics of ASIC expression in iDCs, we further investigated that in mDCs. As shown in Fig. 4A and 4B, the distribution and expression of ASICs were similar in iDCs and mDCs. However, the amplitude of ASIC currents induced by extracellular pH 6 was increased significantly in mDCs (Fig. 4C), suggesting an enhanced function of ASICs in mDCs.

FIGURE 4.

ASIC1, ASIC2, and ASIC3 are functionally expressed in mDCs. A, Localization of ASICs in mDCs was detected by double-staining immunofluorescence (original magnification ×400). Nuclei were counterstained with Hoechst33342 (blue). Green, ASICs; red, CD11c. B, Expression of ASICs in mDCs. iDCs served as control and β-actin was used for normalization. C, ASIC-like currents elicited by pH 6 in iDCs and mDCs (n = 6 for each group). *p < 0.05, significant difference between the two groups.

FIGURE 4.

ASIC1, ASIC2, and ASIC3 are functionally expressed in mDCs. A, Localization of ASICs in mDCs was detected by double-staining immunofluorescence (original magnification ×400). Nuclei were counterstained with Hoechst33342 (blue). Green, ASICs; red, CD11c. B, Expression of ASICs in mDCs. iDCs served as control and β-actin was used for normalization. C, ASIC-like currents elicited by pH 6 in iDCs and mDCs (n = 6 for each group). *p < 0.05, significant difference between the two groups.

Close modal

It was reported that exposure to pH 6.5 for 4 h resulted in a significant maturation of DCs, characterized by a significant increase in CD11c, MHC class II, and CD86 (16). In addition to those molecules, CD80 on DCs was upregulated after exposure to pH 6.5 for 4 h (Fig. 5); however, it is still unknown how acidosis induces DC maturation. Because ASICs are proton-gated cation channels and can be activated by extracellular H+, we hypothesized that the effect of acidosis on DCs is mediated via ASICs. To examine this hypothesis, DCs were incubated at pH 7.3 for 30 min at 37°C in the presence of amiloride (100 μM), a blocker of ASICs, before being exposed to pH 6.5 for 4 h at 37°C. Amiloride had no effect on CD11c, MHC class II, CD80, or CD86 at pH 7.3 (Supplemental Fig. 1); however, upregulation of CD11c, MHC class II, CD80, and CD86 stimulated by acidosis was significantly inhibited by amiloride (Fig. 5), suggesting that ASICs are involved in DC maturation induced by acidosis.

FIGURE 5.

pH 6.5 increases the expression of cell-surface molecules CD11c, MHC class II, CD80, and CD86 in DCs, and this effect was abrogated by amiloride. DCs were incubated with or without amiloride (100 μM) at pH 7.3 for 30 min at 37°C, 5% CO2 before exposure to pH 6.5 for 4 h at 37°C, 7% CO2. AD, Graphs from a representative experiment. EH, MFI values of CD11c, MHC class II, CD80, and CD86 were analyzed by flow cytometry. The data represent mean ± SEM of six independent experiments. #p < 0.05, significant difference for pH 6.5 compared with pH 7.3; *p < 0.05, significant difference for amiloride treatment compared with pH 6.5.

FIGURE 5.

pH 6.5 increases the expression of cell-surface molecules CD11c, MHC class II, CD80, and CD86 in DCs, and this effect was abrogated by amiloride. DCs were incubated with or without amiloride (100 μM) at pH 7.3 for 30 min at 37°C, 5% CO2 before exposure to pH 6.5 for 4 h at 37°C, 7% CO2. AD, Graphs from a representative experiment. EH, MFI values of CD11c, MHC class II, CD80, and CD86 were analyzed by flow cytometry. The data represent mean ± SEM of six independent experiments. #p < 0.05, significant difference for pH 6.5 compared with pH 7.3; *p < 0.05, significant difference for amiloride treatment compared with pH 6.5.

Close modal

Previous studies demonstrated that nonsteroidal anti-inflammatory drugs (NSAIDs) significantly inhibited ASIC currents in hippocampus and sensory neurons (30, 31). To investigate the effect of NSAIDs on ASICs in iDCs, the responses of ASIC currents to NSAIDs (ibuprofen or diclofenac) were tested. Extracellular application of 200 μM ibuprofen significantly reduced ASIC currents from 364.17 ± 57.15 pA to 176.07 ± 28.77 pA (Fig. 6A, 6B). The inhibitory percentage was 51.49 ± 3.75%, and the inhibition could be reversed by extensive washing with bath solution. Similarly, diclofenac (200 μM) significantly reduced ASIC currents from 353.97 ± 51.58 pA to 166.7 ± 18.57 pA (Fig. 6C, 6D). The inhibition was also reversible, and the inhibitory percentage was 51.67 ± 8.3%. These data suggested that ASIC currents in iDCs are also sensitive to NSAIDs. Therefore, ASICs in DCs are possible targets for NSAIDs in inflammation.

FIGURE 6.

Inhibitory effect of NSAIDs on ASICs in iDCs. A and B, ASIC currents in iDCs were reversibly inhibited by ibuprofen (200 μM). C and D, ASIC currents in iDCs were reversibly inhibited by diclofenac (200 μM). Data shown are mean ± SEM (n = 6 for each group). *p < 0.05 versus control.

FIGURE 6.

Inhibitory effect of NSAIDs on ASICs in iDCs. A and B, ASIC currents in iDCs were reversibly inhibited by ibuprofen (200 μM). C and D, ASIC currents in iDCs were reversibly inhibited by diclofenac (200 μM). Data shown are mean ± SEM (n = 6 for each group). *p < 0.05 versus control.

Close modal

After determining the inhibitory effect of NSAIDs on acid-sensing channels in DCs, we examined the effect of NSAIDs on DC maturation induced by acidosis. DCs were preincubated with ibuprofen or diclofenac at pH 7.3 for 30 min at 37°C and were exposed to pH 6.5 for 4 h. Ibuprofen and diclofenac had no effect on the expression of CD11c, MHC II, CD80, or CD86 at pH 7.3 (Supplemental Fig. 2), but they significantly abrogated the upregulation of CD11c, MHC class II, CD80, and CD86 molecules induced by acidosis (Fig. 7), indicating that NSAIDs can inhibit the maturation of DCs stimulated by acidosis.

FIGURE 7.

Upregulation of cell-surface molecules CD11c, MHC class II, CD80, and CD86 induced by pH 6.5 in DCs is inhibited by ibuprofen and diclofenac. DCs were incubated with or without ibuprofen (200 μM) or diclofenac (200 μM) at pH 7.3 for 30 min at 37°C, 5% CO2 before exposure to pH 6.5 for 4 h at 37°C, 7% CO2. AD, Graphs from a representative experiment. EH, The MFI values for CD11c, MHC class II, CD80, and CD86 were analyzed by flow cytometry. Data shown are mean ± SEM of six independent experiments. #p < 0.05, significant difference for pH 6.5 compared with pH 7.3; *p < 0.05, **p < 0.01, significant difference for treatment with ibuprofen or diclofenac compared with pH 6.5.

FIGURE 7.

Upregulation of cell-surface molecules CD11c, MHC class II, CD80, and CD86 induced by pH 6.5 in DCs is inhibited by ibuprofen and diclofenac. DCs were incubated with or without ibuprofen (200 μM) or diclofenac (200 μM) at pH 7.3 for 30 min at 37°C, 5% CO2 before exposure to pH 6.5 for 4 h at 37°C, 7% CO2. AD, Graphs from a representative experiment. EH, The MFI values for CD11c, MHC class II, CD80, and CD86 were analyzed by flow cytometry. Data shown are mean ± SEM of six independent experiments. #p < 0.05, significant difference for pH 6.5 compared with pH 7.3; *p < 0.05, **p < 0.01, significant difference for treatment with ibuprofen or diclofenac compared with pH 6.5.

Close modal

The data above demonstrated that ASICs are involved in the upregulation of cell-surface molecules related to Ag presentation induced by acidosis in DCs. MLR was used to examine the effect of acidosis on the Ag-presenting ability of DCs and the role of ASICs. DCs were incubated in pH 7.3 medium for 30 min in the presence or absence of amiloride (100 μM), ibuprofen (200 μM), or diclofenac (200 μM) before being exposed to pH 6.5 for 4 h; DCs incubated at pH 7.3 for 4.5 h served as controls. After washing, DCs were pretreated with mitomycin C and cultured with T cells stained with CFSE at pH 7.3 for 5 d, and T cell proliferation was detected by flow cytometry. As expected, pH 6.5 increased the Ag-presenting ability of DCs, whereas amiloride and NSAIDs (ibuprofen or diclofenac) abrogated this effect (Fig. 8), suggesting that ASICs play a critical role in the regulation of the Ag-presenting ability of DCs by acidosis.

FIGURE 8.

Effect of pH 6.5 on the Ag-presenting ability of DCs is inhibited by amiloride, ibuprofen, and diclofenac. DCs (from C57BL/6 mice) were incubated or not with amiloride (100 μM), ibuprofen (200 μM), or diclofenac (200 μM) at pH 7.3 for 30 min before exposure to pH 6.5 for 4 h; subsequently, the DCs were treated with mitomycin C and cultured with allogeneic T cells (from BALB/c mice) stained with CFSE at pH 7.3 for 5 d. Results are expressed as proliferating index and are shown as mean ± SEM of six independent experiments. ##p < 0.01, significant difference for pH 6.5 versus pH 7.3; **p < 0.01, significant difference for drug treatment compared with pH 6.5.

FIGURE 8.

Effect of pH 6.5 on the Ag-presenting ability of DCs is inhibited by amiloride, ibuprofen, and diclofenac. DCs (from C57BL/6 mice) were incubated or not with amiloride (100 μM), ibuprofen (200 μM), or diclofenac (200 μM) at pH 7.3 for 30 min before exposure to pH 6.5 for 4 h; subsequently, the DCs were treated with mitomycin C and cultured with allogeneic T cells (from BALB/c mice) stained with CFSE at pH 7.3 for 5 d. Results are expressed as proliferating index and are shown as mean ± SEM of six independent experiments. ##p < 0.01, significant difference for pH 6.5 versus pH 7.3; **p < 0.01, significant difference for drug treatment compared with pH 6.5.

Close modal

The results demonstrated that ASICs are expressed in DCs and are involved in the maturation of DCs induced by acidosis.

As the most potent APC, DCs are specialized to capture and process Ags and present them to T cells to induce immune responses. Therefore, DCs are critical for the induction of immune responses triggered by microorganisms, tumors, and autoantigens. Recently, there is increasing evidence to suggest that inflammation plays an important role in various diseases, such as tumors and autoimmune diseases (3235). Inflammation is always accompanied by tissue acidification, which means that the microenvironment of pathological sites is acidic compared with normal sites; however, the effect of an acidic microenvironment on the physiological functions of DCs is less well known. Although a few studies have focused on the role of acidosis in DC function, the mechanism underlying the effect remains unclear (16, 36).

Because ASICs are H+-gated cation channels and are activated by extracellular protons, and the expression of ASICs in T cells, B cells, and macrophages of mice was reported recently (37), we proposed that ASICs serve as sensors for extracellular acidosis and mediate the responses of DCs to acidosis.

To test this hypothesis, the expression of ASICs in mouse iDCs and mDCs was investigated. As expected, iDCs expressed ASIC1, ASIC2, and ASIC3 at the mRNA and protein levels (Fig. 1A, 1B). Then, immunofluorescence confocal microscopy and electron microscopy were used to examine their localization in iDCs. Unlike the distribution in neurons, ASIC1 and ASIC3 are found mainly in cytoplasm rather than on the cell membrane, whereas ASIC2 is located exclusively on the cell membrane (Fig. 1C, 1D). Consistently, the distribution and expression of ASICs in mDCs were similar to those in iDCs (Fig. 4A, 4B).

We further examined the electrophysiological characteristics of ASICs in DCs. We found that the inward ASIC-like currents in DCs were reversibly inhibited by a blocker of ASICs, amiloride (Fig. 2), suggesting that ASICs are functionally expressed in DCs. Interestingly, the amplitude of ASIC currents was larger in mDCs than in iDCs (Fig. 4C), indicating that the function of ASICs is enhanced in mDCs.

TRPV1 is a nonselective cation channel with high permeability for Ca2+. It was reported to be responsive to a variety of stimuli, such as noxious heat, Cap, endovanilloids, and acid in neurons (38). A recent study suggested that TRPV1 is expressed in human iDCs and plays a role in the differentiation and activation of iDCs (39). However, previous studies reported conflicting data on the existence and function of TRPV1 in mouse iDCs (26, 40). Using Cap, a specific agonist for TRPV1, we tried to elicit currents in iDCs but were unsuccessful (Fig. 3A). Furthermore, we could not detect TRPV1 expression in iDCs using RT-PCR (Fig. 3B). These results suggested that currents evoked by acid in iDCs are not via TRPV1.

In periphery tissues, DCs are in an immature stage and have phagocytic abilities. After encountering pathogens, they undergo a process of maturation to become efficient APCs and migrate from periphery tissues into the draining lymph nodes to induce primary immune responses. One important feature of mDCs is the increased expression of MHC molecules and costimulatory molecules on the cell membrane. Vermeulen et al. (16) reported that extracellular acidosis upregulates the expression of cell-surface proteins that are involved in Ag presentation of DCs, indicating that acidosis has an important role in the maturation of DCs. Consistent with the data of Vermeulen et al., we found that the surface expression of CD11c, MHC class II, and CD86 on DCs was upregulated in an acidic environment (Fig. 5). In addition to those molecules, the expression of costimulatory molecule CD80 was increased at pH 6.5 (Fig. 5). The upregulations were inhibited by amiloride, a blocker for ASICs (Fig. 6), suggesting that ASICs mediate the maturation of DCs induced by acidosis. Enhanced migration is another characteristic of DC maturation. However, the results suggested that extracellular acidosis does not influence the expression of CCR7 on DCs (Supplemental Fig. 2).

It was suggested that NSAIDs inhibit ASIC currents in various neurons and may protect neurons from damage associated with acidosis. Although Dorofeeva et al. (30) demonstrated that NSAIDs reduced the maximal response to a decrease in pH but did not induce a significant decrease in ASICs’ sensitivity to protons in hippocampal interneurons, the mechanism of the NSAID action is still not clear. Similar to previous reports, the results showed that ibuprofen and diclofenac significantly inhibited ASIC currents in DCs (Fig. 6). Moreover, we found that the maturation of DCs induced by acidosis was also inhibited by ibuprofen and diclofenac (Fig. 7), indicating that the inhibitory effect of NSAIDs on acidosis-induced maturation is mediated by ASICs. However, the mechanism by which NSAIDs inhibit ASICs of DCs needs to be investigated further. Additionally, acidosis increased the ability of DCs to stimulate the proliferation of allogeneic T cells; this effect was significantly inhibited by amiloride, ibuprofen, and diclofenac (Fig. 8), indicating that ASICs mediate the maturation of DCs induced by acidosis and that ASICs in DCs may be a novel target for NSAIDs in the anti-inflammation reaction.

In conclusion, the results showed that ASICs play an important role in the physiologic function of mouse bone marrow-derived DCs as receptors for extracellular acidosis. Functional ASICs are expressed in DCs and mediate the upregulation of CD11c, MHC class II, CD80, and CD86 induced by acidosis. ASICs also mediate the Ag-presenting ability of DCs induced by acidosis. ASICs may be a new therapeutic target for regulating the function of DCs in inflammation, tumors, and autoimmune diseases. However, further efforts will be made to clarify the precise mechanism by which ASICs transduce the signal of extracellular acidosis to DCs and induce their maturation.

We thank Dr. Jixin Zhong, Center for Biotechnology and Genomic Medicine, Medical College of Georgia, for assistance with preparing the manuscript.

This work was supported by National Development Program (973) for Key Basic Research of China Grant 2007CB512402 (to F.G.), Key Project of National Natural Science Foundation of China Grant 30930104 (to J.C.), and National Natural Science Foundation of China Grant 30700736 (to Z.T.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ASIC

acid-sensing ion channel

Cap

capsaicin

DC

dendritic cell

iDC

immature dendritic cell

mDC

mature dendritic cell

MFI

mean fluorescence intensity

NSAID

nonsteroidal anti-inflammatory drug

TRPV1

transient receptor potential vanilloid-1.

1
Banchereau
J.
,
Briere
F.
,
Caux
C.
,
Davoust
J.
,
Lebecque
S.
,
Liu
Y. J.
,
Pulendran
B.
,
Palucka
K.
.
2000
.
Immunobiology of dendritic cells.
Annu. Rev. Immunol.
18
:
767
811
.
2
Guermonprez
P.
,
Valladeau
J.
,
Zitvogel
L.
,
Théry
C.
,
Amigorena
S.
.
2002
.
Antigen presentation and T cell stimulation by dendritic cells.
Annu. Rev. Immunol.
20
:
621
667
.
3
Niess
J. H.
,
Reinecker
H. C.
.
2005
.
Lamina propria dendritic cells in the physiology and pathology of the gastrointestinal tract.
Curr. Opin. Gastroenterol.
21
:
687
691
.
4
Fong
L.
,
Engleman
E. G.
.
2000
.
Dendritic cells in cancer immunotherapy.
Annu. Rev. Immunol.
18
:
245
273
.
5
Radstake
T. R.
,
van Lieshout
A. W.
,
van Riel
P. L.
,
van den Berg
W. B.
,
Adema
G. J.
.
2005
.
Dendritic cells, Fcgamma receptors, and Toll-like receptors: potential allies in the battle against rheumatoid arthritis.
Ann. Rheum. Dis.
64
:
1532
1538
.
6
Blanco
P.
,
Palucka
A. K.
,
Pascual
V.
,
Banchereau
J.
.
2008
.
Dendritic cells and cytokines in human inflammatory and autoimmune diseases.
Cytokine Growth Factor Rev.
19
:
41
52
.
7
Dubos
R. J.
1955
.
The micro-environment of inflammation or Metchnikoff revisited.
Lancet
269
:
1
5
.
8
Abbot
N. C.
,
Spence
V. A.
,
Swanson-Beck
J.
,
Carnochan
F. M.
,
Gibbs
J. H.
,
Lowe
J. G.
.
1990
.
Assessment of the respiratory metabolism in the skin from transcutaneous measurements of pO2 and pCO2: potential for non-invasive monitoring of response to tuberculin skin testing.
Tubercle
71
:
15
22
.
9
Simmen
H. P.
,
Blaser
J.
.
1993
.
Analysis of pH and pO2 in abscesses, peritoneal fluid, and drainage fluid in the presence or absence of bacterial infection during and after abdominal surgery.
Am. J. Surg.
166
:
24
27
.
10
Månsson
B.
,
Geborek
P.
,
Saxne
T.
,
Björnsson
S.
.
1990
.
Cytidine deaminase activity in synovial fluid of patients with rheumatoid arthritis: relation to lactoferrin, acidosis, and cartilage proteoglycan release.
Ann. Rheum. Dis.
49
:
594
597
.
11
Hunt
J. F.
,
Fang
K.
,
Malik
R.
,
Snyder
A.
,
Malhotra
N.
,
Platts-Mills
T. A.
,
Gaston
B.
.
2000
.
Endogenous airway acidification. Implications for asthma pathophysiology.
Am. J. Respir. Crit. Care Med.
161
:
694
699
.
12
Tannock
I. F.
,
Rotin
D.
.
1989
.
Acid pH in tumors and its potential for therapeutic exploitation.
Cancer Res.
49
:
4373
4384
.
13
Ojugo
A. S.
,
McSheehy
P. M.
,
McIntyre
D. J.
,
McCoy
C.
,
Stubbs
M.
,
Leach
M. O.
,
Judson
I. R.
,
Griffiths
J. R.
.
1999
.
Measurement of the extracellular pH of solid tumours in mice by magnetic resonance spectroscopy: a comparison of exogenous (19)F and (31)P probes.
NMR Biomed.
12
:
495
504
.
14
van Sluis
R.
,
Bhujwalla
Z. M.
,
Raghunand
N.
,
Ballesteros
P.
,
Alvarez
J.
,
Cerdán
S.
,
Galons
J. P.
,
Gillies
R. J.
.
1999
.
In vivo imaging of extracellular pH using 1H MRSI.
Magn. Reson. Med.
41
:
743
750
.
15
Lardner
A.
2001
.
The effects of extracellular pH on immune function.
J. Leukoc. Biol.
69
:
522
530
.
16
Vermeulen
M.
,
Giordano
M.
,
Trevani
A. S.
,
Sedlik
C.
,
Gamberale
R.
,
Fernández-Calotti
P.
,
Salamone
G.
,
Raiden
S.
,
Sanjurjo
J.
,
Geffner
J. R.
.
2004
.
Acidosis improves uptake of antigens and MHC class I-restricted presentation by dendritic cells.
J. Immunol.
172
:
3196
3204
.
17
Wemmie
J. A.
,
Price
M. P.
,
Welsh
M. J.
.
2006
.
Acid-sensing ion channels: advances, questions and therapeutic opportunities.
Trends Neurosci.
29
:
578
586
.
18
Wemmie
J. A.
,
Chen
J.
,
Askwith
C. C.
,
Hruska-Hageman
A. M.
,
Price
M. P.
,
Nolan
B. C.
,
Yoder
P. G.
,
Lamani
E.
,
Hoshi
T.
,
Freeman
J. H.
 Jr.
,
Welsh
M. J.
.
2002
.
The acid-activated ion channel ASIC contributes to synaptic plasticity, learning, and memory.
Neuron
34
:
463
477
.
19
Jahr
H.
,
van Driel
M.
,
van Osch
G. J.
,
Weinans
H.
,
van Leeuwen
J. P.
.
2005
.
Identification of acid-sensing ion channels in bone.
Biochem. Biophys. Res. Commun.
337
:
349
354
.
20
Grifoni
S. C.
,
Jernigan
N. L.
,
Hamilton
G.
,
Drummond
H. A.
.
2008
.
ASIC proteins regulate smooth muscle cell migration.
Microvasc. Res.
75
:
202
210
.
21
Inaba
K.
,
Inaba
M.
,
Romani
N.
,
Aya
H.
,
Deguchi
M.
,
Ikehara
S.
,
Muramatsu
S.
,
Steinman
R. M.
.
1992
.
Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J. Exp. Med.
176
:
1693
1702
.
22
Lutz
M. B.
,
Kukutsch
N.
,
Ogilvie
A. L.
,
Rössner
S.
,
Koch
F.
,
Romani
N.
,
Schuler
G.
.
1999
.
An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow.
J. Immunol. Methods
223
:
77
92
.
23
Xiong
Z.
,
Liu
C.
,
Wang
F.
,
Li
C.
,
Wang
W.
,
Wang
J.
,
Chen
J.
.
2006
.
Protective effects of breviscapine on ischemic vascular dementia in rats.
Biol. Pharm. Bull.
29
:
1880
1885
.
24
Wang
T.
,
Gu
J.
,
Wu
P. F.
,
Wang
F.
,
Xiong
Z.
,
Yang
Y. J.
,
Wu
W. N.
,
Dong
L. D.
,
Chen
J. G.
.
2009
.
Protection by tetrahydroxystilbene glucoside against cerebral ischemia: involvement of JNK, SIRT1, and NF-kappaB pathways and inhibition of intracellular ROS/RNS generation.
Free Radic. Biol. Med.
47
:
229
240
.
25
Page
A. J.
,
Brierley
S. M.
,
Martin
C. M.
,
Hughes
P. A.
,
Blackshaw
L. A.
.
2007
.
Acid sensing ion channels 2 and 3 are required for inhibition of visceral nociceptors by benzamil.
Pain
133
:
150
160
.
26
O’Connell
P. J.
,
Pingle
S. C.
,
Ahern
G. P.
.
2005
.
Dendritic cells do not transduce inflammatory stimuli via the capsaicin receptor TRPV1.
FEBS Lett.
579
:
5135
5139
.
27
Yu
G.
,
Fang
M.
,
Gong
M.
,
Liu
L.
,
Zhong
J.
,
Feng
W.
,
Xiong
P.
,
Wang
C. Y.
,
Gong
F.
.
2008
.
Steady state dendritic cells with forced IDO expression induce skin allograft tolerance by upregulation of regulatory T cells.
Transpl. Immunol.
18
:
208
219
.
28
Chen
J.
,
Daggett
H.
,
De Waard
M.
,
Heinemann
S. H.
,
Hoshi
T.
.
2002
.
Nitric oxide augments voltage-gated P/Q-type Ca(2+) channels constituting a putative positive feedback loop.
Free Radic. Biol. Med.
32
:
638
649
.
29
Yermolaieva
O.
,
Chen
J.
,
Couceyro
P. R.
,
Hoshi
T.
.
2001
.
Cocaine- and amphetamine-regulated transcript peptide modulation of voltage-gated Ca2+ signaling in hippocampal neurons.
J. Neurosci.
21
:
7474
7480
.
30
Dorofeeva
N. A.
,
Barygin
O. I.
,
Staruschenko
A.
,
Bolshakov
K. V.
,
Magazanik
L. G.
.
2008
.
Mechanisms of non-steroid anti-inflammatory drugs action on ASICs expressed in hippocampal interneurons.
J. Neurochem.
106
:
429
441
.
31
Voilley
N.
,
de Weille
J.
,
Mamet
J.
,
Lazdunski
M.
.
2001
.
Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of acid-sensing ion channels in nociceptors.
J. Neurosci.
21
:
8026
8033
.
32
Le Bitoux
M. A.
,
Stamenkovic
I.
.
2008
.
Tumor-host interactions: the role of inflammation.
Histochem. Cell Biol.
130
:
1079
1090
.
33
Gonda
T. A.
,
Tu
S.
,
Wang
T. C.
.
2009
.
Chronic inflammation, the tumor microenvironment and carcinogenesis.
Cell Cycle
8
:
2005
2013
.
34
Centonze
D.
,
Muzio
L.
,
Rossi
S.
,
Cavasinni
F.
,
De Chiara
V.
,
Bergami
A.
,
Musella
A.
,
D’Amelio
M.
,
Cavallucci
V.
,
Martorana
A.
, et al
.
2009
.
Inflammation triggers synaptic alteration and degeneration in experimental autoimmune encephalomyelitis.
J. Neurosci.
29
:
3442
3452
.
35
Eizirik
D. L.
,
Colli
M. L.
,
Ortis
F.
.
2009
.
The role of inflammation in insulitis and beta-cell loss in type 1 diabetes.
Nat. Rev. Endocrinol.
5
:
219
226
.
36
Martínez
D.
,
Vermeulen
M.
,
von Euw
E.
,
Sabatté
J.
,
Maggíni
J.
,
Ceballos
A.
,
Trevani
A.
,
Nahmod
K.
,
Salamone
G.
,
Barrio
M.
, et al
.
2007
.
Extracellular acidosis triggers the maturation of human dendritic cells and the production of IL-12.
J. Immunol.
179
:
1950
1959
.
37
Friese
M. A.
,
Craner
M. J.
,
Etzensperger
R.
,
Vergo
S.
,
Wemmie
J. A.
,
Welsh
M. J.
,
Vincent
A.
,
Fugger
L.
.
2007
.
Acid-sensing ion channel-1 contributes to axonal degeneration in autoimmune inflammation of the central nervous system.
Nat. Med.
13
:
1483
1489
.
38
Pingle
S. C.
,
Matta
J. A.
,
Ahern
G. P.
.
2007
.
Capsaicin receptor: TRPV1 a promiscuous TRP channel
.
Handb. Exp. Pharmacol.
:
155
171
.
39
Tóth
B. I.
,
Benko
S.
,
Szöllosi
A. G.
,
Kovács
L.
,
Rajnavölgyi
E.
,
Bíró
T.
.
2009
.
Transient receptor potential vanilloid-1 signaling inhibits differentiation and activation of human dendritic cells.
FEBS Lett.
583
:
1619
1624
.
40
Basu
S.
,
Srivastava
P.
.
2005
.
Immunological role of neuronal receptor vanilloid receptor 1 expressed on dendritic cells.
Proc. Natl. Acad. Sci. USA
102
:
5120
5125
.

The authors have no financial conflicts of interest.

Supplementary data