The pH-sensing receptor OGR1 improves barrier function of epithelial cells and inhibits migration in an acidic environment
Abstract
The pH-sensing receptor ovarian cancer G protein-coupled receptor 1 (OGR1; GPR68) is expressed in the gut. Inflammatory bowel disease is typically associated with a decrease in local pH, which may lead to altered epithelial barrier function and subsequent gastrointestinal repair involving epithelial cell adhesion and migration. As the mechanisms underlying the response to pH changes are not well understood, we have investigated OGR1-mediated, pH-dependent signaling pathways in intestinal epithelial cells. Caco-2 cells stably overexpressing OGR1 were created and validated as tools to study OGR1 signaling. Barrier function, migration, and proliferation were measured using electric cell-substrate impedance-sensing technology. Localization of the tight junction proteins zonula occludens protein 1 and occludin and the rearrangement of cytoskeletal actin were examined by confocal microscopy. Paracellular permeability and protein and gene expression analysis using DNA microarrays were performed on filter-grown Caco-2 monolayers. We report that an acidic pH shift from pH 7.8 to 6.6 improved barrier function and stimulated reorganization of filamentous actin with prominent basal stress fiber formation. Cell migration and proliferation during in vitro wound healing were inhibited. Gene expression analysis revealed significant upregulation of genes related to cytoskeleton remodeling, cell adhesion, and growth factor signaling. We conclude that acidic extracellular pH can have a signaling function and impact the physiology of intestinal epithelial cells. The deconstruction of OGR1-dependent signaling may aid our understanding of mucosal inflammation mechanisms.
mucosal inflammation is a common feature of the two major forms of inflammatory bowel disease (IBD): Crohn's disease (CD) and ulcerative colitis (UC). The intestinal mucosa forms a barrier that separates the luminal content of the gastrointestinal tract from the internal milieu of the organism. Under normal condition, the intestinal epithelium and its apical tight junctions prevent entry of luminal microorganisms, harmful microbial byproducts, and other noxious contents to the host's internal milieu (22). In UC and CD, epithelial barrier function is impaired, and changes in tight junction protein expression and distribution are observed (57). Other diseases associated with loss of intestinal barrier function are celiac disease, ischemic colitis, and intestinal graft-vs.-host disease (GVHD) (4, 12, 53). Barrier function in intestinal epithelia is differentially influenced by dynamic changes in filamentous actin (F-actin), which subsequently alter tight junction protein distribution and expression (39). Typically, inflammation leads to disruption of epithelial junctions (18, 34).
Signaling pathways involved in the regulation of intestinal barrier function are only beginning to be understood. Assembly, disassembly, and maintenance of tight junctions are influenced by signaling proteins, such as PKC, MAPKs, myosin light-chain kinase, and the Ras superfamily of small GTPases (5, 43, 46). The transcription factor serum response factor (SRF) is a master regulator of the actin cytoskeleton and of contractile homeostasis (25, 36).
Gut-wall inflammation was found associated with extracellular acidification of tissue (9, 29, 37), resulting in a shift of the immunological balance and dysregulation of the intestinal barrier [reviewed in Baumgart and Sandborn (3) and Salim and Soderholm (40)]. Whereas low extracellular pH can be seen as the consequence of a pathophysiological process, it also becomes apparent that extracellular protons can act as messengers, activating specific sensory pathways, and thereby, directly influencing tissue homeostasis. In particular, a small family of three G protein-coupled receptors (GPCRs) has emerged that is able to sense mild-to-moderate extracellular acidification just below the physiological set point of pH 7.4. Maximal receptor activation is observed at pH 6.8–7.0 (21). pH-sensing receptors appear involved, e.g., in vascular homeostasis, inflammation, and bone metabolism (7, 27, 28, 33, 55, 56).
One of these GPCRs, ovarian cancer GPCR 1 (OGR1; alias GPR68), is expressed in the intestinal epithelium and upregulated during inflammation (6). Earlier work has established that this receptor can trigger calcium release from intracellular stores, activate PKC, and stimulate SRF-dependent promoter activity, potentially linking it to epithelial function (21, 28).
Therefore, we set out to examine the role of OGR1 by studying phenotypic and molecular responses to acidic pH in Caco-2 cells, a human epithelial cell line derived from a colon adenocarcinoma of a Caucasian male. These cells represent a widely used model system for the study of signal transduction pathways implicated in epithelial function. Our findings suggest that activation of OGR1 signaling by acidic pH causes actin cytoskeleton modification via intracellular calcium/PKC, ERK, and SRF signaling cascades, with pronounced basal stress fiber formation and subsequent enhanced barrier function, in addition to increased protein and RNA expression of cell adhesion and ECM protein-binding genes.
MATERIALS AND METHODS
Reagents and antibodies.
Primary antibodies used for immunocytochemistry included the following: rabbit anti-E-cadherin (24E10; #3195; Cell Signaling Technology, Danvers, MA), rabbit anti-occludin (#31721; Abcam, Cambridge, UK), mouse anti-vinculin (#11194; Abcam), and rabbit anti-zonula occludens protein 1 (ZO-1; N-term; #40-2300; Invitrogen, Basel, Switzerland). Secondary antibodies used for immunocytochemistry included the following: Alexa goat anti-mouse 488 (#A11029; Invitrogen), Alexa goat anti-rabbit 633 (#A21070; Invitrogen), donkey anti-rabbit 488 (#703-505-155; Jackson ImmunoResearch, West Grove, PA), and goat anti-mouse 549 (#115-506-068; Jackson ImmunoResearch). Dyes and chemicals used for cellular staining included the following: Alexa Fluor 488 Phalloidin (#A12379; Invitrogen) and nuclear stain Hoechst 34580 (#H21486; Invitrogen).
Primary antibodies used for Western blotting included the following: mouse anti-claudin-2 (CLDN2) antibody (#32-5600; Invitrogen), mouse anti-lamin A/C (LMNA) antibody (#612162; BD Biosciences, Allschwil, Switzerland), rabbit anti-NF-κB p65 (Ser276) antibody (#3034; Cell Signaling Technology), rabbit anti-IκB-α antibody (#9242; Cell Signaling Technology), and mouse anti-human GAPDH (monoclonal; #MCA4740; AbD Serotec, Oxford, UK). Secondary antibodies used for Western blotting and conjugated to horseradish peroxidase included the following: donkey anti-rabbit IgG (#NA934; GE Healthcare, Glattbrugg, Switzerland) and goat anti-mouse (#A8924; Sigma-Aldrich, Buchs, Switzerland).
Cell culture.
Caco-2 cells (LGC Promochem, Molsheim, Switzerland) and derived clones were cultured in 5% CO2, 37°C, in DMEM (GlutaMAX; Gibco, Invitrogen), with an additional 100 U/ml penicillin and 100 μg/ml streptomycin (Sigma-Aldrich), geneticin-selective antibiotic, 400 μg/ml (G418 Sulfate; Gibco, Invitrogen), and 10% FCS (2-01F120-I; Amimed; BioConcept, Allschwil, Switzerland).
pH shift experiments were carried out in serum-free RPMI medium (1-41F24-I; Amimed; BioConcept), supplemented with 2 mM GlutaMAX (35050-038; Gibco, Invitrogen) and 20 mM HEPES. For pH adjustment of the RPMI medium, the appropriate quantities of NaOH or HCl were added, and the medium was left to equilibrate in the CO2 incubator for at least 36 h before using it for culture.
OGR1 overexpression.
The OGR1 construct (National Center for Biotechnology Information reference sequence NM_003485) was obtained through PCR using the following oligonucleotide primers: human (h)OGR1 forward 1, 5′ GGA TCC ATG AGG AGT GTG GCC CCT TCA GGC C; hOGR1 reverse 1, 5′ GAA TTC TAG GCC AAC CTG CCC GTG GGG AAC C (initiation and stop codons are underlined).
The OGR1 cDNA was obtained from human genomic DNA (Clontech, Mountain View, CA) using PCR and cloned into the mammalian expression plasmid pcDNA3.1(+) using the BamHI and EcoRI sites. All constructs were verified by DNA sequencing (Microsynth, Balgach, Switzerland). Cells were transfected with the OGR1 expression construct in serum-free DMEM by using the FuGENE HD Transfection Reagent (Roche Diagnostics, Basel, Switzerland). Stable transfectants were selected by treatment with 400 μg/ml G-418 for 2–3 wk. Cells exhibiting intracellular calcium signaling upon acidification were selected for further expansion. Further selection was based on significant inositol phosphate (IP) formation at acidic pH and confirmation of robust OGR1 mRNA expression compared with Caco-2 cells harboring the empty vector (Caco-2 vector control).
Calcium flux.
Calcium flux and IP formation experiments were carried out in HBSS containing 130 mM NaCl, 0.9 mM NaH2PO4, 5.4 mM KCl, 0.8 mM MgSO4, 1.0 mM CaCl2, and 25 mM glucose, buffered with 20 mM HEPES (15630-056; Invitrogen). The pH of all solutions was adjusted using a calibrated pH meter (Metrohm, Herisau, Switzerland). All data presented in this paper are referenced to pH measured at room temperature. To obtain pH at 37°C, 0.15 pH unit should be subtracted for HEPES buffers in the range of pH 6.8–7.8, according to our calibration experiments.
Cells were seeded in uncoated, 384-well, clear-bottom, black-walled imaging plates (#3712; Corning, Lowell, MA). Seeding density was 30,000 cells/well, resulting in ∼90% confluent monolayers on the day of Ca2+ imaging. After 24–36 h, cell culture medium was removed, and cells were loaded for 1 h at 37°C in a non-CO2 incubator, with 20 μl Calcium 4 dye (prepared according to the manufacturer's instructions; Molecular Devices, Sunnyvale, CA) in pH 7.8 HBSS buffer, containing 2.5 mM Probenecid (#P-8761; Sigma-Aldrich), to reduce the baseline signal. Plates were then transferred to the FLIPR (Molecular Devices) or FDSS7000 (Hamamatsu, Shizuoka Prefecture, Japan) fluorescence plate reader and Ca2+ responses measured using a charge-coupled device camera with excitation at 480 nM and emission at 540 nM. Before any stimulation, a baseline was recorded over 5 s (Fb). For pH shift, another 10 μl pH 7.8 HBSS buffer was added as control, followed by 20 μl appropriate pH agonist buffers, resulting in the desired final pH. Fluorescence was read over 1.5 min. In routine experiments, peak fluorescence (Fpeak) was determined and normalized to baseline. Data are reported as ΔF/F = (Fpeak − Fb)/Fb. In specific cases, a calibration step was added to obtain maximum and minimum fluorescence (Fmax and Fmin, respectively) readings for calculation of free intracellular Ca2+ concentration ([Ca2+]i; see Fig. 1B). To this end, ionomycin and EDTA were added as indicated. [Ca2+]i was calculated using the following equation, with Kd = 345 nM for the Calcium 4 dye: [Ca2+]i = Kd (F − Fmin)/(Fmax − F) (48).
IP formation assay.
Cells were seeded in 48-well plates (#3548; Corning) at an approximate density of 100,000 cells/well and grown to near confluence. IP formation was measured as described previously (21), using a 20-min incubation period and 20 mM LiCl to block inositol monophosphatase.
RNA extraction and quantitative real-time PCR.
Total RNA was isolated using the RNeasy Mini Kit in the automated QIAcube following the manufacturer's recommendations (Qiagen, Hombrechtikon, Switzerland). For removal of residual DNA, a DNase treatment was performed, according to the manufacturer's instructions, for 15 min at room temperature as part of the QIAcube program. Determination of mRNA expression was performed by quantitative real-time PCR on a 7900HT real-time PCR system (Applied Biosystems, Foster City, CA), under the following cycling conditions: 20 s at 95°C, then 45 cycles of 95°C for 1 s, and 60°C for 20 s with the TaqMan Fast Universal Master Mix. Samples were analyzed as triplicates. Relative mRNA expression was determined the by the ΔΔCt Method (20), which calculates the quantity of the target sequences relative to the endogenous control β-actin or GAPDH and a reference sample.
TaqMan Gene Expression Assays (all from Applied Biosystems), used in this study, were Hs 00268858-S1 GPR68 (OGR1), Hs 00270999-S1 GPR4, Hs 00269247-S1 GPR65 T cell death-associated gene 8 (TDAG8), human GAPDH Vic minor groove binder and nonfluor (4326317E), and human β-actin Vic TAMRA (4310881E).
Quantification of phosphorylated ERK1/2.
ERK1/2 phosphorylation levels were determined by cell-based sandwich immunoassay and quantified by homogenous time-resolved fluorescence (Cellul'erk phospho-Erk1/2; Cisbio Bioassays, Codolet, France). Caco-2 OGR1 clones U1 and U18 and vector control cells were seeded in 96-well plates at a density of 25,000 cells/well and cultured overnight at 37°C/5% CO2. Cells were starved for 5 h in pH 7.6 serum-free medium, followed by treatment with pH 6.8 pre-equilibrated-buffered medium for 10 or 30 min at 37°C. Levels of phosphorylated ERK1/2 were determined following the protocol provided by Cisbio Bioassays. Data were normalized to the signals obtained at nonstimulating pH (pH 7.8 = 100%).
Luciferase reporter assays.
We used a serum response element (SRE)-luciferase reporter construct (Clontech) to study the effect of proton activation in OGR1-overexpressing Caco-2 cells on the SRF activation pathway. The SRE reporter construct, used here, encodes a firefly luciferase gene under the control of a minimal cytomegalovirus (CMV) promoter and tandem repeats of the SRE.
Cells were seeded in 48-well plates (#3548; Corning) at a density of 120,000 cells/well. The next day, cells were cotransfected with 500 ng each of the SRE-luciferase reporter construct or the control reporter plasmid pTAL-luc (Clontech) at a ratio of 3 μl FuGENE HD (Roche Diagnostics)/1 μg DNA. To control for variations in transfection efficiency, 100 ng of the pRL Renilla luciferase reporter plasmid (pRL-CMV; Promega, Madison, WI) was cotransfected in each well. Eighteen hours after transfection, the cells were starved in serum-free pH 7.8 RPMI medium for at least 5 h and then treated for 3 h in pH 6.8 or 7.8 pre-equilibrated serum-free RPMI medium. Cells were harvested in 1× passive lysis buffer (Promega), and luciferase activities were measured by using a GloMax-Multi Detection System (Promega). The pH 7.8 control condition values were set to 100%. The pCMV empty vector and the pTAL luciferase reporter vector were used as negative controls. Triplicate wells were measured for each transfection, and experiments were performed at least three times.
Transepithelial electrical resistance and epithelial cell permeability (paracellular flux).
Cells were seeded on noncoated Transwell Corning clear polyester membrane filters in 12-well format, 0.4 μm pore size (#3460; Sigma-Aldrich) at 125,000 cells/well. Cells were allowed to grow and differentiate for 3 wk before assays were performed. Culture medium was replaced 3×/wk and daily after 14 days.
Transepithelial electrical resistance (TEER) was measured using an equilibrated Epithelial Voltohmmeter (World Precision Instruments, Berlin, Germany) with “chopstick” electrodes.
Epithelial permeability was quantified by measuring the transepithelial flux of FITC-labeled dextran (40 kDa; Sigma-Aldrich), following confirmation of monolayer integrity by TEER. Starvation in serum-free RPMI medium for 24 h at pH 7.6 preceded simulation by acidic pH. For acidification, the apical medium was replaced with serum-free, pre-equilibrated RPMI at pH 6.7, containing 0.2 mg/ml FITC-labeled dextran, and incubated at 37°C, 5% CO2. After 5 h incubation, 90 μl medium samples from the basal chambers were collected and analyzed for FITC-dextran by fluorometry (EnVision Multilabel Reader; 2104-0010; PerkinElmer, Waltham, MA), excitation 485 nM, and emission 540 nM. Data were normalized by defining flux at pH 7.8 as 100%.
Cell impedance measurements by electric cell-substrate impedance sensing.
Impedance of Caco-2 monolayers was monitored using electric cell-substrate impedance-sensing (ECIS) instruments (Applied BioPhysics, Troy, NY). Experiments were conducted at 37°C and 5% CO2 in a humidified incubator. Cells were seeded in 8- or 96-well plates with electrodes embedded at the bottom of each well (ECIS 8W, 10E or 10E+; ECIS 96W, 10E+). Wells were pretreated with 10 mM L-cysteine. Seeding density was 150,000–250,000 cells/350 μl for eight-well arrays and 150,000–170,000 cells/200 μl for 96-well arrays.
Monolayers were subjected to a pH shift, at the earliest, 3 days after confluence was reached; thus total time after plating was 6–12 days. Growth medium was replaced with starvation medium (pH 7.8 serum-free RPMI medium plus glutamate, 20 mM HEPES) for 4–12 h to keep OGR1 in an inactive state. The baseline resistance was measured for 1 h before treatment. Starvation medium was removed and replaced with pre-equilibrated serum-free RPMI at different pH values in the absence or presence of PMA, as indicated, and impedance was measured in real time at multiple frequencies at a constant current of 1 μA at 20-s intervals.
The ECIS Z Theta instrument (Applied BioPhysics) allows impedance to be split into two components: resistance and capacitance. To determine the most suitable frequency for our experimental setup, multiple frequency scans were performed. As observed for other epithelial cell cultures, maximal effects were observed between 2,000 and 8,000 Hz, and we chose a frequency of 4,000 Hz for our standard experiments.
Automated wound-healing/cell-migration assay with ECIS.
ECIS allows the induction of a defined lesion into a confluent cell monolayer using an electric-field pulse; the repopulation and functional reorganization of the wounded area can be monitored by noninvasive impedance measurements using parameters, as described above. Lesions were set by applying three alternating current voltage pulses of 5.0 V amplitude, 64,000 Hz frequency, 20-s duration, at 2-min intervals. Impedance, resistance, and capacitance were measured at multiple frequencies every 20 s for 24 h.
Immunocytochemistry and confocal laser-scanning microscopy.
Cells were seeded at a density of 150,000–250,000 cells/350 μl per well in eight-well plates (#161107; ibidi, Martinsried, Germany). Plating was done in parallel when seeding the ECIS impedance arrays. Medium and culture conditions, including starvation before the pH change, were as described for the ECIS arrays, unless otherwise stated. Treatment of the epithelial cell monolayers was typically at 7–10 days and at least 3 days after confluency was reached. After low pH treatment, monolayers were rinsed three times with PBS and fixed in 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 (Mallinckrodt, Paris, KY), and blocked for 30 min in blocking solution (5% BSA and 5% goat serum in 0.1% Triton X-100). Cell monolayers were blocked overnight in blocking solution (5% BSA and 5% goat serum) in 0.1% Triton X-100 in PBS. Primary antibodies were diluted (2 mg/ml) in blocking solution and incubated with monolayers for 1 h at room temperature. After washing three times, cell monolayers were incubated with secondary antibodies for 1 h, followed by incubation for 5 min with phalloidin conjugated to Alexa Fluor 488, diluted 1:100 (Alexa Fluor 488 Phalloidin; A12379; Invitrogen), which targets polymerized actin. Nuclei were counterstained with Hoechst 34580. Slides were stored at 4°C in the dark until imaged. Confocal data sets were sampled on a Zeiss LSM 700 inverted microscope; image acquisition and postprocessing were performed with proprietary Zeiss software, Zen 2012. Three random z-stacks/well were acquired and analyzed for each experimental condition. Each experiment contained duplicate wells and was carried out at least three times independently. F-Actin was quantified by measuring fluorescence intensity in ImageJ (42).
Protein extraction and Western blot analysis.
Caco-2 cells were seeded at 250,000 cells/well in six-well Transwell plates (#3450; Sigma-Aldrich) and cultured as described for the paracellular flux assays. Following stimulation as indicated, monolayers were washed twice with ice-cold PBS and scraped directly into ice-cold radioimmunoprecipitation buffer (Thermo Fisher Scientific, Waltham, MA), supplemented with complete protease inhibitor cocktail (Roche Diagnostics). Samples were stored at −20°C until further processing. After thawing, material was centrifuged for 10 min at 13,800 g, and the protein content of the cell lysate supernatants was determined by the bicinchoninic acid protein assay (Thermo Fisher Scientific). Western blotting was performed according to standard procedures, as published (47). Protein bands were visualized using the ECL Plus detection kit from Amersham Biosciences (Arlington Heights, IL). Equal loading of the samples was demonstrated by reprobing membranes with a GAPDH antibody (AbD Serotec). The level of protein expression was quantified by OptiQuant Analysis Software (Microsoft XP). Details on the antibodies used for Western blotting are provided above.
Expression profiling by microarrays.
Caco-2 cells were grown on Transwell plates, as described above for protein extraction, and monolayer integrity was determined by TEER. For pH shift, medium was replaced in the apical compartment with pre-equilibrated serum-free RPMI medium at indicated pH. The control condition was pH 7.8. Medium in the basal compartment was also replaced and maintained at nonstimulating pH 7.8. Treatment time for microarray samples was 24 h. Cells were collected and RNA samples prepared with Ovation PicoSL WTA System V2 (#3312-12; NuGEN, San Carlos, CA). Reverse transcription was carried out from 100 ng total RNA using single primer isothermal amplification, and a linear isothermal strand-displacement DNA amplification process was used to prepare micrograms of amplified cDNA (Ovation PicoSL WTA System V2; #3312-12; NuGEN). cDNA quality and quantity were determined using NanoDrop ND 1000 and Bioanalyzer 2100. Fragmented and biotin-labeled cDNA targets were generated starting with 3.5 μg cDNA with the Encore Biotin Module (#4200-12; NuGEN).
Biotin-labeled cDNA samples were hybridized to the GeneChip Human Gene 1.1 ST Array Strip (P/N 901626; Affymetrix, Santa Clara, CA), following protocols provided by Affymetrix. Data were summarized on gene level using Robust Multi-array Average. For each experimental group (vector control and OGR1-overexpressing clone U1), data quality was assessed using the bioconductor/R package “arrayQualityMetrics” (17), and reproducibility between replicates was assessed using Pearson's correlation for all of the filtered expression values and hierarchical clustering. For all pairwise comparisons, differentially expressed genes were selected using fold change and P value, as determined using ANOVA (as implemented by the R package, Linear Models for Microarray Data) and F-test for the complete experimental design. The results were compared on Venn diagrams and analyzed in the context of existing biological knowledge using MetaCore software for pathway and Gene Ontology Enrichment.
Data were deposited at the Gene Expression Omnibus under GSE60294 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=mzqjsmgmjdmvvef&acc=GSE60294).
Statistical analysis.
Statistical analyses were performed using GraphPad Prism 5 (GraphPad Software, San Diego, CA). Data are presented as means ± SE or SD (ECIS), and statistical significance was determined using unpaired two-tailed Student's t-test. P < 0.05 was considered significant. Where indicated, one-way ANOVA was performed, followed by the Tukey post hoc test.
RESULTS
Caco-2 cells overexpressing OGR1.
Overexpression of the pH-sensing receptor OGR1 in human Caco-2 epithelial cells and isolation of cell clones were performed, as described in materials and methods. OGR1 is a Gq-coupled receptor and is known to stimulate IP formation and intracellular calcium signaling upon exposure to neutral or slightly acidic pH. The cell clones U1 and U18 were selected based on their robust IP formation at pH 6.8 (Fig. 1A). As a consequence of stimulated phosphoinositide turnover, calcium is released from intracellular stores, and this response can be measured using calcium-sensitive fluorescent dyes. In cells expressing OGR1, a shift in extracellular pH to slightly acidic values induced robust increases of intracellular-free calcium concentrations that peaked at >100 nM within 2 min (Fig. 1B). The EC50 for calcium release observed in our experiments was at pH 7.2 (Fig. 1C), well in line with earlier results for OGR1 (21). A significantly weaker response was observed in the vector control cells, which may be attributable to the endogenous expression of low levels of OGR1 in Caco-2 cells. Kinetics and amplitudes of intracellular pH (pHi) changes in response to pH shift were similar in cells expressing OGR compared with vector control, indicating that OGR1 does not interfere significantly with regulation of pHi (data not shown).

Fig. 1.Validation of the Caco-2 ovarian cancer G protein-coupled receptor 1 (OGR1)-overexpressing cell clones U1 and U18. A: OGR1 overexpression leads to pH-dependent inositol phosphate (IP) formation, which was measured in serum-free medium at indicated pH in the absence (n.a.) or presence (Li) of lithium for 20 min. DPM, disintegrations/min; VC, Caco-2_Vector Control; U1, Caco-2_OGR1 clone U1; U18, Caco-2_OGR1 clone U18. B: a shift in pH toward more acidity triggers intracellular calcium transients. Addition of ionomycin and EDTA was performed to obtain maximum and minimum fluorescence values for calibration. A scale for intracellular Ca2+ concentrations ([Ca2+]i), as derived from calibration, is shown on the left. C: pH-dependent mobilization of intracellular calcium. Values shown are normalized fluorescence units [baseline fluorescence (Fb)] taken at peak response, ΔF/F = (Fpeak − Fb)/Fb. D: OGR1 mRNA expression in clones U1 and U18 was confirmed by RT-PCR. Expression data were normalized to β-actin and are expressed relative to vector control (=1). E: ERK1/2 phosphorylation levels following stimulation with low pH for 30 min were determined by immunoassay and quantified by homogenous time-resolved fluorescence. F: serum response factor (SRF)-dependent induction of luciferase activity was assessed at 6 h following pH shift to indicated pH. SRE, serum response element. Statistical significance was determined by 1-way ANOVA, followed by Tukey's post hoc test. A and E: figure elements are representative of 2 independent experiments; B, C, and F: elements are representative of 3 independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
To assess the level of OGR1 mRNA expression in cells, we performed RT-PCR. Clones U1 and U18 showed a several hundred-fold increased mRNA expression relative to vector control (Fig. 1D). We also analyzed expression of the related pH sensors GPR4 and TDAG8 and found unchanged levels when comparing control cells with OGR1 transfectants (data not shown). Transfection of Caco-2 cells with an OGR1 receptor construct bearing a C-terminal myc tag showed expression of the protein predominantly at the plasma membrane (data not shown), as observed before for human embryonic kidney cells and fibroblasts (21).
We next proceeded to test for the level of ERK phosphorylation and SRF-dependent transcription, as OGR1-dependent stimulation of these signaling events was reported in other cell types (50).
OGR1 overexpression leads to increased phosphorylation of ERK1/2 and increased SRF-dependent gene transcription upon acidification.
Exposure of Caco-2 cells overexpressing OGR1 to acidic pH led to a significant phosphorylation of ERK1 and -2 isoforms within minutes (Fig. 1E). This was followed by increased expression of the SRF-dependent luciferase reporter (Fig. 1F). In clones U1 and U18, the signal increased 4.9- and 7.4-fold (P < 0.05 and P < 0.01), respectively, compared with the vector control.
Decreased paracellular permeability at acidic pH in cells overexpressing OGR1.
To investigate the influence of acidic pH on epithelial permeability, the diffusion of fluorescently labeled dextran was measured across the Caco-2 monolayers. We observed a significant decrease in the permeability of the OGR1-overexpressing clone U1 compared with the vector control monolayer, 6 h following the shift of extracellular pH from pH 7.6 to 6.7 (Fig. 2A).

Fig. 2.Improved barrier function at acidic pH. A: acidification significantly decreased FITC-dextran flux across OGR1-overexpressing clone U1 monolayers compared with VC. Measurements of 40 kDa FITC-dextran flux were performed at pH 7.8 and 6.7, over 6 h. Experiments were performed between 21 to 27 days after cell seeding. Data are normalized to pH 7.8 = 100%, and the normalized flux at pH 6.7 is presented. Conditions within experiments were performed in triplicate. Data are shown as average from 5 independent experiments ± SE. OGR1 overexpression in Caco-2 cells [clones U1 (B) and U18 (C)] led to improved impedance at acidic pH. Changes in impedance of cell monolayers were monitored in real time by electric cell-substrate impedance-sensing (ECIS) and reported at frequency 4,000 Hz. Representative experiments are shown. B: data were collected in the 8-well array format. Conditions within experiments were in duplicate or triplicate. Impedance was measured every 20 s for 14 h at pH 7.6 or 6.65 or with PMA (2 nM, pH 7.6). Data are shown as means ± SD. C: data were collected using the 96-well array format. Impedance was measured every 30 s for 20 h, and results are shown as means ± SD of 6 replicates for each condition: pH 7.6 or 6.77 or PMA (1 nM, pH 7.6). D: acidification significantly increased impedance in OGR1-overexpressing clones U1 and U18 compared with VC. Results from 6 independent experiments, performed in the 8-well array format, are presented as average impedance ± SE. Values were taken at 5 h and normalized by the calculation (Max − Min)/Min. Statistical significance was determined by 1-way ANOVA, followed by Tukey's post hoc test. **P < 0.01, ***P < 0.001.
We carried out further experiments to monitor epithelial function using measurements of cellular impedance. This technology allows for noninvasive monitoring of epithelial function over time under several experimental conditions in parallel. Typical time traces of cellular impedance for clones Caco-2 U1 and U18 compared with vector control are shown in Fig. 2, B and C, respectively. For both clones, exposure to acidic pH led to increased impedance that developed slowly over time, peaked ∼3–6 h, and thereafter declined. To exclude possible artifacts due to cell cloning, we confirmed an impedance increase in response to acidification in nonclonal cell populations isolated directly following transfection. As expected, impedance changes measured at 6 h after pH shift were smaller but significant (OGR1 population pH 7.7: 44.8 ± 20.5; OGR1 population pH 6.8: 197.8 ± 36.3; means + SE; n = 4; P < 0.05). For vector control, there was no significant change.
PMA, a potent and reversible PKC activator, was used as a positive control. PMA was shown to activate PKC and increase TEER in Caco-2 cells (52). As expected, PMA, applied at pH 7.6, induced an increase in impedance in all cells, including the vector control, with similar kinetics and amplitude, as observed for the response to low pH in the OGR1-transfected clones.
Figure 2D shows normalized impedance responses at 5 h, compiled from several experiments carried out independently. The response of OGR1-transfected Caco-2 cells to acidic pH was highly significant. The small response to pH shift in vector control cells may be due to the presence of low levels of endogenous OGR1.
Inhibition of cell migration at acidic pH in cells overexpressing OGR1.
The effect of stably expressed OGR1 on cell migration was assessed by an automated wound-healing/cell-migration assay with ECIS. Wounds were introduced as described in materials and methods, following cell culture at acidic pH for 6 h. Impedance, resistance, and capacitance readings were recorded at multiple frequencies to monitor how cells reoccupy the electrode area in culture wells. Figure 3A shows the vector control, where no significant change in impedance was observed among the experimental conditions tested (pH 7.6, 7.0, and 6.8), and no difference in the migration rates after wounding could be measured. On the other hand, cells expressing OGR1 exhibited the pH-dependent impedance increase, as described before, and following wounding, the recovery of impedance was inhibited at pH 7.0 and 6.8 compared with pH 7.6, indicating reduced cell migration (Fig. 3B).

Fig. 3.In vitro wound-healing assay. Automated wound-healing/cell-migration assays were performed with ECIS, where an electric field is used to introduce a defined lesion into the monolayer, and the repopulation of this wounded area is monitored. Wounds were induced 6 h after the pH shift. A: vector control; B: OGR1-overexpressing clone U1. A and B: each is representative of at least 3 independent experiments. Conditions within experiments were performed in duplicate (control pH 7.6) or triplicate (pH 7.0, pH 6.8). All conditions, with the exception of VC, pH 7.6, are presented as means ± SD.
Increased F-actin stress fibers and focal adhesions but no effect on tight junctions in OGR1-transfected Caco-2 cells exposed to acidic pH.
To gain a better understanding of the role of OGR1 in regulating cell–cell and cell–matrix communication in Caco-2 monolayers, we examined the status of actin and several cell junction proteins by confocal microscopy. Exposure of Caco-2 cells expressing OGR1 to slightly acidic pH led to a striking increase of F-actin stress fiber formation, which was nearly absent in vector control cells (Fig. 4). These fibers are basolateral, as shown in the orthogonal view, constructed from a series of Z-stack images of monolayers (Fig. 4B).

Fig. 4.Increased filamentous actin (F-actin) stress fibers and focal adhesions at acidic pH. Caco-2 cell monolayers were acidified for 6 h, at least 3 days after confluency was reached. A: representative images show maximum intensity projections from confocal z-stacks (magnification, 20×; 60–80 slices). Nuclei are identified by Hoechst staining (blue), F-actin cytoskeleton by fluorescein-conjugated phalloidin (green), and occludin by immunofluorescence labeling with anti-occludin (red). Data shown are representative of at least 3 independent experiments. Original scale bar, 20 μm. B: a representative cell monolayer of OGR1-overexpressing clone U18 after 6 h, at pH 6.8, is shown. Basal stress fibers are highly prominent. B, left and bottom: orthogonal views (OV) of the monolayer. F, F-actin stress fibers on the basolateral surface of the cells; N, nuclei in cross-section. C: quantification of F-actin was performed in ImageJ. Data are representative of 4–5 independent experiments. Data are presented as means ± SE. ***P < 0.001.
In contrast to the findings for polymerized actin, no significant increase or redistribution of occludin or ZO-1 was obvious by confocal microscopy analysis after low pH treatment (Figs. 4 and 5, and data not shown).

Fig. 5.Distribution of zonula occludens protein 1 (ZO-1) in monolayers. Experiments were performed as described for Fig. 4. ZO-1 is detected following incubation with anti-ZO-1 (red). No significant differences between the vector control and OGR1 clones U1 or U18 were observed for ZO-1 distribution after treatment at low pH. Data shown are representative of at least 3 independent experiments. Original bar, 20 μm. Caco-2 cell monolayers were acidified for 6 h, at least 3 days after confluency was reached.
We also analyzed the status of cell junction proteins using Western blotting, following our standard experimental conditions for TEER measurements. In contrast to the observations by confocal microscopy, OGR1-overexpressing cells showed a significant increase in both occludin and ZO-1 overall protein content vs. control cells, reflecting the long-term growth of cells under standard cell culture conditions (pH 7.4 and lower), where OGR1 is partially activated. The reasons for this discrepancy are not clear; it may, in part, be due to the fact that Western blotting measures the overall protein pool, whereas immunocytochemistry, as applied here, only detects proteins expressed at the plasma membrane. No difference in expression of either of the tight junction proteins at nonstimulating vs. acidic pH was observed following the relatively short culture time under differentiating pH conditions (Fig. 6, A and B).

Fig. 6.Western blotting for tight junction proteins. Filter-grown Caco-2 vector control and OGR1 clone U1 monolayers, grown for 21 days, were acidified (pH 6.8) for 5–6 h following a 4-h incubation in serum-free medium at pH 7.8. Protein expression from whole-cell lysates was normalized to GAPDH expression by densitometry. PLAUR, urokinase-type plasminogen activator receptor.
Interestingly, there was a significant reduction in expression of the pore-forming protein CLDN2 in cells overexpressing OGR1, which appears nicely complementary to the increase in tight junction proteins described above. Again, no difference in expression levels was detected following 5–6 h after pH shift (Fig. 6E).
Vinculin has been reported to couple integrins to the actomyosin cytoskeleton of focal adhesion proteins to the ECM (26, 32). In agreement with the striking upregulation of F-actin fibers, Western blotting for vinculin revealed a slight increase in protein expression at pH 6.8 vs. 7.7 (Fig. 6C). This result is also in agreement with the microarray gene expression analysis reported below, where an expression fold change of 2.29 was shown for vinculin mRNA upon OGR1 activation by acidic pH (Table 1).
Entrez Gene ID | Gene Symbol | Gene Name |
---|---|---|
6059 | ABCE1 | ATP-binding cassette, subfamily E (OABP), member 1 |
27063 | ANKRD1 | Ankyrin repeat domain 1 (cardiac muscle) |
301 | ANXA1 | Annexin A1 |
330 | BIRC3 | Baculoviral inhibitor of apoptosis repeat containing 3 |
654 | BMP6 | Bone morphogenetic protein 6 |
10917 | BTNL3 | Butyrophilin-like 3 |
80323 | CCDC68 | Coiled-coil domain containing 68 |
634 | CEACAM1 | Carcinoembryonic antigen-related cell adhesion molecule 1 (biliary glycoprotein) |
1048 | CEACAM5 | Carcinoembryonic antigen-related cell adhesion molecule 5 |
4680 | CEACAM6 | Carcinoembryonic antigen-related cell adhesion molecule 6 (nonspecific crossreacting antigen) |
1087 | CEACAM7 | Carcinoembryonic antigen-related cell adhesion molecule 7 |
1081 | CGA | Glycoprotein hormones, alpha polypeptide |
53405 | CLIC5 | Chloride intracellular channel 5 |
1465 | CSRP1 | Cysteine and glycine-rich protein 1 |
2919 | CXCL1 | Chemokine (C-X-C motif) ligand 1 (melanoma growth-stimulating activity, alpha) |
2920 | CXCL2 | Chemokine (C-X-C motif) ligand 2 |
22943 | DKK1 | Dickkopf homolog 1 (Xenopus laevis) |
1843 | DUSP1 | Dual-specificity phosphatase 1 |
1846 | DUSP4 | Dual-specificity phosphatase 4 |
1847 | DUSP5 | Dual-specificity phosphatase 5 |
1848 | DUSP6 | Dual-specificity phosphatase 6 |
1906 | EDN1 | Endothelin 1 |
1948 | EFNB2 | Ephrin-B2 |
2012 | EMP1 | Epithelial membrane protein 1 |
2069 | EREG | Epiregulin |
54206 | ERRFI1 | ERBB receptor feedback inhibitor 1 |
2118 | ETV4 | ets Variant 4 |
388666 | FLJ36116 | Hypothetical locus LOC388666 |
9245 | GCNT3 | Glucosaminyl (N-acetyl) transferase 3, mucin type |
11010 | GLIPR1 | GLI pathogenesis-related 1 |
2877 | GPX2 | Glutathione peroxidase 2 (gastrointestinal) |
1839 | HBEGF | Heparin-binding EGF-like growth factor |
3399 | ID3 | Inhibitor of DNA binding 3, dominant-negative helix-loop-helix protein |
3673 | ITGA2 | Integrin, alpha 2 (CD49B, alpha 2 subunit of VLA-2 receptor) |
3655 | ITGA6 | Integrin, alpha 6 |
3918 | LAMC2 | Laminin, gamma 2 |
112597 | LINC00152 | Long intergenic nonprotein coding RNA 152 |
112597 | LINC00152 | Long intergenic nonprotein coding RNA 152 |
200879 | LIPH | Lipase, member H |
388796 | LOC388796 | Hypothetical LOC388796 |
9645 | MICAL2 | Microtubule-associated monoxygenase, calponin, and LIM domain containing 2 |
406991 | MIR21 | microRNA 21 |
4609 | MYC | v-myc Myelocytomatosis viral oncogene homolog (avian) |
115677 | NOSTRIN | Nitric oxide synthase trafficker |
27035 | NOX1 | NADPH oxidase 1 |
4879 | NPPB | Natriuretic peptide B |
3164 | NR4A1 | Nuclear receptor subfamily 4, group A, member 1 |
4938 | OAS1 | 2′-5′-Oligoadenylate synthetase 1, 40/46 kDa |
5329 | PLAUR | Plasminogen activator, urokinase receptor |
10769 | PLK2 | Polo-like kinase 2 |
5366 | PMAIP1 | PMA-induced protein 1 |
5473 | PPBP | Pro-platelet basic protein [chemokine (C-X-C motif) ligand 7] |
11098 | PRSS23 | Protease, serine, 23 |
5743 | PTGS2 | Prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase) |
5865 | RAB3B | RAB3B, member RAS oncogene family |
29984 | RHOD | ras Homolog gene family, member D |
57494 | RIMKLB | Ribosomal modification protein rimK-like family member B |
6039 | RNASE6 | Ribonuclease, RNase A family, k6 |
6286 | S100P | S100 Calcium-binding protein P |
150094 | SIK1 | Salt-inducible kinase 1 |
28231 | SLCO4A1 | Solute carrier organic anion transporter family, member 4A1 |
677838 | SNORA61 | Small nucleolar RNA, H/ACA box 61 |
9301 | SNORD27 | Small nucleolar RNA, C/D box 27 |
26774 | SNORD80 | Small nucleolar RNA, C/D box 80 |
6876 | TAGLN | Transgelin |
64129 | TINAGL1 | Tubulointerstitial nephritis antigen-like 1 |
4071 | TM4SF1 | Transmembrane 4 L six family member 1 |
10626 | TRIM16 | Tripartite motif containing 16 |
Because lamin A/C was reported to regulate SRF activity via actin polymerization (13), we were interested in evaluating the status of lamin A/C protein levels under our experimental conditions. As observed for ZO-1 and occludin, lamin A/C was increased significantly in cells overexpressing OGR1; however, no influence of the short-term acidic pH shift was observed (Fig. 6D).
Microarray analysis.
To obtain a more comprehensive overview of gene and pathway regulation by acidic pH in Caco-2 cells expressing OGR1, we performed a DNA microarray analysis.
We compared gene expression patterns of vector control cells with those of OGR1-overexpressing cells (clone U1), following 24 h treatment under differentiating pH conditions. Figure 7A shows a heatmap of expression patterns, demonstrating a significant response to acidosis. A volcano plot, indicating the most significant expression changes, is presented in Fig. 7B. Highly regulated genes include genes involved in cell adhesion (carcinoembryonic antigen-related cell adhesion molecule 1 and 5); integrin alpha 2 (ITGA2); laminin gamma 2; vasoconstriction (endothelin-1); inflammatory processes (CXCL2); prostaglandin-endoperoxide synthase 2 (PTGS2), also known as cyclooxygenase-2 (COX-2); nuclear receptor subfamily 4, group A, member 1 (NR4A1); and the WNT signaling pathway inhibitor Dickkopf-related protein 1. Many differentially regulated genes are SRF target genes involved in regulation of the cytoskeleton [Miano et al. (25) and personal communication with J. M. Miano, 2013] (Tables 1 and 2).

Fig. 7.Microarray analysis. A: heatmap showing the expression values obtained in the 4 experimental conditions for all genes with an absolute fold change >1.5. Data are shown as log2 Robust Multi-array Average normalized values; pink = highest, white = average, blue = lowest. B: volcano plot showing the results of fitting a linear model (differential response to stimulation between the OGR1-overexpressing and control cell lines) to the expression values [x-axis, fold change; y-axis, negative log10 adjusted P value (log. adj. P. val)]. NR4A1, nuclear receptor subfamily 4, group A, member 1; PTGS2, prostaglandin-endoperoxide synthase 2; DUSP1/6, dual-specificity phosphatase 1/6; CEACAM1/5, carcinoembryonic antigen-related cell adhesion molecule 1/5; OAS1, 2′-5′-oligoadenylate synthetase 1; ANKRD1, ankyrin repeat domain 1; LAMC2, laminin, gamma 2; DKK1, Dickkopf-related protein 1; EDN1, endothelin 1; ETV4, ets variant 4; BIRC3, baculoviral inhibitor of apoptosis repeat containing 3; PPBP, pro-platelet basic protein.
Cytoskeleton Internal | Membrane | Contractile | Growth | Atypical | Transcription Factors | Validated Unconserved | Hypo/Cyto-Contractile |
---|---|---|---|---|---|---|---|
ACTBa | ACTN1 | ACTA1 | AGPT1 | ANKRD1 | |||
ACTG1 | AOC3 | ACTA2 | BCL2 | ABCA4 | BARX2 | APOD | ANLN |
ACTR3 (ARP3)b | ARC | ACTC | BOK | ADM | E2F5 | AR | ANXA2 |
AKAP12A | CD248 | ACTG2 | CDK16 | AMD1 | EGR1 | CDH5 (mouse) | ANXA3 |
BIN1 | CNP | ATP2A2 | CTGF | ASB5 | EGR2 | CDKN1A (human) | ARPC4 |
CAP1 | DMD | CALB1 | CYR61 | AZI2 | EGR3 | CPVL | BVES |
CAPZA3b | DTNA | CALD1 | DUSP5 | BATF | EGR4 | DOC1 | CDV1 |
CFL1 | ENAH | CASQ1 | DUSP6 | BICC1 | EHOX | DSCR1 | DSP |
CFL2 | ITGA1 | CASQ2 | EDN1 | BPGM | ELF5 | ERRFI1 | FLNB |
CNN1 | ITGA5 | CKM | FGFR3 | BRD3 | ELK1 | HOXB7 | FNBP1 |
CORO1A | ITGB1 | CPT1B | GLIPR1 | CA3 | ETV1 | HSP70 | FNBP3 |
CSRP1 | ITGB1BP2 | CRYAB | GRK6 | CD48 | FHL1 | MTMR6 | GJA1 |
CSRP2 | MOG | DMPK | IER2 (PIP92) | CENPB | FHL2 | OR10T2 | JPH2 |
DES | PLP1 | EFHA1 | IGF2 | CFTR | FOS | OR2B3 | KRT14 |
DSTN (ADF) | SDC2 | KCNMB1 | IL2RA | COL1A2 | FOSB | PGHS2 | KRT18 |
ELMO1 | SDC4 | MYH11 | INS (rat, mouse only) | CTNNBL1 | GPATCH4 | PLAU | KRT19 |
EPLIN | SDPR | MYH4 | MAP3K14 | DNAJB1 | GTF2H5 | PPGB | KRT7 |
FLNA | SPTB | MYH6 | MAP4K4 | EEF1B2 | HES6 | PTMS | KRT8 |
FLNC | TGFB1I1 | MYH7 | MAPK10 | GADD45G | HIF3A | RACK1 | LDB3 |
HSPB7 | TJP1 | MYH9 | MCL1 | GALNT3 | HOXB5 | RNF12 | LRRC10 |
KRT17 | TLN | MYL3 | PRM1 | GPC4 | HOXC6 | SLTRK3 | MSN |
LMOD1 | TMEM34 | MYL4 | PTN | HIPK1 | ING4 | SORBS1 | MYO1B |
LPP | TSPAN2 | MYL9 | SHKBP1 | HSPG2 | JUNB | SREBP2 | MYOM1 |
OPHN1 | VCL | MYLKv6 | TGFBR1 | IMPACT | KLF5 | NEBL | |
PDLIM5 (ENH) | VIL1 | MYLKv7 | THBS1 | INTS9 | KLF6 | NES | |
PFN1 | MYPT1 | VEGFR3 | IPF1 | LEF1 | OCLN | ||
RND3 | PLN | ZFP36 | LCP2 | LZF | PALLD | ||
SNX2 | SLC8A1 | LDLR | MYOD1 | PFN2 | |||
TAGLN1 | SMTNA | LRP4 | NFATC4 | RHOQ | |||
TPM1 | TNNC1 | MAT1A | NFYB | RHOU | |||
TRIP6 | TNNT2 | MBP | NKX2-5 | SVIL | |||
TTN | TPM2 | MGST1 | NR4A1 | SYNM | |||
TUFT1 | MRGPRF | NR4A2 | TCAP | ||||
MRRF | RING1 | TMEM30A | |||||
MRVIL | SKI | TMOD3 | |||||
NPM1 | SMYD1 | TNS1 | |||||
NPPA | SRF | TPM4 | |||||
OPN3 | TCFAP2B | VASP | |||||
P2RX1 | WWTR1 | ZYX | |||||
PAFAH1B1 | |||||||
PGM2 | |||||||
PLAC8 | |||||||
PMFBP1 | |||||||
PTGS2 | |||||||
RAF1 | |||||||
RAI2 | |||||||
RBBP6 | |||||||
RG9MTD2 | |||||||
RRAD | |||||||
RSU1 | |||||||
SCYL1 | |||||||
SEMA3A | |||||||
SERPIND1 | |||||||
SLC2A1 | |||||||
TERA | |||||||
TRAPPC2 | |||||||
TSPAN13 | |||||||
TXNL2 | |||||||
UROD | |||||||
FXN |
Of particular interest is the highly significant regulation of the urokinase-type plasminogen activator receptor (PLAUR or uPAR); this receptor binds the serine protease uPA and regulates localized ECM remodeling. PLAUR is a SRF target gene also involved in regulation of the cytoskeleton (25). We identified antibodies useful to detect the protein in Western blot, and we could indeed demonstrate strong upregulation in cells overexpressing OGR1 compared with vector-transfected controls (Fig. 6F).
DISCUSSION
IBDs are conditions that result from destructive inflammation of the intestinal tract and are characterized by inappropriate and exaggerated mucosal immune responses that can span the entire mucosal wall, as in CD, or be confined to the submucosa, as in UC (18, 35). Mucosal damage and loss of intestinal barrier are also linked to systemic diseases involving other organ systems, such as Type 1 diabetes, GVHD, acquired immunodeficiency syndrome, multiple sclerosis, rheumatoid arthritis, and autism (12, 30).
Enhanced local acidification in the gut lumen as well as in the mucosa has been observed during intestinal inflammation and implicated in the pathogenesis and progression of IBD (9, 29, 37). Mammalian cells have the ability to sense extracellular acidification via acid-sensing ion channels (14, 54) and proton-sensing GPCRs (16, 21).
Today, three pH-sensing GPCRs are described (44). All sense extracellular pH in a narrow range, which appears physiologically relevant; at pH 7.6, the receptors are almost silent, and maximal activation is observed at pH 6.8. The first receptor, GPR4, is predominantly expressed and functional in endothelial cells but may exert functions in other cell types as well (55, 56). GPR65, also known as TDAG8, appears confined to the immune system and bone, where it was shown to regulate cytokine release in macrophages and T cells (27, 31) and to regulate osteoclast activity (11). OGR1, also known as GPR68, the subject of the present study, was found expressed in a variety of cell types, including osteoblasts, airway smooth muscle cells, and neutrophils, and has been associated with, e.g., bone metabolism and inflammatory lung disease (15, 33).
Little information is available on expression and function of OGR1 in epithelial cells. Our earlier work has shown that OGR1 mRNA is expressed in human small intestine and colon (6). We notably observed that OGR1 expression is significantly higher in human colon biopsies from CD and UC patient groups compared with the non-IBD control group. Whereas these results can, in part, be explained by influx of immune cells into the inflamed tissue, there clearly is an epithelial component of OGR1 expression. In agreement with this notion, we detect regulated OGR1 expression in freshly isolated human crypts. We used Caco-2 cells, a well-established model system, to study the potential role of OGR1 in intestinal epithelial biology. Caco-2 cells spontaneously differentiate, polarize, and form tight junctions and an epithelial membrane similar to the human colon with high TEER and low paracellular permeability (2, 10). We found Caco-2 cells to express rather low levels of OGR1 endogenously and therefore, decided to express the receptor ectopically at robust levels. While the present manuscript was in preparation, Thongon and colleagues (49) described a modest upregulation of endogenous OGR1 in Caco-2 cells (approximately twofold at the protein level) by the proton pump inhibitor omeprazole and showed OGR1-dependent regulation of Mg2+ transport.
In our study, we could show that OGR1 acts as a pH sensor in the Caco-2 cell system, as could be expected from earlier work carried out in different cellular backgrounds (21, 28, 41, 50). With respect to epithelial physiology, our results show that activation of OGR1 by acidic pH increases epithelial barrier function strikingly. This effect is linked to pronounced F-actin basal stress fiber formation and correlates with inhibition of cell migration and proliferation during epithelial “wound healing” in vitro. Gene expression analysis using DNA microarrays revealed stimulation of a broad range of inflammatory response genes, as well as induction of a comprehensive set of cytoskeletal and ECM-associated genes described to be regulated by SRF-dependent transcription.
Interestingly, research carried out over the past decade has established that SRF-dependent gene transcription can occur through two parallel pathways: one activated through the originally identified raf/ERK/ternary complex factor cascade and one mediated as a consequence of F-actin assembly, due to the lowering of nuclear globular actin (G-actin) and activation of G-actin-binding proteins of the myocardin-related transcription factor (MRTF) family (51). A recent analysis of serum-induced gene transcription in murine fibroblasts has concluded that the larger number of gene-induction events is linked to the MRTF pathway (8), and indeed, several of the highly regulated genes in the Caco-2 cells exposed to low pH fall into this category [e.g., PTGS2, CXCL2, ITGA2, NR4A1; see Table 2 in Esnault et al. (8)].
The nuclear matrix protein lamin A/C, a product of the LMNA gene, was recently reported as a critical mediator of MRTF-dependent gene transcription and regulator of cell–matrix interaction (13). This protein was strongly induced in Caco-2 cells expressing OGR1, suggesting facilitation of the MRTF axis of signal transduction. It will be interesting to test this conjecture in other cell types responding to OGR1 activation by low pH and to dissect the role of G protein signaling pathways triggered by OGR1 (Gq vs. G13).
Of note, other groups have reported reduced cell motility and increased interaction with ECM in cancer cells expressing OGR1 (19, 38), as well as regulation of cell matrix interaction and contractile function in vascular smooth muscle cells (23, 41).
A proinflammatory function of the OGR1 receptor is well documented (1, 15). Our data obtained with Caco-2 cells confirm this notion, as exemplified by the upregulation of genes, such as PTGS2 = COX2, NR4A, and PLAUR in response to extracellular acidification (24, 45).
In summary, our data show that activation of OGR1 by a slightly acidic pH in Caco-2 epithelial cells leads to an acute barrier enhancement over several hours, reduced cell motility, and a long-term induction of proteins involved in cell junction assembly and cell/matrix interaction, as well as induction of genes associated with tissue inflammation. Figure 8 shows a schematic representation of the proton-induced, OGR1-mediated pathway based on consolidation of our data.

Fig. 8.Schematic representation of proposed OGR1-mediated pathways in intestinal epithelial cells. Activation of OGR1 by extracellular protons results in acute barrier enhancement, F-actin basal stress fiber formation, and induction of actin and microtubule cytoskeleton, cell matrix, cell–cell junctions, and ECM genes, in addition to inflammatory response genes (e.g., NR4A). DAG, diacylglycerol; IP3, inositol triphosphate; TJ, tight junction; AJ, adherens junction; P-ERK, phosphorylated ERK; uPAR, PLAUR.
Whereas stabilization of the epithelial barrier has beneficial effects during acute infections, epithelial cell motility is important for the normal homeostasis of the intestinal mucosa. Therefore, it is expected that chronic activation of OGR1 may exacerbate tissue damage in IBD, which may be sustained further by increased release of inflammatory mediators from myeloid cells exposed to acidic pH (6). Indeed, we could show that deficiency of OGR1 protects female mice from disease manifestations in a murine model of IBD (6).
GRANTS
Support for this research was provided by the University of Zurich Center for Integrative Human Physiology (ZIHP) and grants from the
DISCLOSURES
G. Rogler discloses grant support from Abbott, Ardeypharm, Essex, FALK, Flamentera, Novartis, Tillots, Union Chimique Belge, and Zeller. The other authors declare no competing financial interests.
AUTHOR CONTRIBUTIONS
G.A.K-U., C.A.W., G.R., and K.S. conception and design of research; C.d.V., S.V., G.J., I.T., S.L., and M-G.L. performed experiments; C.d.V., S.V., I.C., G.J., I.T., M-G.L., M.O., and K.S. analyzed data; C.d.V., J.J.E., and K.S. interpreted results of experiments; C.d.V. and K.S. prepared figures; C.d.V. and K.S. drafted manuscript; C.d.V., I.C., and K.S. edited and revised manuscript; C.d.V., S.V., I.C., G.J., I.T., S.L., M-G.L., M.O., J.J.E., G.A.K-U., C.A.W., G.R., and K.S. approved final version of manuscript.
ACKNOWLEDGMENTS
The authors thank Dr. Jelena Kühn Georgijevic, Mr. Lennart Opitz, and Dr. Hubert Rehrauer from the Functional Genomics Center Zurich for the microarray service/analysis; Drs. Ulf Rädler, Christian Renken, and Charles Keese from Applied Biophysics for their expert advice; and Nathalie and Louis de Vallière for preparation of the pathway diagram. Christian Hiller and Barbara Wilmering-Wetter are gratefully acknowledged for their technical assistance.
REFERENCES
- 1. . Proton-sensing ovarian cancer G protein-coupled receptor 1 on dendritic cells is required for airway responses in a murine asthma model. PloS One 8: e79985, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 2. . Selective paracellular permeability in two models of intestinal absorption: cultured monolayers of human intestinal epithelial cells and rat intestinal segments. Pharm Res 10: 1123–1129, 1993.
Crossref | PubMed | Web of Science | Google Scholar - 3. . Crohn's disease. Lancet 380: 1590–1605, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 4. . The two faces of IKK and NF-kappaB inhibition: prevention of systemic inflammation but increased local injury following intestinal ischemia-reperfusion. Nat Med 9: 575–581, 2003.
Crossref | PubMed | Web of Science | Google Scholar - 5. . Small GTPases of the Ras superfamily regulate intestinal epithelial homeostasis and barrier function via common and unique mechanisms. Tissue Barriers 1: e26938, 2013.
Crossref | PubMed | Google Scholar - 6. . G Protein-coupled pH-sensing receptor OGR1 is a regulator of intestinal inflammation. Inflamm Bowel Dis 21: 1269–1281, 2015.
PubMed | Web of Science | Google Scholar - 7. . Acidosis activation of the proton-sensing GPR4 receptor stimulates vascular endothelial cell inflammatory responses revealed by transcriptome analysis. PloS One 8: e61991, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 8. . Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts. Genes Dev 28: 943–958, 2014.
Crossref | PubMed | Web of Science | Google Scholar - 9. . Very low intraluminal colonic pH in patients with active ulcerative colitis. Dig Dis Sci 38: 1989–1993, 1993.
Crossref | PubMed | Web of Science | Google Scholar - 10. . Epithelial properties of human colonic carcinoma cell line Caco-2: electrical parameters. Am J Physiol 247: C260–C267, 1984.
Link | Web of Science | Google Scholar - 11. . TDAG8 activation inhibits osteoclastic bone resorption. FASEB J 28: 871–879, 2014.
Crossref | PubMed | Web of Science | Google Scholar - 12. . The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood 95: 2754–2759, 2000.
Crossref | PubMed | Web of Science | Google Scholar - 13. . Lamin A/C and emerin regulate MKL1-SRF activity by modulating actin dynamics. Nature 497: 507–511, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 14. . Acid-sensitive ion channels and receptors. Handb Exp Pharmacol 283–332, 2009.
Crossref | PubMed | Google Scholar - 15. . Extracellular acidification stimulates IL-6 production and Ca2+ mobilization through proton-sensing OGR1 receptors in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 299: L567–L577, 2010.
Link | Web of Science | Google Scholar - 16. . Acidic tumor microenvironment and pH-sensing G protein-coupled receptors. Front Physiol 4: 354, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 17. . arrayQualityMetrics—a bioconductor package for quality assessment of microarray data. Bioinformatics 25: 415–416, 2009.
Crossref | PubMed | Web of Science | Google Scholar - 18. . The life and death of epithelia during inflammation: lessons learned from the gut. Annu Rev Pathol 7: 35–60, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 19. . Ovarian cancer G protein coupled receptor 1 suppresses cell migration of MCF7 breast cancer cells via a Galpha12/13-Rho-Rac1 pathway. J Mol Signal 8: 6, 2013.
Crossref | PubMed | Google Scholar - 20. . Analysis of relative gene expression data using real-time quantitative PCR and the 2ΔΔCT method. Methods 25: 402–408, 2001.
Crossref | PubMed | Web of Science | Google Scholar - 21. . Proton-sensing G-protein-coupled receptors. Nature 425: 93–98, 2003.
Crossref | PubMed | Web of Science | Google Scholar - 22. . Epithelial barriers in homeostasis and disease. Annu Rev Pathol 5: 119–144, 2010.
Crossref | PubMed | Web of Science | Google Scholar - 23. . Extracellular acidification induces connective tissue growth factor production through proton-sensing receptor OGR1 in human airway smooth muscle cells. Biochem Biophys Res Commun 413: 499–503, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 24. . Inflammation: a role for NR4A orphan nuclear receptors? Biochem Soc Trans 39: 688–693, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 25. . Serum response factor: master regulator of the actin cytoskeleton and contractile apparatus. Am J Physiol Cell Physiol 292: C70–C81, 2007.
Link | Web of Science | Google Scholar - 26. . The role of vinculin in the regulation of the mechanical properties of cells. Cell Biochem Biophys 53: 115–126, 2009.
Crossref | PubMed | Web of Science | Google Scholar - 27. . Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J Immunol 182: 3243–3251, 2009.
Crossref | PubMed | Web of Science | Google Scholar - 28. . The proton-activated G protein coupled receptor OGR1 acutely regulates the activity of epithelial proton transport proteins. Cell Physiol Biochem 29: 313–324, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 29. . Intestinal luminal pH in inflammatory bowel disease: possible determinants and implications for therapy with aminosalicylates and other drugs. Gut 48: 571–577, 2001.
Crossref | PubMed | Web of Science | Google Scholar - 30. . Intestinal permeability defects: is it time to treat? Clin Gastroenterol Hepatol 11: 1075–1083, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 31. . Activation of T cell death-associated gene 8 regulates the cytokine production of T cells and macrophages in vitro. Eur J Pharmacol 683: 325–331, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 32. . New insights into vinculin function and regulation. Int Rev Cell Mol Biol 287: 191–231, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 33. . Extracellular acidification enhances osteoclast survival through an NFAT-independent, protein kinase C-dependent pathway. Bone 42: 150–161, 2008.
Crossref | PubMed | Web of Science | Google Scholar - 34. . Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol 14: 141–153, 2014.
Crossref | PubMed | Web of Science | Google Scholar - 35. . Inflammatory bowel disease. N Engl J Med 347: 417–429, 2002.
Crossref | PubMed | Web of Science | Google Scholar - 36. . Actin' together: serum response factor, its cofactors and the link to signal transduction. Trends Cell Biol 16: 588–596, 2006.
Crossref | PubMed | Web of Science | Google Scholar - 37. . Gastrointestinal pH profiles in ulcerative colitis. Gastroenterology 102: A681, 1992.
Google Scholar - 38. . Effects of ovarian cancer G protein coupled receptor 1 on the proliferation, migration, and adhesion of human ovarian cancer cells. Chin Med J 124: 1327–1332, 2011.
PubMed | Web of Science | Google Scholar - 39. . Regulation of epithelial permeability by the actin cytoskeleton. Cytoskeleton (Hoboken) 68: 653–660, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 40. . Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm Bowel Dis 17: 362–381, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 41. . The GPCR OGR1 (GPR68) mediates diverse signalling and contraction of airway smooth muscle in response to small reductions in extracellular pH. Br J Pharmacol 166: 981–990, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 42. . NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9: 671–675, 2012.
Crossref | PubMed | Web of Science | Google Scholar - 43. . Intestinal infection with Giardia spp. reduces epithelial barrier function in a myosin light chain kinase-dependent fashion. Gastroenterology 123: 1179–1190, 2002.
Crossref | PubMed | Web of Science | Google Scholar - 44. . Receptors for protons or lipid messengers or both? J Recept Signal Transduct Res 26: 599–610, 2006.
Crossref | PubMed | Web of Science | Google Scholar - 45. . Regulation of cell signalling by uPAR. Nat Rev Mol Cell Biol 11: 23–36, 2010.
Crossref | PubMed | Web of Science | Google Scholar - 46. . Regulation of epithelial transport and barrier function by distinct protein kinase C isoforms. Am J Physiol Cell Physiol 281: C649–C661, 2001.
Link | Web of Science | Google Scholar - 47. . Loss of protein tyrosine phosphatase nonreceptor type 22 regulates interferon-γ-induced signaling in human monocytes. Gastroenterology 144: 978–988.e910, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 48. . Measurement of intracellular calcium. Physiol Rev 79: 1089–1125, 1999.
Link | Web of Science | Google Scholar - 49. . The roles of acid-sensing ion channel 1a and ovarian cancer G protein-coupled receptor 1 on passive Mg2+ transport across intestinal epithelium-like Caco-2 monolayers. J Physiol Sci 64: 129–139, 2014.
Crossref | PubMed | Web of Science | Google Scholar - 50. . Prostaglandin I(2) production and cAMP accumulation in response to acidic extracellular pH through OGR1 in human aortic smooth muscle cells. J Biol Chem 280: 34458–34464, 2005.
Crossref | PubMed | Web of Science | Google Scholar - 51. . Shedding light on nuclear actin dynamics and function. Trends Biochem Sci 38: 376–377, 2013.
Crossref | PubMed | Web of Science | Google Scholar - 52. . PKC-dependent regulation of transepithelial resistance: roles of MLC and MLC kinase. Am J Physiol 277: C554–C562, 1999.
Link | Web of Science | Google Scholar - 53. . Tight junctions, intestinal permeability, and autoimmunity: celiac disease and type 1 diabetes paradigms. Ann N Y Acad Sci 1165: 195–205, 2009.
Crossref | PubMed | Web of Science | Google Scholar - 54. . A proton-gated cation channel involved in acid-sensing. Nature 386: 173–177, 1997.
Crossref | PubMed | Web of Science | Google Scholar - 55. . Reduced pathological angiogenesis and tumor growth in mice lacking GPR4, a proton sensing receptor. Angiogenesis 14: 533–544, 2011.
Crossref | PubMed | Web of Science | Google Scholar - 56. . Vascular abnormalities in mice deficient for the G protein-coupled receptor GPR4 that functions as a pH sensor. Mol Cell Biol 27: 1334–1347, 2007.
Crossref | PubMed | Web of Science | Google Scholar - 57. . Changes in expression and distribution of claudin 2, 5 and 8 lead to discontinuous tight junctions and barrier dysfunction in active Crohn's disease. Gut 56: 61–72, 2007.
Crossref | PubMed | Web of Science | Google Scholar