Skip to main content
American Journal of Physiology - Cell Physiology logoLink to American Journal of Physiology - Cell Physiology
. 2023 May 8;324(6):C1263–C1273. doi: 10.1152/ajpcell.00523.2022

SLC26A3 (DRA) is stimulated in a synergistic, intracellular Ca2+-dependent manner by cAMP and ATP in intestinal epithelial cells

Rafiquel Sarker 1, Ruxian Lin 1, Varsha Singh 1, Mark Donowitz 1,2, Chung-Ming Tse 1,
PMCID: PMC10243534  PMID: 37154494

graphic file with name c-00523-2022r01.jpg

Keywords: adenosine 5′-triphosphate, Ca2+, cAMP, DRA, SLC26A3, synergy

Abstract

In polarized intestinal epithelial cells, downregulated in adenoma (DRA) is an apical Cl/HCO3 exchanger that is part of neutral NaCl absorption under baseline conditions, but in cyclic adenosine monophosphate (cAMP)-driven diarrheas, it is stimulated and contributes to increased anion secretion. To further understand the regulation of DRA in conditions mimicking some diarrheal diseases, Caco-2/BBE cells were exposed to forskolin (FSK) and adenosine 5′-triphosphate (ATP). FSK and ATP stimulated DRA in a concentration-dependent manner, with ATP acting via P2Y1 receptors. FSK at 1 µM and ATP at 0.25 µM had minimal to no effect on DRA given individually; however, together, they stimulated DRA to levels seen with maximum concentrations of FSK and ATP alone. In Caco-2/BBE cells expressing the Ca2+ indicator GCaMP6s, ATP increased intracellular Ca2+ (Ca2+i) in a concentration-dependent manner, whereas FSK (1 µM), which by itself did not significantly alter Ca2+i, followed by 0.25 µM ATP produced a large increase in Ca2+ that was approximately equal to the elevation caused by 1 µM ATP. 1,2-Bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM) pretreatment prevented the ATP and FSK/ATP synergistically increased the DRA activity and the increase in Ca2+i caused by FSK/ATP. FSK/ATP synergistic stimulation of DRA was similarly observed in human colonoids. In Caco-2/BBE cells, subthreshold concentrations of FSK (cAMP) and ATP (Ca2+) synergistically increased Ca2+i and stimulated DRA activity with both being blocked by BAPTA-AM pretreatment. Diarrheal diseases, such as bile acid diarrhea, in which both cAMP and Ca2+ are elevated, are likely to be associated with stimulated DRA activity contributing to increased anion secretion, whereas separation of DRA from Na+/H+ exchanger isoform-3 (NHE3) contributes to reduced NaCl absorption.

NEW & NOTEWORTHY The BB Cl/HCO3 exchanger DRA takes part in both neutral NaCl absorption and stimulated anion secretion. Using intestinal cell line, Caco-2/BBE high concentrations of cAMP and Ca2+ individually stimulated DRA activity, whereas low concentrations, which had no/minimal effect, synergistically stimulated DRA activity that required a synergistic increase in intracellular Ca2+. This study increases understanding of diarrheal diseases, such as bile salt diarrhea, in which both cAMP and elevated Ca2+ are involved.

INTRODUCTION

SLC26A3 [downregulated in adenoma (DRA)] is an intestinal brush border (BB) Cl/HCO3 exchange protein that is linked to the BB SLC9A3 [Na+/H+ exchanger-3 (NHE3)] to produce the neutral NaCl absorptive process, the major way intestinal Na+ absorption occurs in the period between meals (19). DRA is expressed in the largest amount in the human proximal colon but is also present in significant amounts in the human duodenum and ileum (3, 6, 7, 10, 55). It is localized to the upper crypt and villus in the small intestine and upper crypt and surface cells of the colon (3, 7, 10); in these cells, it is colocalized with NHE3, and in many of these cells also with cystic fibrosis transmembrane conductance regulator (CFTR; 3, 1114). DRA is functionally and physically linked to NHE3 and takes part in neutral NaCl absorption under baseline conditions. However, in addition, in some conditions associated with elevated cyclic adenosine monophosphate (cAMP), DRA also carries out increased rates of HCO3 secretion and in the same cases, physically interacts with the BB Cl channel, CFTR (3, 12), and dissociates from NHE3. However, the role of DRA in diarrheal diseases is unclear; DRA is inhibited in inflammatory bowel disease (IBD) and Salmonella-related diarrheas, whereas its failure to function or failure to be produced causes congenital chloride diarrhea (2, 5, 15, 16).

There is increasing evidence that increased second messengers, as occurs with most diarrheal diseases, alter DRA activity (2, 3, 5, 8, 9, 12, 13, 1623). Elevation in intracellular cyclic nucleotides and/or Ca2+ is part of the pathophysiology of many diarrheal diseases; for instance, cAMP in cholera toxin-related diarrhea, guanosine 3′,5′-cyclic monophosphate (cGMP) in E. coli heat-stable enterotoxin-related diarrhea, and Ca2+ in rotavirus-related diarrhea, whereas in bile salt-induced diarrhea both elevated cAMP and Ca2+ contribute to the pathogenesis (2430). In addition to the role of cAMP and elevated intracellular Ca2+ signaling pathways in pathological processes, they also take part in the regulation of multiple physiological processes, in which their actions are independent, additive, synergistic, or inhibitory. In the current studies, we tested the hypotheses that both elevated cAMP and Ca2+ stimulated DRA activity and examined the interactions of elevated cAMP and elevated Ca2+ in DRA stimulation to determine whether they affected DRA entirely independently or if there were interactions that were additive or synergistic in polarized intestinal epithelia model cell lines.

MATERIALS AND METHODS

Chemicals, Reagents, and Antibodies

Reagents were obtained from Sigma Aldrich (St. Louis, MO) unless otherwise stated. 2,7-Bis(2-carboxyethyl)-5-carboxyfluorescein acetoxymethyl ester (BCECF-AM) was from Thermo Fisher. Alexa Fluor 488 and 568-conjugated goat anti-mouse and anti-rabbit secondary antibodies were from LI-COR. IRDye 800 (No. 926–32210) and 680 (No. 926–68071) were used with mouse monoclonal and rabbit polyclonal antibodies, respectively (1:10,000 dilutions). Rabbit polyclonal antibodies to human influenza hemagglutinin (HA; C29F4, 1:1,000) and rabbit monoclonal antibodies to HA (No. 3724, 1:1,000) were from Cell Signaling. β-Actin antibodies (No. A2228. 1:3,000) were from Sigma Aldrich. HA-cholinergic M3 receptor cDNA was from Addgene (No. 40753). BPTU N-[2-[2-(1,1-Dimethylethyl)phenoxy]-3-pyridinyl]-N′-[4-(trifluoromethoxy)phenyl]urea was from Tocris Bioscience (No. 6078). DRAinh-A250 was a gift from Dr. Alan Verkman (University of California, San Francisco).

Cell Culture

The Caco-2/BBE cell line originally derived from a human colon adenocarcinoma was obtained from M. Mooseker (Yale University) and J. Turner (Harvard University). Cells were grown on 0.4-µm membrane Transwell inserts in DMEM containing 25 mM NaHCO3 supplemented with 0.1 mM nonessential amino acids, 10% fetal bovine serum (heat inactivated, 55°C for 30 min), 4 mM glutamine, 50 U/mL penicillin, and 50 μg/mL streptomycin, pH 7.4, in 5% CO2/air at 37°C.

Endoscopic specimens of the human proximal colon from healthy human subjects were used to establish primary cultures of human colonoids as previously described (13). Colonoids were expanded and plated on Transwell inserts (0.4 µm, Νo. 3470, Corning Inc.) to form monolayers. For differentiation, colonoids were maintained in a medium that lacked WNT3A, R-spondin1, and SB202190 for 5 days. The procurement and study of human colonoids were approved by the Institutional Review Board of Johns Hopkins University School of Medicine (NA_00038329; 13).

Caco-2/BBE Cells and Human Colonoids Stably Transduced with Lentivirus GCaMP6s and Transiently Transduced with Adenovirus-HA-NHE3

Caco-2/BBE wild-type cells and human proximal colonoids were transduced with GCaMP6s lentiviral particles and stable Caco-2/GCaMP6s and colonic GCaMP6s cell lines were generated with puromycin selection (10 μg/mL) as described previously (31). In some cases, adenovirus-HA-NHE3 was prepared as described and studied ∼48 h after transduction in Caco-2 cells (31).

Measurement of Cl/HCO3 Exchange Activity

Cl/HCO3 exchange activity was measured in Caco-2/BBE cells and colonoid monolayers fluorometrically using the intracellular pH (pHi)-sensitive dye BCECF-AM and a custom chamber allowing separate apical and basolateral perfusion, as previously described (13, 32). Cells were incubated with 10 mmol/L BCECF-AM in NaCl solution (138 mmol/L NaCl, 5 mmol/L KCl, 2 mmol/L CaCl2, 1 mmol/L MgSO4, 1 mmol/L NaH2PO4, 10 mmol/L glucose, 20 mmol/L HEPES, pH 7.4) for 30 min at 37°C and mounted in a fluorometer (Photon Technology International, Birmingham, NJ). Cells were perfused on the apical surfaces with Cl solution (110 mmol/L NaCl, 5 mmol/L KCl, 1 mmol/L CaCl2, 1 mmol/L MgSO4, 10 mmol/L glucose, 25 mmol/L NaHCO3, 1 mmol/L amiloride, 5 mmol/L HEPES, 95% O2/5% CO2) or Cl-free solution (110 mmol/L Na-gluconate, 5 mmol/L K-gluconate, 5 mmol/L Ca-gluconate, 1 mmol/L Mg-gluconate, 10 mmol/L glucose, 25 mmol/L NaHCO3, 1 mmol/L amiloride, 5 mmol/L HEPES, 95% O2/5% CO2) at a flow rate of 1 mL/min; the basolateral perfusion contained the Cl solution throughout the experiment. The switch from Cl-containing to Cl-free solution generates a Cl-out concentration gradient that stimulates HCO3 entry across the cell membrane performed by Cl/HCO3 exchanger(s), and the resulting rate of change in pHi was recorded. Multiple rounds of removing (indicated by closed arrowheads)/replenishing (indicated by open arrowheads) extracellular Cl were performed to determine the Cl/HCO3 exchange activity under basal conditions as a time control as well as in the presence of several compounds, including FSK (1–10 µM, apical and basolateral, 7 min), adenosine 5′-triphosphate (ATP; 0.25–10 µM, apical), uridine-5′-triphosphate (UTP; 1 µM, apical), and carbachol (1 µM, basolateral). At the end of each experiment, pHi was calibrated using K-clamp solutions with 10 mmol/L nigericin (Cayman Chemical, Ann Arbor, MI) that were set at pH 6.8 and 7.8. The initial rate of alkalinization after the switch from Cl solutions to Cl-free solutions was calculated using Origin 8.0 software (OriginLab, Northampton, MA) and in some cases, the stable pHi reached after alkalinization was determined. To determine the effect of the DRA inhibitor, DRAinh-A250 on DRA activity, the control DRA activity was determined by one cycle of Cl removal and readdition to colonoids grown on monolayers. The colonoids were then exposed to 3 µM A250 in both apical and basolateral Cl perfusate for 20 min. Apical Cl perfusate was then switched to Cl-free perfusate containing the same concentration of A250 for determining Cl/HCO3 exchange activity. For determining FSK/ATP-stimulated Cl/HCO3 exchange activity, FSK was included in Cl/A250 perfusate for the last 7 min with A250. ATP was included in the apical Cl-free/A250 perfusate.

Apical Cl/HCO3 exchange in these Caco-2/BBE cells was shown to be contributed by DRA based on prevention of the change in apical Cl removal-induced alkalinization by pretreatment with DRA inhibitor-A250 (5 µM; 13). We previously reported that the IC50 values of DRAinh-A250 in HEK293/DRA cells, Caco-2/BBE cells, and human colonoids were similar and were 0.12 ± 0.04 µmol/L, 0.53 ± 0.10 µmol/L, and 0.22 ± 0.08 µmol/L, respectively (13). DRA knockout in Caco-2 cells completely eliminated Cl-dependent pHi changes (13). We further confirmed that DRAinh-A250 inhibited basal DRA activity and FSK (0.5 µM)/ATP (10 µM) stimulated DRA activity in colonoids in a similar manner (81 ± 3% vs. 76 ± 8% inhibition, respectively) (Supplemental Fig. S1, AC).

Intracellular Ca2+ Measurements

Plasmid pGP-CMV-GCaMP6s (http://addgene.org/40753) was cloned into lentiviral vector pCDH-EF1-MCS-IRES (puro) and lentiviral particles were produced. Caco-2/BBE wild-type cells were transduced with GCaMP6s lentiviral particles and stable Caco-2/GCaMP6s cell lines were generated with puromycin selection (10 μg/mL). Caco-2 cells stably expressing GCaMP were seeded into 12-well Transwell (polyester membrane, Corning No. 3460) plates (1 × 105 cells/well). Cells grown for 12–15 days postconfluency were serum-deprived for 2–3 h before the study. Live images of GCaMP fluorescence were obtained every second with an Olympus FV30000RS confocal microscope with ×10/0.75 numerical aperture (NA) objective lens using the resonance mode at 37°C in the OkoLab stage top plus transparent shroud incubator. ATP (0.25 µM or 1 μM) was added apically at specific time points without interrupting the live cell imaging. Time series were acquired with 512 × 512 pixels, 300 × 300-μm field of view for 600 time points at ∼1 s per time point. In some studies, EGTA (2 μM) was added ∼1 min before FSK and/or ATP addition. In other studies, cells were treated with 25 µM 1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM) for 20–30 min before FSK and/or ATP addition. Intensity of GCaMP6s fluorescence was quantitated by MetaMorph. To determine changes in Ca2+, ATP-induced changes in GCaMP6s fluorescence (F) were divided by baseline fluorescence (F0) and compared with maximum F/F0 that occurred with 1 µM ionomycin.

Statistical Analyses

Results are expressed as means ± SE. Statistical evaluation was done by Student’s t tests or ANOVA with Bonferroni correction when three or more comparisons were made. P values ≤ 0.05 were considered significant.

RESULTS

DRA Is Acutely Stimulated by Elevated Intracellular Ca2+

We previously demonstrated that cAMP (FSK) acutely stimulated DRA activity in Caco-2/BBE cells and human colonoids (13). Here, we determined the effect of elevating intracellular Ca2+ on DRA activity in these cells. Receptor-mediated Ca2+ release from intracellular calcium stores, including endoplasmic reticulum (ER), occurs through both intestinal P2Y purinergic receptors and cholinergic M3 receptors that includes activation of phospholipase C (PLC; 33, 34). As shown in Fig. 1, A and B, in which DRA was measured in the same Caco-2/BBE monolayer under basal conditions and after ATP (1 µM) exposure on the apical surface, ATP rapidly stimulated DRA activity. ATP (1 µM) caused ∼40% stimulation of basal BB DRA activity. Similar experiments determined the effects of 10 and 0.25 µM ATP. The ATP-induced stimulation at 10 µM ATP was slightly but not significantly greater than that at 1 µM ATP, whereas minimal (<10%) ATP stimulation occurred with apical 0.25 µM ATP exposure (Fig. 1C). The effect of apical addition of 1 µM UTP was also determined. Similar to the effect of ATP, apical UTP rapidly stimulated DRA activity (Fig. 1D). Because rotavirus, which causes diarrhea and inhibits Na+ absorption as well as induces intestinal fluid secretion, was shown to cause calcium waves by activation of P2Y1 receptors (33), it was determined if pretreatment with the P2Y1 receptor antagonist N-[2-[2-(1,1-dimethylethyl)phenoxy]-3-pyridinyl]-N′-[4-(trifluoromethoxy)phenyl]urea (BPTU) prevented the ATP (1 µM) stimulation of DRA activity. As shown in Fig. 1E, BPTU (10 µM) pretreatment prevented ATP stimulation of DRA activity.

Figure 1.

Figure 1.

ATP and UTP acutely stimulate DRA activity in Caco-2/BBE cells. A: Cl/HCO3 activity was determined by measuring intracellular pH with BCECF/fluorometry as removing apical bathing Cl solution stimulated intracellular alkalinization that was reversed by readdition of Cl after a constant pHi was reached. Polarized Caco-2/BBE cells with apical and basolateral surface perfused with Cl reached a steady-state pHi and then Cl was removed only from the apical media and initial rates of intracellular alkalinization were determined and in some cases, the steady state of pHi reached was also determined. Multiple cycles of removal (closed arrowhead)/readdition (open arrowhead) of Cl were performed and the effect of apical ATP (1 µM) was determined. Results from a single monolayer are shown with increased initial rate and steady-state pHi after ATP exposure. The Cl/HCO3 exchange activity is considered to be due to DRA activity since Cl removal-induced alkalinization in these cells was entirely prevented by treatment with DRA inhibitor A250 (3–5 µM) (13, 56 and Supplemental Fig. S1). B: effect of ATP (1 µM) on Cl/HCO3 exchange activity, determined as in A. C: experiments as in A and B studying the effects of multiple concentrations of ATP, which were compared with DRA activity in the same monolayer before ATP perfusion; results were calculated with untreated DRA activity set as 100% for each experiment and shown as the horizontal dashed line. ATP (0.25 µM) caused a minimal stimulation of DRA, whereas 1 and 10 µM ATP caused similar significant stimulation compared with basal activity. D: apical UTP (1 µM) acutely stimulated apical Cl/HCO3 exchange activity. The magnitude of increased DRA activity (compare B and D) caused by UTP was similar to that caused by ATP. E: acute ATP stimulation of DRA activity was prevented by pretreatment with 10 µM BPTU, a specific P2Y1 inhibitor, added apically and basolaterally. Results in BE are means ± SE. In all experiments, n represents the number of separate experiments. BCECF, 2,7-Bis(2-carboxyethyl)-5-carboxyfluorescein; BPTU, N-[2-[2-(1,1-dimethylethyl)phenoxy]-3-pyridinyl]-N′-[4-(trifluoromethoxy)phenyl]urea; DRA, downregulated in adenoma; pHi, intracellular pH.

Carbachol is known to elevate intracellular Ca2+ in intestinal epithelial cells via the M3 cholinergic receptor, acting on the endoplasmic reticulum (ER) through phospholipase C (PLC)-inositol 1,4,5-trisphosphate (IP3) signaling (34). The effect of carbachol was determined on DRA activity in Caco-2/BBE cells stably transduced to express the cholinergic M3 receptor, which is not present endogenously (Fig. 2A). Addition of 1 µM carbachol to the basolateral surface rapidly stimulated DRA activity (Fig. 2, B and C) by ∼45% above basal activity in the same cells. These results support that receptor-mediated intracellular elevation of Ca2+ stimulates DRA activity in polarized human intestinal epithelial cells.

Figure 2.

Figure 2.

Carbachol (CCH) acutely stimulates DRA activity in Caco-2/BBE-M3 receptor cells. A: Caco-2 cells were stably transduced with the cholinergic M3 receptor epitope tagged with HA. The M3 receptors were lacking endogenously in these cells. Lysates from these cells were immunoblotted for the epitope-labeled M3 receptor using anti-HA antibody, along with the loading control, β-actin. B and C: experiments similar to those described in Fig. 1A were performed, but examining the effect of basolateral carbachol on Cl/HCO3 exchange activity in Caco-2/BBE-M3 cholinergic receptor cells. B: single experiment. C: means ± SE of multiple experiments, comparing basal and carbachol effects on Cl/HCO3 activity from the same monolayers. n = number of separate experiments. DRA, downregulated in adenoma; HA, human influenza hemagglutinin.

Having shown that acutely elevated Ca2+ stimulates DRA activity, it was determined whether there was an interaction in the DRA stimulation by elevated cAMP and elevated Ca2+; specifically, whether Ca2+ elevation and increased cAMP affected DRA entirely independently or whether there were interactions such that their effects were additive, synergistic, or competitive.

cAMP and ATP Acutely Stimulate DRA in a Synergistic Manner

We previously reported that 10 µM FSK acutely stimulated DRA in Caco-2 cells and in human colonoids (13) and confirmed that finding (Fig. 3, A and B). To evaluate the interactions of cAMP and elevated Ca2+ in the acute stimulation of DRA, lower concentrations of FSK were evaluated for effects on DRA activity in comparison with the effects of the higher concentration. Figure 3C demonstrates that in comparison with DRA stimulation by 10 µM FSK, exposure to 1 µM did not have a significant effect on DRA activity.

Figure 3.

Figure 3.

Acute DRA stimulation by FSK in Caco-2/BBE cells. Apical and basolateral FSK (10 µM) was exposed to Caco-2 cells and ∼7 min later, effects on DRA activity were determined as in Fig. 1A by apical Cl removal. A: Cl/HCO3 activity in a single experiment. Arrowheads as in Fig. 1. B: DRA activity, means ± SE from multiple experiments comparing the effects on DRA activity of 1 and 10 µM FSK, demonstrating significant stimulation of DRA by 10 µM but not by 1 µM FSK. n = number of separate experiments. Ap, apical; BL, basolateral; FSK, forskolin.

SLC26A6 [putative anion transporter 1 (PAT-1)] and CFTR were previously shown to be stimulated in a synergistic manner by elevation of cAMP and Ca2+ (12, 35). As 0.25 µM ATP caused a very minimal stimulation of DRA while 1 µM FSK did not alter DRA activity, the additive effects at these concentrations were determined. In these studies, FSK (1 µM) was exposed to the Caco-2/BBE cells for ∼7 min in Cl-containing media and then ATP (0.25 µM) was added in a Cl-free solution to determine DRA activity. Although 1 µM FSK did not alter DRA activity, this combination of FSK and ATP significantly increased DRA activity by ∼70% above baseline (Fig. 4, AC). These results demonstrate that concentrations of FSK and ATP, which themselves cause minimal or no effects on DRA activity, together cause a robust stimulation, thus demonstrating a synergistic interaction of cAMP and Ca2+. Further studies examined the effects of maximal concentrations of FSK (10 µM) and ATP (10 µM) alone and together on DRA activity. Although both 10 µM ATP (140 ± 2%) and 10 µM FSK (164 ± 6%) alone acutely stimulated DRA, added sequentially, as done in Fig. 4A, their DRA stimulation was not additive (184 ± 11%; Fig. 4D).

Figure 4.

Figure 4.

Concentrations of FSK and ATP that have minimal effects on DRA activity cause synergistic stimulation of DRA in Caco-2/BBE cells. A: single experiment in which Cl/HCO3 exchange activity was determined under basal, untreated conditions and after FSK (1 µM) addition to the apical and basolateral perfusions followed in ∼7 min by apical addition of ATP (0.25 µM). B: multiple experiments as in A, showing DRA activity as means ± SE. n = number of separate experiments. While neither FSK (1 µM) nor ATP (0.25 µM) alone significantly altered DRA activity, together they can stimulate DRA activity. C: multiple experiments as in A and B showing DRA activity as means ± SE but with concentrations of FSK (10 µM) and ATP (10 µM) that each maximally stimulate DRA activity and studied together with a time course as in A. Maximum concentrations of DRA plus ATP did not demonstrate additive or synergistic DRA stimulation. In all individual experiments, results were normalized to untreated DRA activity, which was set as 100% and is shown as the horizontal line. Results are means ± SE. n = number of separate experiments. P values calculated by comparison to untreated conditions for each experiment set as 100%. DRA, downregulated in adenoma; FSK, forskolin.

As DRA has been shown to take part in both neutral NaCl absorption, interacting with NHE3, and anion secretion, while the Caco-2/BBE cells used in these studies express a minimal amount of endogenous NHE3, we determined whether increased NHE3 expression altered the FSK/ATP synergistic DRA stimulation. Adenoviral-NHE3 transduction was used to transiently increase NHE3 expression. The trace of a representative experiment in Fig. 5A demonstrates that DRA stimulation in NHE3 expressing Caco-2/BBE cells with ATP (1 µM) is characterized by both an increase in initial rate of DRA activity and an increase in the steady-state pHi reached and this is similar to the response in wild-type cells (Fig. 1A). Stimulation of DRA by FSK and ATP using concentrations that produced synergistic stimulation, as described in Fig. 4, also caused synergistic stimulation of DRA activity in Caco-2/NHE3 cells, which was slightly but not significantly smaller than that which occurred without the increase in NHE3 expression [compare Fig. 5B, 49 ± 5% and Fig. 4B, 64 ± 8%, not significant (ns)].

Figure 5.

Figure 5.

FSK/ATP-mediated synergistic stimulation of DRA is similar in Caco-2/BBE cells acutely transduced with HA-NHE3. Experiments were performed to determine whether the presence of HA-NHE3 in Caco-2/BBE cells affected the FSK/ATP-induced synergistic stimulation of DRA. Caco-2 cells were transduced with adenovirus-HA-NHE3 and studied for Cl/HCO3 exchange activity ∼48 h later. A: single experiment from the same Caco-2 monolayer with DRA activity determined under basal conditions and after apical addition of 1 µM ATP. The traces are superimposed starting at time 0, although they were performed sequentially. Note that ATP stimulated the initial rate of DRA as well as increased the steady-state pHi achieved. B: similar studies as those performed in Fig. 4 with basal DRA activity and effects of adding FSK (1 µM) followed by ATP (0.25 µM) demonstrated. Results are means ± SE. n = number of separate experiments. DRA, downregulated in adenoma; FSK, forskolin; HA, human influenza hemagglutinin; NHE3, Na+/H+ exchanger isoform-3.

Cytosolic-Free Ca2+ Is Increased by ATP Alone and Synergistically Increased by the Low Concentrations of FSK and ATP, Which Synergistically Stimulate DRA

Changes in free intracellular Ca2+ caused by ATP alone and following FSK were determined in Caco-2/BBE cells stably expressing the genetically encoded Ca2+ indicator, GCaMP6s, that responds to elevation in free Ca2+ with an increase in fluorescent intensity. GCaMP6s was stably expressed in Caco-2/BBE cells by lentiviral transduction with puromycin selection, which created a cell line with GCaMP6s expression in nearly all cells, as demonstrated by confocal microscopy (Fig. 6A). ATP, when added apically to these cells, caused a very rapid increase in intracellular Ca2+ that occurred in a concentration-dependent manner between 0.25 and 10 µM ATP. ATP (0.25 µM) caused a single small peak in Ca2+ elevation that occurred in some cells and even this partial response only occurred in some but not all experiments. One and 10 µM ATP caused higher initial peaks in Ca2+ but also induced several small Ca2+ peaks that occurred at later times (Fig. 6B). In contrast to the effect of ATP, FSK at 1 (Fig. 7) and 10 µM (data not shown) did not alter Ca2+.

Figure 6.

Figure 6.

Apical ATP causes a concentration-dependent increase in intracellular Ca2+ in Caco-2/BBE cells stably expressing the Ca2+ indicator GCaMP6s. A: the Ca2+ sensor protein GCaMP6s was stably expressed in Caco-2/BBE cells grown on glass bottom dishes and the effects of several concentrations of apical ATP were determined on intracellular Ca2+ in cells in an OkoLab stage top plus transparent shroud incubator, studied with Olympus FV3000RS confocal microscope [×20/0.75 numerical aperture (NA) objective] using the resonance mode at 37°C; exposure to ionomycin (1 µM) produced an intracellular fluorescence signal demonstrating expression of GCaMP6s in nearly all cells. There were similar findings when this experiment was repeated three times. Scale bar is 100 µm. B: exposure to several concentrations of apical ATP was quantitated as the ratio of the maximum fluorescence signal achieved to that originally present (F/Fo); ionomycin (1 µM) was added as a positive control at the end of study of each monolayer. ATP caused a concentration-dependent increase in intracellular Ca2+. Single traces of each concentration of ATP are shown. These studies were repeated 7–14 times, with quantitation for 0.25 µM ATP shown in Fig. 7.

Figure 7.

Figure 7.

FSK and ATP synergistically increase intracellular Ca2+ in Caco-2/BBE-GCaMP6s cells. A: intracellular Ca2+was determined as in Fig. 6 in Caco-2/BBE-GCaMP6s cells exposed to 0.25 µM ATP followed by 1 µM ATP and then after 1 µM ionomycin as a positive control. In parallel, the FSK (1 µM)/ATP (0.25 µM) conditions that led to synergistic stimulation of DRA were studied. Synergistic FSK/ATP produced elevated Ca2+ similar to that produced by 1 µM ATP and that produced by 1 µM ionomycin. B: intracellular Ca2+ in Caco-2/BBE-GCaMP6s cells showing maximum increased Ca2+ caused by ATP (0.25 and 1 µM), FSK (1 µM), and synergistic concentrations of FSK (1 µM)/ATP (0.25 µM); studies also performed after pretreatment with BAPTA-AM (25 µM, 20–30 min) or EGTA (2 µM, 1 min) added before ATP and/or FSK. Ionomycin (1 µM) was added as a positive control. Results are means ± SE of maximum Ca2+ level. n = number of monolayers studied. BAPTA-AM prevented any changes in intracellular Ca2+, whereas EGTA pretreatment did not alter the increase in Ca2+ caused by 1 µM ATP and by FSK (1 µM)/ATP (0.25 µM). BAPTA-AM, 1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis(acetoxymethyl ester); FSK, forskolin.

Whether the synergistic stimulation of DRA by FSK and ATP was reflected by a synergistic increase in intracellular Ca2+ was determined. Addition of FSK (1 µM) and ATP (0.25 µM) with the same time sequence as in Fig. 4 caused a rapid increase in intracellular Ca2+ following ATP addition (Fig. 7A). This increase was similar in magnitude to the effect of 1 µM ATP and to the increase caused by 1 µM ionomycin, which was used as a positive control (Fig. 7, A and B). The elevation of intracellular Ca2+ by ATP (0.25 and 1 µM) and the FSK/ATP concentrations causing the synergistic increase in Ca2+ were totally prevented by pretreatment of the Caco-2/BBE cells with BAPTA-AM (25 µM, 20–30 min pretreatment; Fig. 7B), whereas exposure to 2 µM ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA) for 1 min before ATP exposure did not alter the 1-µM ATP-induced increase in Ca2+ or the synergistic increase caused by FSK (1 µM)/ATP(0.25 µM; Fig. 7B).

Elevated Ca2+ Is Necessary for Synergistic Stimulation of DRA by FSK/ATP

Whether the synergistic stimulation of DRA activity was Ca2+ dependent was determined, using the same conditions that were demonstrated to prevent the FSK/ATP increase in intracellular Ca2+. Pretreatment of Caco-2/BBE cells with 25 µM BAPTA-AM for 20–30 min abolished the synergistic stimulation of FSK (1 µM)/ATP (0.25 µM). In addition, BAPTA-AM pretreatment decreased basal DRA activity, although there was a wide range of basal DRA activities in different experiments (Fig. 8).

Figure 8.

Figure 8.

BAPTA-AM prevents the FSK/ATP synergistic increase in DRA activity. A: Cl/HCO3 exchange activity was determined in Caco-2/BBE cells under basal conditions and when stimulated by FSK (1 µM)/ATP(0.25 µM) as in Fig. 4. Similar studies were performed in monolayers exposed to BAPTA-AM (25 µM, 20–30 min pretreatment). BAPTA-AM pretreatment prevented the synergistic stimulation of DRA. Results are means ± SE. n = number of separate experiments. B: comparison of basal DRA activity from experiments including those in A, demonstrating that BAPTA-AM significantly reduced basal DRA activity. Results are means ± SE. n = number of separate experiments. BAPTA-AM, 1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis(acetoxymethyl ester); DRA, downregulated in adenoma; FSK, forskolin.

Human Colonoids Exhibit cAMP/ATP Synergy

Since the low concentrations of FSK and ATP synergistically stimulated DRA in Caco-2/BBE cells and we previously reported that 10 µM FSK acutely stimulated DRA in human colonoids, we tested whether FSK and ATP also synergistically stimulated DRA activity in human colonoids. Low concentrations of FSK and ATP were evaluated for the stimulation of DRA. As shown in Fig. 9A, 0.5 µM FSK had no significant effect on colonoid DRA activity. Similarly, concentrations of ATP up to 10 µM had no significant effect (Fig. 9B). The additive/synergistic effects of FSK and ATP at these concentrations on DRA activity were similarly determined as in Caco-2/BBE cells. In these studies, FSK (0.5 µM) was exposed to the colonoid cells for ∼7 min in Cl-containing media and then ATP (10 µM) was included in Cl-free solution to determine DRA activity (Fig. 9C). This combination of FSK and ATP significantly increased DRA activity by 76 ± 22% (Fig. 9, C and D), confirming that FSK and ATP synergistically stimulate DRA in colonoids.

Figure 9.

Figure 9.

Concentrations of FSK and ATP that have minimal effects on DRA activity cause synergistic stimulation of DRA in human colonoid monolayers. A: single experiment in which Cl/HCO3 exchange activity was determined under basal, untreated conditions and after FSK (0.5 µM) addition to the apical and basolateral perfusions. Cycles of Cl removal and readdition to apical surfaces were indicated by closed and open arrowheads. The basolateral surfaces were always perfused with Cl medium. At this concentration of FSK, there was no effect on colonoid DRA. B: representative experiment showed that apical ATP (10 µM) had no effect on DRA activity. After the basal DRA activity was determined, ATP was included in apical Cl-free medium for studying its effect on DRA. C: single experiment showed that FSK (0.5 µM)/ATP (10 µM) synergistically stimulated DRA activity. After the basal DRA activity was determined, FSK was included in both apical and basolateral Cl-containing perfusates for ∼7min and the apical perfusate was switched to Cl free with ATP. D: multiple experiments as in A, B, and C showing DRA activity as means ± SE. The control untreated DRA activity was set at 100% and is indicated by a horizontal line. DRA activities measured with 0.5 µM FSK or 10 µM ATP are not statistically different from control. n = number of separate experiments. DRA, downregulated in adenoma; FSK, forskolin.

To identify differences in ATP effects on DRA activity and the requirement for higher ATP concentrations to cause synergistic stimulation of DRA activity in human colonoids compared with Caco-2 cells, colonoids stably expressing GCaMPs were studied. As shown in Supplemental Fig. S2, 0.5 µM FSK and 0.25 µM ATP did not significantly alter intracellular Ca2+, whereas 1 and 10 µM ATP similarly increased Ca2+.

DISCUSSION

This study demonstrates that the FSK and apical ATP induced elevated Ca2+ to act synergistically to acutely stimulate DRA in polarized intestinal epithelial cells. Synergistic interaction of cyclic nucleotide and Ca2+ signaling is not unique for the regulation of DRA in intestinal epithelial cells. In fact, cAMP/Ca2+ synergy occurs in many aspects of signal transduction across species, organs, and physiologic responses, as well as is involved in transcriptional and posttranscriptional signaling. Several examples include effects on odor detection in drosophila (36), synergistic GH release from chicken pituitary (37), carbaprostacyclin (cPG12) induction of differentiation of preadipocytes to mature adipocytes (38), stimulation of c-FOS in Jurkat cells (39), and prostaglandin E2 (PGE2)-induced stimulation of prolactin transcription in Jurkat cells (40). Although cAMP/Ca2+ synergy occurs in many signaling pathways, the cross talk between the two-second messenger systems does not always involve synergy; for example, cAMP and Ca2+ are additive but not synergistic in extracellular signal-regulated kinase (ERK) activation in a mouse model of long-lasting long-term potentiation, related to learning (41), and there are examples in which cAMP and elevated Ca2+ have opposing effects on the same signaling pathway, such as in metabotropic glutamate receptor signaling (42).

Intracellular Ca2+ was necessary for the cAMP/Ca2+ synergistic acute stimulation of DRA in Caco-2 cells since both the effect of ATP alone on DRA and the synergistic stimulation were inhibited by pretreatment with BAPTA-AM but not by extracellular EGTA at a concentration that greatly lowered the extracellular free Ca2+ but was only present for a minute before ATP addition so as not to lower intracellular Ca2+. Concerning the upstream signaling, a P2Y1 receptor inhibitor prevented ATP stimulation of DRA activity. Of interest, this is the same signaling pathway involved in spreading the Ca2+ effect across many cells following rotavirus infection of small intestinal villus enterocytes (33).

Multiple mechanisms for cross talk/synergy between cAMP/Ca2+ signaling have been previously defined in detail. Given the involvement of apical P2Y1 receptors with PLC-IP3 generation, the studies by Muallem and colleagues (43) are most relevant to our demonstration in Caco-2 cells that synergistic stimulation of DRA requires the magnitude of elevation of intracellular Ca2+ also to increase synergistically. In the studies by Muallem and colleagues, cAMP increased the binding and sensitivity of IP3 to its ER Ca2+ channel receptor, IP3R, increasing the amount of Ca2+ released per molecule of bound IP3, to synergistically elevate cytosolic Ca2+. However, multiple other mechanisms of cAMP/Ca2+ synergy have been identified. These include, among others, Ca2+-dependent adenylyl cyclase activity (44) and direct effects of calmodulin (CaM) binding to the CFTR R domain, with Ca2+-CaM binding increasing the open probability similar to the effects of cAMP-induced stimulation via phosphorylation, with the overall effects of Ca2+/CaM and cAMP being additive but together achieving the maximum activity caused by either alone (45). The latter results indicate effects via a common pathway. We have previously reported that FSK/cAMP elevates intracellular Ca2+ via Epac1 in T84 cells (46). Of note, in the present study, we showed that FSK failed to increase intracellular Ca2+ in Caco-2 cells and in human colonoids (Fig. 7A and Supplemental Fig. S2).

The current studies appear to help explain previous studies in the model cell, Caco-2/BBE, in which DRA seemed to be both stimulated and inhibited by cAMP (8, 13, 14). We previously reported, based on use of proximity ligation assays in Caco-2/BBE cells expressing transduced NHE3 and endogenous DRA and CFTR, that under baseline conditions, DRA bound NHE3 and CFTR and NHE3 bound DRA and CFTR. These associations were dynamic. With elevation of cAMP, the interaction of NHE3 with DRA and CFTR was reduced and that between DRA and CFTR was increased, with overall activity of DRA increasing (3). The seeming contradiction of both DRA stimulation and inhibition can be explained by occurrence of reduced neutral NaCl absorption related to the previously demonstrated cAMP-induced decrease in NHE3 on the plasma membrane (8) that reduced the pool that interacts with BB DRA. In addition, the elevation of cAMP leads to increased DRA trafficking to the apical membrane where DRA interacts with CFTR and leads to increased anion secretion, as previously shown by us (13). Assuming that this occurs in cAMP/Ca2+ synergy in normal intestinal epithelial cells as well as in Caco-2 cells, it will be important to define the magnitude of the separation of NHE3 from DRA (which is part of the mechanism of inhibition of intestinal Na absorption that occurs in diarrhea as NHE3 trafficking from the plasma membrane to the early endosomes) and that of increased binding of DRA with CFTR (which is part of the stimulation of anion secretion). In the present studies, we showed that synergistic stimulation of DRA was observed whether NHE3 was present or not, and interestingly the magnitude of synergistic stimulation in the presence of NHE3 appeared to be slightly less than when it is minimally present or absent.

The results presented here differ from the detailed studies of Lamprecht and Seidler, also using Caco-2/BBE cells (23, 47). Those studies acutely elevated intracellular Ca2+ with ionomycin or UTP and found that both did not alter initial rates of DRA activity but lowered the steady-state pHi achieved after ionomycin/UTP. This was interpreted to indicate that elevated intracellular Ca2+ probably changed the pHi sensitivity of DRA as the major way Ca2+ altered DRA activity. What explains the differences in our findings is that both FSK and apical ATP, as well as synergistic concentrations of FSK and ATP, stimulate DRA activity that can only be speculated about. The current studies, however, show DRA stimulation that involves both initial rates and steady-state pHi, effects that were reversed by BAPTA-AM, demonstrating that potential injury from elevated Ca2+ was not occurring. It must be noted that another major difference in these two Caco-2/BBE studies relates to the mechanism of DRA stimulation. In the study by Lamprecht et al. (23), neither ionomycin nor UTP altered the plasma membrane expression of DRA, while we previously reported that, using super-resolution combined with confocal microscopy, changes in surface expression of DRA and NHE3 supported that synergist stimulation of DRA was associated with increased apical membrane DRA expression and reduced NHE3 surface expression (3). Of note, there was a major difference in the ways our studies were performed, consisting of the support the Caco-2 cells were grown on. In our studies, Caco-2 cells were grown on semipermeable supports (Transwell filters) allowing access to both apical and basolateral surfaces to bathing solutions with standardized polarization based on days postconfluency studied. Previous demonstration of Caco-2 polarization and differentiation has generally been done with cells grown on such semipermeable supports rather than the solid supports apparently used in the Lamprecht studies (23, 47).

We expanded our studies to normal human colonoids. While similar to Caco-2/BBE cells in that in colonoids, 0.5 µM FSK failed to stimulate DRA activity (Fig. 9A) and 10 µM FSK significantly stimulated it (13), 10 µM ATP failed to significantly affect colonoid DRA activity (Fig. 9B). Nonetheless, the combination 0.5 µM FSK/10 µM ATP synergistically stimulated DRA activity. In parallel studies, the 1-µM ATP-induced increase in Ca2+ (peak F/Fo reached as percent of ionomycin induced maximum increase in F/Fo) was similar in both cell models (Caco-2 cells 69 ± 10% and human colonoids 66 ± 7%) (compare Fig. 7B and Supplemental Fig. S2). While we can conclude that ATP and cAMP synergistically stimulate DRA activity, the human colonoid studies demonstrate that the elevated Ca2+ is not sufficient to stimulate DRA activity and indicate that there are additional downstream factors that are necessary. Further studies are needed to identify the additional factors and to understand the differences in mechanisms of synergy in Caco-2 cells and human colonoids.

The demonstration that cAMP and Ca2+ synergistically stimulate DRA activity has relevance to pathophysiological mechanisms of multiple diarrheal diseases in which both cAMP and Ca2+ are elevated. In each of these, there is a potential role for cAMP/Ca2+ cross talk/synergy but in none have detailed evaluation of these interactions been achieved. PGE2 acutely elevates both intracellular Ca2+ and cAMP in intestinal epithelial cells and is part of the pathophysiology of IBD (48, 49), with effects on multiple intestinal cell types that likely contribute to the IBD pathophysiology (enteric nervous system, macrophages, dendritic cells, others). Bile salts signal by cAMP and protein kinase C (PKC) as part of cholerrheic enteropathy in which increased bile salt concentrations in the lumen of the colon stimulate CFTR activity while inhibiting NaCl absorption (50, 51). In mouse B cells, cross linking the B cell antigen receptor using anti-IgM antibody initially elevates cAMP and subsequently increases intracellular Ca2+, which leads to synergistic increase in CD80 transcription. The latter is part of B cell activation/interferon γ production that participates in multiple viral diarrheas, including rotavirus (52). In addition, low levels of apical ATP are known to be present in some polarized cells (53, 54) with the cause being less certain, although potential explanations include cell turnover/death, due to translocation of immunological and inflammatory cells through the tight junctions, and effects of interactions with the microbiome, among others. These low levels of ATP potentially are able to act synergistically with changes in cAMP that occur as part of digestion, as well as in diarrheal diseases, to synergistically participate in what is considered normal physiological regulation of intestinal salt and water transport. Future studies evaluating the relevance of increases in both cAMP and Ca2+ in the same intestinal cells or in the multiple intestinal cells beyond the enterocytes that contribute to diarrheal disease pathophysiology are likely to provide a new understanding of the pathophysiology of diarrhea and to provide drug targets to develop for treating diarrheal diseases.

DATA AVAILABILITY

Data will be made available upon reasonable request.

SUPPLEMENTAL DATA

Supplemental figure legends: https://doi.org/10.6084/m9.figshare.22306264.

Supplemental Figs. S1 and S2: https://doi.org/10.6084/m9.figshare.22306429.

GRANTS

Studies were supported in part by the NIH/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) RO1 DK026523 (to M.D.), RO1 DK116352 (to C.-M.T./M.D.), R24 DK099803 (to M.D.), PO1AI125181(to M.D.), and Digestive Disease Research Core Center Grant P30 DK089502 (to M.D.).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

R.S., V.S., M.D., and C.-M.T. conceived and designed research; R.S., R.L., and C.-M.T. performed experiments; R.S., R.L., M.D., and C.-M.T. analyzed data; R.S., V.S., M.D., and C.-M.T. interpreted results of experiments; R.S., R.L., M.D., and C.-M.T. prepared figures; R.S., M.D., and C.-M.T. drafted manuscript; R.S., V.S., M.D., and C.-M.T. edited and revised manuscript; R.S., R.L., V.S., M.D., and C.-M.T. approved final version of manuscript.

ACKNOWLEDGMENTS

We acknowledge the helpful input of Dr. Shmuel Muallem in the designing and interpretation of these studies. Graphical abstract created with BioRender and published with permission.

REFERENCES

  • 1. Walker NM, Simpson JE, Yen PF, Gill RK, Rigsby EV, Brazill JM, Dudeja PK, Schweinfest CW, Clarke LL. Down-regulated in adenoma Cl/HCO3 exchanger couples with Na/H exchanger 3 for NaCl absorption in murine small intestine. Gastroenterology 135: 1645–1653.e3, 2008. doi: 10.1053/j.gastro.2008.07.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Anbazhagan AN, Priyamvada S, Alrefai WA, Dudeja PK. Pathophysiology of IBD associated diarrhea. Tissue Barriers 6: e1463897, 2018. doi: 10.1080/21688370.2018.1463897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Donowitz M, Sarker R, Lin R, McNamara G, Tse CM, Singh V. Identification of intestinal NaCl absorptive-anion secretory cells: potential functional significance. Front Physiol 13: 892112, 2022. doi: 10.3389/fphys.2022.892112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Malakooti J, Saksena S, Gill RK, Dudeja PK. Transcriptional regulation of the intestinal luminal Na+ and Cl transporters. Biochem J 435: 313–325, 2011. doi: 10.1042/BJ20102062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Barrett KE. Epithelial transport in digestive diseases: mice, monolayers, and mechanisms. Am J Physiol Cell Physiol 318: C1136–C1143, 2020. doi: 10.1152/ajpcell.00015.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Hayashi H, Nagai H, Ohba KI, Soleimani M, Suzuki Y. Segmental differences in Slc26a3-dependent Cl- absorption and HCO3- secretion in the mouse large intestine in vitro in ussing chambers. J Physiol Sci 71: 5, 2021. doi: 10.1186/s12576-020-00784-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Talbot C, Lytle C. Segregation of Na/H exchanger-3 and Cl/HCO3 exchanger SLC26A3 (DRA) in rodent cecum and colon. Am J Physiol Gastrointest Liver Physiol 299: G358–G367, 2010. doi: 10.1152/ajpgi.00151.2010. [DOI] [PubMed] [Google Scholar]
  • 8. Musch MW, Arvans DL, Wu GD, Chang EB. Functional coupling of the downregulated in adenoma Cl-/base exchanger DRA and the apical Na+/H+ exchangers NHE2 and NHE3. Am J Physiol Gastrointest Liver Physiol 296: G202–G210, 2009. doi: 10.1152/ajpgi.90350.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Lamprecht G, Heil A, Baisch S, Lin-Wu E, Yun CC, Kalbacher H, Gregor M, Seidler U. The down regulated in adenoma (dra) gene product binds to the second PDZ domain of the NHE3 kinase A regulatory protein (E3KARP), potentially linking intestinal Cl-/HCO3- exchange to Na+/H+ exchange. Biochemistry 41: 12336–12342, 2002. doi: 10.1021/bi0259103. [DOI] [PubMed] [Google Scholar]
  • 10.The Human Protein Atlas. Proteinatlas.org.
  • 11. Rossmann H, Jacob P, Baisch S, Hassoun R, Meier J, Natour D, Yahya K, Yun C, Biber J, Lackner KJ, Fiehn W, Gregor M, Seidler U, Lamprecht G. The CFTR associated protein CAP70 interacts with the apical Cl-/HCO3- exchanger DRA in rabbit small intestinal mucosa. Biochemistry 44: 4477–4487, 2005. doi: 10.1021/bi048828b. [DOI] [PubMed] [Google Scholar]
  • 12. Ko SB, Zeng W, Dorwart MR, Luo X, Kim KH, Millen L, Goto H, Naruse S, Soyombo A, Thomas PJ, Muallem S. Gating of CFTR by the STAS domain of SLC26 transporters. Nat Cell Biol 6: 343–350, 2004. doi: 10.1038/ncb1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Tse CM, Yin J, Singh V, Sarker R, Lin R, Verkman AS, Turner JR, Donowitz M. cAMP stimulates SLC26A3 activity in human colon by a CFTR-dependent mechanism that does not require CFTR activity. Cell Mol Gastroenterol Hepatol 7: 641–653, 2019. doi: 10.1016/j.jcmgh.2019.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Singh AK, Riederer B, Chen M, Xiao F, Krabbenhöft A, Engelhardt R, Nylander O, Soleimani M, Seidler U. The switch of intestinal Slc26 exchangers from anion absorptive to HCOFormula secretory mode is dependent on CFTR anion channel function. Am J Physiol Cell Physiol 298: C1057–C1065, 2010. doi: 10.1152/ajpcell.00454.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Moseley RH, Höglund P, Wu GD, Silberg DG, Haila S, de la Chapelle A, Holmberg C, Kere J. Downregulated in adenoma gene encodes a chloride transporter defective in congenital chloride diarrhea. Am J Physiol Gastrointest Liver Physiol 276: G185–G192, 1999. doi: 10.1152/ajpgi.1999.276.1.G185. [DOI] [PubMed] [Google Scholar]
  • 16. Marchelletta RR, Gareau MG, McCole DF, Okamoto S, Roel E, Klinkenberg R, Guiney DG, Fierer J, Barrett KE. Altered expression and localization of ion transporters contribute to diarrhea in mice with Salmonella-induced enteritis. Gastroenterology 145: 1358–1368.e1-4, 2013. doi: 10.1053/j.gastro.2013.08.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Yu Q. Slc26a3 (DRA) in the gut: expression, function, regulation, role in infectious diarrhea and inflammatory bowel disease. Inflamm Bowel Dis 27: 575–584, 2021. doi: 10.1093/ibd/izaa256. [DOI] [PubMed] [Google Scholar]
  • 18. Singla A, Kumar A, Priyamvada S, Tahniyath M, Saksena S, Gill RK, Alrefai WA, Dudeja PK. LPA stimulates intestinal DRA gene transcription via LPA2 receptor, PI3K/AKT, and c-Fos-dependent pathway. Am J Physiol Gastrointest Liver Physiol 302: G618–G627, 2012. doi: 10.1152/ajpgi.00172.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Chatterjee I, Kumar A, Castilla-Madrigal RM, Pellon-Cardenas O, Gill RK, Alrefai WA, Borthakur A, Verzi M, Dudeja PK. CDX2 upregulates SLC26A3 gene expression in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 313: G256–G264, 2017. doi: 10.1152/ajpgi.00108.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Kumar A, Jayawardena D, Anbazhagan AN, Chatterjee I, Priyamvada S, Alrefai WA, Borthakur A, Dudeja PK. Decreased SLC26A3 expression and function in intestinal epithelial cells in response to Cryptosporidium parvum infection. Am J Physiol Cell Physiol 317: C1205–C1212, 2019. doi: 10.1152/ajpcell.00278.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Saksena S, Singla A, Goyal S, Katyal S, Bansal N, Gill RK, Alrefai WA, Ramaswamy K, Dudeja PK. Mechanisms of transcriptional modulation of the human anion exchanger SLC26A3 gene expression by IFN-{gamma}. Am J Physiol Gastrointest Liver Physiol 298: G159–G166, 2010. doi: 10.1152/ajpgi.00374.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Saksena S, Tyagi S, Goyal S, Gill RK, Alrefai WA, Ramaswamy K, Dudeja PK. Stimulation of apical Cl/HCO3(OH) exchanger, SLC26A3 by neuropeptide Y is lipid raft dependent. Am J Physiol Gastrointest Liver Physiol 299: G1334–G1343, 2010. doi: 10.1152/ajpgi.00039.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Lamprecht G, Hsieh CJ, Lissner S, Nold L, Heil A, Gaco V, Schäfer J, Turner JR, Gregor M. Intestinal anion exchanger down-regulated in adenoma (DRA) is inhibited by intracellular calcium. J Biol Chem 284: 19744–19753, 2009. doi: 10.1074/jbc.M109.004127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Taub M, Bonorris G, Chung A, Coyne MJ, Schoenfield LJ. Effect of propranolol on bile acid- and cholera enterotoxin-stimulated cAMP and secretion in rabbit intestine. Gastroenterology 72: 101–105, 1977. doi: 10.1016/S0016-5085(77)80311-9. [DOI] [PubMed] [Google Scholar]
  • 25. Ao M, Sarathy J, Domingue J, Alrefai WA, Rao MC. Chenodeoxycholic acid stimulates Cl(-) secretion via cAMP signaling and increases cystic fibrosis transmembrane conductance regulator phosphorylation in T84 cells. Am J Physiol Cell Physiol 305: C447–C456, 2013. doi: 10.1152/ajpcell.00416.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Bijvelds MJ, Jorna H, Verkade HJ, Bot AG, Hofmann F, Agellon LB, Sinaasappel M, de Jonge HR. Activation of CFTR by ASBT-mediated bile salt absorption. Am J Physiol Gastrointest Liver Physiol 289: G870–G879, 2005. doi: 10.1152/ajpgi.00226.2005. [DOI] [PubMed] [Google Scholar]
  • 27. Domingue JC, Ao M, Sarathy J, Rao MC. Chenodeoxycholic acid requires activation of EGFR, EPAC, and Ca2+ to stimulate CFTR-dependent Cl- secretion in human colonic T84 cells. Am J Physiol Cell Physiol 311: C777–C792, 2016. doi: 10.1152/ajpcell.00168.2016. [DOI] [PubMed] [Google Scholar]
  • 28. Potter GD, Sellin JH, Burlingame SM. Bile acid stimulation of cyclic AMP and ion transport in developing rabbit colon. J Pediatr Gastroenterol Nutr 13: 335–341, 1991. doi: 10.1097/00005176-199111000-00001. [DOI] [PubMed] [Google Scholar]
  • 29. Fondacaro JD, Garvey JL. Bile acid uptake and calcium flux in brush border membrane vesicles. Life Sci 32: 1449–1454, 1983. doi: 10.1016/0024-3205(83)90910-4. [DOI] [PubMed] [Google Scholar]
  • 30. Maenz DD, Forsyth GW. Ricinoleate and deoxycholate are calcium ionophores in jejunal brush border vesicles. J Membr Biol 70: 125–133, 1982. doi: 10.1007/BF01870222. [DOI] [PubMed] [Google Scholar]
  • 31. Sarker R, Valkhoff VE, Zachos NC, Lin R, Cha B, Chen TE, Guggino S, Zizak M, de Jonge H, Hogema B, Donowitz M. NHERF1 and NHERF2 are necessary for multiple but usually separate aspects of basal and acute regulation of NHE3 activity. Am J Physiol Cell Physiol 300: C771–C782, 2011. doi: 10.1152/ajpcell.00119.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Yin J, Tse CM, Avula LR, Singh V, Foulke-Abel J, de Jonge HR, Donowitz M. Molecular basis and differentiation-associated alterations of anion secretion in human duodenal enteroid monolayers. Cell Mol Gastroenterol Hepatol 5: 591–609, 2018. doi: 10.1016/j.jcmgh.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chang-Graham AL, Perry JL, Engevik MA, Engevik KA, Scribano FJ, Gebert JT, Danhof HA, Nelson JC, Kellen JS, Strtak AC, Sastri NP, Estes MK, Britton RA, Versalovic J, Hyser JM. Rotavirus induces intercellular calcium waves through ADP signaling. Science 370: eabc3621, 2020. doi: 10.1126/science.abc3621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Przyborski SA, Levin RJ. Cholinergic modulation of electrogenic ion transport in different regions of the rat small intestine. J Pharm Pharmacol 49: 691–697, 1997. doi: 10.1111/j.2042-7158.1997.tb06094.x. [DOI] [PubMed] [Google Scholar]
  • 35. Shcheynikov N, Wang Y, Park M, Ko SB, Dorwart M, Naruse S, Thomas PJ, Muallem S. Coupling modes and stoichiometry of Cl-/HCO3- exchange by slc26a3 and slc26a6. J Gen Physiol 127: 511–524, 2006. doi: 10.1085/jgp.200509392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Murmu MS, Martin JR. Interaction between cAMP and intracellular Ca(2+)-signaling pathways during odor-perception and adaptation in Drosophila. Biochim Biophys Acta 1863: 2156–2174, 2016. doi: 10.1016/j.bbamcr.2016.05.014. [DOI] [PubMed] [Google Scholar]
  • 37. Perez FM, Malamed S, Scanes CG. Possible participation of calcium in growth hormone release and in thyrotropin-releasing hormone and human pancreatic growth hormone-releasing factor synergy in a primary culture of chicken pituitary cells. Gen Comp Endocrinol 75: 481–491, 1989. doi: 10.1016/0016-6480(89)90184-6. [DOI] [PubMed] [Google Scholar]
  • 38. Vassaux G, Far DF, Gaillard D, Ailhaud G, Negrel R. Inhibition of prostacyclin-induced Ca2+ mobilization by phorbol esters in Ob1771 preadipocytes. Prostaglandins 46: 441–451, 1993. doi: 10.1016/0090-6980(93)90080-q. [DOI] [PubMed] [Google Scholar]
  • 39. Kaptein JS, Yang CL, Lin CK, Nguyen TT, Chen FS, Lad PM. Synergy between signal transduction pathways is obligatory for expression of c-fos in B and T cell lines: implication for c-fos control via surface immunoglobulin and T cell antigen receptors. Immunobiology 193: 465–485, 1995. doi: 10.1016/S0171-2985(11)80431-6. [DOI] [PubMed] [Google Scholar]
  • 40. Gerlo S, Verdood P, Gellersen B, Hooghe-Peters EL, Kooijman R. Mechanism of prostaglandin (PG)E2-induced prolactin expression in human T cells: cooperation of two PGE2 receptor subtypes, E-prostanoid (EP) 3 and EP4, via calcium- and cyclic adenosine 5'-monophosphate-mediated signaling pathways. J Immunol 173: 5952–5962, 2004. doi: 10.4049/jimmunol.173.10.5952. [DOI] [PubMed] [Google Scholar]
  • 41. Miningou Zobon NT, Jędrzejewska-Szmek J, Blackwell KT. Temporal pattern and synergy influence activity of ERK signaling pathways during L-LTP induction. eLife 10: e64644, 2021. doi: 10.7554/eLife.64644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Francesconi A, Duvoisin RM. Opposing effects of protein kinase C and protein kinase A on metabotropic glutamate receptor signaling: selective desensitization of the inositol trisphosphate/Ca2+ pathway by phosphorylation of the receptor-G protein-coupling domain. Proc Natl Acad Sci USA 97: 6185–6190, 2000. doi: 10.1073/pnas.97.11.6185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Park S, Shcheynikov N, Hong JH, Zheng C, Suh SH, Kawaai K, Ando H, Mizutani A, Abe T, Kiyonari H, Seki G, Yule D, Mikoshiba K, Muallem S. Irbit mediates synergy between ca(2+) and cAMP signaling pathways during epithelial transport in mice. Gastroenterology 145: 232–241, 2013. doi: 10.1053/j.gastro.2013.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Cooper DM. Molecular and cellular requirements for the regulation of adenylate cyclases by calcium. Biochem Soc Trans 31: 912–915, 2003. doi: 10.1042/bst0310912. [DOI] [PubMed] [Google Scholar]
  • 45. Bozoky Z, Ahmadi S, Milman T, Kim TH, Du K, Di Paola M, Pasyk S, Pekhletski R, Keller JP, Bear CE, Forman-Kay JD. Synergy of cAMP and calcium signaling pathways in CFTR regulation. Proc Natl Acad Sci USA 114: E2086–E2095, 2017. doi: 10.1073/pnas.1613546114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hoque KM, Woodward OM, van Rossum DB, Zachos NC, Chen L, Leung GP, Guggino WB, Guggino SE, Tse CM. Epac1 mediates protein kinase A-independent mechanism of forskolin-activated intestinal chloride secretion. J Gen Physiol 135: 43–58, 2010. doi: 10.1085/jgp.200910339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Hianik T, Laputková G, Bajci A. Prostaglandin E2 and F2 alpha induced calcium transport across lipid bilayers. Gen Physiol Biophys 7: 317–323, 1988. [PubMed] [Google Scholar]
  • 48. Asbóth G, Phaneuf S, Europe-Finner GN, Tóth M, Bernal AL. Prostaglandin E2 activates phospholipase C and elevates intracellular calcium in cultured myometrial cells: involvement of EP1 and EP3 receptor subtypes. Endocrinology 137: 2572–2579, 1996. doi: 10.1210/endo.137.6.8641211. [DOI] [PubMed] [Google Scholar]
  • 49. Hegyi P, Maléth J, Walters JR, Hofmann AF, Keely SJ. Guts and gall: bile acids in regulation of intestinal epithelial function in health and disease. Physiol Rev 98: 1983–2023, 2018. doi: 10.1152/physrev.00054.2017. [DOI] [PubMed] [Google Scholar]
  • 50. Peregrin AT, Ahlman H, Jodal M, Lundgren O. Involvement of serotonin and calcium channels in the intestinal fluid secretion evoked by bile salt and cholera toxin. Br J Pharmacol 127: 887–894, 1999. doi: 10.1038/sj.bjp.0702615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Natarajan K, Sahoo NC, Rao KV. Signal thresholds and modular synergy during expression of costimulatory molecules in B lymphocytes. J Immunol 167: 114–122, 2001. doi: 10.4049/jimmunol.167.1.114. [DOI] [PubMed] [Google Scholar]
  • 52. Schwiebert EM, Egan ME, Hwang TH, Fulmer SB, Allen SS, Cutting GR, Guggino WB. CFTR regulates outwardly rectifying chloride channels through an autocrine mechanism involving ATP. Cell 81: 1063–1073, 1995. doi: 10.1016/s0092-8674(05)80011-x. [DOI] [PubMed] [Google Scholar]
  • 53. Devidas S, Guggino WB. The cystic fibrosis transmembrane conductance regulator and ATP. Curr Opin Cell Biol 9: 547–552, 1997. doi: 10.1016/s0955-0674(97)80032-4. [DOI] [PubMed] [Google Scholar]
  • 54. Inami A, Kiyono H, Kurashima Y. ATP as a pathophysiologic mediator of bacteria-host crosstalk in the gastrointestinal tract. Int J Mol 19: 2371, 2018. doi: 10.3390/ijms19082371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Simpson JE, Schweinfest CW, Shull GE, Gawenis LR, Walker NM, Boyle KT, Soleimani M, Clarke LL. PAT-1 (Slc26a6) is the predominant apical membrane Cl-/HCO3- exchanger in the upper villous epithelium of the murine duodenum. Am J Physiol Gastrointest Liver Physiol 292: G1079–G1088, 2007. doi: 10.1152/ajpgi.00354.2006. [DOI] [PubMed] [Google Scholar]
  • 56. Haggie PM, Cil O, Lee S, Tan JA, Rivera AA, Phuan PW, Verkman AS. SLC26A3 inhibitor identified in small molecule screen blocks colonic fluid absorption and reduces constipation. JCI Insight 3: e121370, 2018. doi: 10.1172/jci.insight.121370. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental figure legends: https://doi.org/10.6084/m9.figshare.22306264.

Supplemental Figs. S1 and S2: https://doi.org/10.6084/m9.figshare.22306429.

Data Availability Statement

Data will be made available upon reasonable request.


Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

RESOURCES