Abstract
Zinc is a commonly used antidiarrheal supplement; however, its exact mechanism of action is not well understood. Calcium-sensing receptor (CaSR) is a regulator of intestinal ion transport and a therapeutic target for secretory diarrhea. CaSR is activated by various cations and here we investigated the roles of CaSR in the antidiarrheal effects of the divalent metal zinc (Zn2+). In human intestinal T84 cells expressing CaSR, zinc (100 μM) inhibited forskolin-induced secretory Isc by 60% and its effect was comparable to CaSR activator cinacalcet. Zinc effect was via inhibition of apical CFTR Cl− channel and basolateral K+ channels. In cell models, zinc was a CaSR agonist and its antisecretory effects were CaSR-dependent. Similarly, 100 μM zinc inhibited forskolin-induced secretory Isc by 40% in wildtype mouse intestine with no antisecretory effects in intestinal epithelia-specific CaSR knockout mice (Casrflox/flox;Vil1-cre). Zinc inhibited Isc induced by clinically-relevant cAMP agonists (cholera toxin and vasoactive intestinal peptide) by 65% in T84 cells. Interestingly, zinc had no effect on cGMP agonists (heat-stable E. coli enterotoxin and linaclotide)-induced secretory Isc, suggesting its antisecretory effects are specific to cAMP. The mechanisms of zinc effect in T84 cells involved intracellular Ca2+ release via ryanodine receptors and inhibition of cAMP synthesis. Our findings suggest that CaSR activation is a major mechanism for the antidiarrheal effects of zinc which specifically reduces cAMP levels. In addition to its use in cholera, zinc can be effective in other cAMP-mediated secretory diarrheas.
Keywords: CFTR, ion transport, cholera, secretory diarrhea
NEW & NOTEWORTHY
Zinc is a commonly used antidiarrheal supplement; however, its exact mechanisms of antisecretory effects remain unknown. In this study, we demonstrated that zinc is an agonist of the extracellular calcium-sensing receptor (CaSR) and its antidiarrheal effects are via reduced cAMP synthesis in intestinal epithelial cells. In addition to elucidating the mechanism of action of this antidiarrheal supplement, our data support the use of zinc as a simple and effective treatment option for all cAMP-mediated diarrheas.
Graphical Abstract:

INTRODUCTION
Diarrhea is a major cause of global morbidity and mortality, particularly in young children (1). Secretory diarrhea is a common type with diverse etiologies including infections, inflammation, drugs, tumors, and genetic disorders (2). Cyclic nucleotide (cAMP and cGMP) elevation in intestinal epithelial cells is the key driver of intestinal fluid loss in certain secretory diarrheas. cAMP elevation is the major pathology in cholera and vasoactive intestinal peptide-secreting tumors (VIPoma), whereas cGMP elevation is the main driver of fluid loss in traveler’s diarrhea and GUCY2C mutations (2–4). In all these diseases, elevated cyclic nucleotides result in activation of luminal membrane Cl− channel CFTR (cystic fibrosis transmembrane conductance regulator) and basolateral membrane K+ channels which result in Cl− and fluid secretion (2). In addition, inhibition of apical membrane Na+/H+ exchanger 3 (NHE3) contributes to intestinal fluid losses by impairing fluid absorption (5). Current treatment for secretory diarrhea is primarily supportive, and fluid replacement with oral rehydration solution (ORS) is the mainstay of cholera treatment (6). In addition to ORS, World Health Organization recommends oral zinc treatment in children with diarrhea (7). Despite its extensive clinical use in diarrhea, the mechanism of antidiarrheal effects of zinc are not well understood. Zinc was shown to improve intestinal epithelial barrier and immune system function, which are thought to in part contribute in its antidiarrheal effects (8). Some studies also suggested that zinc might have direct antisecretory effects in the intestine (9), however the exact mechanisms of this effect are unclear. Although some studies attributed the effects of zinc on GPR39 (also known as “zinc-sensing receptor”) which might promote Cl− absorption in enterocytes (10), the mechanisms of effects of zinc on hypersecretion (the key pathology in secretory diarrheas) are largely unknown (11).
Extracellular Ca2+-sensing receptor (CaSR) is a G protein-coupled receptor (GPCR) expressed in many tissues including parathyroid, kidney, brain, bone and gut (12). CaSR activation by approved drug cinacalcet inhibits cAMP and cGMP agonists-induced Cl− secretion in human intestinal cells and mouse intestine by promoting cyclic nucleotide hydrolysis via phosphodiesterases (PDE) (13, 14). In addition to small molecules, CaSR is physiologically activated by di- and trivalent cations (15, 16). Since zinc (Zn2+) is a divalent metal, we postulated that CaSR activation might be responsible from its antisecretory effects in intestinal epithelial cells.
MATERIALS AND METHODS
Chemicals
All chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA), except CFTRinh-172 (MedChemExpress).
Cell culture
T84 (CCL-248) and HEK-293 (CRL-1573) were obtained from American Type Culture Collection (Gaithersburg, MD). HEK-293 cells overexpressing CaSR (HEK-CaSR) were purchased from AddexBio (San Diego, CA). Fischer rat thyroid cells stably expressing human wild-type CFTR (FRT-CFTR) were obtained from UCSF Cystic Fibrosis Drug Discovery Core Center. T84 and FRT-CFTR cells were cultured as described (13). HEK-293 and HEK-CaSR cells were cultured in DMEM medium containing a high concentration of glucose, 10%fetal bovine serum, 100 U/ml penicillin, and 100 ug/ml streptomycin. HEK CaSR cells were continuously selected by adding G418 sulfate.
Short-circuit current (Isc) measurements
Cells were grown on snapwell inserts (12 mm diameter, 0.4 μm polyester membrane; Corning Life Sciences, Tewksbury, MA) at 37°C in 5% CO2 /95% air and used for short-circuit current experiments 5–7 days after plating. Inserts were mounted in Ussing chambers with each hemichamber containing bicarbonate-buffered Ringer’s solution (pH 7.4, in mM: 120 NaCl, 5 KCl, 1 MgCl2, 1 CaCl2, 10 D-glucose, 5 HEPES, and 25 NaHCO3). Unless otherwise specified, all compounds were added to both apical and basolateral bathing solutions. The solutions were aerated with 95% O2 /5% CO2 and maintained at 37°C during experiments. Isc was measured using an EVC4000 multichannel voltage clamp (World Precision Instruments, Sarasota, FL, USA) via Ag/AgCl electrodes and 3 M KCl agar bridges, as described (14, 17). In parallel experiments, T84 cells were grown on permeable filters as described above, and bathed with Ringer’s solution (±100 μM ZnCl2) for 60 min. To test the effects of zinc on epithelial barrier function, transepithelial electrical resistance (TEER) was measured using a Millicell-ERS Resistance System with dual electrode volt-ohmmeter (Millipore, Bedford, MA). Net TEER (ohms × cm2) was calculated by subtracting the resistance of cell-free media from measured resistance as described (14).
To measure apical Cl− conductance, the basolateral membrane was permeabilized with 500 μg/ml amphotericin B for 30 min and 60 mM basolateral-to-apical Cl− gradient was applied (13, 16). For these experiments, Ringer’s was the basolateral bathing solution (120 mM NaCl) and the apical solution contained 60 mM NaCl and 60 mM sodium gluconate. To measure basolateral membrane K+ conductance, the apical membrane was permeabilized with 20 μM amphotericin B for 30 min and apical-to-basolateral potassium gradient was applied (13, 14). The apical solution (pH 7.4) contained in mM: 142.5 K-gluconate, 1 CaCl2, 1 MgCl2, 0.43 KH2PO4, 0.35 Na2HPO4, 10 HEPES, and 10 D-glucose. In the basolateral solution (pH 7.4) 142.5 mM K-gluconate was replaced by 5.5 mM K-gluconate and 137 mM N-methylglucamine.
Animals
Wild-type, Casrflox/flox (030647) and vil1-cre (004586) mice (all in C57BL/6 background) were obtained from Jackson Laboratories (Bar Harbor, ME). Intestinal epithelia-specific Casr knockout mice (Casrflox/flox;Vil1-cre) were generated by crossbreeding and genotype was confirmed by PCR. Animals were bred in UCSF Laboratory Animal Resource Center and experiments were done in adherence with NIH Guide for the Care and Use of Laboratory Animals. All animal experiments were approved by IACUC. Both male and female mice (10–16 weeks old) were used in all experiments.
Intestinal Isc measurements
Jejunum of mice was excised under isoflurane anesthesia and soaked in isoosmolar solution (300 mM mannitol in water) with 10 μM indomethacin to inhibit prostaglandin production. Seromuscular layer was stripped under a dissection microscope. As done before (14, 16), mucosal segments were mounted on Ussing chambers containing Ringer’s solution (120 mM NaCl and 10 mM glucose) on the basolateral side. In apical chamber NaCl was replaced with 60 mM NaCl and 60 mM sodium gluconate, and glucose was replaced with 10 mM mannitol. Isc was measured as described above.
CaSR activity measurements
For intracellular Ca2+ measurements, T84 cells were plated in 96-well, black-walled microplates (Corning), HEK-293 and HEK-CaSR cells were plated in Poly-D-Lysine-treated plates (Greiner Bio-One). Confluent cells were loaded with calcium indicator Fluo-4 NW (Invitrogen) per manufacturer’s instructions and Fluo-4 fluorescence was measured in each well continuously with a plate reader (Tecan Infinite M1000) at excitation/emission wavelengths of 495/516 nm after addition of 100 or 300 μM ZnCl2. The experiments were done using Hank’s buffered salt solution (HBSS) as the assay buffer. In some experiments, Ca2+ and Mg2+-free HBSS was used to avoid CaSR activation by these physiological agonists. In some experiments, cells were pretreated with the PLC inhibitor U73122 (10 μM), sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor thapsigargin (1 μM) or ryanodine receptor (RyR) inhibitor dantrolene (10 μM) for 10 minutes prior to addition of ZnCl2. For quantification of IP1 (stable downstream metabolite of IP3), T84 cells were grown in 384-well opaque plates (PerkinElmer) and treated with 0–100 μM ZnCl2 for 30 minutes. Next, cells were lysed and the IP1 concentration in each well was quantified using the IP-One Gq kit (Cisbio) according to the manufacturer’s instructions.
cAMP and cGMP measurements
T84 cells were grown in clear 24-well plates (Corning) and pretreated for 20 minutes with 100 μM ZnCl2 or vehicle control. For cAMP measurement, cells were treated with 10 μM forskolin (for 5 minutes) and lysed by repeated freeze/thaw and centrifuged to remove cell debris. In some experiments, cells were treated with PDE inhibitor IBMX (500 μM) together with forskolin. The supernatant was assayed for cAMP using the cAMP Parameter immunoassay kit according to the manufacturer’s instructions (R&D Systems, Bio-Techne). For cGMP measurement, cells were treated with 0.1 μg/ml STa toxin (for 5 minutes) and lysed by repeated freeze/thaw and centrifuged to remove cell debris. The supernatant was assayed for cGMP using the cGMP immunoassay kit according to the manufacturer’s instructions (Cell Signaling Technology, Danvers, MA).
Statistical Analysis
Experiments with two groups were analyzed with two-tailed Student’s t-test. For experiments with three or more groups, one-way analysis of variance and post hoc Newman-Keuls multiple comparisons test was used. P value < 0.05 was considered statistically significant.
Study approval
All animal experiments were approved by IACUC.
RESULTS
Zinc inhibits CFTR-mediated Cl− secretion in T84 cells
Human intestinal T84 cells are a commonly used model to study intestinal ion transport which natively express CaSR and respond to CaSR agonists (13, 16, 18). In T84 cells, zinc concentration-dependently inhibited cAMP agonist forskolin-induced secretory short-circuit current (Isc) by up to 60%, which was comparable to the effect of small molecule CaSR activator cinacalcet (Fig. 1A and B). Since CFTR is the major cAMP-gated ion Cl− channel in intestinal epithelia, we tested the roles of CFTR in zinc effect using its selective inhibitor CFTRinh-172. Forskolin-induced secretory Isc in T84 cells was partially reversed by CFTRinh-172. Zinc treatment concentration-dependently reduced CFTR activity in T84 cells as suggested by reduced CFTRinh-172 responses, and its effect was comparable to cinacalcet (Fig. 1A and C). To rule out any potential effects on barrier permeability, TEER was measured with and without 100 μM zinc treatment. Under the same conditions Isc studies are performed, zinc had no effect on TEER (Fig. 1D). These results collectively suggest that zinc reduces CFTR-mediated Cl− secretion in human intestinal epithelial cells similar to CaSR agonists.
Figure 1. Zinc inhibits cAMP-induced Cl− secretion and CFTR activity in T84 cells.

A. Short-circuit current (Isc) traces showing forskolin (10 μM) and CFTRinh-172 (10 μM) responses in the presence of 20 min ZnCl2 pretreatment at various concentrations. Cinacalcet (30 μM) was used as a positive control for CaSR activation. B. Summary data for forskolin response for studies in A. C. Summary data for CFTRinh-172 response for studies in A. D. Transepithelial electrical resistance (TEER) measurements in T84 cells after 60 min bathing in Ringer solution with and without 100 μM ZnCl2. Mean ± S.E.M., n= 5–11 experiments per group. One-way analysis of variance and post hoc Newman-Keuls multiple comparisons test, *p<0.05, **p<0.01, ***p<0.001, ns: not significant.
Zinc inhibits apical CFTR Cl− channel and basolateral K+ channels in T84 cells
Although CFTR is the major Cl− channel in intestinal epithelia, forskolin-induced secretory Isc in T84 cells is due to combined actions of apical membrane Cl− and basolateral membrane K+ channels (19). To selectively study the effects of zinc on apical vs. basolateral ion transport, we used selective membrane permeabilization and ion gradients. In T84 cells with basolateral membrane permeabilization and basolateral-to-apical Cl− gradient, zinc inhibited forskolin-induced apical Cl− secretion and CFTR activity by 65% (Fig. 2A and B). To directly study basolateral membrane K+ conductance we used apical membrane permeabilization and apical-to-basolateral K+ gradient. In this setting, zinc inhibited basolateral K+ conductance as suggested by 65% lower responses to forskolin and cAMP-activated K+ channel inhibitor BaCl2 (Fig. 2C and D). These studies suggest that zinc inhibits both apical Cl− and basolateral K+ conductance in human intestinal epithelial cells, similar to CaSR agonists (13, 16).
Figure 2. Zinc inhibits apical membrane Cl− conductance and basolateral membrane K+ conductance in T84 cells.

A. Isc traces in T84 cells showing responses to forskolin (10 μM) and CFTRinh-172 (10 μM, apical) with basolateral membrane permeabilization (250 μg/ml amphotericin B) and 60 mM basolateral-to-apical Cl− gradient. Experiments were done with and without 100 μM ZnCl2 pre-treatment for 20 min. B. Summary data for forskolin and CFTRinh-172 responses for studies in A. C. Isc traces showing responses to forskolin (10 μM) and BaCl2 (5 mM, basolateral) with apical membrane permeabilization (20 μM amphotericin B) and apical-to-basolateral K+ gradient. Experiments were done with and without 100 μM ZnCl2 pre-treatment for 20 min. D. Summary data for forskolin and BaCl2 responses for studies in C. Mean ± S.E.M., n=5–6 experiments per group, Student’s t-test, **p<0.01, ***p<0.001.
Antisecretory effect of zinc is dependent on CaSR in cells and mouse intestine
To study the roles of CaSR in zinc effect, we initially used FRT-CFTR cells that have robust CFTR currents, but do not express CaSR or respond to CaSR agonists (13, 16, 20). In this setting, zinc had no effect on forskolin or CFTRinh-172 responses suggesting that zinc does not have a direct CFTR inhibitory effect (Fig. 3A and B). GPR39 is proposed as the “zinc-sensing receptor” responsible from some of its biological effects (21). To test whether GPR39 activation has any roles in antisecretory effects of zinc, we used its selective agonist GPR39-C3 in T84 cells, which natively express GPR39 (22). Unlike zinc and CaSR agonists, GPR39-C3 pretreatment had no effects on forskolin or CFTRinh-172-induced Isc changes in T84 cells (Fig. 3C and D), which suggests that antisecretory effects of zinc is independent of GPR39. To test the effects of zinc on CaSR directly, we used HEK cells transfected with CaSR (HEK-CaSR). Zinc treatment resulted in intracellular Ca2+ elevation in HEK-CaSR (Fig. 4A), which is the known signaling pathway for CaSR activation in these cells (23, 24). Zinc treatment had no effect on intracellular Ca2+ in non-transfected HEK-293 cells which do not express CaSR (23) even at a high concentration of 300 μM (Fig. 4A). These experiments were done using regular assay buffer containing Ca2+ and Mg2+, which are known physiological CaSR agonists. To directly test the effects of zinc on CaSR activity in the absence of any other agonists, we performed similar experiments in Ca2+ and Mg2+-free assay buffer. In this setting, zinc treatment concentration-dependently elevated intracellular Ca2+ and activated CaSR with an EC50 of ~100 μM (Fig. 4B). To test whether zinc has any allosteric activating effects of on CaSR, we tested Ca2+-induced CaSR activation in HEK-CaSR cells in the presence and absence of zinc. Extracellular Ca2+ activated CaSR with similar potency in the presence and absence of extracellular zinc (Fig. 4C). These results collectively suggest that zinc is a direct CaSR agonist and its antisecretory effects are via CaSR activation in intestinal epithelial cells. To further determine the role of CaSR in zinc effect, we did Isc studies in mouse intestine. Pretreatment of jejunal mucosa with 100 μM zinc decreased forksolin-induced Isc change by 40% in wild-type mice (Fig. 5A). Zinc had no antisecretory effects in intestinal epithelia-specific CaSR knockout mice (Casrflox/flox;Vil1-cre, Fig. 5B) which further suggests that its antisecretory effects are via CaSR activation in the intestine.
Figure 3. Antisecretory effect of zinc is CaSR-dependent.

A. Isc traces showing forskolin (10 μM) and CFTRinh-172 (10 μM) responses in FRT-CFTR cells with basolateral membrane permeabilization (500 μg/ml amphotericin B) and 60 mM basolateral-to-apical Cl− gradient. Experiments were done with and without 100 μM ZnCl2 pre-treatment for 20 min. B. Summary data for forskolin and CFTRinh-172 responses for studies in A. C. Isc traces showing forskolin (10 μM) and CFTRinh-172 (10 μM) responses with and without GPR39-C3 pretreatment (10 μM for 20 min) in T84 cells. D. Summary data for forskolin and CFTRinh-172 responses for studies in C. Mean ± S.E.M., n=5–6 experiments per group, Student’s t-test, ns: not significant.
Figure 4. Zinc is a CaSR agonist.

A. (left) Representative intracellular Ca+2 traces measured by Fluo-4 fluorescence and in HEK-CaSR and HEK-293 cells with ZnCl2 (100–300 μM) treatment. Experiments were done in HBSS which contains 1.2 mM Ca2+ and 0.9 mM Mg2+. Summary data for Fluo-4 fluorescence change is presented on the right. B. Zinc concentration-response for intracellular Ca+2 elevation in HEK-CaSR cells. Experiments were done in Ca2+ and Mg2+-free HBSS. C. Extracellular Ca2+ concentration-response curves for CaSR activation in the absence and presence of 100 μΜ ZnCl2 in the assay buffer (Ca2+ and Mg2+-free HBSS). Mean ± S.E.M., n=4–8 experiments per group, Student’s t-test, **p<0.01, ***p<0.001.
Figure 5. Antisecretory effect of zinc in mouse intestine is CaSR-dependent.

A. Isc traces and summary data showing responses to 10 μM forskolin in jejunal mucosa of wildtype mice with and without 100 μM ZnCl2 pre-treatment for 20 min. B. Isc traces and summary data showing responses to 10 μM forskolin in jejunal mucosa of intestinal epithelia-specific CaSR knock out mice (Vil1-Cre; Casr-flox) with and without 100 μM ZnCl2 pre-treatment for 20 min. Mean ± S.E.M., n=7–9 experiments per group, Student’s t-test, *p<0.05, ns: not significant.
Zinc inhibits Cl− secretion induced by cAMP agonists, but not cGMP agonists in T84 cells
Next, we tested the effects of zinc on Cl− secretion induced by clinically relevant secretagogues. Similar to the experiments with forskolin, zinc pretreatment inhibited secretory Isc induced by other cAMP agonists cholera toxin (Fig. 6A and B) and VIP (Fig. 6C and D) by 65%. 100 μM zinc treatment similarly inhibited CFTRinh-172 responses in both settings (Fig. 6). In certain secretory diarrheas such as Traveler’s diarrhea, cytoplasmic cGMP elevation is the key driver of intestinal Cl− secretion. To test the antisecretory efficacy of zinc in cGMP-mediated diarrheas, we used heat-stable E. coli enterotoxin (STa) as the cGMP agonist. Interestingly, zinc treatment had no effects on Isc changes induced by STa-toxin or CFTRinh-172 in this setting (Fig. 7A and B). Similarly, zinc treatment had no effect on secretory Isc and CFTRinh-172 response when guanylate cyclase-C receptor agonist linaclotide was used as the cGMP agonist (Fig. 7C and D). These results suggest that the antisecretory effects of zinc are specific for cAMP agonists.
Figure 6. Zinc inhibits Cl− secretion induced by cAMP agonists.

A. Isc traces showing responses to 1 μg/ml cholera toxin and 10 μM CFTRinh-172 with and without 100 μM ZnCl2 pre-treatment for 20 min. B. Summary data for cholera toxin and CFTRinh-172 responses for studies in A. C. Isc traces showing responses to 10 nM vasoactive intestinal peptide (VIP) and 10 μM CFTRinh-172 with and without 100 μM ZnCl2 pre-treatment for 20 min. D. Summary data for VIP and CFTRinh-172 responses for studies in C. Mean ± S.E.M., n=5–6 experiments per group, Student’s t-test, *p<0.05, **p<0.01, ***p<0.001.
Figure 7. Zinc has no effect on Cl− secretion induced by cGMP agonists.

A. Isc traces showing responses to 0.1 μg/ml heat-stable E. coli enterotoxin (STa toxin) and 10 μM CFTRinh-172 with and without 100 μM ZnCl2 pre-treatment for 20 min. B. Summary data for STa toxin and CFTRinh-172 responses for studies in A. C. Isc traces showing responses to 1 μM linaclotide and 10 μM CFTRinh-172 with and without 100 μM ZnCl2 pre-treatment for 20 min. D. Summary data for linaclotide and CFTRinh-172 responses for studies in C. Mean ± S.E.M., n=6 experiments per group, Student’s t-test, ns: not significant.
Zinc inhibits cAMP synthesis and stimulates intracellular Ca2+ release via RyR
We previously showed that CaSR activation by other agonists (cinacalcet and Mg2+) stimulates hydrolysis of cyclic nucleotides via PDE in intestinal epithelial cells. Consistent with this mechanism, cinacalcet and Mg2+ were effective in both cAMP and cGMP-mediated diarrhea models (13, 16). To test the roles of this signaling mechanism in zinc effect, we measured cAMP levels. In T84 cells, forskolin treatment resulted in marked elevation in cellular cAMP, which was largely reduced by zinc pretreatment (Fig. 8A) consistent with its antisecretory effects in the presence of cAMP agonists. Interestingly, co-treatment with PDE inhibitor IBMX did not reverse the effect of zinc on cAMP, suggesting that zinc does not stimulate PDE activity and it likely inhibits cAMP synthesis. Consistent with lack of its antisecretory effects, zinc pretreatment had no effects on STa toxin-induced cGMP elevation in T84 cells (Fig. 8B). These results suggest that zinc activates a distinct signaling pathway to specifically reduce cAMP synthesis in intestinal epithelial cells.
Figure 8. Zinc inhibits cAMP synthesis and stimulates intracellular Ca2+ release by RyR.

A. cAMP concentration in T84 cell lysates with 10 μM forskolin (±500 μM IBMX, phosphodiesterase inhibitor) treatment in the presence of 20 min pretreatment with and without 100 μM ZnCl2. B. cGMP concentration in T84 cell lysates with 0.1 μg/ml heat-stable E. coli enterotoxin (STa toxin) treatment in the presence of 20 min pretreatment with and without 100 μM ZnCl2. C. Changes in intracellular Ca2+ measured by Fluo-4 NW fluorescence in T84 cells with 300 μM ZnCl2 or vehicle treatment. In some experiments, T84 cells were pretreated with PLC inhibitor U73122 (10 μM), sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor thapsigargin (1 μM) or ryanodine receptor inhibitor dantrolene (10 μM) for 10 min before Zn2+ addition. D. IP1 (stable downstream metabolite of IP3) accumulation after 30 min of treatment with 100 μM ZnCl2 or vehicle.
CaSR activation by other agonists (cinacalcet and Mg2+) result in Ca2+ release from intracellular stores via PLC and IP3 receptors in intestinal epithelial cells (13, 16). To study the roles of this signaling mechanism in zinc effect, we performed studies using intracellular Ca2+ indicator Fluo-4 NW. Similar to other CaSR agonists, zinc treatment resulted in intracellular Ca2+ elevation in T84 cells. The source of this Ca2+ response was intracellular stores as suggested by abolishment of zinc effect with sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor thapsigargin pretreatment (Fig. 8C). Interestingly, PLC inhibitor U73122 had minimal effect on zinc-induced intracellular Ca2+ release. Similarly, zinc treatment had no effects on IP1 (stable downstream metabolite of IP3) levels in T84 cells (Fig. 8D), suggesting that zinc effect is independent of PLC-IP3 pathway. Ryanodine receptor (RyR) is another major pathway for intracellular Ca2+ release (25) and pretreatment of T84 cells with RyR inhibitor dantrolene essentially abolished zinc-induced intracellular Ca2+ elevation (Fig. 8C). Since some adenylate cyclase isoforms are known to be inhibited by Ca2+ (26), this mechanism can potentially explain the effects of zinc on reducing cAMP synthesis. These results collectively suggest that activation of CaSR by zinc stimulates a unique signaling pathway in intestinal epithelial cells which inhibits cAMP agonists-induced Cl− secretion.
DISCUSSION
Here we showed that zinc has profound antisecretory effects in human intestinal epithelial cells and mouse intestine. Several lines of evidence suggest that antisecretory effects of zinc effect are via activation of CaSR. In addition to its current use in cholera, our findings suggest that zinc can be an effective targeted treatment for other forms of cAMP-mediated secretory diarrheas.
Although ORS is the mainstay for secretory diarrhea treatment, the World Health Organization also recommends oral zinc treatment for 10–14 days in children with diarrhea (7). Randomized controlled trials showed that zinc can reduce stool weight and diarrhea duration in children with acute diarrhea and cholera (27, 28). Based on these, a zinc-fortified ORS was developed which appears to have greater efficacy than traditional ORS in pediatric diarrhea patients which further supports utility of zinc in diarrhea (29). The efficacy of zinc in diarrhea have traditionally been attributed to its effects on epithelial barrier and immune system, since zinc deficiency can impair these processes (8). However, some studies suggested the possibility of direct antisecretory effects of zinc. Zinc was shown to inhibit basolateral cAMP-activated K+ channel activity in rat intestine (9) and human colonic crypts (30). However, the exact mechanisms of this observed phenomenon remained unclear. Here we showed that zinc inhibits all cAMP-gated ion channels in intestinal epithelia including apical CFTR Cl− channel and basolateral K+ channels. Importantly, we found that reduced cAMP levels via CaSR activation is the underlying mechanisms of the antisecretory effects of zinc.
Here we found that zinc has profound antisecretory effects at 100 μM. To the best of our knowledge, there are no studies reporting stool or intestinal fluid zinc concentrations in diarrhea patients on zinc treatment, thus it is difficult to directly ascertain translational relevance of our findings. An earlier clinical study in pediatric cholera patients showed an average of 1.6–1.8 kg per day watery stool output (27). Assuming the WHO recommended daily zinc dose of 20 mg (~300 μmoles) (31) and 50% intestinal absorption (32), the zinc concentration in stool of cholera patients is predicted to be 85–95 μM. Thus, it is plausible that the zinc doses currently used in clinic may yield sufficient zinc levels in intestinal fluid for CaSR activation and antisecretory effects. However, we acknowledge that these calculations are indirect, and clinical studies quantifying stool zinc concentration are needed to test validity of these estimates. A previous study showed that during acute diarrhea in infants, fecal zinc loss is increased by 2.5 folds and plasma zinc concentration is reduced by 50% compared to healthy controls (33). It is plausible that this acute zinc loss might be exacerbating diarrhea via reduced CaSR activity in the intestinal epithelia. However, future clinical studies quantifying zinc concentrations in intestinal fluid and plasma of diarrhea patients on zinc treatment are needed to better understand clinical relevance of our findings.
Our findings suggest that antisecretory effect of zinc is dependent on CaSR in human intestinal epithelial cells and mouse intestine. Some earlier studies proposed GPR39 as the “zinc-sensing receptor” responsible from its biological actions (21, 34). Here we showed that, unlike zinc, selective GPR39 agonist has no antisecretory effects in human intestinal T84 cells. Previous studies in transfected cell models also showed that GPR39 activation results in elevated cAMP (35), contrary to zinc effect found here which is reduced cAMP. These collectively suggest that the antisecretory effects of zinc is independent of GPR39 in the intestinal epithelial cells. Although zinc was listed as a potential CaSR ligand (36), there has been limited research in this subject and an earlier study claimed that zinc does not activate CaSR (21). However, several lines of evidence presented herein including experiments in transfected cell models and knockout mice suggest that zinc is a CaSR agonist with antidiarrheal effects similar to other CaSR activators.
Another interesting finding of this study is that zinc inhibits Cl− secretion induced by cAMP agonists with no effect on cGMP agonists-induced Cl− secretion. These findings are in agreement with a previous study which reported that zinc inhibits cholera toxin-induced secretion but not STa toxin-induced secretion in human intestinal Caco-2 cells (37). We recently showed that CaSR agonists cinacalcet and Mg2+ inhibit Cl− secretion induced by both cAMP and cGMP agonists (13, 14, 16). Cinacalcet and Mg2+ increases intracellular Ca2+ via Gq-PLC-IP3 pathway, which leads to activation of PDE and hydrolysis of cyclic nucleotides, which explains their efficacy in both cAMP and cGMP-mediated diarrhea models. Here we found that zinc also elevates intracellular Ca2+ via CaSR activation, however its effects are through RyR and not PLC. Consistent with its selective efficacy in cAMP agonist-induced secretion, zinc reduced cAMP levels by inhibiting its synthesis without any effects on cGMP. Since some adenylate cyclase isoforms are known to be inhibited by calcium (26), this mechanism can potentially explain the effects of zinc on reducing cAMP synthesis. These results collectively suggest a potential distinct signaling pathway when CaSR is activated by zinc. Consistent with this, CaSR activation in certain settings and cell types is known to reduce cAMP synthesis (38). Future studies elucidating the mechanisms of CaSR activation by different agonists might be informative for better understanding of intestinal ion transport and its modulation by CaSR. Some earlier studies using membrane preparations and recombinant enzymes suggested that zinc can inhibit adenylate cyclase at micromolar concentrations (39, 40). However, it is unlikely that the zinc effect observed in our study is due to direct inhibition of adenylate cyclase, since free zinc levels in cytoplasm are at tightly kept at pico-nanomolar range and higher levels of cytoplasmic zinc are toxic (41).
In conclusion, our findings suggest that CaSR activation in intestinal epithelial cells is a key mechanism for antidiarrheal effects of zinc. In addition to its use in cholera, our findings suggest that zinc can be used in other forms of cAMP-mediated secretory diarrheas including VIPoma and bile acid diarrhea.
ACKNOWLEDGMENTS
The graphical abstract was prepared using Biorender.com.
GRANTS
This study was supported by grants from NIH (DK126070, EY036139) and Cystic Fibrosis Foundation (CIL24G0) to OC.
Footnotes
DISCLOSURES
The authors have declared that no conflict of interest exists.
DATA AVAILABILITY
Source data for this study will be available at [doi: 10.17632/8kjkd6g2jj.1] following an embargo from the date of publication (no more than 3 months) to allow for commercialization of research findings.
REFERENCES
- 1.Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect Dis. 2018;18(11):1211–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Thiagarajah JR, Donowitz M, and Verkman AS. Secretory diarrhoea: mechanisms and emerging therapies. Nat Rev Gastroenterol Hepatol. 2015;12(8):446–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ao M, Sarathy J, Domingue J, Alrefai WA, and 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. 2013;305(4):C447–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Müller T, Rasool I, Heinz-Erian P, Mildenberger E, Hülstrunk C, Müller A, et al. Congenital secretory diarrhoea caused by activating germline mutations in GUCY2C. Gut. 2016;65(8):1306–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Yun CH, Oh S, Zizak M, Steplock D, Tsao S, Tse CM, et al. cAMP-mediated inhibition of the epithelial brush border Na+/H+ exchanger, NHE3, requires an associated regulatory protein. Proc Natl Acad Sci U S A. 1997;94(7):3010–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pietroni MAC. Case management of cholera. Vaccine. 2020;38 Suppl 1:A105–a9. [DOI] [PubMed] [Google Scholar]
- 7.https://www.who.int/selection_medicines/committees/expert/22/s17.5_ORS-zinc.pdf.
- 8.Shankar AH, and Prasad AS. Zinc and immune function: the biological basis of altered resistance to infection. The American Journal of Clinical Nutrition. 1998;68(2):447S–63S. [DOI] [PubMed] [Google Scholar]
- 9.Hoque KM, Rajendran VM, and Binder HJ. Zinc inhibits cAMP-stimulated Cl secretion via basolateral K-channel blockade in rat ileum. Am J Physiol Gastrointest Liver Physiol. 2005;288(5):G956–63. [DOI] [PubMed] [Google Scholar]
- 10.Sunuwar L, Asraf H, Donowitz M, Sekler I, and Hershfinkel M. The Zn2+-sensing receptor, ZnR/GPR39, upregulates colonocytic Cl− absorption, via basolateral KCC1, and reduces fluid loss. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2017;1863(4):947–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Das S, Jayaratne R, and Barrett KE. The Role of Ion Transporters in the Pathophysiology of Infectious Diarrhea. Cell Mol Gastroenterol Hepatol. 2018;6(1):33–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chavez-Abiega S, Mos I, Centeno PP, Elajnaf T, Schlattl W, Ward DT, et al. Sensing Extracellular Calcium - An Insight into the Structure and Function of the Calcium-Sensing Receptor (CaSR). Adv Exp Med Biol. 2020;1131:1031–63. [DOI] [PubMed] [Google Scholar]
- 13.Oak AA, Chhetri PD, Rivera AA, Verkman AS, and Cil O. Repurposing calcium-sensing receptor agonist cinacalcet for treatment of CFTR-mediated secretory diarrheas. JCI Insight. 2021;6(4). [Google Scholar]
- 14.Chu T, Yottasan P, Goncalves LS, Oak AA, Lin R, Tse M, et al. Calcium-sensing receptor activator cinacalcet for treatment of cyclic nucleotide-mediated secretory diarrheas. Transl Res. 2024;263:45–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jensen AA, and Bräuner-Osborne H. Allosteric modulation of the calcium-sensing receptor. Curr Neuropharmacol. 2007;5(3):180–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.de Souza Goncalves L, Chu T, Master R, Chhetri PD, Gao Q, and Cil O. Mg2+ supplementation treats secretory diarrhea in mice by activating calcium-sensing receptor in intestinal epithelial cells. J Clin Invest. 2024;134(2). [Google Scholar]
- 17.Yottasan P, Chu T, Chhetri PD, and Cil O. Repurposing calcium-sensing receptor activator drug cinacalcet for ADPKD treatment. Transl Res. 2024;265:17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gama L, Baxendale-Cox LM, and Breitwieser GE. Ca2+-sensing receptors in intestinal epithelium. Am J Physiol. 1997;273(4):C1168–75. [DOI] [PubMed] [Google Scholar]
- 19.Oak AA, Chu T, Yottasan P, Chhetri PD, Zhu J, Du Bois J, et al. Lubiprostone is non-selective activator of cAMP-gated ion channels and Clc-2 has a minor role in its prosecretory effect in intestinal epithelial cells. Mol Pharmacol. 2022;102(2):106–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Garrett JE, Tamir H, Kifor O, Simin RT, Rogers KV, Mithal A, et al. Calcitonin-secreting cells of the thyroid express an extracellular calcium receptor gene. Endocrinology. 1995;136(11):5202–11. [DOI] [PubMed] [Google Scholar]
- 21.Hershfinkel M, Moran A, Grossman N, and Sekler I. A zinc-sensing receptor triggers the release of intracellular Ca2+ and regulates ion transport. Proc Natl Acad Sci U S A. 2001;98(20):11749–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pongkorpsakol P, Buasakdi C, Chantivas T, Chatsudthipong V, and Muanprasat C. An agonist of a zinc-sensing receptor GPR39 enhances tight junction assembly in intestinal epithelial cells via an AMPK-dependent mechanism. Eur J Pharmacol. 2019;842:306–13. [DOI] [PubMed] [Google Scholar]
- 23.Nemeth EF, Steffey ME, Hammerland LG, Hung BC, Van Wagenen BC, DelMar EG, et al. Calcimimetics with potent and selective activity on the parathyroid calcium receptor. Proc Natl Acad Sci U S A. 1998;95(7):4040–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Song S, Babicheva A, Zhao T, Ayon RJ, Rodriguez M, Rahimi S, et al. Notch enhances Ca(2+) entry by activating calcium-sensing receptors and inhibiting voltage-gated K(+) channels. Am J Physiol Cell Physiol. 2020;318(5):C954–c68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Woll KA, and Van Petegem F. Calcium-release channels: structure and function of IP(3) receptors and ryanodine receptors. Physiol Rev. 2022;102(1):209–68. [DOI] [PubMed] [Google Scholar]
- 26.Halls ML, and Cooper DM. Regulation by Ca2+-signaling pathways of adenylyl cyclases. Cold Spring Harb Perspect Biol. 2011;3(1):a004143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Roy SK, Hossain MJ, Khatun W, Chakraborty B, Chowdhury S, Begum A, et al. Zinc supplementation in children with cholera in Bangladesh: randomised controlled trial. Bmj. 2008;336(7638):266–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Dhingra U, Kisenge R, Sudfeld CR, Dhingra P, Somji S, Dutta A, et al. Lower-Dose Zinc for Childhood Diarrhea - A Randomized, Multicenter Trial. N Engl J Med. 2020;383(13):1231–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bahl R, Bhandari N, Saksena M, Strand T, Kumar GT, Bhan MK, et al. Efficacy of zinc-fortified oral rehydration solution in 6- to 35-month-old children with acute diarrhea. J Pediatr. 2002;141(5):677–82. [DOI] [PubMed] [Google Scholar]
- 30.Medani M, Bzik VA, Rogers A, Collins D, Kennelly R, Winter DC, et al. Zinc sulphate attenuates chloride secretion in human colonic mucosae in vitro. Eur J Pharmacol. 2012;696(1–3):166–71. [DOI] [PubMed] [Google Scholar]
- 31.Lazzerini M, and Wanzira H. Oral zinc for treating diarrhoea in children. Cochrane Database of Systematic Reviews. 2016(12). [Google Scholar]
- 32.Maares M, and Haase H. A Guide to Human Zinc Absorption: General Overview and Recent Advances of In Vitro Intestinal Models. Nutrients. 2020;12(3). [Google Scholar]
- 33.Castillo-Duran C, Vial P, and Uauy R. Trace mineral balance during acute diarrhea in infants. J Pediatr. 1988;113(3):452–7. [DOI] [PubMed] [Google Scholar]
- 34.Levaot N, and Hershfinkel M. How cellular Zn(2+) signaling drives physiological functions. Cell Calcium. 2018;75:53–63. [DOI] [PubMed] [Google Scholar]
- 35.Holst B, Egerod KL, Schild E, Vickers SP, Cheetham S, Gerlach L-O, et al. GPR39 Signaling Is Stimulated by Zinc Ions But Not by Obestatin. Endocrinology. 2007;148(1):13–20. [DOI] [PubMed] [Google Scholar]
- 36.Díaz-Soto G, Rocher A, García-Rodríguez C, Núñez L, and Villalobos C. The Calcium-Sensing Receptor in Health and Disease. Int Rev Cell Mol Biol. 2016;327:321–69. [DOI] [PubMed] [Google Scholar]
- 37.Canani RB, Cirillo P, Buccigrossi V, Ruotolo S, Passariello A, De Luca P, et al. Zinc inhibits cholera toxin-induced, but not Escherichia coli heat-stable enterotoxin-induced, ion secretion in human enterocytes. J Infect Dis. 2005;191(7):1072–7. [DOI] [PubMed] [Google Scholar]
- 38.Di Mise A, Tamma G, Ranieri M, Centrone M, van den Heuvel L, Mekahli D, et al. Activation of Calcium-Sensing Receptor increases intracellular calcium and decreases cAMP and mTOR in PKD1 deficient cells. Scientific Reports. 2018;8(1):5704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Klein C, Heyduk T, and Sunahara RK. Zinc inhibition of adenylyl cyclase correlates with conformational changes in the enzyme. Cell Signal. 2004;16(10):1177–85. [DOI] [PubMed] [Google Scholar]
- 40.Klein C, Sunahara RK, Hudson TY, Heyduk T, and Howlett AC. Zinc inhibition of cAMP signaling. J Biol Chem. 2002;277(14):11859–65. [DOI] [PubMed] [Google Scholar]
- 41.Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, et al. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther. 2024;9(1):6. [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.
Data Availability Statement
Source data for this study will be available at [doi: 10.17632/8kjkd6g2jj.1] following an embargo from the date of publication (no more than 3 months) to allow for commercialization of research findings.
