Cholinergic ion secretion in human colon requires coactivation by cAMP

1998 ◽  
Vol 275 (6) ◽  
pp. G1274-G1281 ◽  
Author(s):  
M. Mall ◽  
M. Bleich ◽  
M. Schürlein ◽  
J. Kühr ◽  
H. H. Seydewitz ◽  
...  

Cl−secretion in the colon can be activated by an increase of either intracellular Ca2+ or cAMP. In this study we examined a possible interdependence of the two second-messenger pathways in human colonic epithelium. When measured in a modified Ussing chamber, carbachol (CCH; 100 μmol/l, basolateral), via an increase in cytosolic Ca2+concentration ([Ca2+]i), activated a transient lumen-negative equivalent short-circuit current ( I sc) [change (Δ) in I sc = −79.4 ± 7.5 μA/cm2]. Previous studies indicated that intracellular Ca2+ directly acts on basolateral K+ channels, thus enhancing driving force for luminal Cl− exit. Increased intracellular cAMP (by basolateral addition of 100 μmol/l IBMX and 1 μmol/l forskolin) activated a sustained lumen-negative current (Δ I sc = −42.4 ± 7.2 μA/cm2) that was inhibited by basolateral trans-6-cyano-4-( N-ethylsulfonyl- N-methylamino)-3-hydroxy-2,2-dimethyl&2-chromane (10 μmol/l), a blocker of KvLQT1 channels. In the presence of elevated cAMP, the CCH-activated currents were augmented (Δ I sc = 167.7 ± 32.7 μA/cm2), suggesting cooperativity of the Ca2+- and cAMP-mediated responses. Inhibition of endogenous cAMP production by indomethacin (10 μmol/l) significantly reduced CCH-activated currents and even reversed the polarity in 70% of the experiments. The transient lumen-positive I sc was probably due to activation of apical K+channels because it was blocked by luminal Ba2+ (5 mmol/l) and tetraethylammonium (10 mmol/l). In the presence of indomethacin (10 μmol/l, basolateral), an increase of cAMP activated a sustained negative I sc. Under these conditions, CCH induced a large further increase in lumen-negative I sc(Δ I sc = −100.0 ± 21.0 μA/cm2). We conclude that CCH acting via [Ca2+]ican induce Cl− secretion only in the presence of cAMP, i.e., when luminal Cl− channels are already activated. The activation of a luminal and basolateral K+ conductance by CCH may be essential for transepithelial KCl secretion in human colon.

2000 ◽  
Vol 279 (1) ◽  
pp. G132-G138 ◽  
Author(s):  
Lane L. Clarke ◽  
Matthew C. Harline ◽  
Lara R. Gawenis ◽  
Nancy M. Walker ◽  
John T. Turner ◽  
...  

The loss of cystic fibrosis transmembrane conductance regulator (CFTR)-mediated transepithelial HCO3 − secretion contributes to the pathogenesis of pancreatic and biliary disease in cystic fibrosis (CF) patients. Recent studies have investigated P2Y2 nucleotide receptor agonists, e.g., UTP, as a means to bypass the CFTR defect by stimulating Ca2+-activated Cl− secretion. However, the value of this treatment in facilitating transepithelial HCO3 − secretion is unknown. Gallbladder mucosae from CFTR knockout mice were used to isolate the Ca2+-dependent anion conductance during activation of luminal P2Y2receptors. In Ussing chamber studies, UTP stimulated a transient peak in short-circuit current ( I sc) that declined to a stable plateau phase lasting 30–60 min. The plateau I sc after UTP was Cl− independent, HCO3 − dependent, insensitive to bumetanide, and blocked by luminal DIDS. In pH stat studies, luminal UTP increased both I sc and serosal-to-mucosal HCO3 − flux ( J s→m) during a 30-min period. Substitution of Cl− with gluconate in the luminal bath to inhibit Cl−/HCO3 −exchange did not prevent the increase in J s→mand I sc during UTP. In contrast, luminal DIDS completely inhibited UTP-stimulated increases in J s→m and I sc. We conclude that P2Y2 receptor activation results in a sustained (30–60 min) increase in electrogenic HCO3 − secretion that is mediated via an intracellular Ca2+-dependent anion conductance in CF gallbladder.


2008 ◽  
Vol 294 (6) ◽  
pp. C1443-C1453 ◽  
Author(s):  
Dong Wang ◽  
Ying Sun ◽  
Wei Zhang ◽  
Pingbo Huang

In airway epithelial cells, apical adenosine regulates transepithelial anion secretion by activation of apical cystic fibrosis transmembrane conductance regulator (CFTR) via adenosine receptors and cAMP/PKA signaling. However, the potent stimulation of anion secretion by adenosine is not correlated with its modest intracellular cAMP elevation, and these uncorrelated efficacies have led to the speculation that additional signaling pathways may be involved. Here, we showed that mucosal adenosine-induced anion secretion, measured by short-circuit current ( Isc), was inhibited by the PLC-specific inhibitor U-73122 in the human airway submucosal cell line Calu-3. In addition, the Isc was suppressed by BAPTA-AM (a Ca2+ chelator) and 2-aminoethoxydiphenyl borate (2-APB; an inositol 1,4,5-trisphosphate receptor blocker), but not by PKC inhibitors, suggesting the involvement of PKC-independent PLC/Ca2+ signaling. Ussing chamber and patch-clamp studies indicated that the adenosine-induced PLC/Ca2+ signaling stimulated basolateral Ca2+-activated potassium (KCa) channels predominantly via A2B adenosine receptors and contributed substantially to the anion secretion. Thus, our data suggest that apical adenosine activates contralateral K+ channels via PLC/Ca2+ and thereby increases the driving force for transepithelial anion secretion, synergizing with its modulation of ipsilateral CFTR via cAMP/PKA. Furthermore, the dual activation of CFTR and KCa channels by apical adenosine resulted in a mixed secretion of chloride and bicarbonate, which may alter the anion composition in the secretion induced by secretagogues that elicit extracellular ATP/adenosine release. Our findings provide novel mechanistic insights into the regulation of anion section by adenosine, a key player in the airway surface liquid homeostasis and mucociliary clearance.


2012 ◽  
Vol 303 (10) ◽  
pp. F1425-F1434 ◽  
Author(s):  
Yu Liu ◽  
Madhumitha Rajagopal ◽  
Kim Lee ◽  
Lorenzo Battini ◽  
Daniel Flores ◽  
...  

Prostaglandin E2 (PGE2) contributes to cystogenesis in genetically nonorthologous models of autosomal dominant polycystic kidney disease (ADPKD). However, it remains unknown whether PGE2 induces the classic features of cystic epithelia in genetically orthologous models of ADPKD. We hypothesized that, in ADPKD epithelia, PGE2 induces proliferation and chloride (Cl−) secretion, two archetypal phenotypic features of ADPKD. To test this hypothesis, proliferation and Cl− secretion were measured in renal epithelial cells deficient in polycystin-1 (PC-1). PC-1-deficient cells increased in cell number (proliferated) faster than PC-1-replete cells, and this proliferative advantage was abrogated by cyclooxygenase inhibition, indicating a role for PGE2 in cell proliferation. Exogenous administration of PGE2 increased proliferation of PC-1-deficient cells by 38.8 ± 5.2% ( P < 0.05) but inhibited the growth of PC-1-replete control cells by 49.4 ± 1.9% ( P < 0.05). Next, we tested whether PGE2-specific E prostanoid (EP) receptor agonists induce intracellular cAMP and downstream β-catenin activation. PGE2 and EP4 receptor agonism (TCS 2510) increased intracellular cAMP concentration and the abundance of active β-catenin in PC-1-deficient cells, suggesting a mechanism for PGE2-mediated proliferation. Consistent with this hypothesis, antagonizing EP4 receptors reverted the growth advantage of PC-1-deficient cells, implicating a central role for the EP4 receptor in proliferation. To test whether PGE2-dependent Cl− secretion is also enhanced in PC-1-deficient cells, we used an Ussing chamber to measure short-circuit current ( Isc). Addition of PGE2 induced a fivefold higher increase in Isc in PC-1-deficient cells compared with PC-1-replete cells. This PGE2-induced increase in Isc in PC-1-deficient cells was blocked by CFTR-172 and flufenamic acid, indicating that PGE2 activates CFTR and calcium-activated Cl− channels. In conclusion, PGE2 activates aberrant signaling pathways in PC-1-deficient epithelia that contribute to the proliferative and secretory phenotype characteristic of ADPKD and suggests a therapeutic role for PGE2 inhibition and EP4 receptor antagonism.


2007 ◽  
Vol 293 (1) ◽  
pp. G335-G346 ◽  
Author(s):  
Jacob G. Kirkland ◽  
Graeme S. Cottrell ◽  
Nigel W. Bunnett ◽  
Carlos U. Corvera

Cholecystitis is one of the most common gastrointestinal diseases. Inflammation induces the activation of proteases that can signal to cells by cleaving protease-activated receptors (PARs) to induce hemostasis, inflammation, pain, and repair. However, the distribution of PARs in the gallbladder is unknown, and their effects on gallbladder function have not been fully investigated. We localized immunoreactive PAR1 and PAR2 to the epithelium, muscle, and serosa of mouse gallbladder. mRNA transcripts corresponding to PAR1 and PAR2, but not PAR4, were detected by RT-PCR and sequencing. Addition of thrombin and a PAR1-selective activating peptide (TFLLRN-NH2) to the serosal surface of mouse gallbladder mounted in an Ussing chamber stimulated an increase in short-circuit current in wild-type but not PAR1 knockout mice. Similarly, serosally applied trypsin and PAR2 activating peptide (SLIGRL-NH2) increased short-circuit current in wild-type but not PAR2 knockout mice. Proteases and activating peptides strongly inhibited electrogenic responses to subsequent stimulation with the same agonist, indicating homologous desensitization. Removal of HCO3− ions from the serosal buffer reduced responses to thrombin and trypsin by >80%. Agonists of PAR1 and PAR2 increase intracellular Ca2+ concentration in isolated and cultured gallbladder epithelial cells. The COX-2 inhibitor meloxicam and an inhibitor of CFTR prevented the stimulatory effect of PAR1 but not PAR2. Thus PAR1 and PAR2 are expressed in the epithelium of the mouse gallbladder, and serosally applied proteases cause a HCO3− secretion. The effects of PAR1 but not PAR2 depend on generation of prostaglandins and activation of CFTR. These mechanisms may markedly influence fluid and electrolyte secretion of the inflamed gallbladder when multiple proteases are generated.


1986 ◽  
Vol 251 (2) ◽  
pp. G253-G262 ◽  
Author(s):  
D. A. Russell

Experiments were performed to determine the ionic basis and physiological messengers of transepithelial ion transport alterations (short-circuit current, Isc) measured during the induction of intestinal anaphylaxis in an Ussing chamber. Antigens derived from Trichinella spiralis, an intestinal parasite, were used to challenge jejunal tissue from guinea pigs immunized by infection with the parasite. Histamine (10(-4) M) caused an increased in Isc that was similar to that induced by antigen. Diphenhydramine (10(-5) M) inhibited the epithelial electrical responses to histamine by 100% and to antigen by 60-70%. Indomethacin (10(-5) M), in combination with diphenhydramine, completely inhibited the antigen-induced rise in Isc. Furosemide (10(-4) M) caused 50-60% inhibition of the increase in Isc induced by antigen and histamine. Antigen challenge of isolated enterocytes did not alter intracellular cAMP content. However, antigen challenge of jejunal segments in which epithelial cells were in contact with sensitized mast cells increased mucosal cAMP content. These results suggest that electrogenic Cl- secretion, mediated in part by cAMP, contributes to antigen-induced jejunal ion transport changes and that histamine and prostaglandins are involved in eliciting these epithelial responses.


1993 ◽  
Vol 264 (6) ◽  
pp. C1542-C1549 ◽  
Author(s):  
M. D. DuVall ◽  
S. M. O'Grady

Porcine gallbladder epithelium from the neck and the fundus of the organ was stripped of serosal muscle and mounted in Ussing chambers to investigate the mechanisms of K secretion. The sensitivity to K channel blockers and regulation by norepinephrine (NE), adenosine 3',5'-cyclic monophosphate (cAMP), and increases in intracellular Ca concentration ([Ca]) were studied. The porcine gallbladder secretes K (approximately 0.8 mu eq/cm2.h) under basal conditions. Mucosal tetraethylammonium (TEA) produced a concentration-dependent increase in short-circuit current (Isc) and inhibited the unidirectional serosal-to-mucosal 86Rb flux JsmRb, resulting in a > 60% reduction in net Rb secretion. In contrast, serosal Ba produced a concentration-dependent decrease in Isc and stimulated JsmRb, resulting in a > 200% increase in net Rb secretion. NE inhibited JsmRb and net Rb secretion in both regions. In the fundic region the mucosal-to-serosal Rb flux (JmsRb) was also significantly increased, suggesting that active K absorption was activated. Exogenous cAMP increased JsmRb and net Rb secretion by > 85% in both regions. This increase in net Rb secretion was blocked by mucosal TEA but unaffected by NE. The Ca ionophore ionomycin also increased JsmRb and net Rb secretion and reduced the Isc by approximately 50%. Neither mucosal TEA nor Ba blocked changes in steady-state Rb secretion induced by ionomycin. Although both serosal Ba and ionomycin produced significant reductions in Isc, the effects of Ba were blocked by ionomycin pretreatment. These findings indicate that basal K secretion occurs through TEA-sensitive apical K channels and is regulated by intracellular cAMP. NE likely reduces K secretion by decreasing intracellular concentration of cAMP.(ABSTRACT TRUNCATED AT 250 WORDS)


2004 ◽  
Vol 72 (8) ◽  
pp. 4848-4858 ◽  
Author(s):  
H. J. Epple ◽  
J. Mankertz ◽  
R. Ignatius ◽  
O. Liesenfeld ◽  
M. Fromm ◽  
...  

ABSTRACT The diarrheal mechanisms in Aeromonas enteritis are not completely understood. In this study we investigated the effect of aeromonads and of their secretory products on ion secretion and barrier function of monolayers of human intestinal cells (HT-29/B6). Ion secretion was determined as a short-circuit current (ISC) of HT-29/B6 monolayers mounted in Ussing-type chambers. Transepithelial resistance (Rt) served as a measure of permeability. A diarrheal strain of Aeromonas hydrophila (strain Sb) added to the mucosal side of HT-29/B6 monolayers induced a significant ISC (39 ± 3 μA/cm2) and decreased the Rt to ∼10% of the initial value. A qualitatively identical response was obtained with sterile supernatant of strain Sb, and Aeromonas supernatant also induced a significant ISC in totally stripped human colon. Tracer flux and ion replacement studies revealed the ISC to be mainly accounted for by electrogenic Cl− secretion. Supernatant applied serosally completely abolished basal ISC. The supernatant-induced ISC was inhibited by the protein kinase C inhibitor chelerythrine, whereas a protein kinase A inhibitor (H8) and a Ca2+ chelator (BAPTA-AM) had no effect. Physicochemical properties indicated that the supernatant's active compound was an aerolysin-related Aeromonas beta-hemolysin. Accordingly, identical ISC and Rt responses were obtained with Escherichia coli lysates harboring the cloned beta-hemolysin gene from strain SB or the aerA gene encoding for aerolysin. Sequence comparison revealed a 64% homology between aerolysin and the beta-hemolysin cloned from Aeromonas sp. strain Sb. In conclusion, beta-hemolysin secreted by pathogenic aeromonads induces active Cl− secretion in the intestinal epithelium, possibly by channel insertion into the apical membrane and by activation of protein kinase C.


2002 ◽  
Vol 283 (2) ◽  
pp. G347-G356 ◽  
Author(s):  
Yutaka Hosoda ◽  
Shin-Ichiro Karaki ◽  
Yukiko Shimoda ◽  
Atsukazu Kuwahara

Interaction between substance P (SP) and PGE2on Cl−secretion in the guinea pig distal colonic epithelia was investigated. A short-circuit current ( Isc) was measured as an index of ion transport. Mucosa preparations deprived of muscle and submucosa of distal colon were mounted in the Ussing flux chamber and treated with TTX and piroxicam to remove the influences of neuronal activity and endogenous PG synthesis, respectively. Although SP (10−7M) itself evoked little increase in Isc, exogenous PGE2concentration dependently enhanced the response of SP. The effect of PGE2on the SP-evoked response was mimicked by forskolin and 8-bromoadenosine cAMP. Depletion of Ca2+from the bathing solution reduced the PGE2-dependent response of SP. Effects of PGE2, SP, and SP in the presence of PGE2on intracellular Ca2+concentration ([Ca2+]i) in isolated crypt cells were measured by the confocal microscope fluorescence imaging system. SP, but not PGE2, temporally evoked an increase in [Ca2+]ibut declined to the baseline within 3 min. A return of the SP-evoked increase in [Ca2+]iwas slower in the presence of PGE2than SP alone. These results suggest that PGE2synergistically enhances SP-evoked Cl−secretion via an interaction between the intracellular cAMP and [Ca2+]iin the epithelial cells. In conclusion, SP and PGE2could cooperatively induce massive Cl−secretion in guinea pig distal colon at epithelial levels.


1999 ◽  
Vol 277 (3) ◽  
pp. G709-G716 ◽  
Author(s):  
M. Mall ◽  
M. Bleich ◽  
J. Kuehr ◽  
M. Brandis ◽  
R. Greger ◽  
...  

Cystic fibrosis (CF) patients show characteristic defects in epithelial ion transport, such as failure in cAMP-dependent Cl−secretion. Because the cystic fibrosis transmembrane conductance regulator (CFTR) also functions as a downregulator of epithelial Na+ channels (ENaC), enhanced Na+ conductance was found in the airways of CF patients. Here, we examined whether enhanced epithelial Na+ conductance is also present in the colonic epithelium of CF patients and examined the underlying mechanisms. Thus transepithelial voltages were measured, and equivalent short-circuit currents ( I sc-eq) were determined by means of a novel type of Ussing chamber. Non-CF tissues demonstrated cAMP-dependent Cl− secretion that was absent in biopsies of CF patients. Correspondingly, Isc-eq was inhibited in non-CF but not in CF epithelia when synthesis of endogenous prostaglandins was blocked by indomethacin. In the presence of indomethacin, a larger portion of amiloride-sensitive Isc-eq was detected in CF tissues, suggesting enhanced ENaC conductance in colonic mucosa of CF patients. Increase of intracellular cAMP by forskolin and IBMX inhibited amiloride-sensitive ENaC currents in non-CF tissues but not in CF biopsies. Therefore, enhanced epithelial Na+ conductance is present in the CF colon and is probably due to missing downregulation by CFTR.


1999 ◽  
Vol 276 (1) ◽  
pp. G64-G72 ◽  
Author(s):  
Richard V. Benya ◽  
Jorge A. Marrero ◽  
Denis A. Ostrovskiy ◽  
Athanasia Koutsouris ◽  
Gail Hecht

Galanin is a peptide hormone widely expressed in the central nervous system and gastrointestinal (GI) tract. Within the GI tract galanin is present in enteric nerve terminals where it is known to modulate intestinal motility by altering smooth muscle contraction. Recent studies also show that galanin can alter intestinal short-circuit current ( I sc) but with differing results observed in rats, rabbits, guinea pigs, and pigs. In contrast, nothing is known about the ability of galanin to alter ion transport in human intestinal epithelial tissues. By RT-PCR, we determined that these tissues express only the galanin-1 receptor (Gal1-R) subtype. To evaluate Gal1-R pharmacology and physiology, we studied T84 cells. Gal1-R expressed by these cells bound galanin rapidly (half time 1–2 min) and with high affinity (inhibitor constant 0.7 ± 0.2 nM). T84 cells were then studied in a modified Ussing chamber and alterations in I sc, a measure of all ion movement across the tissue, were determined. Maximal increases in I sc were observed in a concentration-dependent manner around 2 min after stimulation with peptide, with 1 μM galanin causing I sc to rise more than eightfold and return to baseline occurring within 10 min. The increase in galanin-induced I sc was shown by125I efflux studies to be due to Cl− secretion, which occurred independently of alterations in cAMP and phospholipase C. Rather, Cl− secretion is mediated via a Ca2+-dependent, pertussis toxin-sensitive mechanism. These data suggest that galanin released by enteric nerves may act as a secretagogue in the human colon by activating Gal1-R.


Sign in / Sign up

Export Citation Format

Share Document