Abstract
The physiology of gastric acid secretion is one of the earliest subjects in medical literature and has been continuously studied since 1833. Starting with the notion that neural stimulation alone drives acid secretion, progress in understanding the physiology and pathophysiology of this process has led to the development of therapeutic strategies for patients with acid-related diseases. For instance, understanding the physiology of parietal cells led to the developments of histamine 2 receptor blockers, proton pump inhibitors, and recently, potassium-competitive acid blockers. Furthermore, understanding the physiology and pathophysiology of gastrin has led to the development of gastrin/CCK2 receptor antagonists. The need for refinement of existing drugs in patients have led to 2nd and 3rd generation drugs with better efficacy at blocking acid secretion. Further understanding of the mechanism of acid secretion by gene targeting in mice has enabled us to dissect the unique role for each regulator to leverage and justify the development of new targeted therapeutics for acid-related disorders. Further research on the mechanism of stimulation of gastric acid secretion and the physiological significances of gastric acidity in gut microbiome is needed in the future.
Keywords: Gastrin, gene targeting, netazepide, potassium-competitive acid blockers
Graphical abstract
In the stomach, endocrine pathways include G cell-ECL cell and G cell-parietal cells, paracrine pathways include ECL cell-parietal cell, D cell-ECL cell, D cell-G cell and D cell-parietal cell, neuronal pathways include nerve- all cell types, particularly ECL cell, and luminal pathways include HCl-D cell and food-G cell (not shown). Note: pharmacological targets on parietal cells and ECL cells.
Perspectives from history
In 1833, William Beaumont (1785 – 1853) published a book entitled Experiments and Observations on the Gastric Juice and the Physiology of Digestion, which was based on the observations of a patient named Alexis St. Martin who was accidentally shot by a gun in 1822 and survived with a permeant gastric fistula (1, 2). Subsequently, William Osler (1849–1919), known as the father of modern medicine, wrote an article entitled William Beaumont. A pioneer American physiologist highlighting the important contributions of Beaumont including (a) a more accurate and complete description of the components of gastric juice; (b) confirmation of previous observations that hydrochloric acid was the acid-type contained in gastric juice; (c) recognition that gastric juice and gastric mucus were separate entities secreted by the stomach; and (d) that digestion was dependent on muscular motions of the stomach (3–5).
Neural regulation of gastric acid secretion was demonstrated by Ivan Petrovich Pavlov (1849 –1936). In 1897, Pavlov summarized his work in a book entitled Lectures on the Work of the Principal Digestive Glands (6), which is one of the most important books in neurobiology. Using canine models of gastric fistula and gastric pouches, Pavlov demonstrated the neural control of salivary, gastric, and pancreatic secretions. In describing gastric secretion, Pavlov suggested a role for vagal stimulation in the process of acid secretion, which was verified experimentally by severing vagal nerve innervation to the stomach (Fig. 1). Pavlov also proposed the concept that conditional reflexes regulate digestion; the novel idea that psychology can regulate physiology. In 1904, Pavlov received the Nobel Prize in Physiology or Medicine for his ground-breaking experimental work in neurophysiology (7, 8).
Figure. 1:
The neuroendocrine mechanism of gastric acid secretion (HCl). Gastric wall is rich in blood supply and innervation by parasympathetic nerves from the dorsal motor nucleus of the vagus nerve (DMV), particular Cck neuron and its subtype of acetylcholine-contained Chat+ neurons, and by intrinsic neurons containing pituitary adenylate cyclase-activating polypeptide (PACAP) and galanin. Ingested food (Food) in the gastric lumen stimulates G cells to release gastrin (aminated gastrin) and gly-gastrin into the blood circulation. Gastrin stimulates CCK2 receptors (CCK2R) which are expressed on ECL cells and parietal cells. Activation of CCK2R on the ECL cells induces histamine formation by stimulation of histidine decarboxylase (HDC) and histamine secretion. Through paracrine mechanisms, the released histamine stimulates H2 receptors (H2R) which are located on parietal cells. Activation of H2R triggers parietal cells to secret HCl via H+,K+-ATPase in cooperation with K+ and Ca2+ channels and Na+/H+ and Cl−/HCO3− exchangers. ECL cells are also under neural control, particularly stimulation by PACAP on PAC1 receptor (PAC1R) and inhibition by galanin on Gal1 receptor (Gal1R) and under paracrine control by somatostatin on somatostatin subtype 2 receptor (SSTR2). Parietal cells are also under endocrine (e.g. gly-gastrin), paracrine (histamine, somatostatin) and neural control (acetylcholine released from vagal nerve-chat+ neuron) on muscarinic acetylcholine receptor subtype 3 (M3) receptor. G cells are under neural control (acetylcholine on M3R and galanin on Gal1 receptor) and paracrine control (somatostatin on SSTR2). D cells in the stomach consist of open-type in the antrum and close-type in the oxyntic mucosa and are under control of PACAP on PAC1R. The negative feedback loop between circulating gastrin and gastric luminal HCl is mediated by the opened D cells which are under neural control via PAC1R and M4R.
Once the vagal neurotransmitter acetylcholine was validated as an agonist of gastric acid secretion, other regulators were soon uncovered. For this, the endocrine regulation of gastric acid secretion was proposed by John Sydney Edkins (1863 – 1940). In 1905, Edkins proposed the novel concept in stomach that hormones, like secretin that was discovered by W.M. Bayliss and E.H. Starling (9), could also regulate acid secretion. To test this experimentally, an extract from the stomach pylorus (but not fundus) injected into the jugular vein resulted in gastric acid and pepsin secretion in anesthetized cats (10). It was subsequently discovered that the compound stimulating acid secretion was a hormone, produced specifically in the pyloric mucosa, and was named “gastrin” (11, 12). This original hypothesis was validated by H.J. Tracy and R.A. Gregory in 1964 by demonstrating that a series of synthetic peptides structurally related to gastrin also had agonist properties for acid secretion (13) (Fig. 1).
Paracrine regulation of gastric acid secretion was derived from an initial observation by Leon Popielski (1866 – 1920) who found that gastric acid secretion was induced by histamine when given subcutaneously but not intravenously in the canine model of gastric fistula (14, 15). Endogenous histamine was believed to be secreted by mast cells located in the gastric mucosa until gastric enterochromaffin-like (ECL) cells were identified by Rolf Håkanson. In 1966, Håkanson observed dopamine-containing epithelial cells in the oxyntic mucosa of rats that had been injected with L-DOPA and this cell population was morphologically indistinguishable from 5-hydroxytryptamine-containing EC cells (16). A year later, Håkanson found that the ECL cells contained histamine (17–20) (Fig. 1).
Once the major effectors of acid secretion were uncovered, studies by John G. Forte (1934–2012) and George Sachs (1935–2019), in particular, interrogated the intracellular mechanisms that regulate acid secretion including the structure of parietal cells (21, 22), the physiology and cell biology of parietal cells (23, 24), the physiology of proton pump functions (25, 26), and the location and function of ion pumps that are essential to support acid secretion (27) (Fig. 1). For timely reviews see (28–30).
Based on the understanding of the neural regulation of gastric acid secretion, Lester Dragstedt (1893–1975) demonstrated the therapeutic effects of vagotomy for duodenal ulcer in 1947, and as such, truncal, selective, and highly selective vagotomy were once commonly performed to treat peptic ulcer disease (31–33) (Fig. 1). In 1972 James Black (1924 – 2010) discovered that the effect of histamine on acid secretion was mediated by histamine H2 receptors (H2R), which led to the development of the selective H2R antagonists (H2RA) metiamide (34, 35) and then cimetidine (36), which was launched in 1976 as Tagamet (34, 36–38) (Fig. 1). In 1988 Black, with Ellion and Hitchings, received the Nobel Prize in Physiology or Medicine for pioneering research that led to new classes of drugs: importantly histamine H2RA to address acid hypersecretion-induced gastric and duodenal ulcer diseases. Cimetidine and other H2RAs that followed, such as famotidine, nizatidine, and ranitidine, provided effective medical treatment for acid-related disorders. However, patients began to relapse despite continuing H2RA treatment. Eventually it was realized that H2RA-induced acid suppression results in an increase in serum gastrin (hypergastrinemia) and tolerance to H2RA treatment (39).
The gastrin receptor was cloned and characterized by Alan S. Kopin and co-workers in 1992 using isolated canine parietal cells (40), and was shown to be one of the receptors that bind cholecystokinin (CCK), a hormone secreted by the L cells in duodenum. Two types of CCK receptors have been identified, type A, “alimentary,” and type B, “brain”. Based on recommendations of the International Union of Pharmacology (IUPHAR) committee regarding receptor nomenclature and drug classification, the CCK-A receptor was renamed CCK1 receptor (CCK1R), and the CCK-B receptor was renamed CCK2 receptor (CCK2R). CCK1R binds and responds to sulfated CCK with a 500- to 1,000-fold higher affinity or potency than sulfated gastrin or nonsulfated CCK. The CCK2R binds and responds to gastrin or CCK with almost the same affinity or potency but discriminates poorly between sulfated and nonsulfated peptides (41).
The CCK2R is localized to the ECL cells and the neck zone proliferating cells of the stomach. Physiologically, gastrin stimulates acid secretion by acting directly on the parietal cells and/or by mobilizing histamine release from the ECL cells, which then induces acid secretion by binding to the H2 receptors located on parietal cells (Fig. 1). Gastrin is also a growth factor, particularly for the oxyntic mucosa of the stomach, where hypergastrinemia stimulates proliferations of CCK2R+ progenitors, resulting in increased parietal- and ECL-cell mass. With unchecked gastrin-mediated proliferation, ECLoma and carcinoid tumors develop via the activation of extracellular signal-regulated kinase/mitogen-activated protein kinase signaling (42–45).
With the success of H2RA, pharmaceutical companies such as Merck, Lilly, and Wyeth, became interested in researching CCK2R antagonists (CCK2RAs). Many inhibitors were synthesized, but problems with potency, selectivity for the CCK2R, agonist activity, and oral bioavailability hampered their efficacy. Some CCK2RAs were tested in healthy subjects and patients, but none proved worthy of further development (46–49). In fact, the interest in CCK2RAs waned when substituted benzimadazoles, like omeprazole, were shown to inhibit the H+, K+-ATPase (50, 51), which is the proton pump on gastric parietal cells (52). After decades of investigation about the properties of a pump that secretes acid, Sachs and colleagues (53–55) described the nature of this pump, which facilitated the development of inhibitors to block its activity. Proton pump inhibitors (PPIs) proved superior to H2RAs in healing acid-related conditions and, although PPIs also induce hypergastrinemia, drug tolerance does not develop because of the mechanism of drug action (56). The finding that H. pylori causes most cases of peptic ulcer disease (57), and can be eradicated by a short course of PPI treatment and a cocktail of antibiotics, was likely another reason for disinterest in researching and developing a CCK2RA for clinical use. After H. pylori eradication had been introduced, the need for maintenance therapy for peptic ulcer disease was largely eliminated and gastro-esophageal reflux disease (GERD) became the main indication for prolonged inhibition of gastric acid. Many millions of patients have taken PPI since omeprazole was marketed in the 1980s. Although in recent years there has been a resurgence of interest in the physiology and pathology of gastrin, especially the trophic effects of hypergastrinemia on the stomach, pancreas and colon, there remains no CCK2RA available for clinical use (58, 59). Thus, novel therapeutic strategies are needed to inhibit gastric acid secretion and prevent the trophic/tumor-promoting effects of gastrin when using CCK2RA, as is described below.
Control of gastric acid secretion by gastrin/CCK2 receptor antagonists
Physiology
Gastrin is a peptide hormone produced by a single gene that is synthesised as pre-progastrin, and is processed into progastrin and amidated gastrin peptide fragments, mostly G17 and G34 (60). Both gastrin fragments have a C-terminal amidated tetrapeptide that bind to CCK2R in the stomach, and in the central and peripheral nervous systems.
G cells in the gastric antrum produce gastrin, which stimulates gastric acid secretion, proliferation, migration, differentiation, and have anti-apoptotic effects on gastric epithelial cells. Gastrin also regulates various genes in the gastric mucosa and stimulates paracrine cascades, including cytokines and growth factors (60, 61). Gastrin activates CCK2R on ECL cells in the oxyntic mucosa of the stomach to secrete histamine, which in turn stimulates histamine H2-receptors on adjacent parietal cells to secrete acid via the H+/K+-ATPase (Fig. 1).
The ingestion of food causes secretion of gastrin into the systemic circulation. Initially, the buffering capacity of food neutralises gastric acid, but as acid secretion continues and digestion proceeds and the gastric contents move into the duodenum, the buffering capacity of the food bolus diminishes and intragastric pH falls. This acidic pH stimulates D cells in the gastric antrum to secrete somatostatin, a hormone that switches-off gastrin secretion. Secretion of gastric acid is also under the control of vagal stimulation, which leads to gastrin release. The vagus also controls the release of somatostatin. Experiments in healthy human subjects, in which food or intravenous gastrin 17 was used to stimulate gastric acid secretion, indicate that gastrin accounts for about 90% of acid secreted after a meal (62) (Fig. 1).
Pathophysiology
Ingestion of food at mealtimes leads to physiological increases and decreases in circulating gastrin throughout the day. However, chronic reduction in the acid output by disease, such as autoimmune chronic atrophic gastritis (CAG), genetic mutations of the proton pump, H. pylori infection of the oxyntic mucosa, or by an acid suppressant, such as a PPI or a potassium-competitive acid blocker (P-CAB), causes hypergastrinemia (62), which is the persistent elevation of circulating gastrin, which can be friend or foe depending on the circumstances, as follows:
Hypergastrinemia in patients with CAG stimulates ECL-cell growth and, in some patients, may cause gastric neuroendocrine tumours (g-NETs), which have malignant potential (63, 64). CAG patients also had a seven-fold increase in the risk of gastric cancer in one study (65) and 200-fold in another study (66). Mutations of KCNQ1 or KCNE1 genes, which are potassium channels in parietal cells (67), or mutation of the ATP4A gene, which encodes the α-subunit of the proton pump (68), cause achlorhydria and hypergastrinemia, which leads to g-NETs and gastric carcinoma in some patients. Hypergastrinemia induced by long-term PPI therapy is associated with a 3-fold increased risk of gastric carcinoma (69), even after H. pylori-eradication (70).
Patients with Zollinger–Ellison syndrome (ZES) have hypergastrinemia due to ectopic secretion of gastrin from a NETs (usually in the pancreas/duodenum), which is associated with ECL-cell growth and greater malignant potential than g-NETs in CAG patients (71). In order to control ZES, the majority of ZES patients require lifelong treatment for the marked gastric acid hypersecretion induced by hypergastrinemia, with potent gastric acid anti-secretory agents (72).
Gastrin is known to induce miR-222 overexpression (73), which has been reported to be upregulated in many types of cancer (74–77). Higher levels of miR-222 are associated with more advanced disease and reduced 5year survival (78, 79). miR-222 expression is also increased in gastric cancer tissue-derived mesenchymal stem cells and in the stomach of H. pylori infected individuals (80, 81). The CCK2RA netazepide reduced overexpressed miR-222 in the serum and gastric corpus mucosa of patients with CAG, hypergastrinemia and g-NETs, and miR-222 returned to normal levels after stopping netazepide treatment (73).
The Mongolian gerbil has been used as an efficient, robust, and cost-effective rodent model that recapitulates many features of H. pylori-induced gastric inflammation and carcinogenesis in humans (82). Netazepide prevented oxyntic mucosal inflammation induced by H. pylori infection in Mongolian gerbils, indicating that gastrin may also have inflammatory activity (59).
Preclinical studies
Several benzodiazepine-derived CCK2RA, such as L-364,718 (devazepide), L-365,260, CI-988, YM022, Z-360 and YF476 (netazepide) have been invented (83, 84). Two new classes have recently been synthesized, JB95008 is a substituted imidazole (85) and JNJ-26070109 is a benzamide derivative (86). JNJ-26070109 caused dose-dependent inhibition of pentagastrin-stimulated acid secretion in rats and dogs and prevented rebound hyper-responsiveness of acid secretion to pentagastrin after stopping omeprazole in rats. YM022 also prevented rebound hyper-responsiveness of acid secretion to pentagastrin after stopping omeprazole (87). However, most other CCK2RAs have had problems with potency, selectivity for the CCK2 versus CCK1 receptor, solubility or oral bioavailability. Netazepide, a benzodiazepine derivative, has been the most studied CCK2RA (88–90) with main results as follows:
In in vitro studies, netazepide caused a concentration-dependent inhibition of specific binding of the ligand [125I] CCK-8 to rat and cloned human CCK2R in vitro. The affinity of netazepide for rat gastrin receptors was 264 and 70 times higher than that of two other CCK2RA, L-365,26020 and CI-988, respectively, and 4100 times its affinity for rat pancreatic CCK1 receptors. Netazepide had very little affinity for various other receptors, such as histamine, muscarinic and benzodiazepine receptors.
In anaesthetised rats, intravenous netazepide caused dose-dependent inhibition of pentagastrin-stimulated gastric acid secretion, with an ED50 of 0.0086 mmol/kg, compared with an ED50 of 0.013 mmol/kg for intravenous famotidine.
In Heidenhain pouch dogs, intravenous and oral netazepide each caused dose-dependent inhibition of pentagastrin-stimulated gastric acid secretion, with ED50 values of 0.018 and 0.020 mmol/kg, respectively. ED50 for intravenous and oral famotidine was 0.078 and 0.092 μmol/kg, respectively
In dogs with a gastric fistula, intravenous netazepide dose-dependently inhibited pentagastrin-induced gastric acid secretion, with ED50 0.0023 mmol/kg. Also in gastric fistula dogs, oral netazepide, famotidine, and the PPI, omeprazole, each dose-dependently inhibited peptone-induced gastric acid secretion, with ED50 of 0.11, 0.76 and 4.28 mmol/kg, respectively. Therefore, netazepide was about 7 and 40 times more potent than famotidine and omeprazole, respectively. Comparison of ED50 for intravenous and oral netazepide indicates that the oral bioavailability was 26–28% in rats and 27–50% dogs.
In rats, netazepide prevented the increases in ECL-cell activity and density, oxyntic mucosal thickness, mucosal histamine decarboxylase (HDC) activity and serum pancreastatin caused by PPI-induced hypergastrinemia (91). It should be noted that the ECL cells make up the larger part (60–70%) of the endocrine cells of the stomach. They are rich in pancreastatin/chromogranin (CGA)-related peptides which shared with most other peptide hormone-producing cells (92–94). Surgical removal of the acid-producing part of the stomach eliminates most pancreastatin (70–80%) from the blood stream, and the synthesis and release of pancreastatin from the ECL cells is known to be via the exocytotic proteins under neurohormonal regulation, including gastrin (95–100). Thus, pancreastatin is a biomarker of ECL-cell activity (101–107). ECL cells contain vesicular monoamine transporter 2 (VMAT2) to accumulate histamine into the secretory vesicles (108–110), while EC cells contain VMAT1 (and therefore accumulate serotonin)(111). Reserpine, once-used clinical drug, inhibited histamine secretion from the ECL cells (110, 112).
Thus, these preclinical studies have shown netazepide to be a potent, highly selective, competitive and orally active CCK2RA. Indeed, netazepide has been described as the “gold standard” CCK2RA (83) and has been used in many other experimental animal-based studies, to assess the physiology and pathology of gastrin.
Clinical studies
In early clinical studies, L-365,260 produced modest and short-lasting inhibition of gastrin-stimulated acid secretion (113). Since then, netazepide has been the only CCK2RA used in studies to assess the effect of a CCK2RA on gastric acid production in humans. To date, there have been no studies of a CCK2RA in patients with acid-related diseases. However, the clinical pharmacology of oral netazepide has been characterised in a series of studies in healthy subjects.
In a first-in-human, single-dose study in which netazepide was compared with the H2RA, ranitidine, both increased 24-h gastric pH. The onset of activity was similarly rapid for both, but activity of ranitidine lasted only about 12 h whereas that of netazepide exceeded 24 h. Median tmax and t1/2 were about 1 and 7 h, respectively, and the pharmacokinetics were dose proportional (114). Recent human CCK2R binding studies have shown that netazepide is an insurmountable (non-competitive) CCK2RA, not a competitive antagonist as suggested by the preclinical studies (unpublished data). That would explain the long duration of activity of netazepide in raising 24-h gastric acidity.
Single doses of netazepide caused dose-dependent inhibition of pentagastrin-stimulated gastric acid secretion, which persisted after repeated doses (115). Repeated doses also caused hypergastrinemia, which is consistent with persistent acid suppression.
Netazepide and the PPI, rabeprazole, alone and combined once daily for 6 weeks were compared with respect to their effect on basal and pentagastrin-induced gastric acid secretion and 24-h circulating gastrin and CgA at baseline, start and end of treatment, and to determine if netazepide can prevent the trophic effects of PPI-induced hypergastrinemia (116). Single doses of netazepide alone and rabeprazole alone caused dose-dependent inhibition of pentagastrin-stimulated gastric acid secretion, which persisted after repeated doses. Rabeprazole alone and netazepide alone for 6 weeks were similarly effective in reducing acid and increasing serum gastrin. A combination of rabeprazole and netazepide increased serum gastrin and reduced basal acid secretion more than either treatment alone, suggesting more effective acid suppression. Rabeprazole alone increased plasma CgA – a sign of increased ECL-cell activity - whereas netazepide alone reduced plasma CgA - a sign of reduced ECL-cell activity. When combined with rabeprazole, netazepide prevented the increase in CgA resulting from rabeprazole-induced hypergastrinemia, which is consistent with blockade of CCK2R on ECL cells. Thus, netazepide suppressed pentagastrin-stimulated gastric acid secretion as effectively as rabeprazole; the reduction in basal acid secretion and greater increase in serum gastrin by the combination is consistent with more effective acid suppression. The increase in plasma CgA after rabeprazole is consistent with a trophic effect on ECL cells, which netazepide prevented. Therefore, netazepide with or without a PPI, is a potential treatment for acid-related conditions, and netazepide is a potential treatment for the trophic effects of hypergastrinemia.
In a study to assess whether netazepide can prevent the trophic effect of the PPI, esomeprazole, dosed for 28 days, esomeprazole increased circulating gastrin and CgA, whereas netazepide increased gastrin but not CgA, and inhibited dose-dependently the CgA response to esomeprazole. Gastrin and CgA returned to baseline within 2–3 days of esomeprazole withdrawal; netazepide did not shorten that time. There was no rebound dyspepsia after PPI withdrawal that others have reported (117). Thus, netazepide can prevent the trophic effects of hypergastrinemia, but a study of netazepide in patients on long-term PPI therapy is required to establish whether PPI withdrawal really can lead to rebound hyperacidity, and whether netazepide can prevent it.
Although PPIs are safe medicines, PPI-induced hypergastrinemia is associated with: ECL-cell and parietal-cell hyperplasia; fundic gland polyps; increased risk of gastric cancer and of bone fractures; and rebound hyperacidity and dyspepsia after PPI withdrawal. A CCK2RA should be free of those effects and should prevent them if co-administered with a PPI (118).
Ceclazepide is a new benzodiazepine-derived CCK2RA and prodrug that has advantages over netazepide in terms of selectivity, solubility and bioavailability, and is in early clinical development for acid-related conditions (119).
Control of gastric acid secretion by potassium-competitive acid blockers (P-CABs)
Studies aimed to understand H+ transport by the H+, K+-ATPase and the development of acid pump blockers were accompanied by similar efforts to understand potassium (K+)-transport and making K+-binding site blockers. Potassium transport by the H+, K+-ATPase occurs from the gland lumen to the cytoplasmic face of the apical surface of parietal cells and is required for pump activity. With the development of PPIs moving forward at a rapid pace, the development of P-CABs followed suit and the first successful chemistry was reported in 1981 by Long and colleagues as SCH 28080 (2-methyl-8-(phenylmethoxy)imidazo[1 ,2-a]pyrindine-3-acetonitrile)(120). There were subsequently a number of P-CABs in development, but SCH 28080 moved rapidly to investigations in numerous acid secretion models including in human patients (121–123). SCH 28080 was shown to be a competitive but reversible inhibitor of H+, K+-ATPase activity, was highly selective for the H+, K+-ATPase pump, and had a rapid onset of action (121, 124, 125). The original P-CABs had unwanted side-effects in human clinical trials that hindered further development until Takeda Pharmaceuticals, under the scientific leadership of Tadataka Yamada (1945–2021), resurrected this class of drugs. P-CAB development was re-initiated to respond to several unmet needs that emerged from PPI usage, including suppression of night-time acid secretion and rapid symptom relief.
Takeda synthesized a new pyrrole derivative in 2010, 1-[5-(2-fluorophenyl)-1-(pyridin-3- ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine monofumarate ((126), TAK-438 or vonoprazan fumarate), that was a completely different formulation from other P-CABs including SCH 28080 (127). In 2011 this drug was fully characterized and found to be K+-competitive, accumulated more effectively than SCH 28080 in parietal cells, had a slow disassociation rate and very high affinity for the H+, K+-ATPase pump, thus providing faster onset and longer-lasting inhibition of acid secretion compared to SCH 28080 (128) In many clinical trials, TAK-438 has proven to be effective at maintaining a high night-time pH with a good safety profile (129–131), and is effective or superior to PPI treatment with a similar safety profile for treating ulcer diseases, erosive esophagitis, H. pylori infection, and GERD (132–134). Greater acid suppression results in much higher levels of serum gastrin and the associated risks compared with PPIs (135). Tegoprazan and fexuprazan are the latest generation of P-CABs, with similar efficacy profiles to TAK-438 (136–138). A recent study demonstrated the molecular structure of the H+, K+-ATP with TAK-438 bound (vonoprazan)(139). P-CAB therapy for the treatment of acid-related disorders has been reviewed recently by others (140–142).
Control of gastric acid secretion revealed through genetic dissection
The stomach is primarily innervated by parasympathetic nerves from the dorsal motor nucleus of the vagus nerve (DMV, Fig. 1). Studies using immunohistochemistry showed that the preganglionic vagal nerve terminals act at cholinergic ganglia in the stomach, to then stimulate intrinsic cholinergic neurons to fire (via nicotinic receptor activation) and release acetylcholine at muscarinic receptors on cell targets such as parietal cells and G cells (143). Both pituitary adenylate cyclase-activating polypeptide (PACAP)-containing fibers and galanin-containing fibers were found in the gastric wall (Fig. 1), including mucosa, submucosa, myenteric plexus and muscularis (144–147).
A recent study using single-cell transcriptomics, recombinase-expressing mouse lines and recombinase-dependent antegrade neural projection tracing techniques showed that DMV-Cck neurons and DMV-Pdyn neurons exclusively innervate the glandular stomach (Fig. 1), targeting distinct enteric neuron subtypes through acetylcholine, respectively. The enteric neuron subtypes were Chat+, which release acetylcholine to induce gastric contraction, and Nos1+, which release nitric oxide to induce gastric relaxation (148)(Fig. 1).
Utilization of various mouse gene knockout (KO) models have enabled us to better understand the functional role of each regulatory peptide, neurotransmitter, neuroendocrine-mediated receptor, pump, or transporter in the process of gastric acid secretion (Fig. 1-inset).
Gastrin KO in mice led to impaired gastric acid secretion at basal level as well as in response to stimuli such as gastrin, histamine, or carbachol that mimics the effect of acetylcholine on both the muscarinic and nicotinic receptors (149, 150). Glycine-extended gastrin synergized with amidated gastrin 17 to stimulate parietal cells in gastrin KO mice (151). Furthermore, chronic H. pylori infection impaired parietal-cell function with low acid output, hypergastrinemia and hyperplasia of ECL cells in wild-type mice but led to vagally-induced hypersecretion of acid in gastrin KO mice, providing a partial explanation that H. pylori infection is a causal factor for either gastric cancer (which is associated with low gastric acid secretion) or duodenal ulcer (high acid secretion)(152).
CCK2R (but not CCK1R) KO led to altered differentiation of ECL cells that were characterized by a lack of secretory vesicles and histamine formation, markedly impaired gastric acid secretion, atrophy of the oxyntic mucosa and hypergastrinemia (153–156). CCK1+2R double KO mice exhibited impaired acid secretion in response to vagal stimulation but partially normalized the ECL cells, likely by a compensative mechanism via up-regulating PACAP type 1 receptor (PAC1) on the ECL cells (156).
Acid secretion stimulated directly by vagal neurons, which mostly secrete acetylcholine in the gastric body, is mediated by muscarinic receptor subtypes 3 and 5 (M3R and M5R) in mice, probably via Chat+ neurons (157, 158). M3R KO mice had an impaired gastric secretion and lack of trophic responses to hypergastrinemia (159). However, it was also shown in M4R KO mice that M3R activation mediated vagal stimulation of acid secretion in concert with M4R, which indirectly modulated the rate of acid secretion after stimulation (160). These results suggested that the activation of M4R after vagal stimulation inhibited somatostatin release from D cells, thus potentiating the rate of acid secretion occurring via M3R activation (160).
Parietal cells secrete HCl by a mechanism involving at the least H+,K+-ATPase, voltage-gated potassium (Kv) channel, Ca++ channels, and Cl−/HCO3− and Na+/H+ exchangers. Achlorhydria was found in mice lacking H+,K+-ATPase (161), the KCNE2 potassium channel (162), Ca2+ channel (163), Cl−/HCO3− exchangers (Slc4a2, or Ae2) (164, 165), and NHE4 Na+/H+ exchangers (166).
G cells, ECL cells, and parietal cells express somatostatin receptor type 2 (SSTR2). SSTR2 KO mice showed a shift from endocrine/paracrine to neural pathways in regulation of gastric acid secretion, i.e., i) a shift in the regulation of the ECL cells from a paracrine somatostatin-SSTR2-dependent pathway to a neural dependent pathway, with galanin-GalR1 pathway being a strong candidate; and ii) a shift in the stimulatory regulation of the parietal cells from the gastrin-ECL cell axis to an enhancement of the direct action of gastrin and vagal pathways (167).
It is known from studies of isolated ECL cells that PACAP stimulates histamine secretion via PAC1R, whereas galanin suppressed histamine via Gal1R (98). However, studies using SSTR2 KO mice showed that peripheral PACAP inhibited gastric acid secretion, probably by activation of PAC1R on gastric D cells to induce somatostatin release which inhibits both ECL and parietal cells, whereas galanin inhibited gastric acid secretion through a somatostatin-independent mechanism (168, 169). Furthermore, PAC1 receptor KO mice had an increased basal gastric acid output which was associated with increased number of parietal cells (170). Taken together, these results suggest that a dynamic interaction exists between PACAP-ECL cells pathway and PACAP-D cells in controlling gastric acid secretion.
In comparison with wild-type mice, HDC KO mice exhibited impaired acid secretion with secondary hypergastrinemia, whereas mast cell-deficient mice had unchanged acid secretion (171). H2R KO mice had impaired acid secretion in response to histamine and gastrin but not carbachol, suggesting an independent cholinergic signaling pathway in controlling the parietal cells (172, 173). This phenotype can be explained by the known mechanism of secretory cells in general and parietal cells in particular, i.e., the crosstalk between intracellular Ca++ level and cAMP signaling system (174–176)(Fig. 1).
Conclusions
There is a rich history of studies that link acid secretion to neural innervation of the stomach via vagal afferents that secrete acetylcholine. The stomach has a complicated circuit of cells that are innervated by the vagus nerve, each of which contribute to acid secretion by providing negative (somatostatin) or positive (acetylcholine, gastrin, histamine) regulators.
Once it was determined that acid hypersecretion was the cause of morbidity and mortality in human patients, surgical vagotomy was used to block neural innervation to the stomach thus blocking acid secretion. With numerous side effects arising from such an intervention, more selective vagal blockade was done to cut vagal innervation as close to parietal cells as possible. Doing so reduced acid secretion while also reducing some side effects of the surgery.
Drugs that serve to block vagal innervation were not possible due to the widespread localization of vagal afferent neurons through the human body. However, this problem resulted in the development of specific and targeted therapeutics that were developed to unique receptors on parietal cells, which have been used successfully since about 1980. These include H2 receptor antagonists to block histamine H2 receptors and PPIs to block the H+, K+-ATPase proton pump.
New classes of drugs are being developed regularly to fill-in needed gaps in efficacy of current therapeutics. For this, potassium-competitive K+-channel blockers (P-CABs) like TAK-438 and gastrin CCK2 receptor antagonists, like netazeptide and ceclazepide, are in the pipeline.
In using various KO mice, the unique role for each regulator and the mechanisms of physiological compensation has been uncovered. This feature has been used to leverage and justify the development of new targeted therapeutics for gastric acid-related disorders.
Perspectives for future
Today, clinical management of gastric acid-related disorders has been revolutionized by the understanding of the mechanism of acid secretion and accordingly, the introduction of potent antisecretory medications (28). However, the statement by William Beaumont in 1838 that “the quantity of gastric juice secreted by the stomach for the solution of its contents does not depend upon the quantity of food introduced into the cavity, but upon the general requirements of the system” (1), should be readdressed.
The stomach, particularly gastric acid, has been thought to play an important role in the regulation of bone metabolism and mineral homeostasis. It was reported that impaired gastric acidification negatively affects calcium homeostasis and bone mass, based on the bone phenotype of CCK2R KO mice (177). However, this was at odds with the bone phenotype of HDC KO mice that displayed the same impaired acidification as CCK2R KO mice but had an increased bone formation and reduced bone resorption (178) Thus, it is unlikely that a lack of gastric acid production per se causes bone loss (179)(see also a separate article by Chen D et al. in this SI). But on the other hand, studies are needed in the future to evidently demonstrate that an adequate gastric acid production is important for bone metabolism and mineral homeostasis, particularly in connection with long-term use of PPIs and aging.
The gut microbiome/microbiota has been demonstrated to play an important role in physiology and diseases, including metabolism, immune system, CNS system, circulation system, and diseases such as obesity, diabetes, inflammatory bowel disease, dementia, chronic fatigue syndrome, several types of cancer, autoimmune diseases, asthma, and mental disorders/diseases (180–185). Studies are needed in the future to demonstrate the importance of gastric acid production upon the requirements and maintenance of a healthy gut microbiome.
While mechanism that regulate gastric acid secretion have been used effectively to inhibit acid secretion, resulting in effective treatments for acid-related disorders/diseases, the physiological mechanisms whereby the activation of brain ganglia by food, metabolic hormones, gastric luminal contents, and elevated pH stimulate acid secretion are poorly understood. In addition to the cells aforementioned, such as opened-type G cells and D cells, tuft cells have been proposed to be a potential chemo-sensor for the acid secretion. Tuft cells have been implicated in muscarinic responses, producing acetyl choline themselves, and chemosensing via taste receptors. The functional significance of Tuft cells is largely unknown and would thus be an important cell to interrogate in future studies (30).
Acknowledgments
The authors thank the grant supports by the Liaison Committee between the Central Norway Regional Health Authority and Norwegian University of Science and Technology (NTNU) and the Joint Program of the Medical Faculty of NTNU and St. Olav’s University Hospital (DC and CMZ) and by the National Institutes of Health (NIH RO1 DK103046 and NIH P30 DK034854) (SJH).
Footnotes
Conflict of Interest
MB owns Trio Medicines Ltd, which is developing netazepide for use in patients with hypergastrinemia. None of the other authors has any conflicts of interest to declare.
References
- 1.Beaumont W, Experiments and Observations on the Gastric Juice, and the Physiology of Digestion. Reprinted from the Plattsburgh Edition, with notes by Andew Combe (Maclachlan & Stewart, South Bridge; and Simpkin, Marshall & Co., London., Edinburgh, 1833), vol. MDCCCXXXVIII, pp. 1–328. [Google Scholar]
- 2.Gale A, Dr. William Beaumont: Founding Father of Gastroenterology. Mo Med 118, 518–519 (2021). [PMC free article] [PubMed] [Google Scholar]
- 3.Osler W, WILLIAM BEAUMONT. A PIONEER AMERICAN PHYSIOLOGIST. JAMA XXXIX, 1223–1231 (1902). [Google Scholar]
- 4.Roberts CS, in Clinical Methods: The History, Physical, and Laboratory Examinations, rd, Walker HK, Hall WD, Hurst JW, Eds. (Boston, 1990). [PubMed] [Google Scholar]
- 5.Skroska P, The William Beaumont papers: a life in letters. Mo Med 111, 419–423 (2014). [PMC free article] [PubMed] [Google Scholar]
- 6.Pavlov IP, The Work of the Digestive Glands. Thompson T. b. W. H., Ed., (Charles Griffin & Company, Limited, London, 1902). [Google Scholar]
- 7.Haas LF, Ivan petrovich pavlov (1849–1936). J Neurol Neurosurg Psychiatry 67, 299 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Smith GP, Pavlov and integrative physiology. Am J Physiol Regul Integr Comp Physiol 279, R743–755 (2000). [DOI] [PubMed] [Google Scholar]
- 9.Bayliss WM, Starling EH, The mechanism of pancreatic secretion. J Physiol 28, 325–353 (1902). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Edkins JS, On the chemical mechanism of gastric secretion. Proc Roy Soc B76, 376 (1905). [Google Scholar]
- 11.Edkins JS, The chemical mechanism of gastric secretion. J Physiol 34, 133–144 (1906). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Modlin IM, Kidd M, Marks IN, Tang LH, The pivotal role of John S. Edkins in the discovery of gastrin. World J Surg 21, 226–234 (1997). [DOI] [PubMed] [Google Scholar]
- 13.Tracy HJ, Gregory RA, Physiological Properties of a Series of Synthetic Peptides Structurally Related to Gastrin I. Nature 204, 935–938 (1964). [DOI] [PubMed] [Google Scholar]
- 14.Tiligada E, Ennis M, Histamine pharmacology: from Sir Henry Dale to the 21st century. Br J Pharmacol 177, 469–489 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Konturek SJ, Gastric secretion--from Pavlov’s nervism to Popielski’s histamine as direct secretagogue of oxyntic glands. J Physiol Pharmacol 54 Suppl 3, 43–68 (2003). [PubMed] [Google Scholar]
- 16.Hakanson R, Owman C, Distribution and properties of amino acid decarboxylases in gastric mucosa. Biochem Pharmacol 15, 489–499 (1966). [DOI] [PubMed] [Google Scholar]
- 17.Hakanson R, Owman C, Concomitant histochemical demonstration of histamine and catecholamines in enterochromaffin-like cells of gastric mucosa. Life Sci 6, 759–766 (1967). [DOI] [PubMed] [Google Scholar]
- 18.Hakanson R, Lilja B, Owman C, Properties of a new system of amine-storing cells in the gastric mucosa of the rat. Eur J Pharmacol 1, 188–199 (1967). [DOI] [PubMed] [Google Scholar]
- 19.Chen D, Zhao CM, Lindstrom E, Hakanson R, Rat stomach ECL cells up-date of biology and physiology. Gen Pharmacol 32, 413–422 (1999). [DOI] [PubMed] [Google Scholar]
- 20.Chen D, Rehfeld JF, Watts AG, Rorsman P, Gundlach AL, History of key regulatory peptide systems and perspectives for future research. Journal of Neuroendocrinology, e13251. [DOI] [PubMed] [Google Scholar]
- 21.Forte TM, Machen TE, Forte JG, Ultrastructural and physiological changes in piglet oxyntic cells during histamine stimulation and metabolic inhibition. Gastroenterology 69, 1208–1222 (1975). [PubMed] [Google Scholar]
- 22.Forte TM, Machen TE, Forte JG, Ultrastructural changes in oxyntic cells associated with secretory function: a membrane-recycling hypothesis. Gastroenterology 73, 941–955 (1977). [PubMed] [Google Scholar]
- 23.Engevik AC, Kaji I, Goldenring JR, The Physiology of the Gastric Parietal Cell. Physiol Rev 100, 573–602 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Baratta V et al. , In Pursuit of the Parietal Cell - An Evolution of Scientific Methodology and Techniques. Front Physiol 10, 1497 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Faller L et al. , Mechanistic aspects of gastric (H+ + K+)-ATPase. Ann N Y Acad Sci 402, 146–163 (1982). [DOI] [PubMed] [Google Scholar]
- 26.Forte JG, Lee HC, Gastric adenosine triphosphatases: a review of their possible role in HCl secretion. Gastroenterology 73, 921–926 (1977). [PubMed] [Google Scholar]
- 27.Shin JM, Munson K, Vagin O, Sachs G, The gastric HK-ATPase: structure, function, and inhibition. Pflugers Arch 457, 609–622 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Schubert ML, Peura DA, Control of gastric acid secretion in health and disease. Gastroenterology 134, 1842–1860 (2008). [DOI] [PubMed] [Google Scholar]
- 29.Schubert ML, Gastric secretion. Curr Opin Gastroenterol 24, 659–664 (2008). [DOI] [PubMed] [Google Scholar]
- 30.Engevik AC, Kaji I, Goldenring JR, The physiology of the gastric parietal cell. Physiological reviews 100, 573–602 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Dragstedt LR, Clarke JS, Harper PV, Woodward ER, Tovee EB, Supradiaphragmatic Section of the Vagus Nerves to the Stomach in Gastrojejunal Ulcer. J Thorac Surg 16, 226–236 (1947). [PubMed] [Google Scholar]
- 32.Woodward ER, Dr. Dragstedt’s first vagotomy. World J Surg 7, 554 (1983). [DOI] [PubMed] [Google Scholar]
- 33.Rabben HL, Zhao CM, Hayakawa Y, Wang TC, Chen D, Vagotomy and Gastric Tumorigenesis. Curr Neuropharmacol 14, 967–972 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Black JW et al. , Metiamide--an orally active histamine H2-receptor antagonist. Agents Actions 3, 133–137 (1973). [DOI] [PubMed] [Google Scholar]
- 35.Black JW, Duncan WA, Durant CJ, Ganellin CR, Parsons EM, Definition and antagonism of histamine H 2 -receptors. Nature 236, 385–390 (1972). [DOI] [PubMed] [Google Scholar]
- 36.Brimblecombe RW et al. , The pharmacology of cimetidine, a new histamine H2-receptor antagonist. 1975. Br J Pharmacol 160 Suppl 1, S52–53 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Stapleton MP, Sir James Black and propranolol. The role of the basic sciences in the history of cardiovascular pharmacology. Tex Heart Inst J 24, 336–342 (1997). [PMC free article] [PubMed] [Google Scholar]
- 38.Ganellin R, Duncan W, Obituary: James Black (1924–2010). Nature 464, 1292 (2010). [DOI] [PubMed] [Google Scholar]
- 39.Wilder-Smith CH et al. , Tolerance to oral H2-receptor antagonists. Dig Dis Sci 35, 976–983 (1990). [DOI] [PubMed] [Google Scholar]
- 40.Kopin AS et al. , Expression cloning and characterization of the canine parietal cell gastrin receptor. Proc Natl Acad Sci U S A 89, 3605–3609 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dufresne M, Seva C, Fourmy D, Cholecystokinin and gastrin receptors. Physiological reviews, (2006). [DOI] [PubMed] [Google Scholar]
- 42.Zhao CM, Chen D, The ECL cell: relay station for gastric integrity. Curr Med Chem 19, 98–108 (2012). [DOI] [PubMed] [Google Scholar]
- 43.Koh TJ, Chen D, Gastrin as a growth factor in the gastrointestinal tract. Regul Pept 93, 37–44 (2000). [DOI] [PubMed] [Google Scholar]
- 44.Hakanson R, Sundler F, Proposed mechanism of induction of gastric carcinoids: the gastrin hypothesis. Eur J Clin Invest 20 Suppl 1, S65–71 (1990). [DOI] [PubMed] [Google Scholar]
- 45.Sheng W et al. , Hypergastrinemia Expands Gastric ECL Cells Through CCK2R(+) Progenitor Cells via ERK Activation. Cell Mol Gastroenterol Hepatol 10, 434–449 e431 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.McDonald IM, CCK2 receptor antagonists. Expert Opin Ther Pat 11, 445–462 (2001). [Google Scholar]
- 47.Black JW, Kalindjian SB, Gastrin agonists and antagonists. Pharmacol Toxicol 91, 275–281 (2002). [DOI] [PubMed] [Google Scholar]
- 48.Herranz R, Cholecystokinin antagonists: pharmacological and therapeutic potential. Med Res Rev 23, 559–605 (2003). [DOI] [PubMed] [Google Scholar]
- 49.Black JW, Reflections on some pilot trials of gastrin receptor blockade in pancreatic cancer. Eur J Cancer 45, 360–364 (2009). [DOI] [PubMed] [Google Scholar]
- 50.Fellenius E et al. , Substituted benzimidazoles inhibit gastric acid secretion by blocking (H+ + K+)ATPase. Nature 290, 159–161 (1981). [DOI] [PubMed] [Google Scholar]
- 51.Wallmark B, Larsson H, Humble L, The relationship between gastric acid secretion and gastric H+,K+-ATPase activity. J Biol Chem 260, 13681–13684 (1985). [PubMed] [Google Scholar]
- 52.Olbe L, Carlsson E, Lindberg P, A proton-pump inhibitor expedition: the case histories of omeprazole and esomeprazole. Nat Rev Drug Discov 2, 132–139 (2003). [DOI] [PubMed] [Google Scholar]
- 53.Fellenius E et al. , Substituted benzimidazoles inhibit gastric acid secretion by blocking (H++ K+) ATPase. Nature 290, 159–161 (1981). [DOI] [PubMed] [Google Scholar]
- 54.Besancon M, Simon A, Sachs G, Shin JM, Sites of reaction of the gastric H, K-ATPase with extracytoplasmic thiol reagents. J Biol Chem 272, 22438–22446 (1997). [DOI] [PubMed] [Google Scholar]
- 55.Sachs G et al. , A nonelectrogenic H+ pump in plasma membranes of hog stomach. J Biol Chem 251, 7690–7698 (1976). [PubMed] [Google Scholar]
- 56.Olbe L, Carlsson E, Lindberg P, A proton-pump inhibitor expedition: the case histories of omeprazole and esomeprazole. Nature reviews drug discovery 2, 132–139 (2003). [DOI] [PubMed] [Google Scholar]
- 57.Marshall BJ, Warren JR, Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet 1, 1311–1315 (1984). [DOI] [PubMed] [Google Scholar]
- 58.Modlin IM, Sachs G, Wright N, Kidd M, Edkins and a century of acid suppression. Digestion 72, 129–145 (2005). [DOI] [PubMed] [Google Scholar]
- 59.Sørdal Ø et al. , The Gastrin Receptor Antagonist Netazepide (YF 476) Prevents Oxyntic Mucosal Inflammation Induced by Helicobacter Pylori Infection in Mongolian Gerbils. Helicobacter 18, 397–405 (2013). [DOI] [PubMed] [Google Scholar]
- 60.Dockray GJ, Moore A, Varro A, Pritchard DM, Gastrin receptor pharmacology. Current gastroenterology reports 14, 453–459 (2012). [DOI] [PubMed] [Google Scholar]
- 61.Dimaline R, Varro A, Novel roles of gastrin. The Journal of physiology 592, 2951–2958 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Blair AJ III, Richardson CT, Walsh JH, Feldman M, Variable contribution of gastrin to gastric acid secretion after a meal in humans. Gastroenterology 92, 944–949 (1987). [DOI] [PubMed] [Google Scholar]
- 63.Grozinsky-Glasberg S et al. , Metastatic type 1 gastric carcinoid: a real threat or just a myth? World journal of gastroenterology: WJG 19, 8687 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sagatun L et al. , Follow-up of patients with ECL cell-derived tumours. Scandinavian Journal of Gastroenterology 51, 1398–1405 (2016). [DOI] [PubMed] [Google Scholar]
- 65.Vannella L, Lahner E, Osborn J, Annibale B, Systematic review: gastric cancer incidence in pernicious anaemia. Alimentary pharmacology & therapeutics 37, 375–382 (2013). [DOI] [PubMed] [Google Scholar]
- 66.Mahmud N et al. , The incidence of neoplasia in patients with autoimmune metaplastic atrophic gastritis: a renewed call for surveillance. Annals of Gastroenterology 32, 67 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Winbo A, Sandström O, Palmqvist R, Rydberg A, Iron-deficiency anaemia, gastric hyperplasia, and elevated gastrin levels due to potassium channel dysfunction in the Jervell and Lange-Nielsen Syndrome. Cardiology in the Young 23, 325–334 (2013). [DOI] [PubMed] [Google Scholar]
- 68.Calvete O et al. , Exome sequencing identifies ATP4A gene as responsible of an atypical familial type I gastric neuroendocrine tumour. Human molecular genetics 24, 2914–2922 (2015). [DOI] [PubMed] [Google Scholar]
- 69.Brusselaers N, Wahlin K, Engstrand L, Lagergren J, Maintenance therapy with proton pump inhibitors and risk of gastric cancer: a nationwide population-based cohort study in Sweden. Bmj Open 7, e017739 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cheung KS et al. , Long-term proton pump inhibitors and risk of gastric cancer development after treatment for Helicobacter pylori: a population-based study. Gut 67, 28–35 (2018). [DOI] [PubMed] [Google Scholar]
- 71.Metz DC, Cadiot G, Poitras P, Ito T, Jensen RT, Diagnosis of Zollinger–Ellison syndrome in the era of PPIs, faulty gastrin assays, sensitive imaging and limited access to acid secretory testing. International journal of endocrine oncology 4, 167–185 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lee L, Ramos-Alvarez I, Ito T, Jensen RT, Insights into effects/risks of chronic hypergastrinemia and lifelong ppi treatment in man based on studies of patients with Zollinger–Ellison syndrome. International Journal of Molecular Sciences 20, 5128 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lloyd KA et al. , Gastrin-induced miR-222 promotes gastric tumor development by suppressing p27kip1. Oncotarget 7, 45462 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhang J et al. , miR-221/222 promote malignant progression of glioma through activation of the Akt pathway. International journal of oncology 36, 913–920 (2010). [DOI] [PubMed] [Google Scholar]
- 75.Zhong C, Ding S, Xu Y, Huang H, MicroRNA-222 promotes human non-small cell lung cancer H460 growth by targeting p27. International journal of clinical and experimental medicine 8, 5534 (2015). [PMC free article] [PubMed] [Google Scholar]
- 76.Quintavalle C et al. , miR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPμ. Oncogene 31, 858–868 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Matsuzaki J, Suzuki H, Role of MicroRNAs-221/222 in digestive systems. Journal of clinical medicine 4, 1566–1577 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fu Z et al. , Circulating miR-222 in plasma and its potential diagnostic and prognostic value in gastric cancer. Medical oncology 31, 164 (2014). [DOI] [PubMed] [Google Scholar]
- 79.Kim Y-K et al. , Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic acids research 37, 1672–1681 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Li N et al. , Increased miR-222 in H. pylori-associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK. FEBS letters 586, 722–728 (2012). [DOI] [PubMed] [Google Scholar]
- 81.Liu W et al. , miR-221 and miR-222 simultaneously target RECK and regulate growth and invasion of gastric cancer cells. Medical science monitor: international medical journal of experimental and clinical research 21, 2718 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Noto JM, Romero-Gallo J, Piazuelo MB, Peek RM, The Mongolian gerbil: a robust model of Helicobacter pylori-induced gastric inflammation and cancer. Gastrointestinal Physiology and Diseases: Methods and Protocols, 263–280 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Black JW, Kalindjian SB, Gastrin agonists and antagonists. Pharmacology & toxicology 91, 275–281 (2002). [DOI] [PubMed] [Google Scholar]
- 84.Herranz R, Cholecystokinin antagonists: pharmacological and therapeutic potential. Medicinal research reviews 23, 559–605 (2003). [DOI] [PubMed] [Google Scholar]
- 85.McDonald IM et al. , 2, 7-Dioxo-2, 3, 4, 5, 6, 7-hexahydro-1 H-benzo [h][1, 4] diazonine as a New Template for the Design of CCK2 Receptor Antagonists. Journal of medicinal chemistry 43, 3518–3529 (2000). [DOI] [PubMed] [Google Scholar]
- 86.Barrett T et al. , The cholecystokinin CCK2 receptor antagonist, JNJ-26070109, inhibits gastric acid secretion and prevents omeprazole-induced acid rebound in the rat. British journal of pharmacology 166, 1684–1693 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Nishida A et al. , Gastrin receptor antagonist YM022 prevents hypersecretion after long-term acid suppression. American Journal of Physiology-Gastrointestinal and Liver Physiology 269, G699–G705 (1995). [DOI] [PubMed] [Google Scholar]
- 88.Semple G et al. , (3 R)-N-(1-(tert-Butylcarbonylmethyl)-2, 3-dihydro-2-oxo-5-(2-pyridyl)-1 H-1, 4-benzodiazepin-3-yl)-N ‘-(3-(methylamino) phenyl) urea (YF476): A Potent and Orally Active Gastrin/CCK-B Antagonist. Journal of medicinal chemistry 40, 331–341 (1997). [DOI] [PubMed] [Google Scholar]
- 89.Takinami Y et al. , YF476 is a new potent and selective gastrin/cholecystokinin-B receptor antagonist in vitro and in vivo. Alimentary pharmacology & therapeutics 11, 113–120 (1997). [DOI] [PubMed] [Google Scholar]
- 90.Takemoto Y et al. , Effects of YF476, a potent and selective gastrin/cholecystokinin-B receptor antagonist, on gastric acid secretion in beagle dogs with gastric fistula. Arzneimittel-forschung 48, 403–407 (1998). [PubMed] [Google Scholar]
- 91.Chen D et al. , Effect of cholecystokinin-2 receptor blockade on rat stomach ECL cells. Cell and tissue research 299, 81–95 (2000). [DOI] [PubMed] [Google Scholar]
- 92.Deftos LJ, Chromogranin A: its role in endocrine function and as an endocrine and neuroendocrine tumor marker. Endocr Rev 12, 181–187 (1991). [DOI] [PubMed] [Google Scholar]
- 93.Zhao CM, Chen D, Lintunen M, Panula P, Hakanson R, Secretory organelles in ECL cells of the rat stomach: an immunohistochemical and electron-microscopic study. Cell Tissue Res 298, 457–470 (1999). [DOI] [PubMed] [Google Scholar]
- 94.Norlen P et al. , Expression of the chromogranin A-derived peptides pancreastatin and WE14 in rat stomach ECL cells. Regul Pept 70, 121–133 (1997). [DOI] [PubMed] [Google Scholar]
- 95.Hakanson R, Ding XQ, Norlen P, Chen D, Circulating pancreastatin is a marker for the enterochromaffin-like cells of the rat stomach. Gastroenterology 108, 1445–1452 (1995). [DOI] [PubMed] [Google Scholar]
- 96.Chen D et al. , Gastrin-evoked secretion of pancreastatin and histamine from ECL cells and of acid from parietal cells in isolated, vascularly perfused rat stomach. Effects of isobutyl methylxanthin and alpha-fluoromethylhistidine. Regul Pept 65, 133–138 (1996). [DOI] [PubMed] [Google Scholar]
- 97.Kimura K, Chen D, Lindstrom E, Zhao CM, Hakanson R, Evidence that rat stomach ECL cells represent the main source of circulating pancreastatin. Regul Pept 68, 177–180 (1997). [DOI] [PubMed] [Google Scholar]
- 98.Lindstrom E et al. , Neurohormonal regulation of histamine and pancreastatin secretion from isolated rat stomach ECL cells. Regul Pept 71, 73–86 (1997). [DOI] [PubMed] [Google Scholar]
- 99.Zhao CM, Jacobsson G, Chen D, Hakanson R, Meister B, Exocytotic proteins in enterochromaffin-like (ECL) cells of the rat stomach. Cell Tissue Res 290, 539–551 (1997). [DOI] [PubMed] [Google Scholar]
- 100.Fakhry J et al. , Relationships of endocrine cells to each other and to other cell types in the human gastric fundus and corpus. Cell Tissue Res 376, 37–49 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lindström E et al. , Neurohormonal regulation of histamine and pancreastatin secretion from isolated rat stomach ECL cells. Regulatory peptides 71, 73–86 (1997). [DOI] [PubMed] [Google Scholar]
- 102.Kimura K, Chen D, Lindström E, Zhao C-M, Håkanson R, Evidence that rat stomach ECL cells represent the main source of circulating pancreastatin. Regulatory peptides 68, 177–180 (1997). [DOI] [PubMed] [Google Scholar]
- 103.Håkanson R, Ding X-Q, Norlén P, Chen D, Circulating pancreastatin is a marker for the enterochromaffin-like cells of the rat stomach. Gastroenterology 108, 1445–1452 (1995). [DOI] [PubMed] [Google Scholar]
- 104.Norlén P et al. , Expression of the chromogranin A-derived peptides pancreastatin and WE14 in rat stomach ECL cells. Regulatory peptides 70, 121–133 (1997). [DOI] [PubMed] [Google Scholar]
- 105.Chen D et al. , Gastrin-evoked secretion of pancreastatin and histamine from ECL cells and of acid from parietal cells in isolated, vascularly perfused rat stomach. Effects of isobutyl methylxanthin and α-fluoromethylhistidine. Regulatory peptides 65, 133–138 (1996). [DOI] [PubMed] [Google Scholar]
- 106.Chen D et al. , Acute responses of rat stomach enterochromaffinlike cells to gastrin: secretory activation and adaptation. Gastroenterology 107, 18–27 (1994). [DOI] [PubMed] [Google Scholar]
- 107.Sanduleanu S et al. , Serum chromogranin A as a screening test for gastric enterochromaffin-like cell hyperplasia during acid-suppressive therapy. European Journal of Clinical Investigation 31, 802–811 (2001). [DOI] [PubMed] [Google Scholar]
- 108.Rindi G et al. , Vesicular monoamine transporter 2 as a marker of gastric enterochromaffin-like cell tumors. Virchows Archiv 436, 217–223 (2000). [DOI] [PubMed] [Google Scholar]
- 109.M Zhao C, Chen D, The ECL cell: relay station for gastric integrity. Current medicinal chemistry 19, 98–108 (2012). [DOI] [PubMed] [Google Scholar]
- 110.Zhao C-M, Chen D, Lintunen M, Panula P, Håkanson R, Secretory organelles in ECL cells of the rat stomach: an immunohistochemical and electron-microscopic study. Cell and tissue research 298, 457–470 (1999). [DOI] [PubMed] [Google Scholar]
- 111.Erickson JD, Schafer M, Bonner TI, Eiden LE, Weihe E, Distinct pharmacological properties and distribution in neurons and endocrine cells of two isoforms of the human vesicular monoamine transporter. Proceedings of the National Academy of Sciences 93, 5166–5171 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zhao C-M, Chen D, Lintunen M, Panula P, Håkanson R, Effects of reserpine on ECL-cell ultrastructure and histamine compartmentalization in the rat stomach. Cell and tissue research 295, 131–140 (1999). [DOI] [PubMed] [Google Scholar]
- 113.Murphy MG et al. , The gastrin-receptor antagonist L-365,260 inhibits stimulated acid secretion in humans. Clinical Pharmacology & Therapeutics 54, 533–539 (1993). [DOI] [PubMed] [Google Scholar]
- 114.Boyce M et al. , Single oral doses of netazepide (YF 476), a gastrin receptor antagonist, cause dose-dependent, sustained increases in gastric pH compared with placebo and ranitidine in healthy subjects. Alimentary pharmacology & therapeutics 36, 181–189 (2012). [DOI] [PubMed] [Google Scholar]
- 115.Boyce M, Warrington S, Black J, Netazepide, a gastrin/CCK2 receptor antagonist, causes dose-dependent, persistent inhibition of the responses to pentagastrin in healthy subjects. British Journal of Clinical Pharmacology 76, 689–698 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Boyce M et al. , Effect of netazepide, a gastrin/CCK2 receptor antagonist, on gastric acid secretion and rabeprazole-induced hypergastrinaemia in healthy subjects. British journal of clinical pharmacology 79, 744–755 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Boyce M, van den Berg F, Mitchell T, Darwin K, Warrington S, Randomised trial of the effect of a gastrin/CCK2 receptor antagonist on esomeprazole-induced hypergastrinaemia: evidence against rebound hyperacidity. European journal of clinical pharmacology 73, 129–139 (2017). [DOI] [PubMed] [Google Scholar]
- 118.Boyce M, Lloyd KA, Pritchard DM, Potential clinical indications for a CCK2 receptor antagonist. Current Opinion in Pharmacology 31, 68–75 (2016). [DOI] [PubMed] [Google Scholar]
- 119.Boyce MJ, Thomsen L, Gilbert DA, Wood D. (Google Patents, 2021). [Google Scholar]
- 120.Long J, Properties of SCH 28080: A novel compound with gastric antisecretory and cytoprotective properties. Gastroenterology 80, 1216 (1981). [Google Scholar]
- 121.Chiu P, Casciano C, Tetzloff G, Long J, Barnett A, Studies on the mechanisms of the antisecretory and cytoprotective actions of SCH 28080. Journal of Pharmacology and Experimental Therapeutics 226, 121–125 (1983). [PubMed] [Google Scholar]
- 122.Long J, P. s. Chiu, M. Derelanko, M. Steinberg, Gastric antisecretory and cytoprotective activities of SCH 28080. Journal of Pharmacology and Experimental Therapeutics 226, 114–120 (1983). [PubMed] [Google Scholar]
- 123.Ene M, Khan-Daneshmend T, Roberts C, A study of the inhibitory effects of SCH 28080 on gastric secretion in man. British Journal of Pharmacology 76, 389 (1982). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Beil W, Hackbarth I, Sewing K-F, Mechanism of gastric antisecretory effect of SCH 28080. British journal of pharmacology 88, 19 (1986). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Wallmark B et al. , Inhibition of gastric H+, K+-ATPase and acid secretion by SCH 28080, a substituted pyridyl (1, 2a) imidazole. J Biol Chem 262, 2077–2084 (1987). [PubMed] [Google Scholar]
- 126.Murakami K et al. , Vonoprazan, a novel potassium-competitive acid blocker, as a component of first-line and second-line triple therapy for Helicobacter pylori eradication: a phase III, randomised, double-blind study. Gut 65, 1439–1446 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hori Y et al. , 1-[5-(2-Fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine monofumarate (TAK-438), a novel and potent potassium-competitive acid blocker for the treatment of acid-related diseases. Journal of Pharmacology and Experimental Therapeutics 335, 231–238 (2010). [DOI] [PubMed] [Google Scholar]
- 128.Shin JM et al. , Characterization of a novel potassium-competitive acid blocker of the gastric H, K-ATPase, 1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine monofumarate (TAK-438). Journal of Pharmacology and Experimental Therapeutics 339, 412–420 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Yang E et al. , Night-time gastric acid suppression by tegoprazan compared to vonoprazan or esomeprazole. British Journal of Clinical Pharmacology, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sakurai Y et al. , Safety, tolerability, pharmacokinetics, and pharmacodynamics of single rising TAK-438 (vonoprazan) doses in healthy male Japanese/non-Japanese subjects. Clinical and translational gastroenterology 6, e94 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Jenkins H et al. , Randomised clinical trial: safety, tolerability, pharmacokinetics and pharmacodynamics of repeated doses of TAK-438 (vonoprazan), a novel potassium-competitive acid blocker, in healthy male subjects. Alimentary pharmacology & therapeutics 41, 636–648 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Simadibrata DM, Syam AF, Lee YY, A comparison of efficacy and safety of potassium-competitive acid blocker and proton pump inhibitor in gastric acid-related diseases: A systematic review and meta-analysis. J Gastroen Hepatol, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Chen S et al. , The efficacy and safety of keverprazan, a novel potassium-competitive acid blocker, in treating erosive oesophagitis: A phase III, randomised, double-blind multicentre study. Alimentary Pharmacology & Therapeutics, (2022). [DOI] [PubMed] [Google Scholar]
- 134.Chen S, Chen M, Xiao Y. (Wiley Online Library, 2022), vol. 56, pp. 1072–1073. [Google Scholar]
- 135.Kojima Y et al. , Does the novel potassium-competitive acid blocker vonoprazan cause more hypergastrinemia than conventional proton pump inhibitors? A multicenter prospective cross-sectional study. Digestion 97, 70–75 (2018). [DOI] [PubMed] [Google Scholar]
- 136.Takahashi N, Take Y, Tegoprazan, a novel potassium-competitive acid blocker to control gastric acid secretion and motility. Journal of Pharmacology and Experimental Therapeutics 364, 275–286 (2018). [DOI] [PubMed] [Google Scholar]
- 137.Sunwoo J et al. , Safety, tolerability, pharmacodynamics and pharmacokinetics of DWP 14012, a novel potassium-competitive acid blocker, in healthy male subjects. Alimentary pharmacology & therapeutics 48, 206–218 (2018). [DOI] [PubMed] [Google Scholar]
- 138.Jeong Y-S et al. , Development of Physiologically Based Pharmacokinetic Model for Orally Administered Fexuprazan in Humans. Pharmaceutics 13, 813 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Abe K, Irie K, Nakanishi H, Suzuki H, Fujiyoshi Y, Crystal structures of the gastric proton pump. Nature 556, 214–218 (2018). [DOI] [PubMed] [Google Scholar]
- 140.Abdel-Aziz Y, Metz DC, Howden CW, potassium-competitive acid blockers for the treatment of acid-related disorders. Alimentary Pharmacology & Therapeutics 53, 794–809 (2021). [DOI] [PubMed] [Google Scholar]
- 141.DeVault KR, potassium-competitive acid blockers-is there a role for more complete acid suppression? Alimentary Pharmacology & Therapeutics 56, 1071–1071 (2022). [DOI] [PubMed] [Google Scholar]
- 142.Scarpignato C et al. , Pharmacologic treatment of GERD: Where we are now, and where are we going? Annals of the New York Academy of Sciences 1482, 193–212 (2020). [DOI] [PubMed] [Google Scholar]
- 143.Anlauf M, Schäfer MKH, Eiden L, Weihe E, Chemical coding of the human gastrointestinal nervous system: cholinergic, VIPergic, and catecholaminergic phenotypes. Journal of Comparative Neurology 459, 90–111 (2003). [DOI] [PubMed] [Google Scholar]
- 144.Reglodi D et al. , Presence and effects of pituitary adenylate cyclase activating polypeptide under physiological and pathological conditions in the stomach. Front Endocrinol 9, 90 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Brzozowska M, Całka J, Occurrence and distribution of galanin in the physiological and inflammatory states in the mammalian gastrointestinal tract. Front Immunol 11, 602070 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ekblad E, Rökaeus Å, Sundler F, Galanin nerve fibers in the rat gut: distribution, origin and projections. Neuroscience 16, 355–363 (1985). [DOI] [PubMed] [Google Scholar]
- 147.Chen D, Kodama Y, Kulseng B, Johannessen H, Zhao CM. , Kastin Galanin. A, Ed., Handbook of Biologically Actively Peptides (Elsevier, 2013), vol. 2nd Edition, pp. 1210–1217. [Google Scholar]
- 148.Tao J et al. , Highly selective brain-to-gut communication via genetically defined vagus neurons. Neuron 109, 2106–2115. e2104 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Koh TJ et al. , Gastrin deficiency results in altered gastric differentiation and decreased colonic proliferation in mice. Gastroenterology 113, 1015–1025 (1997). [DOI] [PubMed] [Google Scholar]
- 150.Friis-Hansen L et al. , Impaired gastric acid secretion in gastrin-deficient mice. Am J Physiol 274, G561–568 (1998). [DOI] [PubMed] [Google Scholar]
- 151.Chen D et al. , Glycine-extended gastrin synergizes with gastrin 17 to stimulate acid secretion in gastrin-deficient mice. Gastroenterology 119, 756–765 (2000). [DOI] [PubMed] [Google Scholar]
- 152.Zhao CM et al. , Chronic Helicobacter pylori infection results in gastric hypoacidity and hypergastrinemia in wild-type mice but vagally induced hypersecretion in gastrin-deficient mice. Regul Pept 115, 161–170 (2003). [DOI] [PubMed] [Google Scholar]
- 153.Chen D, Zhao CM, Hakanson R, Rehfeld JF, Gastric phenotypic abnormality in cholecystokinin 2 receptor null mice. Pharmacol Toxicol 91, 375–381 (2002). [DOI] [PubMed] [Google Scholar]
- 154.Chen D et al. , Differentiation of gastric ECL cells is altered in CCK(2) receptor-deficient mice. Gastroenterology 123, 577–585 (2002). [DOI] [PubMed] [Google Scholar]
- 155.Kanai S et al. , Gastric acid secretion in cholecystokinin-1 receptor, −2 receptor, and −1, −2 receptor gene knockout mice. J Physiol Sci 59, 23–29 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zhao CM et al. , Gene expression profiling of gastric mucosa in mice lacking CCK and gastrin receptors. Regul Pept 192–193, 35–44 (2014). [DOI] [PubMed] [Google Scholar]
- 157.Aihara T, Nakamura Y, Taketo MM, Matsui M, Okabe S, Cholinergically stimulated gastric acid secretion is mediated by M(3) and M(5) but not M(1) muscarinic acetylcholine receptors in mice. Am J Physiol Gastrointest Liver Physiol 288, G1199–1207 (2005). [DOI] [PubMed] [Google Scholar]
- 158.Tao J et al. , Highly selective brain-to-gut communication via genetically defined vagus neurons. Neuron 109, 2106–2115 e2104 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Aihara T et al. , Impaired gastric secretion and lack of trophic responses to hypergastrinemia in M3 muscarinic receptor knockout mice. Gastroenterology 125, 1774–1784 (2003). [DOI] [PubMed] [Google Scholar]
- 160.Takeuchi K, Endoh T, Hayashi S, Aihara T, Activation of muscarinic acetylcholine receptor subtype 4 is essential for cholinergic stimulation of gastric acid secretion: relation to D cell/somatostatin. Frontiers in Pharmacology 7, 278 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Spicer Z et al. , Stomachs of mice lacking the gastric H,K-ATPase alpha -subunit have achlorhydria, abnormal parietal cells, and ciliated metaplasia. J Biol Chem 275, 21555–21565 (2000). [DOI] [PubMed] [Google Scholar]
- 162.Roepke TK et al. , The KCNE2 potassium channel ancillary subunit is essential for gastric acid secretion. J Biol Chem 281, 23740–23747 (2006). [DOI] [PubMed] [Google Scholar]
- 163.Chandra M et al. , A role for the Ca2+ channel TRPML1 in gastric acid secretion, based on analysis of knockout mice. Gastroenterology 140, 857–867 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Recalde S et al. , Inefficient chronic activation of parietal cells in Ae2a,b(−/−) mice. Am J Pathol 169, 165–176 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Gawenis LR et al. , Mice with a targeted disruption of the AE2 Cl-/HCO3- exchanger are achlorhydric. J Biol Chem 279, 30531–30539 (2004). [DOI] [PubMed] [Google Scholar]
- 166.Gawenis LR et al. , Impaired gastric acid secretion in mice with a targeted disruption of the NHE4 Na+/H+ exchanger. J Biol Chem 280, 12781–12789 (2005). [DOI] [PubMed] [Google Scholar]
- 167.Zhao CM et al. , Control of gastric acid secretion in somatostatin receptor 2 deficient mice: shift from endocrine/paracrine to neurocrine pathways. Endocrinology 149, 498–505 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Piqueras L, Tache Y, Martinez V, Peripheral PACAP inhibits gastric acid secretion through somatostatin release in mice. Br J Pharmacol 142, 67–78 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Piqueras L, Tache Y, Martinez V, Galanin inhibits gastric acid secretion through a somatostatin-independent mechanism in mice. Peptides 25, 1287–1295 (2004). [DOI] [PubMed] [Google Scholar]
- 170.Lu Y, Germano P, Ohning GV, Vu JP, Pisegna JR, PAC1 deficiency in a murine model induces gastric mucosa hypertrophy and higher basal gastric acid output. J Mol Neurosci 43, 76–84 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Nakamura E et al. , Lack of histamine alters gastric mucosal morphology: comparison of histidine decarboxylase-deficient and mast cell-deficient mice. Am J Physiol Gastrointest Liver Physiol 287, G1053–1061 (2004). [DOI] [PubMed] [Google Scholar]
- 172.Kobayashi T et al. , Abnormal functional and morphological regulation of the gastric mucosa in histamine H2 receptor-deficient mice. J Clin Invest 105, 1741–1749 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Fukushima Y et al. , Structural and functional characterization of gastric mucosa and central nervous system in histamine H2 receptor-null mice. Eur J Pharmacol 468, 47–58 (2003). [DOI] [PubMed] [Google Scholar]
- 174.Seino S, Shibasaki T, PKA-dependent and PKA-independent pathways for cAMP-regulated exocytosis. Physiological reviews 85, 1303–1342 (2005). [DOI] [PubMed] [Google Scholar]
- 175.Li Z-Q, Mårdh S, Interactions between Ca2+-and cAMP-dependent stimulatory pathways in parietal cells. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 1311, 133–142 (1996). [DOI] [PubMed] [Google Scholar]
- 176.Athmann C, Zeng N, Scott DR, Sachs G, Regulation of parietal cell calcium signaling in gastric glands. American Journal of Physiology-Gastrointestinal and Liver Physiology 279, G1048–G1058 (2000). [DOI] [PubMed] [Google Scholar]
- 177.Schinke T et al. , Impaired gastric acidification negatively affects calcium homeostasis and bone mass. Nat Med 15, 674–681 (2009). [DOI] [PubMed] [Google Scholar]
- 178.Fitzpatrick LA et al. , Targeted deletion of histidine decarboxylase gene in mice increases bone formation and protects against ovariectomy-induced bone loss. Proc Natl Acad Sci U S A 100, 6027–6032 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Chen D, Zhao C-M, The possible existence of a gut-bone axis suggested by studies of genetically manipulated mouse models? Current pharmaceutical design 17, 1552–1555 (2011). [DOI] [PubMed] [Google Scholar]
- 180.de Vos WM, Tilg H, Van Hul M, Cani PD, Gut microbiome and health: mechanistic insights. Gut 71, 1020–1032 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Fan Y, Pedersen O, Gut microbiota in human metabolic health and disease. Nature Reviews Microbiology 19, 55–71 (2021). [DOI] [PubMed] [Google Scholar]
- 182.Sanna S, Kurilshikov A, van der Graaf A, Fu J, Zhernakova A, Challenges and future directions for studying effects of host genetics on the gut microbiome. Nature genetics 54, 100–106 (2022). [DOI] [PubMed] [Google Scholar]
- 183.Kuziel GA, Rakoff-Nahoum S, The gut microbiome. Current Biology 32, R257–R264 (2022). [DOI] [PubMed] [Google Scholar]
- 184.Miyauchi E, Shimokawa C, Steimle A, Desai MS, Ohno H, The impact of the gut microbiome on extra-intestinal autoimmune diseases. Nature Reviews Immunology 23, 9–23 (2023). [DOI] [PubMed] [Google Scholar]
- 185.O’Donnell JA, Zheng T, Meric G, Marques FZ, The gut microbiome and hypertension. Nature Reviews Nephrology, 1–15 (2023). [DOI] [PubMed] [Google Scholar]