Gastric secretion : Current Opinion in Gastroenterology (2024)

Introduction

Gastric acid secretion by parietal cells is a highly intricate and dynamic process regulated by neural (efferent and afferent), hormonal (gastrin), and paracrine (histamine and somatostatin) pathways as well as mechanical (distension) and chemical (protein, glutamate, coffee, and ethanol) stimuli. Histamine, secreted from nearby enterochromaffin-like (ECL) cells, diffuses to parietal cells where it activates histamine-2 (H-2) receptors coupled to generation of intracellular adenosine 3′,5′-cyclic monophosphate (cAMP) and subsequent activation of H+K+-adenosine triphosphatase (H+K+-ATPase), the proton pump.

Parietal cells have a limited life span (54 days in mice) and are continuously renewed from stem cells anchored in the isthmus region of the gastric glands [1•]. The stem cells give rise to three main progenitors: prepit, preneck, and preparietal cells. Parietal cells develop either directly from the preparietal cells or less commonly via differentiation of the prepit and preneck cell progenitors. The formation of a parietal cell involves a process of differentiation whereby there is production of cytoplasmic tubulovesicles, an increase in the number and length of microvilli, an increase in number and size of mitochondria, and expansion and invagin*tion of the apical membrane with the formation of a canalicular system. It has been calculated that six parietal cells are produced in one isthmus each month. The most robust acid-producing parietal cells are located in the isthmus whereas the cells become more senescent as they migrate downward towards the base.

Improved understanding of the regulation of gastric acid secretion led to the development of acid-suppressive therapies, histamine H-2 receptor antagonists in the 1970s, and proton pump inhibitors (PPIs) in the 1980s, which have revolutionized the treatment of acid-related disorders including gastroesophageal reflux disease and peptic ulcer disease. More recently, concerns have been raised regarding potential risks of profound acid suppression including hypergastrinemia, rebound acid secretion, fundic gland polyps, infection, osteoporosis, and alterations in drug absorption [2]. Murray et al.[3•] report that ex-vivo addition of PPIs (omeprazole, esomeprazole, and lansoprazole) to isolated rat gastric corpus increases transmucosal permeability in a dose-dependent manner and allows solutes at least as large as 4000 Da (i.e., polyethylene glycol) to permeate. The precise location and mechanism of the leak as well as the clinical consequences are not known. The authors postulate that PPI-induced unregulated transit of peptides and proteins (e.g., luminal epidermal growth factor, bacterial antigens, and food antigens) may potentially contribute to neoplasia. They further posit that increased diffusion of certain drugs may be deleterious (e.g., digoxin) or beneficial (e.g., antibiotics used to eradicate Helicobacter pylori). It should be noted that the absorption of certain drugs, such as levothyroxin, which is enhanced in an acid environment, is decreased during PPI treatment, despite the apparent leak [4].

Parietal cells secrete hydrochloric acid (HCl) as well as intrinsic factor, transforming growth factor-α, amphiregulin, heparin-binding epidermal growth factor-like growth factor, and sonic hedgehog [5,6]. Autoimmune gastritis is an autoimmune disease characterized by loss of parietal cells and consequently achlorhydria, hypergastrinemia, iron deficiency, and, in late stages, cobalamin deficiency. Autoantibodies directed against the parietal cell H+K+-ATPase (i.e., parietal cell antibodies) and against intrinsic factor are found in 80–90% and 30–50%, respectively; the specificity of intrinsic factor antibodies approaches 100% [7]. The prevalence of autoimmune gastritis is ∼2% in the general population but increased three to five-fold in patients with other autoimmune disorders such as type I diabetes, thyroid disease, and primary biliary cirrhosis [8].

Central regulation of gastric acid secretion

The cephalic phase of acid secretion is induced by sensory inputs driven by the thought, sight, smell, taste, and swallowing of food. It primes the stomach to assist digestion by contributing ∼50% to the overall acid response to a meal. Pavlov's pioneering work studying the influence of conditioned reflexes on salivary secretion has recently been applied to the study of gastric acid secretion. Caboclo et al.[9], using methodology that allows collection of gastric juice in rats in their habitual conditions (without restraining), paired sound as the conditioning stimulus with food as the unconditioning stimulus. After 10 days of conditioning, noise significantly increased acid secretion. The work confirms that interprandial secretion may be elicited physiologically by cortical factors such as memory-dependent learning behavior. Such secretion could conceivably play a role in peptic ulcer disease.

Peripheral regulation of gastric acid secretion

A variety of neurocrine, paracrine, and endocrine signals as well as luminal chemicals regulate gastric acid secretion such as gastrin, histamine, ghrelin, glutamate, coffee, somatostatin, and ethanol [10,11].

Stimulants

The major stimulants of acid secretion are histamine, gastrin, and acetylcholine [10]. Histamine, released from neighboring ECL cells, stimulates the parietal cell directly via H-2 receptors. Gastrin, released from antral G cells, stimulates the parietal cell mainly indirectly via cholecystokinin-2 (CCK-2) receptors on ECL cells coupled to histamine release. Acetylcholine, released from postganglionic neurons, stimulates the parietal cell directly via M3 receptors and indirectly via M2 and M4 receptors coupled to inhibition of somatostatin secretion. Other stimulants include ghrelin, glutamate, and coffee.

Histamine

Histamine, like gastrin, may act not only as a secretagogue but also as a mucosal growth factor. Histidine decarboxylase-deficient mice, which lack endogenous histamine synthesis, exhibit alterations in chief cell differentiation [12]. The effect of histamine loss is, in part, mediated by the reciprocal increase in gastrin secretion induced by the decrease in luminal acidity since breeding of histidine decarboxylase-deficient mice onto the gastrin-null background ameliorates the premature chief cell differentiation phenotype. On the other hand, knockout of the H-2 receptor produces a different phenotype without alteration in chief cell lineages, despite an increase in gastrin secretion, suggesting that histamine may exert some of its influence via another histamine receptor, perhaps the H-3 receptor [13].

Gastrin

Gastrin, a hormone released from antral G cells, stimulates gastric acid secretion and mucosal cell growth. Its actions are mediated primarily through the CCK-2 receptor, a G protein-coupled receptor previously termed the CCKB or gastrin receptor. It is currently believed that gastrin stimulates the ECL cells to release histamine, which, in turn, binds to H-2 receptors on parietal cells to stimulate acid secretion [14].

Gastrin is secreted in response to chemical (e.g., elevated pH and protein) and mechanical (e.g., distension) stimuli that act directly on the G cell and/or indirectly via adjacent neuroendocrine cells and neurons. Capsaicin, the pungent ingredient in hot chilli peppers, is known to excite sensory neurons by binding the vanilloid receptor-1 (VR1). Ericson et al.[15•], using isolated human antral glands, localized VR1 to G cells and showed that capsaicin stimulates gastrin secretion. Similar results were reported by Kidd et al.[16] using isolated rodent G cells. Gastrin release was also stimulated by pituitary adenylate cyclase-activating protein, bombesin, sucralose, glucose, caffeine, and bacterial lipopolysaccharide. Whether luminal substances, in intact organisms, can actually reach and interact with specific gastrin cell surface receptors, however, has not been proven.

Hypergastrinemia, defined as a fasting serum gastrin concentration of >100 pg/ml or >47.7 pM, is frequently encountered in patients. Causes include gastrinoma, antral predominant H. pylori gastritis, gastric outlet obstruction, renal failure, retained antrum, atrophic gastritis, and antisecretory therapy [17].

Acute infection with H. pylori is associated with hypochlorhydria. The mechanisms by which H. pylori inhibits acid secretion are multifactorial and include production of the proinflammatory cytokine interleukin 1β. In gastric carcinoma cells, interleukin 1β inhibits gastrin secretion at the transcriptional level, in part via activation of NFκB [18].

Ghrelin

Although ghrelin has been reported to be present in A-like or Gr cells located in the basal portion of oxyntic glands, a recent study reports its presence abundantly in pyloric glands, as well [19]. Gr cells account for 20–30% of all gastric endocrine cells and the mammalian stomach produces 60–80% of the body's ghrelin. Alternate splicing of the preproghrelin yields ghrelin, des-acyl ghrelin, and obestatin [20]. The major active form of human ghrelin is a 28-amino acid peptide modified by octanoyl esterification of a serine residue at position three of the peptide chain. The acyl transferase enzyme responsible for this modification is ghrelin O-acyltransferase (GOAT) [21]. Using a custom-made anti-GOAT antibody, Stengel et al.[22] report that 95% of GOAT-immunoreactive cells in mice co-labeled with ghrelin whereas in rats only 56% of GOAT-positive cells showed co-expression of ghrelin; the remainder, in rat, co-expressed histidine decarboxylase. Similar findings in mice were reported by Sakata et al.[23].

In the gastrointestinal tract, ghrelin increases appetite and stimulates motility and gastric emptying [24]. Following bariatric surgery, in particular procedures that involve gastric resection or bypass, ghrelin levels are reduced [25]. In rat weanling pups, vitamin B12 deficiency is associated with decreased plasma ghrelin levels and lowered body weights [26]. In piglets, zinc supplementation increased plasma concentration of ghrelin as well as food intake and growth [27].

Iwakura et al.[28] have established a ghrelin-producing cell line, MGN3-1, from a mouse gastric ghrelin-producing tumor. This cell line, which contains GOAT and responds appropriately to somatostatin, may prove useful for studying the production and secretion of ghrelin.

It has been postulated that certain gastric epithelial cells may sense luminal nutrients and function similar to taste receptor cells. Hass et al.[29•] have identified taste receptors of the subtype T1R3, which detect sugar and amino acids, on closed type ghrelin cells and adjacent open type brush cells in mice. This is interesting since ghrelin secretion is decreased by food intake, in particular proteins and carbohydrates. Whether food constituents regulate ghrelin secretion indirectly via paracrine signals emanating from adjacent brush cells or from serum concentrations of amino acids and glucose remains to be investigated.

Glutamate

Using elutriation and gradient centrifugation to enrich various cell fractions from isolated rat gastric mucosa, Nakamura et al.[30] show that parietal cells express the metabotropic glutamate receptors mGluR1, mGluR2, mGluR3, mGluR4, mGluR6, and mGluR7. Smaller endocrine cells, possibly somatostatin-containing D cells, expressed mGluR2, 3, 4, and 7, although expression was low. A decrease in somatostatin secretion may be one mechanism whereby glutamate stimulates acid secretion in dogs equipped with vagally innervated or vagally denervated gastric pouches [31].

Coffee

As coffee stimulates gastric acid secretion and is associated with peptic ulcer and gastroesophageal reflux disease, work has been directed to developing a ‘stomach-friendly’ coffee. Weiss et al.[32] at the German Research Center for Food Chemistry developed a flow cytometric method for measuring the intracellular pH of the human parietal cell line HGT-1 using the pH sensitive dye, 1,5-carboxyseminaphtorhodafluor acetoxymethyl ester (SNARF-AM). Treatment with histamine resulted in an 8% decrease in intracellular proton concentration, indicating increased proton efflux whereas treatment with omeprazole resulted in a 22% increase in intracellular protons. Steam treatment prior to roasting reduced the ability of both caffeinated and decaffeinated coffee to stimulate acid secretion in this preparation.

Inhibitors

Inhibitors include somatostatin, ethanol, and cadmium.

Somatostatin

Somatostatin is present in D cells of the gastric oxyntic and pyloric mucosa. It inhibits acid secretion in a paracrine fashion directly by inhibiting secretion from the parietal cell as well as indirectly by inhibiting histamine secretion from the ECL cell and gastrin secretion from the G cell.

The biological actions of somatostatin are mediated via six G protein-coupled receptors encoded by five different genes, termed sst1–sst5. The sst2 somatostatin receptor exists in two splice variants sst2A and sst2B [33]. The sst2 receptor is the most widely distributed somatostatin receptor subtype and is the subtype involved in the regulation of gastric acid secretion. In isolated vascularly perfused mouse stomach, exogenous adenosine, >1 μM, stimulates somatostatin secretion via the A2A receptor whereas at lower concentrations it inhibits somatostatin secretion via the A1 receptor [34].

Alcohol

Alcoholic beverages stimulate acid secretion but the effect of pure alcohol is controversial with most, but not all, studies showing inhibition. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is activated by an increase in the AMP/ATP-ratio and functions as a cellular energy sensory that prevents ATP-depletion of the cell by down-regulating energy consuming key enzymes and inducing pathways that generate ATP. An increase in AMPK inhibits acid secretion in mouse stomach [35]. In isolated rat gastric glands loaded with the pH-sensitive dye BCECF, low-dose (2%) ethanol inhibits secretagogue-dependent acid secretion by activation of the AMPK pathway [36•].

Cadmium

Cadmium, ingested through contaminated food and water, is a highly toxic nonessential heavy metal. In amphibian oxyntic mucosa mounted in Ussing chamber, addition of cadmium to the serosal, but not luminal side, inhibits histamine-stimulated acid secretion [37]. The precise mechanism, however, is not known.

Intracellular regulation of gastric acid secretion

During acid secretion, there is a morphological transformation of the parietal cell with translocation of cytoplasmic tubulovesicles containing the acid-secreting pump (H+K+-ATPase) to the apical membrane where it transports protons out of the cell in exchange for luminal potassium at the expense of a molecule of ATP. Although H+K+-ATPase is often considered the most important contributor for HCl secretion into the lumen, chloride secretion and potassium recycling are equally necessary.

H+K+-adenosine triphosphatase

The parietal cell secretes isotonic HCl against a huge gradient (greater than 106-fold) by ATP-driven exchange of one H+ for one K+ at the apical plasma membrane via the enzyme known as H+K+-ATPase [38••]. The H+K+-ATPase is composed of a catalytic α-subunit and an accessory β-subunit [39]. In the resting state, the bulk of H+K+-ATPase is sequestered within cytoplasmic tubulovesicles and is inactive (Fig. 1). Stimulation of acid secretion effects a recruitment and fusion of the tubulovesicles with the apical membrane and electrogenic H+ transport. A number of proteins have been implicated in parietal cell membrane targeting, docking, and fusion including soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) such as VAMP, syntaxins 1 and 3, and SNAP25 as well as Rab GTPases such as rab11. Other participating proteins include dynamin, F-actin, and ezrin.

PPIs are prodrugs that become locally activated within the acid environment of the apical canaliculus and covalently bind extracellular cysteine residues of the H+K+-ATPase that are accessible from the gastric lumen. Thus, only the proton pumps inserted into the apical membrane are susceptible to blockade by PPIs and the covalent binding mechanism permits PPIs to continue to act long after their plasma concentrations have dropped [40••].

Clotrimazole, an imidazole derivative antimycotic drug, has been shown to inhibit gastric H+K+-ATPase derived from hog gastric mucosa [41]. As there is a loss of polarity in this preparation, it would be useful to repeat the studies in intact stomach.

Channels and transporters

Acid secretion by the parietal cell requires a functional H+K+-ATPase as well as apical chloride and potassium channels [40••] (Fig. 1). Chloride enters the cell via the basolateral membrane chloride channel, SLC26A7, as well as the Cl/HCO3 exchanger, SLC4A2, and the Na+-2ClK+ cotransporter, NKCCl (Fig. 1) [42]. Chloride is thought to exit the apical membrane via the cystic fibrosis transmembrane regulator (CFTR). The CFTR is present in parietal cells and small-molecule CFTR inhibitors as well as mutations in the CFTR decrease gastric acid secretion. An additional route for chloride secretion may involve the Cl/HCO3 exchanger, SLC26A9; it, too, has been localized to the apical membrane of parietal cells and knockout of this exchanger impairs acid secretion.

Potassium secretion into the gastric lumen provides substrate for reciprocal proton secretion and is necessary for maintaining sustained H+K+-ATPase activity. The main apical potassium channel is KCNQ1 [43] (Fig. 1). KCNQ1 assembles with its β-subunit KCNE2 to function as a constitutively open, voltage-insensitive, and acid-resistant luminal potassium channel. Other luminal potassium channels may include members of the Kir family (e.g., Kir4.1) which are trafficked together with H+K+-ATPase to the apical membrane upon stimulation.

Signaling

Two parietal cell intracellular signaling pathways key to acid secretion are the cAMP-mediated protein kinase activation (PKA) pathway induced by histaminergic activation of the H-2 receptor and the intracellular calcium pathway induced by cholinergic activation of the M3 receptor and gastrin activation of the CCK2 receptor. Intracellular levels of cAMP are, in part, also regulated by phosphodiesterase-4 which hydrolyzes cAMP into inactive metabolites [44] and phosphoinositol-3-kinase (P13K) which activates protein kinase B/Akt and inhibits cAMP formation [45].

Acid secretion is highly energy dependent. The parietal cell is packed with abundant mitochondria to ensure an adequate supply of ATP. A recent study suggests that AMPK may function as a metabolic energy regulator to switch off acid secretion as cellular ATP levels fall [35]. As AMPK is a known modulator of the CFTR in other tissue, it is conceivable that it inhibits acid secretion by inhibiting the function of the apical chloride channel, CFTR. As discussed above, low-dose ethanol inhibits acid secretion via activation of the AMPK pathway [36•].

Risks of profound acid suppression

PPIs are the second most prescribed class of drugs in the United States and, although remarkably well tolerated, concerns have been raised recently regarding potential serious clinical adverse effects. The physiological consequence of acid suppression is hypergastrinemia. Gastrin is not only a secretagogue but also exerts growth promoting effects in normal and neoplastic tissues. In the stomach, hypergastrinemia results in parietal and ECL cell hyperplasia which persists for 2–3 months after stopping the PPI [46••]. The clinical consequence is rebound acid secretion after discontinuation of PPIs which may induce dyspeptic symptoms in healthy individuals, exacerbate reflux symptoms in patients with gastroesophageal reflux disease, and provoke symptoms in patients with duodenal ulcer disease [47••].

Gastrin, acting via the CCK-2 receptor, suppresses apoptosis in a variety of tumors in and outside the gastrointestinal tract [48,49]. A recent study suggests that hypochlorhydria (and hence hypergastrinemia) is a strong independent risk factor for esophageal squamous cell carcinoma [50].

Other purported adverse effects of acid suppression by proton pump inhibitors include infections, bacterial overgrowth, osteoporosis, and drug–drug interactions; these are discussed in Denis McCarthy's article.

Conclusion

Too much acid and too little acid secretion are both important pathogenic factors for a variety of common upper gastrointestinal disorders. We continue to make progress in our understanding of gastric acid secretion in health and disease. In particular, much progress has being made in elucidating the mechanisms and pathways regulating H+K+-ATPase trafficking as well as ion transport in the parietal cell. It is anticipated that this knowledge will lead to the development of new drugs to treat acid-peptic disorders. In addition, we are beginning to take stock of potential adverse effects related to prolonged and profound inhibition of acid secretion.

Acknowledgement

The author is grateful to Mary Beatty-Brooks for the artwork.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest

Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 663–665).

1• Karam SM. A focus on parietal cells as a renewing cell population. World J Gastroenterol 2010; 16:538–546. An excellent review summarizing the origin, differentiation, and migration of parietal cells.

  • Cited Here

2 DeVault KR, Talley NJ. Insights into the future of gastric acid suppression. Nat Rev Gastroenterol Hepatol 2009; 6:524–532.

  • Cited Here

3• Murray L, Gabello M, Rudolph D, et al. Transmucosal gastric leak induced by proton pump inhibitors. Dig Dis Sci 2009; 54:1408–1417. In an ex-vivo rat preparation, PPIs increased transmucosal permeability. The clinical consequences of this finding are not known, but it is speculated that such a leak may contribute to neoplasia and food allergy.

  • Cited Here

4 Liwanpo L, Hershman JM. Conditions and drugs interfering with thyroxine absorption. Best Pract Res Clin Endocrinol Metab 2009; 23:781–792.

  • Cited Here

5 Zavros Y, Orr MA, Xiao C, Malinowska DH. Sonic hedgehog is associated with H+-K+-ATPase-containing membranes in gastric parietal cells and secreted with histamine stimulation. Am J Physiol Gastrointest Liver Physiol 2008; 295:G99–G111.

  • Cited Here

6 Shiotani A, Kamada T, Yamanaka Y, et al. Sonic hedgehog and CDX2 expression in the stomach. J Gastroenterol Hepatol 2008; 23:S161–S166.

  • Cited Here

7 Lahner E, Norman GL, Severini C, et al. Reassessment of intrinsic factor and parietal cell autoantibodies in atrophic gastritis with respect to cobalamin deficiency. Am J Gastroenterol 2009; 104:2071–2079.

  • Cited Here

8 Liaskos C, Norman GL, Moulas A, et al. Prevalence of gastric parietal cell antibodies and intrinsic factor antibodies in primary biliary cirrhosis. Clin Chim Acta 2010; 411:411–415.

  • Cited Here

9 Caboclo J, Cury F, Borin A, et al. Gastric secretion elicited by conditioning in rats. Scand J Gastroenterol 2009; 44:672–679.

  • Cited Here

10 Schubert ML, Peura DA. Control of gastric acid secretion in health and disease. Gastroenterology 2008; 134:1842–1860.

  • Cited Here

11 Kanai S, Hosoya H, Akimoto S, et al. Gastric acid secretion in cholecystokinin-1 receptor,-2 receptor, and -1,-2 receptor gene knockout mice. J Physiol Sci 2009; 59:23–29.

  • Cited Here

12 Nozaki K, Weis V, Wang T, et al. Altered gastric chief cell lineage differentiation in histamine-deficient mice. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1211–G1220.

  • Cited Here

13 Aikou S, f*ckushima Y, Ogawa M, et al. Alterations in gastric mucosal lineages before or after acute oxyntic atrophy in gastrin receptor and H-2 histamine receptor-deficient mice. Dig Dis Sci 2009; 54:1625–1635.

  • Cited Here

14 Ericsson P, Hakanson R, Rehfeld JF, Norlen P. Gastrin release: antrum microdialysis reveals a complex neural control. Regul Pept 2010; 161:22–32.

  • Cited Here

15• Ericson A, Mohammed Nur E, Petersson F, Kechagias S. The effects of capsaicin on gastrin secretion in isolated human antral glands: before and after ingestion of red chilli. Dig Dis Sci 2009; 54:491–498. In isolated human antral glands, capsaicin binds to VR1 receptors on G cells to stimulate gastrin secretion.

  • Cited Here

16 Kidd M, Hauso O, Drozdov I, et al. Delineation of the chemomechanosensory regulation of gastrin secretion using pure rodent G cells. Gastroenterology 2009; 137:231–241.

  • Cited Here

17 Murugesan SVM, Varro A, Pritchard DM. Review article: strategies to determine whether hypergastrinaemia is due to Zollinger–Ellison syndrome rather than a more common benign cause. Aliment Pharmacol Ther 2009; 29:1055–1068.

  • Cited Here

18 Chakravorty M, De D, Choudhury A, Roychoudhury S. IL1B promoter polymorphism regulates the expression of gastric acid stimulating hormone gastrin. Int J Biochem Cell Biol 2009; 41:1502–1510.

  • Cited Here

19 Mizutani M, Atsuchi K, Asakawa A, et al. Localization of acyl ghrelin- and des-acyl ghrelin-immunoreactive cells in the rat stomach and their responses to intragastric pH. Am J Physiol Gastrointest Liver Physiol 2009; 297:G874–G980.

  • Cited Here

20 Seim I, Amorim L, Walpole C, et al. Ghrelin gene-related peptides: multifunctional endocrine/autocrine modulators in health and disease. Clin Exp Pharmacol Physiol 2010; 37:125–131.

  • Cited Here

21 Gualillo O, Lago F, Dieguez C. Introducing GOAT: a target for obesity and antidiabetic drugs? Trends Pharmacol Sci 2008; 29:398–401.

  • Cited Here

22 Stengel A, Goebel M, Wang L, et al. Differential distribution of ghrelin-O-acyltransferase (GOAT) immunoreactive cells in the mouse and rat gastric oxyntic mucosa. Biochem Biophys Res Commun 2010; 392:67–71.

  • Cited Here

23 Sakata I, Yang J, Lee CE, et al. Colocalization of ghrelin O-acyltransferase and ghrelin in gastric mucosal cells. Am J Physiol Endocrinol Metab 2009; 297:E134–E141.

  • Cited Here

24 El-Salhy M. Ghrelin in gastrointestinal diseases and disorders: a possible role in the pathophysiology and clinical implications (Review). Int J Mol Med 2009; 24:727–732.

  • Cited Here

25 Camilleri M, Papathanasopoulos A, Odunsi ST. Actions and therapeutic pathways of ghrelin for gastrointestinal disorders. Nat Rev Gastroenterol Hepatol 2009; 6:343–353.

  • Cited Here

26 Bossenmeyer-Pourie C, Blaise S, Pourie G, et al. Methyl donor deficiency affects fetal programming of gastric ghrelin cell organization and function in the rat. Am J Pathol 2010; 176:270–277.

  • Cited Here

27 Yin JD, Li XL, Li DF, et al. Dietary supplementation with zinc oxide stimulates ghrelin secretion from the stomach of young pigs. J Nutr Biochem 2009; 20:783–790.

  • Cited Here

28 Iwakura H, Li Y, Aryasu H, et al. Establishment of a novel ghrelin-producing cell line. Endocrinology 2010; 151:2940–2945.

  • Cited Here

29• Hass N, Schwarzenbacher K, Breer H. T1R3 is expressed in brush cells and ghrelin-producing cells of murine stomach. Cell Tissue Res 2010; 339:493–504. Taste receptors of the subtype T1R3, which detect sugar and amino acids, were detected on ghrelin cells and adjacent brush cells in mice. It remains to be determined whether food constituents regulate ghrelin secretion indirectly via paracrine signals emanating from adjacent brush cells or from serum concentrations of amino acids and glucose.

  • Cited Here

30 Nakamura E, Hasumura M, SanGabriel A, et al. New frontiers in gut nutrient sensor research: luminal glutamate-sensing cells in rat gastric mucosa. J Pharmacol Sci 2010; 112:13–18.

  • Cited Here

31 Zolotarev V, Khropycheva R, Uneyama H, Torii K. Effect of free dietary glutamate on gastric acid secretion in dogs. Ann N Y Acad Sci 2009; 1170:87–90.

  • Cited Here

32 Weiss C, Rubach M, Lang R, et al. Measurement of the intracellular pH in human stomach cells: a novel approach to evaluate the gastric acid secretory potential of coffee beverages. J Agric Food Chem 2010; 58:1976–1985.

  • Cited Here

33 DenBosch JV, Adriaensen D, VanNassauw L, Timmermans JP. The role(S) of somatostatin, structurally related peptides and somatostatin receptors in the gastrointestinal tract: a review. Regul Pept 2009; 156:1–8.

  • Cited Here

34 Yang G, Chen J, Kieffer T, Kwok Y. Regulation of somatostatin release by adenosine in the mouse stomach. J Pharmacol Exp Ther 2009; 329:729–737.

  • Cited Here

35 Sidani S, Kopic S, Socrates T, et al. AMP-activated protein kinase: a physiological off switch for murine gastric acid secretion. Pflugers Arch Eur J Physiol 2009; 459:39–46.

  • Cited Here

36• Kopic S, Corradini S, Sidani S, et al. Ethanol inhibits gastric acid secretion in rats through increased AMP-kinase activity. Cell Physiol Biochem 2010; 25:195–202. In isolated rat gastric glands loaded with the pH-sensitive dye BCECF, low-dose (2%) ethanol inhibits secretagogue-dependent acid secretion by activation of the AMPK pathway.

  • Cited Here

37 Gerbino A, Debellis L, Caroppo R, et al. Cadmium inhibits acid secretion in stimulated frog gastric mucosa. Toxicol Appl Pharmacol 2010; 245:264–271.

  • Cited Here

38•• Forte JG, Zhu LX. Apical recycling of the gastric parietal cell H,K-ATPase. Annu Rev Physiol 2010; 72:273–296. Excellent and thorough review of apical membrane recycling in the parietal cell with emphasis on H+K+-ATPase.

  • Cited Here

39 Durr KL, Abe K, Tavraz NN, Friedrich T. E2P state stabilization by the N-terminal tail of the H,K-ATPase beta-subunit is critical for efficient proton pumping under in vivo conditions. J Biol Chem 2009; 284:20147–20154.

  • Cited Here

40•• Kopic S, Murek M, Geibel JP. Revisiting the parietal cell. Am J Physiol Cell Physiol 2010; 298:C1–C10. Superb review of ion transport in the parietal cell.

  • Cited Here

41 Witzke A, Lindner K, Munson K, Apell H-J. Inhibition of the gastric H,K-ATPase by clotrimazole. Biochemistry 2010; 49:4524–4532.

  • Cited Here

42 Xu J, Song P, Nakamura S, et al. Deletion of the chloride transporter Slc26a7 causes distal renal tubular acidosis and impairs gastric acid secretion. J Biol Chem 2009; 284:29470–29479.

  • Cited Here

43 Pan Q, Ma J, Zhou QS, et al. KCNQ1 loss-of-function mutation impairs gastric acid secretion in mice. Mol Biol Rep 2010; 37:1329–1333.

  • Cited Here

44 Okuda S, Honda M, Ito Y, et al. Phosphodiesterase isozymes involved in regulating acid secretion in the isolated mouse stomach. J Physiol Pharmacol 2009; 60:183–190.

  • Cited Here

45 Rotte A, Pasham V, Bhandaru M, et al. Regulation of gastric acid secretion by PKB/Akt2. Cell Physiol Biochem 2010; 25:695–704.

  • Cited Here

46•• Waldum HL, Qvigstad G, Fossmark R, et al. Rebound acid hypersecretion from a physiological, pathophysiological and clinical viewpoint. Scand J Gastroenterol 2010; 45:389–394. Excellent review of the physiological consequences of gastric acid inhibition by PPIs with emphasis on hypergastrinemia with resultant rebound acid secretion once PPIs are discontinued.

  • Cited Here

47•• Reimer C, Sondergaard B, Hilsted L, Bytzer P. Proton-pump inhibitor therapy induces acid-related symptoms in healthy volunteers after withdrawal of therapy. Gastroenterology 2009; 137:80–87. After long-term PPI therapy is discontinued, healthy volunteers develop dyspeptic symptoms due to rebound gastric acid secretion.

  • Cited Here

48 Korner M, Waser B, Reubi JC, Miller LJ. CCK2 receptor splice variant with intron 4 retention in human gastrointestinal and lung tumours. J Cell Mol Med 2010; 14:933–943.

  • Cited Here

49 Almeida-Vega S, Catlow K, Kenny S, et al. Gastrin activates paracrine networks leading to induction of PAI-2 via MAZ and ASC-1. Am J Physiol Gastrointest Liver Physiol 2009; 296:G414–G423.

  • Cited Here

50 Lijima K, Koike T, Abe Y, et al. Gastric hyposecretion in esophageal squamous-cell carcinomas. Dig Dis Sci 2010; 55:1349–1355.

  • Cited Here

Keywords:

acid secretion; gastrin; H+K+-ATPase; histamine; parietal cell; proton pump; review; signaling; somatostatin; transduction pathways

© 2010 Lippincott Williams & Wilkins, Inc.
Gastric secretion : Current Opinion in Gastroenterology (2024)
Top Articles
Howlsmovingasshole
How To Make Dinosaur Bones Out Of Clay - Stunningdino.com
Whas Golf Card
Aberration Surface Entrances
Pixel Speedrun Unblocked 76
The Ivy Los Angeles Dress Code
Did 9Anime Rebrand
Us 25 Yard Sale Map
Ribbit Woodbine
Jasmine
Rls Elizabeth Nj
Matthew Rotuno Johnson
Infinite Campus Parent Portal Hall County
Lantana Blocc Compton Crips
Myql Loan Login
The Weather Channel Facebook
Newgate Honda
Miss America Voy Forum
WWE-Heldin Nikki A.S.H. verzückt Fans und Kollegen
978-0137606801
Operation Cleanup Schedule Fresno Ca
The Largest Banks - ​​How to Transfer Money With Only Card Number and CVV (2024)
Weather Rotterdam - Detailed bulletin - Free 15-day Marine forecasts - METEO CONSULT MARINE
Andhrajyothy Sunday Magazine
Jalapeno Grill Ponca City Menu
Candy Land Santa Ana
Lehmann's Power Equipment
Wausau Marketplace
Delaware Skip The Games
Outlet For The Thames Crossword
Busted Mcpherson Newspaper
St Clair County Mi Mugshots
Providence Medical Group-West Hills Primary Care
Hampton University Ministers Conference Registration
Gotcha Rva 2022
Aliciabibs
Finding Safety Data Sheets
Student Portal Stvt
Urban Blight Crossword Clue
Worlds Hardest Game Tyrone
Yoshidakins
Royals op zondag - "Een advertentie voor Center Parcs" of wat moeten we denken van de laatste video van prinses Kate?
Facebook Marketplace Marrero La
The 50 Best Albums of 2023
Husker Football
Live Delta Flight Status - FlightAware
התחבר/י או הירשם/הירשמי כדי לראות.
Exploring the Digital Marketplace: A Guide to Craigslist Miami
Value Village Silver Spring Photos
Bf273-11K-Cl
Verizon Forum Gac Family
Smoke From Street Outlaws Net Worth
Latest Posts
Article information

Author: Jeremiah Abshire

Last Updated:

Views: 6092

Rating: 4.3 / 5 (74 voted)

Reviews: 89% of readers found this page helpful

Author information

Name: Jeremiah Abshire

Birthday: 1993-09-14

Address: Apt. 425 92748 Jannie Centers, Port Nikitaville, VT 82110

Phone: +8096210939894

Job: Lead Healthcare Manager

Hobby: Watching movies, Watching movies, Knapping, LARPing, Coffee roasting, Lacemaking, Gaming

Introduction: My name is Jeremiah Abshire, I am a outstanding, kind, clever, hilarious, curious, hilarious, outstanding person who loves writing and wants to share my knowledge and understanding with you.