Comprehensive Physiology Wiley Online Library

Gastric H+,K+‐ATPase

Full Article on Wiley Online Library



Abstract

The gastric H+,K+‐ATPase is responsible for gastric acid secretion. This ATPase is composed of two subunits, the catalytic α subunit and the structural β subunit. The α subunit with molecular mass of about 100 kDa has 10 transmembrane domains and is strongly associated with the β subunit with a single transmembrane segment and a peptide mass of 35 kDa. Its three‐dimensional structure is based on homology modeling and site‐directed mutagenesis resulting in a proton extrusion and K+ reabsorption model. There are three conserved H3O+‐binding sites in the middle of the membrane domain and H3O+ secretion depends on a conformational change involving Lys791 insertion into the second H3O+ site enclosed by E795, E820, and D824 that allows export of protons at a concentration of 160 mM. K+ countertransport involves binding to this site after the release of protons with retrograde displacement of Lys791 and then K+ transfer to E343 and exit to the cytoplasm. This ATPase is the major therapeutic target in treatment of acid‐related diseases and there are several known luminal inhibitors allowing analysis of the luminal vestibule. One class contains the acid‐activated covalent, thiophilic proton pump inhibitors, the most effective of current acid‐suppressive drugs. Their binding sites and trypsinolysis allowed identification of all ten transmembrane segments of the ATPase. In addition, various K+‐competitive inhibitors of the ATPase are being developed, with the advantage of complete and rapid inhibition of acid secretion independent of pump activity and allowing further refinement of the structure of the luminal vestibule of the E2 form of this ATPase. © 2011 American Physiological Society. Compr Physiol 1:2141‐2153, 2011.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

The structure of the gastric H+,K+‐ATPase. The modeled three‐dimensional structure of the H+,K+‐ATPase, HKzxe based on the Na+,K+‐ATPase in the E2P state. The α subunit has three lobes, N (ATP binding), P (phosphorylation), and A (activation) domains in the cytoplasmic domain, and ten transmembrane segments in the membrane domain. The gastric β subunit has short cytoplasmic region, one transmembrane segment, and a heavily glycosylated extracellular region. In this model, the backbone of protein with carbohydrate attached Asn sites is shown. Amino acid sequence number is based on pig H+,K+‐ATPase.

Figure 2. Figure 2.

The catalytic cycle of the gastric H+,K+‐ATPase. A hydronium ion binds to the cytoplasmic surface of the enzyme, and MgATP phosphorylates the protein at Asp386 to form the first ion transport intermediate in the E1P form. The E1P form then converts by a conformational change to the second ion transport form, E2P, with the ion site now exposed to the exterior and hydronium is released at pH ∼1.0. To this form, K+ binds from the outside surface to the same region from which the hydronium was released, and the enzyme dephosphorylates, and then K+ is trapped within the membrane domain in the occluded form (and a similar form is postulated for the hydronium in the outward step of the cycle). The K+ is then deoccluded allowing reformation of the E1 form of the enzyme with the ion site now again facing the cytoplasm and K+ is displaced when ATP is bound. In this model, the naked H+ is used instead of the hydronium ion (H3O+). This model reflects a stoichiometry of 1H/1K/1ATP.

Figure 3. Figure 3.

A model of the H+,K+ ion exchange at acidic luminal pH based on the three‐dimensional structure of the srCa‐ATPase. In panel 1 the 3 hydronium ion‐binding sites at the carboxylates in TM 5, 6 and TM3 with the exported ion at acidic pH outlined in yellow. In panel 2 after phosphorylation, lys791 displaces the hydronium from site 2 in the E1P form (arrow). Then in panel 3 K+ binds to site 2 displacing lys791 generating the E2P form and in panel 4 K+ moves into site 3 allowing exit to the cytoplasm along TM3.

Figure 4. Figure 4.

SCH28080‐bound E2P structure of the gastric H+,K+‐ATPase analyzed at 7 Å resolution. (A) The structure of the H+,K+‐ATPase showing cytoplasmic‐side up. Surface represents the EM density map with superimposed homology model of H+,K+‐ATPase (ribbon). Bound SCH28080 at the luminal cavity, and ADP at the N domain are shown as sticks. A wheat box indicates approximate location of the lipid bilayer. Color code: A domain, blue; P domain, green; N domain, yellow; TM domain, light blue (surface); and β subunit, red. Color for the TM helices of the homology model gradually changes from M1 (blue) to M10 (red). (B) Conformational change of the cytoplasmic domain seen from the top. Surface shows EM density map of (SCH)E2BeF and orange mesh represents E2AlF EM map of the gastric H+,K+‐ATPase. The intersubunit interaction between the P domain and the N‐terminal tail of the β subunit is observed in E2AlF structure, but not present in (SCH)E2BeF structure. White arrows indicate observation of the conformational changes during E2AlF → (SCH)E2BeF transition. This figure is cited, with permission, from the work of Abe et al. 1 and is similar to the homology model of the H+,K+‐ATPase shown above and below.

Figure 5. Figure 5.

K+ pathway in the H+,K+‐ATPase. The image illustrates the entry path for K+ (illustrated as a series of violet spheres) between M4, M5, M6, and M8 in the E2P conformation as determined by molecular dynamics simulation (M3 in the foreground is omitted for clarity) 35,50. The arrival of K+ at the top of this path is predicted to destabilize the interaction of K791 with E820 and E795, and initiate the conformational changes leading to release of phosphate at the active site and conversion back to E1.

Figure 6. Figure 6.

Proposed K+ pathway through the pump srCa‐ATPase homology model. Red to pink is area of restriction including the occlusion site. White to blue less occlusion. Arrows highlight entry and exit pathways for K+.

Figure 7. Figure 7.

Proton pump inhibitors.

Figure 8. Figure 8.

The mechanism of activation of the PPIs shown in general structural form. The substituent R1, R2, R3, and R4 of the general structure (Bz‐Py) represent a substituent chosen from hydrogen, methoxy, methyl, and substituted alkoxy group. Top part shows the protonation of the pyridine ring and the second row of structures shows protonation also of the benzimidazole ring. The bis‐protonated forms are in equilibrium with the protonated benzimidazole and unprotonated pyridine. In brackets is shown the mechanism of activation whereby the 2C of the protonated benzimidazole reacts with the unprotonated fraction of the pyridine moiety that results in rearrangement to a permanent cationic tetracyclic sulfenic acid which in aqueous solute dehydrates to form a cationic sulfenamide. Either of these thiophilic species can react with the enzyme to form disulfides with one or more enzyme cysteines accessible from the luminal surface of the enzyme.

Figure 9. Figure 9.

The amino acids lining the luminal entrance (vestibule) to the ion‐binding sites of the H+,K+‐ATPase that is the basis for docking pyrrolo‐pyridines suggesting critical amino acid residues responsible for high affinity and slow dissociation.

Figure 10. Figure 10.

Acid pump antagonists.

Figure 11. Figure 11.

The vestibule structure predicted for bound TAK‐438. Selected amino acids in the binding region are predicted to affect binding (white, bold). Also shown is H bonding to E795 ‐COO and ‐C=O and to Y799.



Figure 1.

The structure of the gastric H+,K+‐ATPase. The modeled three‐dimensional structure of the H+,K+‐ATPase, HKzxe based on the Na+,K+‐ATPase in the E2P state. The α subunit has three lobes, N (ATP binding), P (phosphorylation), and A (activation) domains in the cytoplasmic domain, and ten transmembrane segments in the membrane domain. The gastric β subunit has short cytoplasmic region, one transmembrane segment, and a heavily glycosylated extracellular region. In this model, the backbone of protein with carbohydrate attached Asn sites is shown. Amino acid sequence number is based on pig H+,K+‐ATPase.



Figure 2.

The catalytic cycle of the gastric H+,K+‐ATPase. A hydronium ion binds to the cytoplasmic surface of the enzyme, and MgATP phosphorylates the protein at Asp386 to form the first ion transport intermediate in the E1P form. The E1P form then converts by a conformational change to the second ion transport form, E2P, with the ion site now exposed to the exterior and hydronium is released at pH ∼1.0. To this form, K+ binds from the outside surface to the same region from which the hydronium was released, and the enzyme dephosphorylates, and then K+ is trapped within the membrane domain in the occluded form (and a similar form is postulated for the hydronium in the outward step of the cycle). The K+ is then deoccluded allowing reformation of the E1 form of the enzyme with the ion site now again facing the cytoplasm and K+ is displaced when ATP is bound. In this model, the naked H+ is used instead of the hydronium ion (H3O+). This model reflects a stoichiometry of 1H/1K/1ATP.



Figure 3.

A model of the H+,K+ ion exchange at acidic luminal pH based on the three‐dimensional structure of the srCa‐ATPase. In panel 1 the 3 hydronium ion‐binding sites at the carboxylates in TM 5, 6 and TM3 with the exported ion at acidic pH outlined in yellow. In panel 2 after phosphorylation, lys791 displaces the hydronium from site 2 in the E1P form (arrow). Then in panel 3 K+ binds to site 2 displacing lys791 generating the E2P form and in panel 4 K+ moves into site 3 allowing exit to the cytoplasm along TM3.



Figure 4.

SCH28080‐bound E2P structure of the gastric H+,K+‐ATPase analyzed at 7 Å resolution. (A) The structure of the H+,K+‐ATPase showing cytoplasmic‐side up. Surface represents the EM density map with superimposed homology model of H+,K+‐ATPase (ribbon). Bound SCH28080 at the luminal cavity, and ADP at the N domain are shown as sticks. A wheat box indicates approximate location of the lipid bilayer. Color code: A domain, blue; P domain, green; N domain, yellow; TM domain, light blue (surface); and β subunit, red. Color for the TM helices of the homology model gradually changes from M1 (blue) to M10 (red). (B) Conformational change of the cytoplasmic domain seen from the top. Surface shows EM density map of (SCH)E2BeF and orange mesh represents E2AlF EM map of the gastric H+,K+‐ATPase. The intersubunit interaction between the P domain and the N‐terminal tail of the β subunit is observed in E2AlF structure, but not present in (SCH)E2BeF structure. White arrows indicate observation of the conformational changes during E2AlF → (SCH)E2BeF transition. This figure is cited, with permission, from the work of Abe et al. 1 and is similar to the homology model of the H+,K+‐ATPase shown above and below.



Figure 5.

K+ pathway in the H+,K+‐ATPase. The image illustrates the entry path for K+ (illustrated as a series of violet spheres) between M4, M5, M6, and M8 in the E2P conformation as determined by molecular dynamics simulation (M3 in the foreground is omitted for clarity) 35,50. The arrival of K+ at the top of this path is predicted to destabilize the interaction of K791 with E820 and E795, and initiate the conformational changes leading to release of phosphate at the active site and conversion back to E1.



Figure 6.

Proposed K+ pathway through the pump srCa‐ATPase homology model. Red to pink is area of restriction including the occlusion site. White to blue less occlusion. Arrows highlight entry and exit pathways for K+.



Figure 7.

Proton pump inhibitors.



Figure 8.

The mechanism of activation of the PPIs shown in general structural form. The substituent R1, R2, R3, and R4 of the general structure (Bz‐Py) represent a substituent chosen from hydrogen, methoxy, methyl, and substituted alkoxy group. Top part shows the protonation of the pyridine ring and the second row of structures shows protonation also of the benzimidazole ring. The bis‐protonated forms are in equilibrium with the protonated benzimidazole and unprotonated pyridine. In brackets is shown the mechanism of activation whereby the 2C of the protonated benzimidazole reacts with the unprotonated fraction of the pyridine moiety that results in rearrangement to a permanent cationic tetracyclic sulfenic acid which in aqueous solute dehydrates to form a cationic sulfenamide. Either of these thiophilic species can react with the enzyme to form disulfides with one or more enzyme cysteines accessible from the luminal surface of the enzyme.



Figure 9.

The amino acids lining the luminal entrance (vestibule) to the ion‐binding sites of the H+,K+‐ATPase that is the basis for docking pyrrolo‐pyridines suggesting critical amino acid residues responsible for high affinity and slow dissociation.



Figure 10.

Acid pump antagonists.



Figure 11.

The vestibule structure predicted for bound TAK‐438. Selected amino acids in the binding region are predicted to affect binding (white, bold). Also shown is H bonding to E795 ‐COO and ‐C=O and to Y799.

 1. Abe K, Tani K, Fujiyoshi Y. Conformational rearrangement of gastric H(+),K(+)‐ATPase induced by an acid suppressant. Nat Commun 2: 155, 2011.
 2. Abe K, Tani K, Nishizawa T, Fujiyoshi Y. Inter‐subunit interaction of gastric H+,K+‐ATPase prevents reverse reaction of the transport cycle. Embo J 28: 1637‐1637, 2009.
 3. Asano S, Kawada K, Kimura T, Grishin AV, Caplan MJ, Takeguchi N. The roles of carbohydrate chains of the beta‐subunit on the functional expression of gastric H(+),K(+)‐ATPase. J Biol Chem 275: 8324‐8324, 2000.
 4. Asano S, Mizutani M, Hayashi T, Morita N, Takeguchi N. Reversible inhibitions of gastric H+,K(+)‐ATPase by scopadulcic acid B and diacetyl scopadol. New biochemical tools of H+,K(+)‐ATPase. J Biol Chem 265: 22167‐22167, 1990.
 5. Bamberg K, Mercier F, Reuben MA, Kobayashi Y, Munson KB, Sachs G. cDNA cloning and membrane topology of the rabbit gastric H+/K(+)‐ATPase alpha‐subunit. Biochim Biophys Acta 1131: 69‐69, 1992.
 6. Bamberg K, Sachs G. Topological analysis of H+,K(+)‐ATPase using in vitro translation. J Biol Chem 269: 16909‐16909, 1994.
 7. Besancon M, Shin JM, Mercier F, Munson K, Miller M, Hersey S, Sachs G. Membrane topology and omeprazole labeling of the gastric H+,K(+)‐ adenosinetriphosphatase. Biochemistry 32: 2345‐2345, 1993.
 8. 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: 22438‐22438, 1997.
 9. Brzezinski P, Malmstrom BG, Lorentzon P, Wallmark B. The catalytic mechanism of gastric H+/K+‐ATPase: Simulations of pre‐steady‐state and steady‐state kinetic results. Biochim Biophys Acta 942: 215‐215, 1988.
 10. Callaghan JM, Toh BH, Pettitt JM, Humphris DC, Gleeson PA. Poly‐N‐acetyllactosamine‐specific tomato lectin interacts with gastric parietal cells. Identification of a tomato‐lectin binding 60‐90 X 10(3) Mr membrane glycoprotein of tubulovesicles. J Cell Sci 95 (Pt 4): 563‐563, 1990.
 11. Canfield VA, Levenson R. Structural organization and transcription of the mouse gastric H+, K(+)‐ATPase beta subunit gene. Proc Natl Acad Sci U S A 88: 8247‐8247, 1991.
 12. Canfield VA, Okamoto CT, Chow D, Dorfman J, Gros P, Forte JG, Levenson R. Cloning of the H,K‐ATPase beta subunit. Tissue‐specific expression, chromosomal assignment, and relationship to Na,K‐ATPase beta subunits. J Biol Chem 265: 19878‐19878, 1990.
 13. Craig WS. Monomer of sodium and potassium ion activated adenosinetriphosphatase displays complete enzymatic function. Biochemistry 21: 5707‐5707, 1982.
 14. Dantzig AH, Minor PL, Garrigus JL, Fukuda DS, Mynderse JS. Studies on the mechanism of action of A80915A, a semi‐naphthoquinone natural product, as an inhibitor of gastric (H(+)‐K+)‐ATPase. Biochem Pharmacol 42: 2019‐2019, 1991.
 15. De Pont JJ, Swarts HG, Karawajczyk A, Schaftenaar G, Willems PH, Koenderink JB. The non‐gastric H,K‐ATPase as a tool to study the ouabain‐binding site in Na,K‐ATPase. Pflugers Arch 457: 623‐623, 2009.
 16. 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 284: 20147‐20147, 2009.
 17. Fellenius E, Berglindh T, Sachs G, Olbe L, Elander B, Sjostrand SE, Wallmark B. Substituted benzimidazoles inhibit gastric acid secretion by blocking (H+ + K+)ATPase. Nature 290: 159‐159, 1981.
 18. Fujisaki H, Shibata H, Oketani K, Murakami M, Fujimoto M, Wakabayashi T, Yamatsu I, Yamaguchi M, Sakai H, Takeguchi N. Inhibitions of acid secretion by E3810 and omeprazole, and their reversal by glutathione. Biochem Pharmacol 42: 321‐321, 1991.
 19. Hall K, Perez G, Anderson D, Gutierrez C, Munson K, Hersey SJ, Kaplan JH, Sachs G. Location of the carbohydrates present in the HK‐ATPase vesicles isolated from hog gastric mucosa. Biochemistry 29: 701‐701, 1990.
 20. Hall K, Perez G, Sachs G, Rabon E. Identification of H+/K(+)‐ATPase alpha,beta‐heterodimers. Biochim Biophys Acta 1077: 173‐173, 1991.
 21. Han KS, Kim YG, Yoo JK, Lee JW, Lee MG. Pharmacokinetics of a new reversible proton pump inhibitor, YH1885, after intravenous and oral administrations to rats and dogs: Hepatic first‐pass effect in rats. Biopharm Drug Dispos 19: 493‐493, 1998.
 22. Hebert H, Xian Y, Hacksell I, Mardh S. Two‐dimensional crystals of membrane‐bound gastric H,K‐ATPase. FEBS Lett 299: 159‐159, 1992.
 23. Hioki Y, Takada J, Hidaka Y, Takeshita H, Hosoi M, Yano M. A newly synthesized pyridine derivative, (Z)‐5‐methyl‐2‐[2‐(1‐ naphthyl)ethenyl]‐4‐piperidinopyridine hydrochloride (AU‐1421), as a reversible gastric proton pump inhibitor. Arch Int Pharmacodyn Ther 305: 32‐32, 1990.
 24. Hori Y, Imanishi A, Matsukawa J, Tsukimi Y, Nishida H, Arikawa Y, Hirase K, Kajino M, Inatomi N. 1‐[5‐(2‐Fluorophenyl)‐1‐(pyridin‐3‐ylsulfonyl)‐1H‐pyrrol‐3‐yl]‐N‐methylmet hanamine monofumarate (TAK‐438), a novel and potent potassium‐competitive acid blocker for the treatment of acid‐related diseases. J Pharmacol Exp Ther 335: 231‐231, 2011.
 25. Hunt RH, Armstrong D, Yaghoobi M, James C, Chen Y, Leonard J, Shin JM, Lee E, Tang‐Liu D, Sachs G. Predictable prolonged suppression of gastric acidity with a novel proton pump inhibitor, AGN 201904‐Z. Aliment Pharmacol Ther 28: 187‐187, 2008.
 26. Ife RJ, Brown TH, Keeling DJ, Leach CA, Meeson ML, Parsons ME, Reavill DR, Theobald CJ, Wiggall KJ. Reversible inhibitors of the gastric (H+/K+)‐ATPase. 3. 3‐substituted‐4‐ (phenylamino)quinolines. J Med Chem 35: 3413‐3413, 1992.
 27. Jensen AM, Sorensen TL, Olesen C, Moller JV, Nissen P. Modulatory and catalytic modes of ATP binding by the calcium pump. Embo J 25: 2305‐2305, 2006.
 28. Kaminski JJ, Bristol JA, Puchalski C, Lovey RG, Elliott AJ, Guzik H, Solomon DM, Conn DJ, Domalski MS, Wong SC, Gold EH, Long JF, Chiu PJ, Steinberg M, McPhail AT. Antiulcer agents. 1. Gastric antisecretory and cytoprotective properties of substituted imidazo[1,2‐a]pyridines. J Med Chem 28: 876‐876, 1985.
 29. Kaminski JJ, Doweyko AM. Antiulcer agents. 6. Analysis of the in vitro biochemical and in vivo gastric antisecretory activity of substituted imidazo[1,2‐a]pyridines and related analogues using comparative molecular field analysis and hypothetical active site lattice methodologies. J Med Chem 40: 427‐427, 1997.
 30. Katz PO, Koch FK, Ballard ED, Bagin RG, Gautille TC, Checani GC, Hogan DL, Pratha VS. Comparison of the effects of immediate‐release omeprazole oral suspension, delayed‐release lansoprazole capsules and delayed‐release esomeprazole capsules on nocturnal gastric acidity after bedtime dosing in patients with night‐time GERD symptoms. Aliment Pharmacol Ther 25: 197‐197, 2007.
 31. Keeling DJ, Fallowfield C, Underwood AH. The specificity of omeprazole as an (H+ + K+)‐ATPase inhibitor depends upon the means of its activation. Biochem Pharmacol 36: 339‐339, 1987.
 32. Laine L, Katz PO, Johnson DA, Ibegbu I, Goldstein MJ, Chou C, Rossiter G, Lu Y. Randomised clinical trial: A novel rabeprazole extended release 50 mg formulation vs. esomeprazole 40 mg in healing of moderate‐to‐severe erosive oesophagitis ‐ the results of two double‐blind studies. Aliment Pharmacol Ther 33: 203‐203, 2011.
 33. LaMattina JL, McCarthy PA, Reiter LA, Holt WF, Yeh LA. Antiulcer agents. 4‐substituted 2‐guanidinothiazoles: Reversible, competitive, and selective inhibitors of gastric H+,K(+)‐ATPase. J Med Chem 33: 543‐543, 1990.
 34. Lane LK, Kirley TL, Ball WJ, Jr. Structural studies on H+,K+‐ATPase: Determination of the NH2‐terminal amino acid sequence and immunological cross‐reactivity with Na+,K+‐ATPase. Biochem Biophys Res Commun 138: 185‐185, 1986.
 35. Law RJ, Munson K, Sachs G, Lightstone FC. An ion gating mechanism of gastric H,K‐ATPase based on molecular dynamics simulations. Biophys J 95: 2739‐2739, 2008.
 36. Leach CA, Brown TH, Ife RJ, Keeling DJ, Laing SM, Parsons ME, Price CA, Wiggall KJ. Reversible inhibitors of the gastric (H+/K+)‐ATPase. 2. 1‐ Arylpyrrolo[3,2‐c]quinolines: Effect of the 4‐substituent. J Med Chem 35: 1845‐1845, 1992.
 37. Lorentzon P, Jackson R, Wallmark B, Sachs G. Inhibition of (H+ + K+)‐ATPase by omeprazole in isolated gastric vesicles requires proton transport. Biochim Biophys Acta 897: 41‐41, 1987.
 38. Ma JY, Song YH, Sjostrand SE, Rask L, Mardh S. cDNA cloning of the beta‐subunit of the human gastric H,K‐ATPase. Biochem Biophys Res Commun 180: 39‐39, 1991.
 39. Maeda M, Ishizaki J, Futai M. cDNA cloning and sequence determination of pig gastric (H+ + K+)‐ATPase. Biochem Biophys Res Commun 157: 203‐203, 1988.
 40. Maeda M, Oshiman K, Tamura S, Kaya S, Mahmood S, Reuben MA, Lasater LS, Sachs G, Futai M. The rat H+/K(+)‐ATPase beta subunit gene and recognition of its control region by gastric DNA binding protein. J Biol Chem 266: 21584‐21584, 1991.
 41. Maeda M, Oshiman K, Tamura S, Futai M. Human gastric (H+ + K+)‐ATPase gene. Similarity to (Na+ + K+)‐ATPase genes in exon/intron organization but difference in control region. J Biol Chem 265: 9027‐9027, 1990.
 42. Mardh S, Norberg L. A continuous flow technique for analysis of the stoichiometry of the gastric H,K‐ATPase. Acta Physiol Scand Suppl 607: 259‐259, 1992.
 43. Melle‐Milovanovic D, Milovanovic M, Nagpal S, Sachs G, Shin JM. Regions of association between the alpha and the beta subunit of the gastric H,K‐ATPase. J Biol Chem 273: 11075‐11075, 1998.
 44. Mendlein J, Ditmars ML, Sachs G. Calcium binding to the H+,K(+)‐ATPase. Evidence for a divalent cation site that is occupied during the catalytic cycle. J Biol Chem 265: 15590‐15590, 1990.
 45. Mendlein J, Sachs G. The substitution of calcium for magnesium in H+,K+‐ATPase catalytic cycle. Evidence for two actions of divalent cations. J Biol Chem 264: 18512‐18512, 1989.
 46. Mendlein J, Sachs G. Interaction of a K(+)‐competitive inhibitor, a substituted imidazo[1,2a] pyridine, with the phospho‐ and dephosphoenzyme forms of H+, K(+)‐ATPase. J Biol Chem 265: 5030‐5030, 1990.
 47. Moller JV, Olesen C, Winther AM, Nissen P. The sarcoplasmic Ca‐ATPase: Design of a perfect chemi‐osmotic pump. Q Rev Biophys 1‐1, 2010.
 48. Morley GP, Callaghan JM, Rose JB, Toh BH, Gleeson PA, van Driel IR. The mouse gastric H,K‐ATPase beta subunit. Gene structure and co‐ordinate expression with the alpha subunit during ontogeny. J Biol Chem 267: 1165‐1165, 1992.
 49. Munson K, Garcia R, Sachs G. Inhibitor and ion binding sites on the gastric H,K‐ATPase. Biochemistry 44: 5267‐5267, 2005.
 50. Munson K, Law RJ, Sachs G. Analysis of the gastric H,K ATPase for ion pathways and inhibitor binding sites. Biochemistry 46: 5398‐5398, 2007.
 51. Munson K, Vagin O, Sachs G, Karlish S. Molecular modeling of SCH28080 binding to the gastric H,K‐ATPase and MgATP interactions with SERCA‐ and Na,K‐ATPases. Ann N Y Acad Sci 986: 106‐106, 2003.
 52. Munson KB, Gutierrez C, Balaji VN, Ramnarayan K, Sachs G. Identification of an extracytoplasmic region of H+,K(+)‐ATPase labeled by a K(+)‐competitive photoaffinity inhibitor. J Biol Chem 266: 18976‐18976, 1991.
 53. Murakami S, Arai I, Muramatsu M, Otomo S, Baba K, Kido T, Kozawa M. Inhibition of gastric H+,K(+)‐ATPase and acid secretion by cassigarol A, a polyphenol from Cassia garrettiana Craib. Biochem Pharmacol 44: 33‐33, 1992.
 54. Nagaya H, Satoh H, Kubo K, Maki Y. Possible mechanism for the inhibition of gastric (H+ + K+)‐adenosine triphosphatase by the proton pump inhibitor AG‐1749. J Pharmacol Exp Ther 248: 799‐799, 1989.
 55. Newman PR, Greeb J, Keeton TP, Reyes AA, Shull GE. Structure of the human gastric H,K‐ATPase gene and comparison of the 5’‐flanking sequences of the human and rat genes. DNA Cell Biol 9: 749‐749, 1990.
 56. Newman PR, Shull GE. Rat gastric H,K‐ATPase beta‐subunit gene: Intron/exon organization, identification of multiple transcription initiation sites, and analysis of the 5’‐flanking region. Genomics 11: 252‐252, 1991.
 57. Nilsson C, Albrektson E, Rydholm H, Rohss K, Hassan‐Ali M, Hasselgren G. Tolerability, pharmacokinetics and effects on gastric acid secretion after single oral doses of the potassium‐competitive acid blocker AZD0865 in healthy male subjects. Gastroenterology 128 (4 Suppl. 2): A528, 2005.
 58. Nishizawa T, Abe K, Tani K, Fujiyoshi Y. Structural analysis of 2D crystals of gastric H+,K+‐ATPase in different states of the transport cycle. J Struct Biol 162: 219‐219, 2008.
 59. Okamoto CT, Karpilow JM, Smolka A, Forte JG. Isolation and characterization of gastric microsomal glycoproteins. Evidence for a glycosylated beta‐subunit of the H+/K(+)‐ATPase. Biochim Biophys Acta 1037: 360‐360, 1990.
 60. Olesen C, Picard M, Winther AM, Gyrup C, Morth JP, Oxvig C, Moller JV, Nissen P. The structural basis of calcium transport by the calcium pump. Nature 450: 1036‐1036, 2007.
 61. Olesen C, Sorensen TL, Nielsen RC, Moller JV, Nissen P. Dephosphorylation of the calcium pump coupled to counterion occlusion. Science 306: 2251‐2251, 2004.
 62. Post RL, Toda G, Rogers FN. Phosphorylation by inorganic phosphate of sodium plus potassium ion transport adenosine triphosphatase. Four reactive states. J Biol Chem 250: 691‐691, 1975.
 63. Rabon EC, Gunther RD, Bassilian S, Kempner ES. Radiation inactivation analysis of oligomeric structure of the H,K‐ATPase. J Biol Chem 263: 16189‐16189, 1988.
 64. Rabon EC, McFall TL, Sachs G. The gastric [H,K]ATPase:H+/ATP stoichiometry. J Biol Chem 257: 6296‐6296, 1982.
 65. Rabon EC, Reuben MA. The mechanism and structure of the gastric H,K‐ATPase. Annu Rev Physiol 52: 321‐321, 1990.
 66. Rabon EC, Smillie K, Seru V, Rabon R. Rubidium occlusion within tryptic peptides of the H,K‐ATPase. J Biol Chem 268: 8012‐8012, 1993.
 67. Reenstra WW, Forte JG. H+/ATP stoichiometry for the gastric (K+ + H+)‐ATPase. J Membr Biol 61: 55‐55, 1981.
 68. Reuben MA, Lasater LS, Sachs G. Characterization of a beta subunit of the gastric H+/K(+)‐transporting ATPase. Proc Natl Acad Sci U S A 87: 6767‐6767, 1990.
 69. Richter JE, Kahrilas PJ, Johanson J, Maton P, Breiter JR, Hwang C, Marino V, Hamelin B, Levine JG. Efficacy and safety of esomeprazole compared with omeprazole in GERD patients with erosive esophagitis: A randomized controlled trial. Am J Gastroenterol 96: 656‐656, 2001.
 70. Sachs G, Shin JM, Besancon M, Prinz C. The continuing development of gastric acid pump inhibitors. Aliment Pharmacol Ther 7: 4‐4, discussion 29‐31, 1993.
 71. Shin JM, Besancon M, Prinz C, Simon A, Sachs G. Continuing development of acid pump inhibitors: Site of action of pantoprazole. Aliment Pharmacol Ther 8: 11‐11, 1994.
 72. Shin JM, Besancon M, Simon A, Sachs G. The site of action of pantoprazole in the gastric H+/K(+)‐ATPase. Biochim Biophys Acta 1148: 223‐223, 1993.
 73. Shin JM, Cho YM, Sachs G. Chemistry of covalent inhibition of the gastric (H+, K+)‐ATPase by proton pump inhibitors. J Am Chem Soc 126: 7800‐7800, 2004.
 74. Shin JM, Goldshleger R, Munson KB, Sachs G, Karlish SJ. Selective Fe2+‐catalyzed oxidative cleavage of gastric H+,K+‐ATPase: Implications for the energy transduction mechanism of P‐type cation pumps. J Biol Chem 276: 48440‐48440, 2001.
 75. Shin JM, Homerin M, Domagala F, Ficheux H, Sachs G. Characterization of the inhibitory activity of tenatoprazole on the gastric H+,K+‐ATPase in vitro and in vivo. Biochem Pharmacol 71: 837‐837, 2006.
 76. Shin JM, Sachs G. Identification of a region of the H,K‐ATPase alpha subunit associated with the beta subunit. J Biol Chem 269: 8642‐8642, 1994.
 77. Shin JM, Sachs G. Dimerization of the gastric H+, K(+)‐ATPase. J Biol Chem 271: 1904‐1904, 1996.
 78. Shin JM, Sachs G. Differences in binding properties of two proton pump inhibitors on the gastric H(+),K(+)‐ATPase in vivo. Biochem Pharmacol 68: 2117‐2117, 2004.
 79. Shin JM, Sachs G. Functional consequences of the oligomeric form of the membrane‐bound gastric H,K‐ATPase. Biochemistry 44: 16321‐16321, 2005.
 80. Shin JM, Sachs G, Cho YM, Garst M. 1‐Arylsulfonyl‐2‐(pyridylmethylsulfinyl) benzimidazoles as new proton pump inhibitor prodrugs. Molecules 14: 5247‐5247, 2009.
 81. Shull GE. cDNA cloning of the beta‐subunit of the rat gastric H,K‐ATPase. J Biol Chem 265: 12123‐12123, 1990.
 82. Shull GE, Lingrel JB. Molecular cloning of the rat stomach (H+ + K+)‐ATPase. J Biol Chem 261: 16788‐16788, 1986.
 83. Simon WA, Budingen C, Fahr S, Kinder B, Koske M. The H+, K(+)‐ATPase inhibitor pantoprazole (BY1023/SK&F96022) interacts less with cytochrome P450 than omeprazole and lansoprazole. Biochem Pharmacol 42: 347‐347, 1991.
 84. Simon WA, Herrmann M, Klein T, Shin JM, Huber R, Senn‐Bilfinger J, Postius S. Soraprazan: Setting new standards in inhibition of gastric acid secretion. J Pharmacol Exp Ther 321: 866‐866, 2007.
 85. Skrabanja AT, van der Hijden HT, De Pont JJ. Transport ratios of reconstituted (H+ + K+)‐ATPase. Biochim Biophys Acta 903: 434‐434, 1987.
 86. Smith GS, Scholes PB. The H+/ATP stoichiometry of the (H+ +K+)‐ATPase of dog gastric microsomes. Biochim Biophys Acta 688: 803‐803, 1982.
 87. Sorensen TL, Olesen C, Jensen AM, Moller JV, Nissen P. Crystals of sarcoplasmic reticulum Ca(2+)‐ATPase. J Biotechnol 124: 704‐704, 2006.
 88. Swarts HG, Koenderink JB, Willems PH, Krieger E, De Pont JJ. Asn792 participates in the hydrogen bond network around the K+‐binding pocket of gastric H,K‐ATPase. J Biol Chem 280: 11488‐11488, 2005.
 89. Takahashi M, Kondou Y, Toyoshima C. Interdomain communication in calcium pump as revealed in the crystal structures with transmembrane inhibitors. Proc Natl Acad Sci U S A 104: 5800‐5800, 2007.
 90. Toh BH, Gleeson PA, Simpson RJ, Moritz RL, Callaghan JM, Goldkorn I, Jones CM, Martinelli TM, Mu FT, Humphris DC, et al. The 60‐ to 90‐kDa parietal cell autoantigen associated with autoimmune gastritis is a beta subunit of the gastric H+/K(+)‐ATPase (proton pump). Proc Natl Acad Sci U S A 87: 6418‐6418, 1990.
 91. Toyoshima C, Asahi M, Sugita Y, Khanna R, Tsuda T, MacLennan DH. Modeling of the inhibitory interaction of phospholamban with the Ca2+ ATPase. Proc Natl Acad Sci U S A 100: 467‐467, 2003.
 92. Toyoshima C, Nakasako M, Nomura H, Ogawa H. Crystal structure of the calcium pump of sarcoplasmic reticulum at 2.6 Å resolution. Nature 405: 647‐647, 2000.
 93. Toyoshima C, Nomura H. Structural changes in the calcium pump accompanying the dissociation of calcium. Nature 418: 605‐605, 2002.
 94. Toyoshima C, Nomura H, Sugita Y. Crystal structures of Ca2+‐ATPase in various physiological states. Ann N Y Acad Sci 986: 1‐1, 2003.
 95. Tyagarajan K, Lipniunas PH, Townsend RR, Forte JG. The N‐linked oligosaccharides of the beta‐subunit of rabbit gastric H,K‐ATPase: Site localization and identification of novel structures. Biochemistry 36: 10200‐10200, 1997.
 96. Tyagarajan K, Townsend RR, Forte JG. The Beta‐subunit of the rabbit H,K‐ATPase: A glycoprotein with all terminal lactosamine units capped with alpha‐linked galactose residues. Biochemistry 35: 3238‐3238, 1996.
 97. Vagin O, Denevich S, Sachs G. Plasma membrane delivery of the gastric H,K‐ATPase: The role of beta‐subunit glycosylation. Am J Physiol Cell Physiol 285: C968‐C976, 2003.
 98. Vagin O, Kraut JA, Sachs G. Role of N‐glycosylation in trafficking of apical membrane proteins in epithelia. Am J Physiol Renal Physiol 296: F459‐F469, 2009.
 99. Vagin O, Turdikulova S, Sachs G. The H,K‐ATPase beta subunit as a model to study the role of N‐glycosylation in membrane trafficking and apical sorting. J Biol Chem 279: 39026‐39026, 2004.
 100. Vagin O, Turdikulova S, Yakubov I, Sachs G. Use of the H,K‐ATPase beta subunit to identify multiple sorting pathways for plasma membrane delivery in polarized cells. J Biol Chem 280: 14741‐14741, 2005.
 101. Walderhaug MO, Post RL, Saccomani G, Leonard RT, Briskin DP. Structural relatedness of three ion‐transport adenosine triphosphatases around their active sites of phosphorylation. J Biol Chem 260: 3852‐3852, 1985.
 102. Wallmark B, Brandstrom A, Larsson H. Evidence for acid‐induced transformation of omeprazole into an active inhibitor of (H+ + K+)‐ATPase within the parietal cell. Biochim Biophys Acta 778: 549‐549, 1984.
 103. Wallmark B, Briving C, Fryklund J, Munson K, Jackson R, Mendlein J, Rabon E, Sachs G. Inhibition of gastric H+,K+‐ATPase and acid secretion by SCH 28080, a substituted pyridyl(1,2a)imidazole. J Biol Chem 262: 2077‐2077, 1987.
 104. Wallmark B, Stewart HB, Rabon E, Saccomani G, Sachs G. The catalytic cycle of gastric (H+ + K+)‐ATPase. J Biol Chem 255: 5313‐5313, 1980.
 105. Zimmermann PJ, Buhr W, Brehm C, Palmer AM, Feth MP, Senn‐Bilfinger J, Simon WA. Novel indanyl‐substituted imidazo[1,2‐a]pyridines as potent reversible inhibitors of the gastric H+/K+‐ATPase. Bioorg Med Chem Lett 17: 5374‐5374, 2007.
Further Reading
 1. Ogawa H, Shinoda T, Cornelius F, Toyoshima C. Crystal structure of the sodium‐potassium pump (Na+,K+‐ATPase) with bound potassium and ouabain. Proc Natl Acad Sci U S A 106: 13742‐13742, 2009.
 2. Shin JM, Munson K, Vagin O, Sachs G. The gastric HK‐ATPase: Structure, function, and inhibition. Pflugers Arch 457: 609‐609, 2009.
 3. Shin JM, Vagin O, Munson K, Kidd M, Modlin IM, Sachs G. Molecular mechanisms in therapy of acid‐related diseases. Cell Mol Life Sci 65: 264‐264, 2008.
 4. Shinoda T, Ogawa H, Cornelius F, Toyoshima C. Crystal structure of the sodium‐potassium pump at 2.4 Å resolution. Nature 459: 446‐446, 2009.
 5. Vagin O, Tokhtaeva E, Yakubov I, Shevchenko E, Sachs G. Inverse correlation between the extent of N‐glycan branching and intercellular adhesion in epithelia. Contribution of the Na,K‐ATPase beta1 subunit. J Biol Chem 283: 2192‐2192, 2008.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Jai Moo Shin, Keith Munson, George Sachs. Gastric H+,K+‐ATPase. Compr Physiol 2011, 1: 2141-2153. doi: 10.1002/cphy.c110010