Comprehensive Physiology || Gastric H + ,K + ...

13
Gastric H + ,K + -ATPase Jai Moo Shin, *1 Keith Munson, 1 and George Sachs 1 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 H 3 O + -binding sites in the middle of the membrane domain and H 3 O + secretion depends on a conformational change involving Lys 791 insertion into the second H 3 O + 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 Lys 791 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 E 2 form of this ATPase. C 2011 American Physiological Society. Compr Physiol 1:2141-2153, 2011. Introduction About 160-mM HCl acid is secreted by the parietal cells of the stomach. Two acid-stimulatory receptors, the H 2 -receptor and the muscarinic M 3 receptor, are present on the parietal cell. After histamine or acetylcholine binds to the corresponding receptor, the morphology of parietal cell changes. This mor- phological change drives gastric H + ,K + -ATPase and vesi- cles containing a K + conductance pathway from cytoplas- mic membranes to the (apical) membrane of the secretory canaliculus, where the pump exchanges cytoplasmic protons for extracytoplasmic potassium ions in an electroneutral man- ner. This activity is responsible for the elaboration of HCl by the parietal cell of the gastric mucosa. The transport of H + across the apical membrane is by means of conforma- tional changes in the protein driven by cyclic phosphoryla- tion and dephosphorylation of the catalytic subunit of the ATPase. This mechanism places the gastric H + ,K + -ATPase in the P 2 ATPase family, containing the Ca 2+ - and the Na + , K + -ATPases. The gastric H + ,K + -ATPase is composed of two subunits, the α subunit and the β subunit. The α subunit with molecular mass of about 100 kDa has the catalytic site and the β subunit with peptide mass of 35 kDa is strongly but noncovalently associated with the α subunit. The β subunit has six or seven N-glycosylated sites exposed to the extracytoplasmic surface (Figure 1). Since the gastric H + ,K + -ATPase is responsible for the final step of acid secretion, this enzyme has been a therapeutic target in treatment of acid-related disease for the last 20 years. The first class of the pump enzyme inhibitor contains the substituted benzimidazoles which are the most effective class of anti-ulcer drugs used in the treatment of a variety of acid- related diseases of the upper gastrointestinal tract. A second class of inhibitor which competes with extracytoplasmic K + is being developed to get better inhibition. Structure of the Gastric H + ,K + -ATPase The primary sequences of the α subunits deduced from cDNA have been defined for rat (82), pig (39), and rabbit (5). The gene sequence for human H + ,K + -ATPase α sub- units have also been determined (41, 55). The human gastric H + ,K + -ATPase gene has 22 exons and encodes a protein of 1035 residues including the initiator methionine residue (Mr = 114,047). These H + ,K + -ATPase α subunits show high homology (60 % identity) with the Na + ,K + -ATPase cat- alytic α subunits and lesser homology with the sarcoplamic * Correspondence to [email protected] 1 Department of Physiology and Medicine, University of California at Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California Published online, October 2011 (comprehensivephysiology.com) DOI: 10.1002/cphy.c110010 Copyright C American Physiological Society Volume 1, October 2011 2141

Transcript of Comprehensive Physiology || Gastric H + ,K + ...

Page 1: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPaseJai Moo Shin,*1 Keith Munson,1 and George Sachs1

ABSTRACTThe gastric H+,K+-ATPase is responsible for gastric acid secretion. This ATPase is composed of twosubunits, the catalytic α subunit and the structural β subunit. The α subunit with molecular mass ofabout 100 kDa has 10 transmembrane domains and is strongly associated with the β subunit witha single transmembrane segment and a peptide mass of 35 kDa. Its three-dimensional structureis based on homology modeling and site-directed mutagenesis resulting in a proton extrusionand K+ reabsorption model. There are three conserved H3O+-binding sites in the middle of themembrane 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 ofprotons at a concentration of 160 mM. K+ countertransport involves binding to this site after therelease of protons with retrograde displacement of Lys791 and then K+ transfer to E343 and exitto the cytoplasm. This ATPase is the major therapeutic target in treatment of acid-related diseasesand there are several known luminal inhibitors allowing analysis of the luminal vestibule. Oneclass contains the acid-activated covalent, thiophilic proton pump inhibitors, the most effectiveof current acid-suppressive drugs. Their binding sites and trypsinolysis allowed identification ofall ten transmembrane segments of the ATPase. In addition, various K+-competitive inhibitors ofthe ATPase are being developed, with the advantage of complete and rapid inhibition of acidsecretion independent of pump activity and allowing further refinement of the structure of theluminal vestibule of the E2 form of this ATPase. C© 2011 American Physiological Society. ComprPhysiol 1:2141-2153, 2011.

IntroductionAbout 160-mM HCl acid is secreted by the parietal cells of thestomach. Two acid-stimulatory receptors, the H2-receptor andthe muscarinic M3 receptor, are present on the parietal cell.After histamine or acetylcholine binds to the correspondingreceptor, the morphology of parietal cell changes. This mor-phological change drives gastric H+,K+-ATPase and vesi-cles containing a K+ conductance pathway from cytoplas-mic membranes to the (apical) membrane of the secretorycanaliculus, where the pump exchanges cytoplasmic protonsfor extracytoplasmic potassium ions in an electroneutral man-ner. This activity is responsible for the elaboration of HClby the parietal cell of the gastric mucosa. The transport ofH+ across the apical membrane is by means of conforma-tional changes in the protein driven by cyclic phosphoryla-tion and dephosphorylation of the catalytic subunit of theATPase. This mechanism places the gastric H+,K+-ATPasein the P2 ATPase family, containing the Ca2+- and the Na+,K+-ATPases.

The gastric H+,K+-ATPase is composed of two subunits,the α subunit and the β subunit. The α subunit with molecularmass of about 100 kDa has the catalytic site and the β subunitwith peptide mass of 35 kDa is strongly but noncovalentlyassociated with the α subunit. The β subunit has six or sevenN-glycosylated sites exposed to the extracytoplasmic surface(Figure 1).

Since the gastric H+,K+-ATPase is responsible for thefinal step of acid secretion, this enzyme has been a therapeutic

target in treatment of acid-related disease for the last 20 years.The first class of the pump enzyme inhibitor contains thesubstituted benzimidazoles which are the most effective classof anti-ulcer drugs used in the treatment of a variety of acid-related diseases of the upper gastrointestinal tract. A secondclass of inhibitor which competes with extracytoplasmic K+

is being developed to get better inhibition.

Structure of the GastricH+,K+-ATPaseThe primary sequences of the α subunits deduced fromcDNA have been defined for rat (82), pig (39), and rabbit(5). The gene sequence for human H+,K+-ATPase α sub-units have also been determined (41, 55). The human gastricH+,K+-ATPase gene has 22 exons and encodes a proteinof 1035 residues including the initiator methionine residue(Mr = 114,047). These H+,K+-ATPase α subunits show highhomology (∼60 % identity) with the Na+,K+-ATPase cat-alytic α subunits and lesser homology with the sarcoplamic

*Correspondence to [email protected] of Physiology and Medicine, University of California atLos Angeles, and VA Greater Los Angeles Healthcare System, LosAngeles, California

Published online, October 2011 (comprehensivephysiology.com)

DOI: 10.1002/cphy.c110010

Copyright C© American Physiological Society

Volume 1, October 2011 2141

Page 2: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

N domain A domain

Membrane domain

P domain

Ion-binding site

Beta subunit (pink)

Cytoplasmic domain

Luminal domainN146

N193

N130 N221

N161

N99

M5/M6 loop

Figure 1 The structure of the gastric H+,K+-ATPase. The modeledthree-dimensional structure of the H+,K+-ATPase, HKzxe based on theNa+,K+-ATPase in the E2P state. The α subunit has three lobes, N(ATP binding), P (phosphorylation), and A (activation) domains in thecytoplasmic domain, and ten transmembrane segments in the mem-brane domain. The gastric β subunit has short cytoplasmic region,one transmembrane segment, and a heavily glycosylated extracellularregion. In this model, the backbone of protein with carbohydrate at-tached Asn sites is shown. Amino acid sequence number is based onpig H+,K+-ATPase.

reticulum Ca2+-ATPase (SERCA) (22%) but more structuresare known for the latter hence more homology modeling hasbeen done with this pump than with the Na+,K+-ATPase.The hog gastric H+,K+-ATPase α subunit sequence deducedfrom its cDNA consists of 1034 amino acids and has a Mrof 114,285. The rat gastric H+,K+-ATPase consists of 1033amino acids and has a Mr of 114,012, and the rabbit gastricH+,K+-ATPase consists of 1035 amino acids, with a Mr of114,201. The sequence based on the known N-terminal aminoacid sequence is one less than the cDNA-derived sequence dueto loss of the N-terminal methionine and this numbering isadopted in this review (34). The homology of the H+,K+-ATPase α subunits among the animal species is extremelyhigh (over 97% identity).

The gastric α subunit has conserved sequences along withthe other P2-type ATPases, the SERCA and the Na+,K+-ATPase, for the ATP-binding site and the phosphorylationsite. The phosphorylation site was observed to be at Asp386

(101), which is well conserved in other P-type ATPases. Themembrane topology of the α subunit has been shown to con-tain 10 membrane spanning segments (5, 6).

The β subunit was first identified in work analyzing thecarbohydrates of gastric membranes (19). Postembeddingelectron microscopic staining techniques showed that wheatgerm agglutinin staining occurred on the inside face of the

inside out gastric vesicles. The same β subunit was found bydetermining the nature of the antigen in autoimmune gastri-tis (90). Using a lectin affinity chromatography, the H+,K+-ATPase α subunit was copurified with the β subunit, showingthat the α subunit interacts with the β subunit (10, 59). Bycross-linking with low concentrations of glutaraldehyde, theα subunit was shown to be closely associated with the β

subunit (20, 65).The primary sequences of the β subunits have been

reported for rabbit (68), hog (90), rat (12, 40, 56, 81),mouse (11, 48), and human (38). The β subunit con-sists of about 290 amino acids with a single transmem-brane segment, which is located at the region near the N-terminus. There are three disulfide bridges in the luminalregion of the β subunit. Seven putative N-glycosylated sites(AsnXaaSer and AsnXaaThr) are found in rabbit (68), thatare conserved in rat and human, but only six putative N-glycosylation sites are present in the hog gastric β subunit.The structure of N-linked oligosaccharides of the β sub-unit of rabbit gastric H+,K+-ATPase was identified (95, 96).All seven N-linked AsnX(Ser/Thr) sites at positions 99,103, 130, 146, 161, 193, and 222 were fully glycosylated.Asn99 was modified exclusively with oligomannosidic-typestructures, Man6GlcNAc2-Man8GlcNAc2, and Asn193 hasMan5GlcNAc2-Man8GlcNAc2 and lactosamine-type struc-tures. Asn 103, 146, 161, and 222 contain lactosamine-typestructures. All the branches of the lactosamine-type structureterminate with Galα-Galβ-GlcNAc extensions (95). The roleof the carbohydrate chains in membrane trafficking has beenstudied in HEK-293 cells and in polarized cells such as LillyLaboratories Cell – Porcine Kidney (LLC-PK) and Madin-Darby canine kidney (MDCK) cells (3, 97-100). In polarizedcells, the steady-state distribution of the H+,K+-ATPase β

subunit depends on the balance between (i) direct sorting fromthe trans-Golgi network (TGN), (ii) secondary associativesorting with a partner protein, and (iii) selective trafficking.

Studies on the expression of the β subunit of the gas-tric H+,K+-ATPase in LLC-PK1 cells showed that mutationof each of the seven N-glycosylation sites of rabbit gastricH+,K+-ATPase provided differential distribution of the sub-unit in endoplasmic reticulum (ER), Golgi, or plasma mem-brane. For example, the β subunit of wild type expressed inLLC-PK1 cells is distributed between the plasma membraneand the intracellular compartments even in the absence ofthe α subunit of the H+,K+-ATPase. At steady state, ∼40%of the protein is detected in the ER, and the remaining 60%is distributed between the plasma membrane, Golgi, TGN,and endosomes. Removal of the seventh N-glycosylation site,Asn222, leads to virtually total retention of the subunit inthe ER. Mutation of N1 increases the amount retained inthe ER to 75%. Similarly, mutation of Asn130 and Asn161also significantly increases the relative amount of the ER-resident fraction, but to a lesser extent. However, mutation ofAsn103, Asn146, or Asn193 does not appreciably alter thequantity of protein retained in the ER. These findings indicatethat N-glycans at Asn99, Asn130, Asn161, and Asn222 are

2142 Volume 1, October 2011

Page 3: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

important for apical membrane trafficking of this β subunit. Itis remarkable that the same N-glycans (Asn99, Asn130, andAsn161) of the H+,K+-ATPase β subunit that are importantfor ER-to-Golgi trafficking are also important for apical de-livery and membrane retention of the protein. These resultssuggest that, similar to ER-to-Golgi trafficking, both apicaldelivery and apical membrane retention of the H+,K+-ATPaseβ subunit are lectin dependent and only specific N-glycansare accessible to these lectins (99).

Another role of the H+,K+-ATPase β subunit is stabi-lizing the E2 P conformation. The N-terminal cytoplasmicregion of the β subunit is known to be responsible for this(16). Two-dimensional crystals of the enzyme at 6.5 A res-olution showed that the N-terminal tail of the β subunit isin direct contact with the phosphorylation domain of the α

subunit. This interaction may hold the phosphorylation do-main in place, thus stabilizing the enzyme conformation andpreventing the reverse reaction of the transport cycle. Indeed,truncation of the β subunit N-terminus did allow reversal oftransport. These results suggest that the β subunit N-terminusprevents the reverse reaction from E2P to E1P, which is likelyto be relevant for the generation of the large in vivo protongradient (2).

Regions of Association Between theα and β SubunitsThe α subunit of the H+,K+-ATPase is strongly associatedwith the β subunit (76). Specifically, the region of the se-quence Arg898 to Arg922 in the α subunit has strong inter-action with the extracytoplasmic domain of the β subunit.By using yeast two-hybrid analysis, two different sequencesin the β subunit Gln64 to Asn130 and Ala156 to Arg188 wereidentified as associated with the extracytoplasmic sequenceof the β subunit. These data enabled identification of majorassociative regions of the α-β subunits of the H+,K+-ATPase(43, 77). There has been much suggestive evidence that theα-β heterodimeric H+,K+-ATPase exists in the membrane asan (α-β)2 oligomer. Such evidence includes target size (63) inthe membrane and unit cell size of the enzyme in membraneimbedded two-dimensional crystals (22). More recent evi-dence showed that the gastric H+,K+-ATPase may functionas an (α-β)2 dimeric heterodimer (79). The key observationin these studies was that the membrane preparation showedfull stoichiometry with respect to ATP binding (1 mol/molα-β) and half stoichiometry with respect to inhibitor bind-ing and phosphorylation suggesting an (α-β)2 oligomer. Theactual oligomeric state of the Na+,K+- and H+,K+-ATPasesnevertheless remains controversial. It was shown that the α-βprotomer of the Na+,K+-ATPase is active in its detergent-solubilized form but the membrane oligomeric form is un-proven (13). It should be noted that the SERCA pump crys-tallizes as a monomer after detergent solubilization but thisof course is unrelated to the oligomeric state in the membrane(92).

Kinetics of the gastric H+,K+-ATPaseThe H+,K+-ATPase exchanges intracellular hydrogen ionsfor extracellular potassium ions by consuming ATP. TheH+/ATP ratio was independent of external KCl and ATP con-centration (64). However, the stoichiometry of H+ per ATPwas different depending on luminal pH. The H+ for K+ stoi-chiometry of the H+,K+-ATPase per ATP hydrolyzed was one(42, 67, 86) at low pH, and two (64, 85) at neutral pH or nearneutral pH. This could be explained by retained protonationat one of the ion binding site carboxylic acids at a pH wellbelow the pKa of the carboxylic amino acid in this site.

The H+,K+-ATPase has many reaction steps, similar tothose of the Na+,K+-ATPase (104). The H+,K+-ATPasebinds the hydronium ion on the cytoplasmic side at highaffinity (E1 conformation). The initial step is the reversiblebinding of ATP to the enzyme in the absence of added K+

ion, followed by a Mg2+ (and hydronium)-dependent trans-fer of γ-phosphate of ATP to Asp386 of the catalytic sub-unit (E1P•H+). Following phosphorylation, the conformationchanges from E1P•H3O+ to the E2P•H30+ form, which hashigh affinity for K+ and low affinity for H3O+ allowing re-lease of H3O+ and binding of K+ from the extracytoplasmicsurface of the enzyme. Breakdown of the E2P form requiresK+ or its congeners on the outside face of the enzyme. Withdephosphorylation, the E1K+ conformation is produced witha low affinity for K+, releasing K+ to the cytoplasmic side,allowing rebinding of H3O+ (Figure 2).

The addition of K+ to the enzyme-bound acyl phosphateresults in a biphasic dephosphorylation. The faster initial stepis dependent on the concentration of K+, whereas the slowerstep is not affected by K+ concentration. The second phase

E1•Mg

E1•Mg•(K+) E1[ATP]•Mg•(H+)

E1P•Mg•(H+)

E2P•Mg•(H+)H+ K+

E2•Mg•(Kocc+)

E2P•Mg•(K+)

K+ ATP

Pi

ADP

Figure 2 The catalytic cycle of the gastric H+,K+-ATPase. A hydro-nium ion binds to the cytoplasmic surface of the enzyme, and MgATPphosphorylates the protein at Asp386 to form the first ion transportintermediate in the E1P form. The E1P form then converts by a confor-mational change to the second ion transport form, E2P, with the ion sitenow exposed to the exterior and hydronium is released at pH ∼1.0. Tothis form, K+ binds from the outside surface to the same region fromwhich the hydronium was released, and the enzyme dephosphorylates,and then K+ is trapped within the membrane domain in the occludedform (and a similar form is postulated for the hydronium in the outwardstep of the cycle). The K+ is then deoccluded allowing reformation ofthe E1 form of the enzyme with the ion site now again facing the cy-toplasm and K+ is displaced when ATP is bound. In this model, thenaked H+ is used instead of the hydronium ion (H3O+). This modelreflects a stoichiometry of 1H/1K/1ATP.

Volume 1, October 2011 2143

Page 4: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

of EP breakdown is accelerated in the presence of K+ but,at K+ concentrations exceeding 500 μM, the rate becomesindependent of K+ concentration. This shows that two formsof EP exist. The first form, presumably E1P is K+ insensitiveand converts spontaneously in the rate-limiting step to E2P, theK+ sensitive form. ATP binding to the H+,K+-ATPase occursin both the E1 and the E2 state, but with a lower affinity in E2

state (9). The Mg2+ remains occluded in the P domain nearAsp730 (74) until dephosphorylation (44, 45, 66) in contrastto the Na+,K+-ATPase, where the Mg ion is released fromE2P (62).

The sequence and mechanistic similarity among the P2-type ATPases, the H+,K+-ATPase, the Na+,K+-ATPase, andthe Ca-ATPase allows extension of the structural results foundfor any individual pump to the other family members. Becausemany crystal structures of the SERCA ATPase have beensolved (27, 60, 61, 87, 89, 91-94), the conformational interme-diates of the catalytic cycle of Ca-ATPase have become wellunderstood at a molecular level. Like the H+,K+-ATPase al-pha subunit, SERCA1a is composed of 10 transmembrane he-lices (M1-M10) and 3 cytosolic domains: N (nucleotide bind-ing), P (phosphorylation), and A (actuator). In its E1 form,

SERCA1a binds two Ca ions with high affinity in the middleof the 10 transmembrane helices, whereas in the E2 form thecation binding sites are associated with protons. Transitionstates of phosphorylation and dephosphorylation are coupledto occlusion of bound calcium and protons, respectively. Inthe forward direction of the functional cycle, the Ca2 E1∼Pstate, formed by reaction of Ca2 E1 with ATP, is converted toan E2P state, accompanied by translocation of Ca2+ from thecytosolic to luminal orientation of the ion-binding site. TheE2P state is then dephosphorylated along with counter trans-port of protons. All steps in the reaction cycle are reversible;thus ADP stimulates the dephosphorylation of the Ca2 E1∼Pstate through regeneration of ATP, and Ca2+/Ca2+ exchangeis observed at high vesicular levels of Ca2+ (60).

A similar set of steps was generated for the H+,K+-ATPase using this ATPase as the basis for homology modeling(35, 49-51).

A detailed model of H+ and K+ transport was proposed asfollows. In panel 1 of Figure 3, Lys-NH3

+-791 is above the hy-dronium site H2. With phosphorylation and formation of E1P,K791 moves into the second hydronium site containing E795,E820, and D824 forming E2P and the lys-NH3

+ displaces the

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 panel1 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 2after phosphorylation, lys791 displaces the hydronium from site 2 in the E1P form (arrow). Then in panel 3 K+ binds to site 2 displacing lys791generating the E2P form and in panel 4 K+ moves into site 3 allowing exit to the cytoplasm along TM3.

2144 Volume 1, October 2011

Page 5: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

H3O+ outward (Fig. 3, panel 2). At pH < 3.0, the hydroniumat H1 remains bound due to the higher pKa of this site con-taining so the K+ binds only at H2, displacing K791 upward(Fig. 3, panel 3) to form the E2P·K+

occ occluded state. Withdephosphorylation, K+ moves into H3, where E820, E343,and the carbonyls of V337 and V341 are reoriented to allowexit of K+ along TM3 (arrow, Fig. 3, panel 4). Critical tothe coupling of ATP binding and subsequent phosphorylationis the conformational change, where the A domain rotatesclockwise to bring the N and P domains into apposition withthe bound ATP acting like a hook to retain this conformationuntil dephosphorylation (1, 58, 92, 94).

Recently, two-dimensional crystals of the H+,K+-ATPasewere analyzed in the presence of the phosphate analog BeFand an inhibitor SCH28080 (1). This structure of the E2Pconformation was similar to that of Ca-ATPase. The overallstructure of the present EM map (Fig. 4) shows characteristicfeatures of the ADP-insensitive E2P conformation, to whichSCH28080 is preferentially bound. The BeF-bound phospho-rylation site (Asp385) at the P domain is covered by the A

domain, and the ADP-bound N domain is separated from theP domain, thus the bound phosphate analog seems to be iso-lated from both ADP and the bulk solution. At this resolution,the side chains cannot be identified and their orientation wasbased on the homology model above.

Potassium ion pathway

The model generated for the E2P conformation showed achannel for the passage of K+ from the luminal vestibule tothe ion occlusion site near the middle of the membrane. TheM5/M6 loop presents the first protein segment encounteredfor passage of the ion into the channel. The only pathway tothe site accessed by the ion is between the carbonyl oxygenatoms of L811 and G812 and the sulfur of C813. This leadsto apparent binding to these two carbonyls and two moleculesof water. This appears from the model to be the initial entrysite into the channel.

At close to neutral pH with deprotonation of both E820and D824 at sites 1 and 2, two K+ can bind to provide equal

A-M2

A domain

SCH28080

β-subunit

βNt

αNt

(A) (B)

N

N domain

P

P domain

TM

AADP

ADP

linker

Cytoplasm

Membrane

Lumen

Figure 4 SCH28080-bound E2P structure of the gastric H+,K+-ATPase analyzed at 7 A resolution. (A) The structure of the H+,K+-ATPaseshowing cytoplasmic-side up. Surface represents the EM density map with superimposed homology model of H+,K+-ATPase (ribbon). BoundSCH28080 at the luminal cavity, and ADP at the N domain are shown as sticks. A wheat box indicates approximate location of the lipidbilayer. Color code: A domain, blue; P domain, green; N domain, yellow; TM domain, light blue (surface); and β subunit, red. Color for theTM helices of the homology model gradually changes from M1 (blue) to M10 (red). (B) Conformational change of the cytoplasmic domainseen 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 figureis 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 andbelow.

Volume 1, October 2011 2145

Page 6: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

Figure 5 K+ pathway in the H+,K+-ATPase. The image illustrates theentry path for K+ (illustrated as a series of violet spheres) between M4,M5, M6, and M8 in the E2P conformation as determined by moleculardynamics simulation (M3 in the foreground is omitted for clarity) (35,50). The arrival of K+ at the top of this path is predicted to destabilizethe interaction of K791 with E820 and E795, and initiate the confor-mational changes leading to release of phosphate at the active siteand conversion back to E1.

stoichiometry to the two extruded protons.## With the absenceof deprotonation of D824 due to its higher pKa, the stoichiom-etry will drop to 1K+/1ATP at low pH, with the initial siteof K+ occupancy at E820 and E795 and then movement toE343 with reprotonation of E820. Such a model would beconsistent with the K+ independence of turnover of the E820mutant (88).

A notable feature of the luminal face of the enzyme mod-eled in the E2 form is the presence of a luminal vestibulebounded by TM4, TM5, and TM6 and the connecting exo-plasmic loops between TM3-TM4 and TM5-TM6 containingcysteine 813 as part of the loop between TM5 and TM6.This vestibule is a natural exit and entry point for transportedcations and also provides access to the different classes ofinhibitors of the gastric H+,K+-ATPase and is illustrated inthe model of Figure 5. Analysis of the SERCA Ca2+-ATPaseshows that this vestibule is closed in the E1 form and open inthe E2 form (47).

A three-dimensional (3D) model of the K+ pathway fromlumen to cytoplasm based on molecular dynamics is shownin Figure 6.

Inhibitors of the H+,K+-ATPaseTwo types of H+,K+ ATPase inhibitors have been devel-oped that react exclusively with the extracytoplasmic surface

Figure 6 Proposed K+ pathway through the pump srCa-ATPase ho-mology model. Red to pink is area of restriction including the occlusionsite. White to blue less occlusion. Arrows highlight entry and exit path-ways for K+.

of the gastric H+,K+-ATPase. One commercially availableclass of inhibitors is the substituted benzimidazoles such asomeprazole, lansoprazole, rabeprazole, pantoprazole, and es-omeprazole, the proton pump inhibitor class (PPI). The otherclass contains a series of K+-competitive reagents, includingimidazo-pyridines that are also known to react on the outsidesurface of the pump competing with potassium ion. This typeis called either the acid pump antagonist (APA) or potassium-competitive acid blocker (P-CAB) class.

The substituted benzimidazole inhibitorsThe first antisecretory inhibitor of this class was 2-(pyridylmethylsulfinyl)benzimidazole (17). This compoundis a weak base ∼pKa 4. The H+,K+-ATPase in the parietalcell secretes acid into the secretory canaliculus generating apH of <1.0 in lumen of this structure. The acidity of thisspace allows accumulation of weak bases of this pKa. Weakbases of a pKa less than 4.0 can be accumulated only in thisacidic space and no other acidic space in the body. Then, thiscompound is rapidly activated by the high acidity and inhibitsacid secretion by binding to the cysteines accessible to theactivated form.

Since a substituted benzimidazole was first reportedto inhibit the H+,K+-ATPase (17), many inhibitors of theH+,K+-ATPase have been synthesized (Figure 7). The

2146 Volume 1, October 2011

Page 7: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

Figure 7 Proton pump inhibitors.

first pump inhibitor used clinically was 2-[[3,5-dimethyl-4-methoxypyridin-2-yl]methylsulfinyl]-5-methoxy-1H-benzimidazole, omeprazole (102), and this was followed byother covalent binding inhibitors all belonging to the substi-tuted benzimidazole family (18, 54, 83). These reagents areweak base, acid activated compounds, which form cationicsulfenamides or sulfenic acids in highly acidic environments.The thiophilic compounds formed react with the SH groupof cysteines in the ATPase to form relatively stable disulfides(Figure 8) (7, 8, 71-73). Since the pump generates acid onits extracytoplasmic surface, only those cysteines availablefrom that surface are accessible to these reagents if labelingis carried out under acid transporting conditions.

Omeprazole has a stoichiometry of 2 mol inhibitor boundper mol phosphoenzyme under acid transporting conditionsand is bound only to the α subunit even in vivo (31, 37, 73).The binding sites of omeprazole are cysteine 813 and 892(7). The sites of reaction of the different PPIs on the enzymediffer according to the particular PPI. However, all PPIs reactwith cysteine 813 in the loop between TM5 and TM6 thatfixes the enzyme in the E2 configuration. Lansoprazole reactswith cysteine 813 and cysteine 321, these being in the luminalvestibule (70), whereas pantoprazole and tenatoprazole reactwith cysteines 813 and 822 (72,73, 75, 78). The reaction withcysteine 822 confers a rather special property to the covalentlyinhibited enzyme, namely, irreversibility to reducing agentsin vitro and in vivo due to lack of accessibility of Cys822.

An innovation was introduced by the S-enantiomer ofomeprazole, esomeprazole, which has a slightly delayedmetabolism compared to the R-enantiomer. In comparativestudies, 40 mg esomeprazole was compared to 20 mg of theracemic omeprazole and some improvement in outcome was

observed (69). However, the active compound generated inacid from esomeprazole has no chiral center and is identicalto that formed from omeprazole.

The key issue in efficacy of this class of drug is their short60-90 min plasma half-life so that there is no drug presentat night even with twice a day meal time-related adminis-tration. Recently, the R-enantiomer of lansoprazole has beenintroduced with a delayed release formulation (30) and a sim-ilar strategy has been adopted for rabeprazole (32) to try toprovide night time drug levels.

There are several conceptual means of improving acid in-hibition with the H+,K+-ATPase as the target. Firstly, a newcore structure can be generated with slower metabolism toimprove the residence time in the blood hence exposure to ac-tive pumps. A delayed release formulation can be establishedalso with the aim of increasing exposure. A prodrug of a PPIcan be synthesized so as to delay absorption and thus also toincrease exposure. Or, a novel mechanism can be used wherepump inhibition is reversible.

Recently a novel formulation of the R-enantiomer of lan-soprazole has been introduced. This is a dual release formula-tion of 60 mg of the PPI with normal enteric coating releasingat around pH 5.0 and a coating labile at pH 7 releasing dexlan-soprazole some hours later.

A biphasic PK profile for this formulation is observed forfirst day treatment that is the same as day 5, showing clearsuperiority 4 to 8 h after dose but not during the night. Thisprofile might explain the small increase in median 24 h pHsince this reflects mainly the low pH at night.

There was no statistically significant difference in healingbetween the 60 mg dose and regular 30 mg lansoprazole.However, there was an improvement in the time above pH

Volume 1, October 2011 2147

Page 8: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

R1 R2

R4Bz-Py

Bz-PyH+

BzH+

Sulfenic acid Sulfenamide

A1

-PyH+

-PyBzH+

R3

N

+H+

+H+

-H+

-H+

NH

SON

R1 R2

R4

R3

N

NH

H

S

ON

R1H2O

H3OR2

R4

R3NNH

HS O

N +

+

-

-

+H+

-H+

R1

R2

R4 R3

N

NH

H

SON

+

+

R1

R1

H2O

R2

R4

R3

N

NH

Enz–SS

Enz-SH Enz-SH

N+

R2

R4

R3NN

S

N +

R1 R2

R4

R3

N

N

H

H

H

S

O

N

++

+

+

R1 R2

R4

R3

NNH

H

H

SON

+

+

Figure 8 The mechanism of activation of the PPIs shown in general struc-tural form. The substituent R1, R2, R3, and R4 of the general structure (Bz-Py)represent a substituent chosen from hydrogen, methoxy, methyl, and substi-tuted alkoxy group. Top part shows the protonation of the pyridine ring andthe second row of structures shows protonation also of the benzimidazolering. The bis-protonated forms are in equilibrium with the protonated ben-zimidazole and unprotonated pyridine. In brackets is shown the mechanismof activation whereby the 2C of the protonated benzimidazole reacts with theunprotonated fraction of the pyridine moiety that results in rearrangement toa permanent cationic tetracyclic sulfenic acid which in aqueous solute dehy-drates to form a cationic sulfenamide. Either of these thiophilic species canreact with the enzyme to form disulfides with one or more enzyme cysteinesaccessible from the luminal surface of the enzyme.

2148 Volume 1, October 2011

Page 9: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

4.0, 71% relative to 60% but mean pH was 4.55 relative tolansoprazole at 4.13 (30). This improvement is similar to whathas been claimed for esomeprazole.

Another method, not requiring alternative formulation, isto make a slowly absorbed derivative of a PPI that is absorbedthroughout the small intestine and not just the duodenumlike all the PPIs. Of various derivatives tested, a sulfonamidederivative, the phenoxyacetic acid sodium salt derivative ofomeprazole seems to be a candidate drug with several de-sirable properties (80). This compound is slowly absorbedthroughout the small intestine but then rapidly hydrolyzed inthe blood to omeprazole and the sulfonic acid. Only tracequantities of the intact molecule are found in humans henceits safety profile should resemble that of omeprazole. The ef-fect of various doses in man showed a linear dose relationshipwith no evidence for saturation hence movement across thegut is by simple diffusion.

There is great prolongation of the blood level of omepra-zole and as found for omeprazole there is increased bioavail-ability after 5 days dosing. But significantly, there is prolon-gation of the residence time of omeprazole in the blood sothat there is drug present at sufficient levels over 24 h after 5days administration (25).

Acid Pump Antagonists (orPotassium-Competitive Acid Blockers)This class of the inhibitor blocks acid pumping by inhibit-ing K+ competitively, so this class is called either APAsor P-CABs. The first core structure of an APA developedin 1980s was an imidazo-pyridine (28). A typical struc-ture of this class is SCH28080. Following SCH28080, manydifferent core structures were made such as piperidinopy-ridines (23), substituted 4-phenylaminoquinolines (26),pyrrolo[3,2-c]quinolines (36), guanidino-thiazoles (33), sub-stituted pyrimidines (21), and scopadulcic acid (4). Some nat-ural products such as cassigarol A (53) and naphthoquinone(14) also showed inhibitory activity.

SCH28080 was the first compound extensively studiedfor its inhibitory mechanism. SCH28080 inhibits the H+,K+-ATPase activity by competing with K+ (103). SCH28080binds to free enzyme extracytoplasmically in the absence of

Figure 9 The amino acids lining the luminal entrance (vestibule) tothe ion-binding sites of the H+,K+-ATPase that is the basis for dockingpyrrolo-pyridines suggesting critical amino acid residues responsiblefor high affinity and slow dissociation.

substrate to form E2 (SCH28080) complexes. SCH28080 in-hibits ATPase activity with high affinity in the absence ofK+. SCH28080 has no effect on spontaneous dephospho-rylation but inhibits K+-stimulated dephosphorylation, pre-sumably by forming a E2−P•[I] complex. Hence SCH28080inhibits K+-stimulated ATPase activity by also competingwith K+ for binding to E2P (46). SCH28080 does notinhibit the Na+,K+-ATPase though there are high similari-ties of the cation-binding region between the Na+,K+- andthe H+,K+-ATPase.

Generally speaking, APA compounds show similar in-hibitory mechanism to that shown by SCH28080. They de-pend for their action on binding to the open form of thevestibule in the H+,K+-ATPase as shown in Figure 9.

Vestibule structureThe structural characteristics of this space are viewed in theorientation of Figure 10 with the M1/M2 segments to the right,

Figure 10 Acid pump antagonists.

Volume 1, October 2011 2149

Page 10: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

M5 to the left, M4 in the foreground, M6 in the background,the ion site above, and the aqueous lumen to the bottom. Be-tween the ion site above and the lumen is the loop connectingM5 and M6. The left and right sides of the vestibule are linedwith hydrophobic side chains which provide the surface forhydrophobic groups of inhibitors to shed their water of hydra-tion before binding thus providing much of the driving forcefor binding. Above the center of the vestibule is a cluster ofnegative charge density given by carboxylate side chains ofthe ion-binding/transport site. This provides through-spacecharge/charge attraction for any positive charge density pro-vided by an inhibitor, either in the form of a formal chargegroup such as an amine or guanidinium or as in the delocal-ized positive charge ring surrounding an aromatic ring systemsuch as the imidazopyridines. These charge interactions aremade more significant by the displacement of water in thispart of the vestibule by specific groups attached appropriatelyto the inhibitor core such as a pyrrolo-pyridine.

The vestibule is narrow in the space between M4 andM6, specifically between A335 and C813, and mutationalanalysis has shown the inhibitors have to fit between theseside chains. C813 is known to be the side chain to which PPIsbind covalently to inhibit the H+,K+-ATPase (8, 73) showingthat these bulky inhibitors (e.g., omeprazole) can enter thevestibule up to the point directly below the M5M6 loop nextto C813. On the left side of the vestibule the space is restrictedby the close interaction of M5 with M4 and a high densityof bulky side chains whereas on the right side there is moreavailable volume. These conclusions confirm earlier workusing models of the H+,K+-ATPase derived from the firstavailable molecular structures of the srCa-ATPase (Fig. 9).

Many of the side chains lining the vestibules are the samein the Na,K and H+,K+-ATPases. A set of side chain muta-tions that are mostly conservative substitutions from the Na,K-ATPase confer ouabain sensitivity to the H+,K+-ATPasehave been identified, namely, Y799F, A335G, C813T, I331V,M330I, R328E, and E820D (15). All but E820D are in thevestibule region. We have shown that the nonconserved Y799,A335, C813, and additionally, M334 contribute to the bindingaffinity (and specificity) of SCH28080 to the H+,K+-ATPasewhile other conserved residues also make strong contributionsto binding affinity especially L809, P810, and L811. Subtledifferences in this region of the two pumps determine thedifferences between efficacies of pump selective inhibitors.

Binding of planar structuresThe best characterized reversible inhibitor family has a coreplanar imidazo-pyridine structure of which SCH28080 is theprototypical example. Early work with derivatives in this classestablished the extended conformation of the phenyl groupwith respect to the imidazopyridine ring as a requirement forhigh-affinity binding (28, 29). Later it was shown that fixingthis conformation with an ethylene bridge in the homolo-gous naphthyridine (e.g., Byk99) increased binding affinity5- to 10-fold (105). Hence, a model for the binding of these

compounds must provide this conformation for the bound in-hibitor. Also, in early work with a photo affinity derivative ofSCH28080 containing a reactive azido group in the para po-sition of the phenyl ring showed specific labeling only in theM1/M2 segments of the membrane domain (52). This resultput the phenyl ring on the right side of the vestibule towardM1/M2. The observed labeling by omeprazole at C813 estab-lished the existence of a space adjacent to this position foran elongated inhibitor structure. Across from C813 is A335and this orientation was established when we showed that theA335C mutation gave a disulfide linkage between the twoside chains and complete loss of SCH28080 inhibition. TheC813T mutation also resulted in a 10-fold decrease in affinity(50). These results showed that there is a narrow space be-tween C813 and A335 and that at least part of the inhibitorwas bound between them. Positioning the flat imidazopyri-dine in the slot between these side chains then accounted notonly for the decrease in SCH28080 affinity seen in A335Cand C813T but also in the L809F, P810G, L811F, M334I, andY799S mutants.

Some of these APAs are shown in Figure 10.The inhibition by the APAs is expected to be fast and

effective since they do not require acid activation. Furtherthe pump is inhibited at half cycle without any proton se-cretion occurring. Data in human show the expected rapidand virtually complete inhibition by APAs (or P-CABs). Forexample, in healthy volunteers, high doses of AZD0865 re-sulted in over 95% inhibition of acid secretion within 1 hafter oral dosing (57). This inhibitor exhibit a classical (sig-moid) dose-response profile, with the magnitude and durationof effect being determined by dose, pKa, and plasma half-life. AZD0865 demonstrated a dose-effect relationship witha dose-dependent duration of inhibition of acid secretion.

A fused ring system of an imidazopyridine is soraprazan.Soraprazan inhibits the H+,K+-ATPase with an IC50 of 0.1μM, Ki of 6.4 nM, and a Kd of 26.4 nM (84). An APA struc-tural class, 5-aryl-1-(3-pyridinesulfonyl)pyrroles, was devel-oped (24). This member of a nonplanar class of compoundhas an IC50 value of 9 to 30 nM thus is the most potent ofthis type of inhibitor so far. It is noteworthy to mention thatthe 3-methylaminomethyl substituent is crucial to for the in-hibitory activity in this class of compound. One compound ofthis class, TAK-438 has a very slow dissociation rate produc-ing prolonged inhibition with a single dose lasting into thenight. It binds to the luminal domain in the open form whichthen closes to the predicted occluded binding configurationas shown in Figure 11.

ConclusionThe final step of gastric acid secretion in the stomach is per-formed by the gastric H+,K+-ATPase. The gastric H+,K+-ATPase exchanges hydronium ion with potassium using ATPhydrolysis. To transport the hydronium ion, the enzyme under-goes large conformational changes in the transition between

2150 Volume 1, October 2011

Page 11: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

Figure 11 The vestibule structure predicted for bound TAK-438. Se-lected amino acids in the binding region are predicted to affect binding(white, bold). Also shown is H bonding to E795 -COO− and -C=Oand to Y799.

E1 and E2. The functional form of the gastric H+,K+-ATPaseis an oligomeric form, composed of the catalytic α subunitand the glycoprotein β subunit. This enzyme is the majortarget for anti-acid secretory drugs. PPIs are in wide spreadclinical use. Another type of acid blocker is being studied toget better inhibition of acid secretion.

AcknowledgementsThis work was supported by US VA Merit Grant, NIH/NIDDKgrant numbers DK053642 and DK058333.

References1. 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 ofgastric H+,K+-ATPase prevents reverse reaction of the transport cycle.Embo J 28: 1637-1643, 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 functionalexpression of gastric H(+),K(+)-ATPase. J Biol Chem 275: 8324-8330,2000.

4. Asano S, Mizutani M, Hayashi T, Morita N, Takeguchi N. Reversibleinhibitions of gastric H+,K(+)-ATPase by scopadulcic acid B anddiacetyl scopadol. New biochemical tools of H+,K(+)-ATPase. J BiolChem 265: 22167-22173, 1990.

5. Bamberg K, Mercier F, Reuben MA, Kobayashi Y, Munson KB,Sachs G. cDNA cloning and membrane topology of the rabbit gastricH+/K(+)-ATPase alpha-subunit. Biochim Biophys Acta 1131: 69-77,1992.

6. Bamberg K, Sachs G. Topological analysis of H+,K(+)-ATPase usingin vitro translation. J Biol Chem 269: 16909-16919, 1994.

7. Besancon M, Shin JM, Mercier F, Munson K, Miller M, Hersey S,Sachs G. Membrane topology and omeprazole labeling of the gas-

tric H+,K(+)- adenosinetriphosphatase. Biochemistry 32: 2345-2355,1993.

8. Besancon M, Simon A, Sachs G, Shin JM. Sites of reaction of thegastric H,K-ATPase with extracytoplasmic thiol reagents. J Biol Chem272: 22438-22446, 1997.

9. Brzezinski P, Malmstrom BG, Lorentzon P, Wallmark B. The catalyticmechanism of gastric H+/K+-ATPase: Simulations of pre-steady-stateand steady-state kinetic results. Biochim Biophys Acta 942: 215-219,1988.

10. Callaghan JM, Toh BH, Pettitt JM, Humphris DC, Gleeson PA. Poly-N-acetyllactosamine-specific tomato lectin interacts with gastric parietalcells. Identification of a tomato-lectin binding 60-90 X 10(3) Mr mem-brane glycoprotein of tubulovesicles. J Cell Sci 95 (Pt 4): 563-576,1990.

11. Canfield VA, Levenson R. Structural organization and transcription ofthe mouse gastric H+, K(+)-ATPase beta subunit gene. Proc Natl AcadSci U S A 88: 8247-8251, 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-specificexpression, chromosomal assignment, and relationship to Na,K-ATPasebeta subunits. J Biol Chem 265: 19878-19884, 1990.

13. Craig WS. Monomer of sodium and potassium ion activated adenosinet-riphosphatase displays complete enzymatic function. Biochemistry 21:5707-5717, 1982.

14. Dantzig AH, Minor PL, Garrigus JL, Fukuda DS, Mynderse JS. Stud-ies on the mechanism of action of A80915A, a semi-naphthoquinonenatural product, as an inhibitor of gastric (H(+)-K+)-ATPase. BiochemPharmacol 42: 2019-2026, 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 theouabain-binding site in Na,K-ATPase. Pflugers Arch 457: 623-634,2009.

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

17. Fellenius E, Berglindh T, Sachs G, Olbe L, Elander B, Sjostrand SE,Wallmark B. Substituted benzimidazoles inhibit gastric acid secretionby blocking (H+ + K+)ATPase. Nature 290: 159-161, 1981.

18. Fujisaki H, Shibata H, Oketani K, Murakami M, Fujimoto M,Wakabayashi T, Yamatsu I, Yamaguchi M, Sakai H, Takeguchi N. In-hibitions of acid secretion by E3810 and omeprazole, and their reversalby glutathione. Biochem Pharmacol 42: 321-328, 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 29:701-706, 1990.

20. Hall K, Perez G, Sachs G, Rabon E. Identification of H+/K(+)-ATPasealpha,beta-heterodimers. Biochim Biophys Acta 1077: 173-179, 1991.

21. Han KS, Kim YG, Yoo JK, Lee JW, Lee MG. Pharmacokinetics of anew reversible proton pump inhibitor, YH1885, after intravenous andoral administrations to rats and dogs: Hepatic first-pass effect in rats.Biopharm Drug Dispos 19: 493-500, 1998.

22. Hebert H, Xian Y, Hacksell I, Mardh S. Two-dimensional crystalsof membrane-bound gastric H,K-ATPase. FEBS Lett 299: 159-162,1992.

23. Hioki Y, Takada J, Hidaka Y, Takeshita H, Hosoi M, YanoM. A newly synthesized pyridine derivative, (Z)-5-methyl-2-[2-(1-naphthyl)ethenyl]-4-piperidinopyridine hydrochloride (AU-1421), as areversible gastric proton pump inhibitor. Arch Int Pharmacodyn Ther305: 32-44, 1990.

24. Hori Y, Imanishi A, Matsukawa J, Tsukimi Y, Nishida H, ArikawaY, 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 blockerfor the treatment of acid-related diseases. J Pharmacol Exp Ther 335:231-238, 2011.

25. Hunt RH, Armstrong D, Yaghoobi M, James C, Chen Y, Leonard J, ShinJM, Lee E, Tang-Liu D, Sachs G. Predictable prolonged suppressionof gastric acidity with a novel proton pump inhibitor, AGN 201904-Z.Aliment Pharmacol Ther 28: 187-199, 2008.

26. Ife RJ, Brown TH, Keeling DJ, Leach CA, Meeson ML, Parsons ME,Reavill DR, Theobald CJ, Wiggall KJ. Reversible inhibitors of the gas-tric (H+/K+)-ATPase. 3. 3-substituted-4- (phenylamino)quinolines. JMed Chem 35: 3413-3422, 1992.

27. Jensen AM, Sorensen TL, Olesen C, Moller JV, Nissen P. Modulatoryand catalytic modes of ATP binding by the calcium pump. Embo J 25:2305-2314, 2006.

28. Kaminski JJ, Bristol JA, Puchalski C, Lovey RG, Elliott AJ, GuzikH, Solomon DM, Conn DJ, Domalski MS, Wong SC, Gold EH, LongJF, Chiu PJ, Steinberg M, McPhail AT. Antiulcer agents. 1. Gastricantisecretory and cytoprotective properties of substituted imidazo[1,2-a]pyridines. J Med Chem 28: 876-892, 1985.

Volume 1, October 2011 2151

Page 12: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Gastric H+,K+-ATPase Comprehensive Physiology

29. Kaminski JJ, Doweyko AM. Antiulcer agents. 6. Analysis of the invitro biochemical and in vivo gastric antisecretory activity of substi-tuted imidazo[1,2-a]pyridines and related analogues using comparativemolecular field analysis and hypothetical active site lattice methodolo-gies. J Med Chem 40: 427-436, 1997.

30. Katz PO, Koch FK, Ballard ED, Bagin RG, Gautille TC, Checani GC,Hogan DL, Pratha VS. Comparison of the effects of immediate-releaseomeprazole oral suspension, delayed-release lansoprazole capsules anddelayed-release esomeprazole capsules on nocturnal gastric acidityafter bedtime dosing in patients with night-time GERD symptoms.Aliment Pharmacol Ther 25: 197-205, 2007.

31. Keeling DJ, Fallowfield C, Underwood AH. The specificity of omepra-zole as an (H+ + K+)-ATPase inhibitor depends upon the means of itsactivation. Biochem Pharmacol 36: 339-344, 1987.

32. Laine L, Katz PO, Johnson DA, Ibegbu I, Goldstein MJ, Chou C,Rossiter G, Lu Y. Randomised clinical trial: A novel rabeprazole ex-tended release 50 mg formulation vs. esomeprazole 40 mg in healingof moderate-to-severe erosive oesophagitis - the results of two double-blind studies. Aliment Pharmacol Ther 33: 203-212, 2011.

33. LaMattina JL, McCarthy PA, Reiter LA, Holt WF, Yeh LA. Antiul-cer agents. 4-substituted 2-guanidinothiazoles: Reversible, competitive,and selective inhibitors of gastric H+,K(+)-ATPase. J Med Chem 33:543-552, 1990.

34. Lane LK, Kirley TL, Ball WJ, Jr. Structural studies on H+,K+-ATPase:Determination of the NH2-terminal amino acid sequence and immuno-logical cross-reactivity with Na+,K+-ATPase. Biochem Biophys ResCommun 138: 185-192, 1986.

35. Law RJ, Munson K, Sachs G, Lightstone FC. An ion gating mecha-nism of gastric H,K-ATPase based on molecular dynamics simulations.Biophys J 95: 2739-2749, 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: 1845-1852, 1992.

37. Lorentzon P, Jackson R, Wallmark B, Sachs G. Inhibition of (H+ +K+)-ATPase by omeprazole in isolated gastric vesicles requires protontransport. Biochim Biophys Acta 897: 41-51, 1987.

38. Ma JY, Song YH, Sjostrand SE, Rask L, Mardh S. cDNA cloning ofthe beta-subunit of the human gastric H,K-ATPase. Biochem BiophysRes Commun 180: 39-45, 1991.

39. Maeda M, Ishizaki J, Futai M. cDNA cloning and sequence determina-tion of pig gastric (H+ + K+)-ATPase. Biochem Biophys Res Commun157: 203-209, 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 subunitgene and recognition of its control region by gastric DNA bindingprotein. J Biol Chem 266: 21584-21588, 1991.

41. Maeda M, Oshiman K, Tamura S, Futai M. Human gastric (H+ + K+)-ATPase gene. Similarity to (Na+ + K+)-ATPase genes in exon/intronorganization but difference in control region. J Biol Chem 265: 9027-9032, 1990.

42. Mardh S, Norberg L. A continuous flow technique for analysis of thestoichiometry of the gastric H,K-ATPase. Acta Physiol Scand Suppl607: 259-263, 1992.

43. Melle-Milovanovic D, Milovanovic M, Nagpal S, Sachs G, Shin JM.Regions of association between the alpha and the beta subunit of thegastric H,K-ATPase. J Biol Chem 273: 11075-11081, 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 thecatalytic cycle. J Biol Chem 265: 15590-15598, 1990.

45. Mendlein J, Sachs G. The substitution of calcium for magnesium inH+,K+-ATPase catalytic cycle. Evidence for two actions of divalentcations. J Biol Chem 264: 18512-18519, 1989.

46. Mendlein J, Sachs G. Interaction of a K(+)-competitive inhibitor, asubstituted imidazo[1,2a] pyridine, with the phospho- and dephospho-enzyme forms of H+, K(+)-ATPase. J Biol Chem 265: 5030-5036,1990.

47. Moller JV, Olesen C, Winther AM, Nissen P. The sarcoplasmic Ca-ATPase: Design of a perfect chemi-osmotic pump. Q Rev Biophys 1-66,2010.

48. Morley GP, Callaghan JM, Rose JB, Toh BH, Gleeson PA, van DrielIR. The mouse gastric H,K-ATPase beta subunit. Gene structure andco-ordinate expression with the alpha subunit during ontogeny. J BiolChem 267: 1165-1174, 1992.

49. Munson K, Garcia R, Sachs G. Inhibitor and ion binding sites on thegastric H,K-ATPase. Biochemistry 44: 5267-5284, 2005.

50. Munson K, Law RJ, Sachs G. Analysis of the gastric H,K ATPase forion pathways and inhibitor binding sites. Biochemistry 46: 5398-5417,2007.

51. Munson K, Vagin O, Sachs G, Karlish S. Molecular modeling ofSCH28080 binding to the gastric H,K-ATPase and MgATP interactionswith SERCA- and Na,K-ATPases. Ann N Y Acad Sci 986: 106-110,2003.

52. Munson KB, Gutierrez C, Balaji VN, Ramnarayan K, Sachs G. Identi-fication of an extracytoplasmic region of H+,K(+)-ATPase labeled bya K(+)-competitive photoaffinity inhibitor. J Biol Chem 266: 18976-18988, 1991.

53. Murakami S, Arai I, Muramatsu M, Otomo S, Baba K, Kido T, KozawaM. Inhibition of gastric H+,K(+)-ATPase and acid secretion by cas-sigarol A, a polyphenol from Cassia garrettiana Craib. Biochem Phar-macol 44: 33-37, 1992.

54. Nagaya H, Satoh H, Kubo K, Maki Y. Possible mechanism for theinhibition of gastric (H+ + K+)-adenosine triphosphatase by the pro-ton pump inhibitor AG-1749. J Pharmacol Exp Ther 248: 799-805,1989.

55. Newman PR, Greeb J, Keeton TP, Reyes AA, Shull GE. Structure ofthe human gastric H,K-ATPase gene and comparison of the 5’-flankingsequences of the human and rat genes. DNA Cell Biol 9: 749-762, 1990.

56. Newman PR, Shull GE. Rat gastric H,K-ATPase beta-subunit gene:Intron/exon organization, identification of multiple transcription initia-tion sites, and analysis of the 5’-flanking region. Genomics 11: 252-262,1991.

57. Nilsson C, Albrektson E, Rydholm H, Rohss K, Hassan-Ali M, Hassel-gren G. Tolerability, pharmacokinetics and effects on gastric acid secre-tion after single oral doses of the potassium-competitive acid blockerAZD0865 in healthy male subjects. Gastroenterology 128 (4 Suppl. 2):A528, 2005.

58. Nishizawa T, Abe K, Tani K, Fujiyoshi Y. Structural analysis of 2Dcrystals of gastric H+,K+-ATPase in different states of the transportcycle. J Struct Biol 162: 219-228, 2008.

59. Okamoto CT, Karpilow JM, Smolka A, Forte JG. Isolation and charac-terization of gastric microsomal glycoproteins. Evidence for a glyco-sylated beta-subunit of the H+/K(+)-ATPase. Biochim Biophys Acta1037: 360-372, 1990.

60. Olesen C, Picard M, Winther AM, Gyrup C, Morth JP, Oxvig C, MollerJV, Nissen P. The structural basis of calcium transport by the calciumpump. Nature 450: 1036-1042, 2007.

61. Olesen C, Sorensen TL, Nielsen RC, Moller JV, Nissen P. Dephospho-rylation of the calcium pump coupled to counterion occlusion. Science306: 2251-2255, 2004.

62. Post RL, Toda G, Rogers FN. Phosphorylation by inorganic phosphateof sodium plus potassium ion transport adenosine triphosphatase. Fourreactive states. J Biol Chem 250: 691-701, 1975.

63. Rabon EC, Gunther RD, Bassilian S, Kempner ES. Radiation inactiva-tion analysis of oligomeric structure of the H,K-ATPase. J Biol Chem263: 16189-16194, 1988.

64. Rabon EC, McFall TL, Sachs G. The gastric [H,K]ATPase:H+/ATPstoichiometry. J Biol Chem 257: 6296-6299, 1982.

65. Rabon EC, Reuben MA. The mechanism and structure of the gastricH,K-ATPase. Annu Rev Physiol 52: 321-344, 1990.

66. Rabon EC, Smillie K, Seru V, Rabon R. Rubidium occlusion withintryptic peptides of the H,K-ATPase. J Biol Chem 268: 8012-8018, 1993.

67. Reenstra WW, Forte JG. H+/ATP stoichiometry for the gastric (K+ +H+)-ATPase. J Membr Biol 61: 55-60, 1981.

68. Reuben MA, Lasater LS, Sachs G. Characterization of a beta sub-unit of the gastric H+/K(+)-transporting ATPase. Proc Natl Acad SciU S A 87: 6767-6771, 1990.

69. Richter JE, Kahrilas PJ, Johanson J, Maton P, Breiter JR, Hwang C,Marino V, Hamelin B, Levine JG. Efficacy and safety of esomeprazolecompared with omeprazole in GERD patients with erosive esophagi-tis: A randomized controlled trial. Am J Gastroenterol 96: 656-665,2001.

70. Sachs G, Shin JM, Besancon M, Prinz C. The continuing develop-ment of gastric acid pump inhibitors. Aliment Pharmacol Ther 7: 4-12,discussion 29-31, 1993.

71. Shin JM, Besancon M, Prinz C, Simon A, Sachs G. Continuing devel-opment of acid pump inhibitors: Site of action of pantoprazole. AlimentPharmacol Ther 8: 11-23, 1994.

72. Shin JM, Besancon M, Simon A, Sachs G. The site of action of panto-prazole in the gastric H+/K(+)-ATPase. Biochim Biophys Acta 1148:223-233, 1993.

73. Shin JM, Cho YM, Sachs G. Chemistry of covalent inhibition of thegastric (H+, K+)-ATPase by proton pump inhibitors. J Am Chem Soc126: 7800-7811, 2004.

74. Shin JM, Goldshleger R, Munson KB, Sachs G, Karlish SJ. SelectiveFe2+-catalyzed oxidative cleavage of gastric H+,K+-ATPase: Impli-cations for the energy transduction mechanism of P-type cation pumps.J Biol Chem 276: 48440-48450, 2001.

75. Shin JM, Homerin M, Domagala F, Ficheux H, Sachs G. Characteriza-tion of the inhibitory activity of tenatoprazole on the gastric H+,K+-ATPase in vitro and in vivo. Biochem Pharmacol 71: 837-849, 2006.

76. Shin JM, Sachs G. Identification of a region of the H,K-ATPase alphasubunit associated with the beta subunit. J Biol Chem 269: 8642-8646,1994.

77. Shin JM, Sachs G. Dimerization of the gastric H+, K(+)-ATPase. JBiol Chem 271: 1904-1908, 1996.

2152 Volume 1, October 2011

Page 13: Comprehensive Physiology || Gastric H               +               ,K               +               -ATPase

P1: OTA/XYZ P2: ABCJWBT335-c110010 JWBT335/Comprehensive Physiology July 27, 2011 10:3 Printer Name: Yet to Come

Comprehensive Physiology Gastric H+,K+-ATPase

78. Shin JM, Sachs G. Differences in binding properties of two protonpump inhibitors on the gastric H(+),K(+)-ATPase in vivo. BiochemPharmacol 68: 2117-2127, 2004.

79. Shin JM, Sachs G. Functional consequences of the oligomeric formof the membrane-bound gastric H,K-ATPase. Biochemistry 44: 16321-16332, 2005.

80. Shin JM, Sachs G, Cho YM, Garst M. 1-Arylsulfonyl-2-(pyridylmethylsulfinyl) benzimidazoles as new proton pump inhibitorprodrugs. Molecules 14: 5247-5280, 2009.

81. Shull GE. cDNA cloning of the beta-subunit of the rat gastric H,K-ATPase. J Biol Chem 265: 12123-12126, 1990.

82. Shull GE, Lingrel JB. Molecular cloning of the rat stomach (H+ +K+)-ATPase. J Biol Chem 261: 16788-16791, 1986.

83. Simon WA, Budingen C, Fahr S, Kinder B, Koske M. The H+, K(+)-ATPase inhibitor pantoprazole (BY1023/SK&F96022) interacts lesswith cytochrome P450 than omeprazole and lansoprazole. BiochemPharmacol 42: 347-355, 1991.

84. Simon WA, Herrmann M, Klein T, Shin JM, Huber R, Senn-BilfingerJ, Postius S. Soraprazan: Setting new standards in inhibition of gastricacid secretion. J Pharmacol Exp Ther 321: 866-874, 2007.

85. Skrabanja AT, van der Hijden HT, De Pont JJ. Transport ratios ofreconstituted (H+ + K+)-ATPase. Biochim Biophys Acta 903: 434-440, 1987.

86. Smith GS, Scholes PB. The H+/ATP stoichiometry of the (H+ +K+)-ATPase of dog gastric microsomes. Biochim Biophys Acta 688: 803-807, 1982.

87. Sorensen TL, Olesen C, Jensen AM, Moller JV, Nissen P. Crystals ofsarcoplasmic reticulum Ca(2+)-ATPase. J Biotechnol 124: 704-716,2006.

88. Swarts HG, Koenderink JB, Willems PH, Krieger E, De Pont JJ. Asn792participates in the hydrogen bond network around the K+-bindingpocket of gastric H,K-ATPase. J Biol Chem 280: 11488-11494, 2005.

89. Takahashi M, Kondou Y, Toyoshima C. Interdomain communication incalcium pump as revealed in the crystal structures with transmembraneinhibitors. Proc Natl Acad Sci U S A 104: 5800-5805, 2007.

90. Toh BH, Gleeson PA, Simpson RJ, Moritz RL, Callaghan JM, GoldkornI, Jones CM, Martinelli TM, Mu FT, Humphris DC, et al. The 60- to90-kDa parietal cell autoantigen associated with autoimmune gastritisis a beta subunit of the gastric H+/K(+)-ATPase (proton pump). ProcNatl Acad Sci U S A 87: 6418-6422, 1990.

91. Toyoshima C, Asahi M, Sugita Y, Khanna R, Tsuda T, MacLennanDH. Modeling of the inhibitory interaction of phospholamban with theCa2+ ATPase. Proc Natl Acad Sci U S A 100: 467-472, 2003.

92. Toyoshima C, Nakasako M, Nomura H, Ogawa H. Crystal structure ofthe calcium pump of sarcoplasmic reticulum at 2.6 A resolution. Nature405: 647-655, 2000.

93. Toyoshima C, Nomura H. Structural changes in the calcium pumpaccompanying the dissociation of calcium. Nature 418: 605-611, 2002.

94. Toyoshima C, Nomura H, Sugita Y. Crystal structures of Ca2+-ATPasein various physiological states. Ann N Y Acad Sci 986: 1-8, 2003.

95. Tyagarajan K, Lipniunas PH, Townsend RR, Forte JG. The N-linkedoligosaccharides of the beta-subunit of rabbit gastric H,K-ATPase: Sitelocalization and identification of novel structures. Biochemistry 36:10200-10212, 1997.

96. Tyagarajan K, Townsend RR, Forte JG. The Beta-subunit of the rab-bit H,K-ATPase: A glycoprotein with all terminal lactosamine units

capped with alpha-linked galactose residues. Biochemistry 35: 3238-3246, 1996.

97. Vagin O, Denevich S, Sachs G. Plasma membrane delivery of the gastricH,K-ATPase: The role of beta-subunit glycosylation. Am J Physiol CellPhysiol 285: C968-C976, 2003.

98. Vagin O, Kraut JA, Sachs G. Role of N-glycosylation in trafficking ofapical 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 amodel to study the role of N-glycosylation in membrane trafficking andapical sorting. J Biol Chem 279: 39026-39034, 2004.

100. Vagin O, Turdikulova S, Yakubov I, Sachs G. Use of the H,K-ATPasebeta subunit to identify multiple sorting pathways for plasma mem-brane delivery in polarized cells. J Biol Chem 280: 14741-14754,2005.

101. Walderhaug MO, Post RL, Saccomani G, Leonard RT, Briskin DP.Structural relatedness of three ion-transport adenosine triphosphatasesaround their active sites of phosphorylation. J Biol Chem 260: 3852-3859, 1985.

102. Wallmark B, Brandstrom A, Larsson H. Evidence for acid-inducedtransformation of omeprazole into an active inhibitor of (H+ + K+)-ATPase within the parietal cell. Biochim Biophys Acta 778: 549-558,1984.

103. Wallmark B, Briving C, Fryklund J, Munson K, Jackson R, MendleinJ, Rabon E, Sachs G. Inhibition of gastric H+,K+-ATPase and acidsecretion by SCH 28080, a substituted pyridyl(1,2a)imidazole. J BiolChem 262: 2077-2084, 1987.

104. Wallmark B, Stewart HB, Rabon E, Saccomani G, Sachs G. The cat-alytic cycle of gastric (H+ + K+)-ATPase. J Biol Chem 255: 5313-5319, 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: 5374-5378, 2007.

Further Reading1. Ogawa H, Shinoda T, Cornelius F, Toyoshima C. Crystal structure of

the sodium-potassium pump (Na+,K+-ATPase) with bound potassiumand ouabain. Proc Natl Acad Sci U S A 106: 13742-13747, 2009.

2. Shin JM, Munson K, Vagin O, Sachs G. The gastric HK-ATPase: Struc-ture, function, and inhibition. Pflugers Arch 457: 609-622, 2009.

3. Shin JM, Vagin O, Munson K, Kidd M, Modlin IM, Sachs G. Molecularmechanisms in therapy of acid-related diseases. Cell Mol Life Sci 65:264-281, 2008.

4. Shinoda T, Ogawa H, Cornelius F, Toyoshima C. Crystal structure ofthe sodium-potassium pump at 2.4 A resolution. Nature 459: 446-450,2009.

5. Vagin O, Tokhtaeva E, Yakubov I, Shevchenko E, Sachs G. Inversecorrelation between the extent of N-glycan branching and intercellularadhesion in epithelia. Contribution of the Na,K-ATPase beta1 subunit.J Biol Chem 283: 2192-2202, 2008.

Volume 1, October 2011 2153