throbber
Biochimica et Biophysica Acta, 728 (1983) 31-38
`Elsevier Biomedical Press
`
`BBA 71532
`
`31
`
`INHIBITION OF GASTRIC (H + + K + )-ATPase BY THE SUBSTITUTED BENZIMIDAZOLE,
`PICOPRAZOLE
`
`B. WALLMARK •, G. SACHS ••, S. MARDH ***and E. FELLENIUS
`
`AB Hassle, Research Laboratories, S-431 83 Molnda/ (Sweden)
`
`(Received August 13th, 1982)
`
`Key words: ( H + + K +)-A TPase inhibition; Proton transport; Picoprazole; Acid secretion; (Porcine stomach)
`
`The substituted benzimidazole, picoprazole, inhibited the gastric (H + + K + )-A TPase in a concentration- and
`time-dependent manner. Half-maximal inhibition of the (H + + K +)-A TPase activity was obtained at about
`2·10 - 6 M under standard conditions. In addition to the inhibition of ATPase activity, parallel inhibition of
`phosphoenzyme formation and the proton transport activity were achieved. Radiolabelled picoprazole was
`found to bind to 100 kDa peptide; this peptide was shown by phosphorylation experiments to contain the
`catalytic centre of the (H + + K +)·A TPase. Studies on the (Na + + K + )-ATPase indicated that this enzyme
`was unaffected by picoprazole. From the data presented and from other pharmacological studies, it is
`proposed that this compound inhibits acid secretion at the level of the parietal cell by its ability to inhibit the
`gastric proton pump, the (H + + K + )·A TPase.
`
`Introduction
`
`The substituted benzimidazoles inhibit gastric
`acid secretion both in vivo and in vitro [1-3].
`Furthermore, in isolated gastric gland prepara(cid:173)
`tions [4] these inhibitors have been shown to in(cid:173)
`hibit acid production monitored as accumulation
`of a weak base, aminopyrine [5,6]. In this glandu(cid:173)
`lar system and in isolated guinea pig mucosa, the
`
`* To whom correspondence should be addressed.
`** Institute of Medical and Physiological Chemistry Biomedi(cid:173)
`cal Centre, Box 575, Uppsala University, S-751 23 Upp(cid:173)
`sala, Sweden.
`*** Laboratory of Membrane Biology, University of Alabama
`in Birmingham, University Station, Birmingham, AL
`35294, U.S.A.
`Abbreviations: (H + + K + )-A TPase, Proton plus potassium(cid:173)
`stimulated A TP phosphohydrolase; (N a+ K + )-A TPase, Sodium
`plus potassium ion transport A TP phosphohydrolase (EC
`3.6.1.3); Pipes, piperazine-N, N' -bis-(2-ethanesulfonic acid);
`Tris,
`tris(hydroxymethyl)aminornethane; dibutyryl-cAMP,
`N 6,0 21 -dibutyryladenosine 3': 5'-cyclic monophosphate.
`
`substituted benzimidazoles inhibited not only basal
`secretion, but also secretion induced by
`the
`secretatogues histamine and dibutyryl-cAMP. In(cid:173)
`hibition was found to be noncompetitive [1,6].
`Furthermore, aminopyrine accumulation stimu(cid:173)
`lated by high K + concentration was also
`antagonized by the compounds [6]. Based on these
`data, a mechanism of action peripheral to cell
`receptor sites and probably located within the
`parietal cell seemed likely [5,6]. During recent
`years, an (H + + K +)-A TPase has been purified
`from the gastric mucosa of various species [7-9]
`and has been localized to the secretory surface of
`the parietal cell [1 OJ. The addition of A TP to
`isolated vesicles containing the enzyme results in
`H+ uptake into the intravesicular space in ex(cid:173)
`change for intra vesicular K + [ 11, 12]. The trans(cid:173)
`port process is apparently electroneutral [ 13]. En(cid:173)
`try of KCl into the vesicle is rate-limiting for
`proton translocation. The (H + + K +)-A TPase thus
`plays an important role in H+ secretion across the
`membranes of the secretory canaliculus of the
`
`0005-2736/83/0000-0000j$03.00 ® 1983 Elsevier Biomedical Press
`
`

`
`32
`
`parietal cell [13,14]. It seemed possible, therefore,
`that the substituted benzimidazoles affect acid
`secretion by acting directly on the (H + + K + )(cid:173)
`ATPase. This paper presents data for one sub(cid:173)
`stituted benzimidazole, picoprazole, and describes
`its inhibitory action on the (H + + K +)-A TPase in
`more detail.
`
`Material and Methods
`
`Materials. Gastric (H + + K +)-A TPase was pre(cid:173)
`pared from hog stomachs as described by Sac(cid:173)
`comani et al. [15]. In brief, membrane vesicles
`containing the (H+ + K+)-ATPase were prepared
`by differential and zonal density gradient centrifu(cid:173)
`gation. The microsomal fraction was centrifuged
`through a 0.25 M sucrosej0.25 M sucrose-8%
`(w jw) Ficoll ® step gradient in a Kontron TZT 48
`zonal rotor (Basel, Switzerland) for 4 h at 47 000
`rpm. The material retained at the interface was
`used for the experiments.
`For transport experiments, the vesicles were
`equilibrated in a mixture of 150 mM KCl, 2 mM
`MgC1 2 and 2 mM Pipes adjusted to pH 6.7 with
`Tris base. After equilibration, the vesicles were
`stored at 4°C until use on the same day. All
`subsequent mixing of the sample solutions was
`done near isotonicity of the equilibrated sample.
`For experiments in which free access of ions to
`either face of the enzyme was required, the vesicle
`preparation was lyophilized and stored at - 80°C
`until resuspension. The (Na + + K + )-ATPase pre(cid:173)
`paration was obtained as described by J0rgensen
`[ 16].
`Reagents. [ y- 32 P]A TP was obtained from the
`Radiochemical Centre, Amersham, U.K. ATP was
`obtained from Sigma, U.S.A. Acridine orange
`was obtained from Merck, Darmstadt, F.R.G. Pi(cid:173)
`coprazole, (H 149 /94), methyl 6-methyl-2-[[(3-
`methyl-2-pyridinyl)methyl]sulfinyl]-1 H -benzimid(cid:173)
`azole-5-carboxylate and [ 14C]picoprazole were
`synthetized by the Chemical Department, AB
`Hassle, Molndal, Sweden (Fig. 1). Picoprazole was
`dissolved in methanol. The final methanol con(cid:173)
`centration did not exceed 1% which alone had no
`effect on any of the activities measured.
`Enzyme assays. K +-stimulated A TPase activity
`of the (H + + K +)-A TPase is designated K +(cid:173)
`A TPase activity. (N a+ + K +)-stimulated A TPase
`
`Picoprazole
`
`( H 149/94)
`
`Fig. I. Structural formula of picoprazole. • indicates the posi(cid:173)
`tion of the 14 C label.
`
`activity of the (Na + + K + )-ATPase is designated
`(Na + + K + )-ATPase activity. K +-stimulated p(cid:173)
`nitrophenylphosphatase activity of the (H+ +
`K+)-ATPase and (Na+ + K+)-ATPase are desig(cid:173)
`nated p-nitrophenylphosphatase ((H+ + K + )(cid:173)
`ATPase) and p-nitrophenylphosphatase ((Na+ +
`K + )-ATPase), respectively. It should be noted that
`p-nitrophenylphosphatase ((N a+ + K +)-A TPase)
`was assayed in the absence of Na +.
`A TPase activity. This was measured by the
`release of inorganic phosphate, which was assayed
`according to the method of Fiske and SubbaRow
`[I 7].
`K + -ATPase activity. The assay medium con(cid:173)
`sisted of 2 mM MgC1 2 , 75 mM Pipes-Tris buffer
`(pH 7.4), 2 mM Na 2 ATP, with or without 10 mM
`KCl in a total volume of 2 mi. The assay was
`incubated for 15 min at 37°C. The reaction was
`then stopped by the addition of I ml 10% trichlo(cid:173)
`roacetic acid. A maximum of 10% of the substrate
`was hydrolyzed over the incubation period, giving
`linear reaction rates. In lyophilized material and in
`the absence of K + , reaction rates were about 10
`ttmol PJmg protein per h. In the presence of both
`Mg 2 + and K +, reaction rates were about 100
`,umol PJmg protein per h. For all inhibition ex(cid:173)
`periments. reaction rates in the presence of Mg 2 +
`and K + but in the absence of inhibitor were set to
`100%.
`(Na + + K + )-ATPase activity was assayed in a
`medium consisting of 5 mM Pipes-Tris buffer (pH
`7.4), 2 mM MgC1 2 , 2 mM Na 2 ATP with or without
`10 mM KCI plus 100 mM N aCl in a total volume
`of 2 ml, and incubated for 3 min at 37°C. The
`reaction was stopped by the addition of l ml 10%
`trichloroacetic acid. In the absence of K + and
`Na +, reaction rates were about 20 ttmol PJmg
`protein per h. In the presence of N a+ and K + ,
`
`

`
`reaction rates were about 400 p.mol Pjmg protein
`per h. For the inhibition experiments reaction
`rates in the presence of Mg2+, Na+ and K+ but
`in the absence of inhibitor were set to 100%.
`p-Nitrophenylphosphatase activity. This was
`measured by the release of p-nitrophenol from
`p-nitrophenyl phosphate [18].
`((H + + K + )(cid:173)
`p-N itrophenylphosphatase
`ATPase) activity was assayed in a medium consist(cid:173)
`ing of 20 mM imidazole-acetic acid buffer (pH
`7.5), 3 mM MgC1 2 , 3 mM Tris-p-nitrophenyl
`phosphate with or without 10 mM KCI in a total
`volume of 0.5 ml. The assay was incubated for 3
`min at 37°C. The reaction was stopped by the
`addition of 25 p.l 50% trichloroacetic acid. In the
`absence of K +, reaction rates were about 4 p.mol
`p-nitrophenol/mg protein per h. In the presence
`of both Mg+ and K +, reaction rates were about
`75 p.mol p-nitrophenol/mg protein per h. This
`reaction rate was given a value of 100%.
`((Na+ + K + )(cid:173)
`p-Nitrophenylphosphatase
`ATPase) activity. This was assayed as described
`for p-nitrophenylphosphatase (H+ + K + -ATPase).
`In the absence of K + , reaction rates were about 7
`p.mol p-nitrophenol/mg protein per h. In the pres(cid:173)
`ence of both Mg + and K + , reaction rates were
`about 160 p.mol p-nitrophenol/mg protein per h,
`which was taken as the 100% value.
`Phosphorylation of the gastric membranes. (H +
`+ K +)-A TPase was preincubated for 30 min at
`37°C. The medium contained 10- 6 , 10- 5 and
`10- 4 M picoprazole, 40 mM Tris-HCl (pH 7.4)
`and 20 p.g of membrane protein. The phosphory(cid:173)
`lation reaction was then measured by the addition
`of 2 mM MgC1 2 and 5 p.M [y- 32 P]ATP for 15 s, at
`22°C. The reaction was stopped by the addition of
`1 ml ice-cold 10% HC104 containing 5 mM ATP
`and 40 mM Na 2 P04 • The precipitated protein was
`collected on a 4 p.m Millipore ® type SSWP filter.
`The filter was washed with 70 ml of 5% HC104
`containing 10 mM Na 2 P04 • The Cerenkov radia(cid:173)
`tion of the filter was measured in a scintillation
`counter.
`SDS-polyacrylamide gel-electrophoresis. Mem(cid:173)
`branes undergoing SDS-polyacrylarnide gel elec(cid:173)
`trophoresis were either first phosphorylated by
`means of [ y- 32 P]A TP or incubated with C4 C]pico(cid:173)
`prazole. The phosphorylation procedure was car(cid:173)
`ried out in a medium consisting of 200 p.gjml
`
`33
`
`gastric membranes, 2 mM MgC1 2 and 5 p.M [y-
`32 P]A TP in the presence or absence of 0.1 M KCI.
`The final volume was 200 p.l. The phosphorylation
`reaction was stopped with 1% SDS to solubilize
`the membranes. About 40 p.g protein was applied
`to a density gradient polyacrylamide gel with a
`starting density of 4% and a final density of 30%.
`Gels were run in a 0.1 M Tris-borate buffer (pH
`6.0) in 0.1% SDS for 2.5 h. Protein was stained
`with Coomassie blue and scanned
`in a gel
`densitometer at 550 nm. Gels run with 32 P-labelled
`membranes were sectioned manually immediately
`after electrophoresis and each slice was counted.
`14 C-picoprazole,
`For binding studies with
`gastric membranes (10 1-'g/ml) were incubated at
`37°C in 2 mM Pipes-Tris (pH 7.4) with 10- 4 M
`14 C]picoprazole in a total volume of 20 ml for 2 h
`[
`and then centrifuged for 1 h at 100 000 X g. The
`resulting pellet was resuspended in 1% SDS and
`about 40 1-'g protein applied to each gel. Gels were
`then treated as described for the phosphorylation
`experiments.
`Proton transport experiments. H + transport rates
`and the extent of pH gradient formation were
`monitored in an Aminco DW2 spectrophotometer
`(America} Instrument Company, U.S.A.) in the
`dual beam model set at 490 and 535 nm [19].
`Vesicles were preequilibrated at a concentration of
`40 p.gjml in 2 mM Pipes-Tris, 150 mM KCI and 2
`mM MgC1 2 at pH 6.7 for 3 h at 22°C. The equi(cid:173)
`librated sample was mixed with 10- 4 M pico(cid:173)
`prazole and after various incubation times, a sam(cid:173)
`ple was withdrawn and placed in a cuvette. 20 p.M
`Acridine orange was added and a baseline re(cid:173)
`corded, after which 0.6 mM Na 2 ATP was added
`to the incubation and the proton transport rate
`assayed.
`Protein determination. Protein was measured by
`the method of Lowry et al. [20].
`
`Results
`
`Kinetics of the inactivation of K + -A TPase activity
`The time course for the inhibition of K +(cid:173)
`ATPase activity is shown in Fig. 2A. The inhibi(cid:173)
`tion was found to be both time- and concentra(cid:173)
`tion-dependent, since the reduction in enzyme ac(cid:173)
`tivity
`increased progressively with
`increased
`
`

`
`~
`
`>
`.;=
`:;;_
`
`~
`
`1.5
`
`f
`g'
`c
`~
`cr
`~
`F 11
`
`34
`
`~
`
`>
`.;=
`:;;_
`~
`.g 50
`[?'
`c
`~
`&'
`-:.
`
`A
`
`1~M
`
`30
`
`60
`90
`30
`T1me M1nutes
`Time , M1nutes
`Fig. 2. A. Time course for inactivation of (H + + K + )-A TPase.
`Membrane suspensions (10 p.g of protein;ml) were incubated
`in a medium consisting of 5 mM Pipes-Tris (pH 7.4) at
`concentrations of picoprazole as indicated. After incubation
`times, indicated at the abscissa, samples were assayed for
`K +-A TPase activity. The K +-stimulated ATPase activity in the
`absence of picoprazole was given a value of 100%. Each time
`point is the mean of four separate determinations. The bars
`indicate the standard error of the mean (S.E.) for each time
`point. B. Semilogarithmic point of the data in Fig. 2A.
`
`inhibitor concentrations.
`time and
`incubation
`When the data from Fig. 2A were replotted semi(cid:173)
`logarithmically (Fig. 2B), the reaction of pico(cid:173)
`prazole with the enzyme was linear over the first
`75% of the total reaction. However, for the remain(cid:173)
`ing 25% a deviation from a straight-line relation(cid:173)
`ship between the logarithm of the remaining en(cid:173)
`zyme activity and time was obtained. From the
`linear portions of the curves in Fig. 2B, half-lives,
`
`2.0
`
`Stope= -1.1
`
`c
`0
`~
`
`~
`lll
`.s:
`r... .e
`~ -+-
`
`C'l
`.2
`
`1.0
`
`-6
`
`-7
`
`tog [PicoprazoleJ
`Fig. 3. Double logarithmic plot of the t 112 of inactivation
`against picoprazole concentrations. The t 112 was calculated
`from the linear part of Fig. 2B.
`
`.....
`>o
`·s:
`~
`~
`QJ
`E
`>o so
`N c:
`UJ
`Cl c:
`·c:
`'iii
`E
`QJ
`0::
`~ 0
`
`-6
`
`-4
`
`-5
`log [Picoprazolel
`Fig. 4. Concentration dependence for inhibition of (H + + K + )(cid:173)
`I' g of
`A TPase by picoprazole. Gastric membranes ( 10
`protein/ml) were incubated for 30 min in 5 mM Pipes-Tris (pH
`7.4) with picoprazole at concentrations indicated. After incuba(cid:173)
`tion, samples were assayed for K +-A TPase activity. The en(cid:173)
`zyme activity in the absence of picoprazole was given a value of
`100%. Each time point is the mean of two separate determina(cid:173)
`tions.
`
`t 112 , were calculated for the inhibition reaction at
`each inhibitor concentration. These values were
`then plotted in a double logarithmic plot against
`the picoprazole concentrations (Fig. 3). This plot
`was linear with a slope close to one. This allows
`the conclusion that inhibition follows pseudo(cid:173)
`first-order kinetics [21], at least over the first 75%.
`The concentration-effect relationship is shown in
`Fig. 4. Increasing concentrations of picoprazole
`resulted in progressive inhibition of the K +(cid:173)
`ATPase activity. Half-maximal inhibition occurred
`at about 2 ·_10- 6 M.
`
`pH dependence of inhibition
`From the results in Fig. 5, it was found that
`enzyme inhibition increased progressively as the
`proton concentration was increased. Despite the
`greater inhibition at lower pH, however, a pH of
`7.4 was taken as standard for most subsequent
`experiments, since this was the optimal pH for the
`enzyme activity [22].
`
`Binding of picoprazo/e to ( H + + K +)-A TPase
`The binding of picoprazole to (H + + K + )(cid:173)
`A TPase was analyzed by SDS-polyacrylarnide gel
`electrophoresis. A major peak was evident at about
`
`

`
`0
`
`0
`
`100
`
`>-
`......
`·:;::
`~ u
`d
`QJ
`E
`>-
`N c: so
`Cl c:
`:~
`E
`QJ
`0:
`~ 0
`
`QJ
`
`6.0
`
`7.0
`
`8.0
`
`pH
`Fig. 5. Effects of pH on the inactivation of (H + + K + )-ATPase.
`Membrane suspension (10 p.g of proteinjml) was incubated in
`2 mM Pipes-Tris buffer at pH 5.7, 6.3, 6.8, 7.4 and 7.8 for 30
`min, with picoprazole at concentrations of I p.M (0--0);
`5 p.M (0--0) and 10 p.M (L>.--L>.). After incubation,
`samples were adjusted to pH 7.4 by the addition of 75 mM
`Pipes-Tris buffer and K +-A TPase activity was assayed. K +(cid:173)
`A TPase activity in the absence of picoprazole was given a value
`of 100%.
`
`1.7
`
`E
`c:
`0 ...,
`...,
`~ 0.95
`~
`"'
`.Q
`~
`.Q
`<(
`
`Qj
`
`A
`
`100K-
`
`[3Zp)
`dpm
`
`20000
`
`10000
`
`8 9
`
`10
`
`11 12
`
`35
`
`100 kDa (Fig. 6B). This peak was also found to
`contain large amounts of radioactivity. However,
`large quantities of radioactivity that did not corre(cid:173)
`spond to any protein peak were retained at the end
`of the gel. This was probably due to binding of
`14C]picoprazole to lipid components, as the low
`[
`molecular weight region of the gel stained with
`Sudan black, a marker for lipids (data not shown).
`In order to identify the protein containing the
`catalytic peptide of the (H+ + K + )-ATPase, we
`studied the incorporation of 32 P from AT 33 P [23].
`The distribution of 32 P after phosphorylation of
`the membrane preparation is shown in Fig. 6A.
`The 100 kDa peptide was found to contain the
`32 P-label (open bars) and, furthermore, when the
`experiment was performed in the presence of K + ,
`the label of the 100 kDa peptide was lost (shaded
`bars). It can therefore be concluded that both the
`14C- and 32 P-labels are confined to the same
`peptide.
`
`r -
`
`B
`
`r -
`
`[ 14c I
`dpm
`
`6000
`
`100 K ....
`
`r--
`
`.--1)
`
`~
`
`.--
`
`4000
`
`.....-
`
`2000
`
`~ v f-v ~
`~ ~ V1
`v
`v
`
`jA.
`
`'""' h Jli\
`
`1 2 3 4 5 6 7 8 9 10 11 12 13
`Gel slice no.
`
`Fig. 6. A. Binding of [y-nP]ATP to (H+ + K + )-ATPase. Gastric membranes (200 p.g of proteinjml) were incubated and
`phosphorylated as described in Methods. Open bars show the distribution of the 32 P-label in the presence of MgAT 32 P and shaded
`bars in the presence of MgAT 32 P plus 100 mM KCI. Gels were prepared and run as detailed in the Methods section. 100 K indicates
`the position of the 100 kDa peptide. B. Binding of [ 14 C]picoprazole to (H+ + K + )-ATPase. Gastric membranes (10 p.g of proteinjml)
`were incubated with w- 4 M [ 14 C]picoprazole, after which the incubate was centrifuged at 100000 X g for 60 min and solubilized in
`1% sodium dodecyl sulphate. Open bars show the distribution of [14 C]picoprazole. 100 K indicates the position of the 100 kDa
`peptide.
`
`

`
`36
`
`Effect of picoprazole on phosphoenzyme formation
`the (H + + K + )(cid:173)
`During the catalytic cycle,
`ATPase forms a phosphoenzyme intermediate
`[24-26]. Since picoprazole was found to bind to
`the I 00 kDa peptide and also inhibited the A TPase
`activity (Figs. 4 and 6), its effect on steady-state
`phosphoenzyme concentration was studied. Pico(cid:173)
`prazole inhibited the phosphoenzyme levels in a
`concentration-related manner. At w- 6 M, 95% of
`the control level was obtained; at w-s M and
`10- 4 M 61% and 35%, respectively, of the control
`level was reached. The control level was 720 pmol
`of phosphoenzyme per milligram of membrane
`protein. Thus, inhibition of K + -ATPase activity
`and phosphoenzyme formation were found to oc(cid:173)
`cur in parallel.
`
`Inhibition of proton transport by picoprazole
`When the (H + + K +)-A TPase is isolated in
`vesicular form, proton transport can be measured
`[12, 19]. Addition of A TP to vesicles preequi(cid:173)
`librated in a medium containing KCl results in
`vesicles energization, and proton transport rates
`can be monitored. In this experiment, gastric
`vesicles were incubated with picoprazole. After the
`times indicated under each curve in Fig. 7 A, a
`sample was withdrawn and the transport signal
`measured. From Fig. 7 A, three parameters were
`calculated for each curve: the inital rate of proton
`transport, taken as the initial slope; the magnitude
`of the signal, and the time at which the maximal
`magnitude was reached. These parameters are
`shown in Fig. 7B, from which it can be seen that
`both the intial rate and maximal magnitude of the
`transport signal were inhibited in parallel. In addi(cid:173)
`tion, the time required to reach the maximal am(cid:173)
`plitude of the transport curve increased progres(cid:173)
`sively with incubation time.
`
`Effects on p-nitrophenylphosphatase (( H + + K + )(cid:173)
`ATPase)
`As stated earlier, proton transport in the iso(cid:173)
`lated vesicle system required extra vesicular MgA TP
`and intra vesicular K +. However, the (H + + K + )(cid:173)
`A TPase can hydrolyze substrates other
`than
`MgA TP. In the presence of p-nitrophenyl phos(cid:173)
`phate and Mg 2 + , K + stimulates the splitting of
`p-nitrophenyl phosphate into p-nitrophenol and
`inorganic phosphate. Two differences are evident
`
`A
`
`ATP
`I
`
`-
`
`0
`
`j -004
`E
`::0
`
`-OOB
`
`-012
`
`0
`
`B
`
`100
`
`.. c...,
`~"g
`Oi '2 50
`:Eg'
`.s~
`
`Il
`
`Trme,sec
`
`200 "'
`]
`'E
`Cl
`~
`i
`150 .f
`~
`;::
`
`'-/KJID-------:6:'::-0------..,±-___::s...~ 100
`0
`
`Time, min
`
`Fig. 7. A. Inhibition of proton by picoprazole. Gastric mem(cid:173)
`brane vesicles (40 JLg of protein/ml were equilibrated in 150
`mM KCl, 2 mM MgC1 2 and 2 mM Pipes-Tris (pH 6.7) at room
`temperature for 3 h, after which 10- 4 M picoprazole was added
`to the equilibration mixture. At the times indicated under the
`curves (0, 2, 13, 24, 35, 57, 68 and 143 min), a sample was
`withdrawn and the proton transport signal assayed. At time
`zero, indicated on the time axis in the figure, the sample and 20
`JLM Acridine orange were added to the cuvette (volume= I ml)
`and a base-line was recorded for about 25 s after which 0.6 mM
`ATP was added to initiate proton transport. B. Three parame(cid:173)
`ters were calculated from the date in Fig. 7A: Initial slope
`(e--e); maximal magnitude taken as the lowest value of
`each curve (.A--.6) and the time from the addition of ATP
`at which the signal reached a maximum ( 0 - - 0 ). All data
`are given as percent of the control signal, which was taken as
`the transport signal in the absence of inhibitor (lowest curve in
`Fig. 7A).
`
`when p-nitrophenyl phosphate is used as substrate
`instead of MgA TP. First, in order to stimulate
`hydrolysis of p-nitrophenyl phosphate, K + is re(cid:173)
`quired on the outside of the vesicle whereas in(cid:173)
`travesicular K + is required for the hydrolysis of
`
`

`
`37
`
`found that even at concentrations as high as
`w- 4 M, the (Na+ + K+)-ATPase activity was un(cid:173)
`affected by the inhibitor (Fig. 8). Furthermore, the
`p-nitrophenylphosphatase ((Na + + K + )-ATPase)
`activity was not inhibited, even at high picoprazole
`concentrations (Fig. 8).
`
`Discussion
`
`The ability to inhibit acid secretion in mam(cid:173)
`malian gastric mucosa has hitherto depended on
`the availability of H 2-receptor antagonists, such as
`cimetidine, or of anticholinergic agents [28,29].
`The substituted benzimidazoles represent a novel
`group of gastric acid secretion inhibitors. Their
`mechanism of action appears to be distinct from
`those of the H 2-blockers or anticholinergics, since
`pharmacological data for the benzimidazole de(cid:173)
`rivative, picoprazole, indicate that its antisecretory
`activity is exerted at a site distal to the production
`of cAMP in the events leading to acid formation
`[I ,6].
`It has been established that the gastric (H + +
`K +)-A TPase plays an essential role in the secre(cid:173)
`tion of acid by the parietal cells [13]. The data
`presented here show that picoprazole, a known
`inhibitor of gastric acid secretion [1-3], inhibits
`the (H + + K +)-A TPase. In addition, the phos(cid:173)
`phorylation and K +-dependent phosphatase reac(cid:173)
`tions were also inhibited. Moreover, SDS-poly(cid:173)
`acrylamide gel electrophoresis showed that the in(cid:173)
`hibitor was bound to the ATPase peptide. The
`inhibition of the proton transport reaction by pi(cid:173)
`coprazole can thus be attributed to inhibition of
`the A TPase activity.
`Since all partial reactions of the (H + + K + )(cid:173)
`A TPase were inhibited by picoprazole it was not
`possible to define the exact point at which the
`reaction pathway was inhibited. A puzzling feature
`of the inhibition of enzyme activity and of H+
`transport by the A TPase is the long preincubation
`requirement. Picoprazole is a hydrophobic com(cid:173)
`pound, which in the unprotonated form, distrib(cid:173)
`utes as a weak base into acidic compartments [6].
`Accordingly, limited penetration of picoprazole is
`unlikely to account for the time dependence of
`inhibition.
`Acidic pH facilitated the inhibition, but it was
`not determined whether this was due to a change
`
`10"6
`[ Picoprazole J
`
`Fig. 8. Effects of picoprazole on p-nitrophenylphosphatase
`((H + + K + )-A TPase),
`(N a+ + K + )-ATPase and p(cid:173)
`nitrophenylphosphatase ((Na + + K + )-ATPase) activity.
`Gastric membranes (10 1-1g of proteinjml) were incubated with
`picoprazole for 30 min in 5 mM Pipes-Tris buffer, pH 7.4
`<•--•). Samples were assayed as detailed in Methods.
`K + -p-nitrophenylphosphatase ((H + + K + )-ATPase) activity in
`the absence of inhibitor was taken as 100%. Kidney membranes
`(10 1-'g of proteinjml) were incubated with picoprazole in 5
`mM Pipes-Tris buffer (pH 7.4) for 30 min. Samples were
`assayed either for (Na + + K + )-ATPase activity (e--e) or
`for p-nitrophenylphosphatase ((Na + + K + )-ATPase) activity
`(N a+ + K + )-ATPase activity and p(cid:173)
`(& - - &),
`nitrophenylphosphatase ((Na + + K + )-ATPase) activity in the
`absence of picoprazole were taken as 100%.
`
`MgA TP. Secondly, hydrolysis of p-nitrophenyl
`phosphate cannot promote proton transport [23].
`From the experiment presented in Fig. 8, it is
`the p(cid:173)
`evident
`that picoprazole
`inhibited
`nitrophenylphosphatase ((H + + K + )-ATPase) ac(cid:173)
`tivity in a concentration-dependent manner. Half(cid:173)
`maximal inhibition was obtained at a concentra(cid:173)
`tion of 5 · 10- 6 M, which is in agreement with the
`value found for the inhibition of K +-A TPase.
`
`Effects on ( N a+ + K +)-A TPase
`Picoprazole was not able to discriminate be(cid:173)
`tween K + -A TPase, p-nitrophenylphosphatase
`((H + + K +)-A TPase) phosphoenzyme formation
`or proton transport. This finding led us to examine
`whether this lack of discrimination was only true
`of the various (H + + K +)-A TPase reactions. We
`therefore also studied the effect of picoprazole on
`a related enzyme system, namely the (Na + + K + )(cid:173)
`A TPase which was chosen because of its close
`similarities to the (H + + K +)-A TPase [27]. We
`
`

`
`38
`
`in reactivity of the inhibitor or to the protein.
`However, both the enhanced reactivity at low pH
`and the trapping of picoprazole in the secretory
`canaliculus [6] would be expected to enhance the
`activity of the compound in the intact cell.
`From the data presented, it can be concluded
`that picoprazole cannot discriminate between K + -
`ATPase, p-nitrophenylphosphatase ((H+ + K + )(cid:173)
`ATPase), phosphoenzyme levels or transport reac(cid:173)
`tions of the enzyme. However, a closely related
`enzyme, (N a+ + K + )-A TPase, was not affected by
`picoprazole. It therefore appears that local acidic
`compartments within the gastric gland are not the
`only factor governing
`the selectivity of pico(cid:173)
`prazole; when tested on the isolated enzymes, pi(cid:173)
`coprazole inhibited the (H + + K +)-A TPase more
`readily than the (N a+ + K +)-A TPase. This com(cid:173)
`pound thus appears to act selectively on the parietal
`cell, by virtue of its inhibition of the gastric proton
`pump.
`
`Acknowledgement
`
`The authors thank Miss Britt-Marie Jaresten
`for excellent technical assistance, Mrs. Greta Tebr(cid:173)
`ing for typing the manuscript and Mrs. Berit
`Elander for drawing the graphs.
`
`References
`
`Sjostrand, S.-E., Ryberg, B. and Olbe, L. (1978) in Gastric
`Ion Transport (Obrink, K.J. and F1emstrom, G., eds.), Acta
`Physiol. Scand. Spec, Suppl., 181-185
`2 Olbe, L., Sjostrand, S.-E. and Fellenius, E. (1979) in Gastrins
`and the Vagus (Rehfeld, R.F. and Amdrup, E., eds.), pp.
`245-250 Academic Press, New York
`3 Olbe, L., Berglindh, T., E1ander, B., Helander, H., Fellenius,
`E., Sjostrand, S.-E., Sundell, G. and Wallmark, B. (1979)
`Scand. J. Gastroenterol. Supp. 55, 131-133
`4 Berglindh, T. and Obrink, K.J. (1976) Acta Physiol. Scand.
`96, 150-159
`5 Fellenius, E., Berglindh, T., Sachs, G., Olbe, L., Elander, B.,
`
`Sjostrand, S.-E. and Wallmark, B. (1981) Nature 290,
`159-161
`6 Fellenius, E., Elander, B., Wallmark, B., Helander, H.F.
`and Berglindh, T. (1982) Am. J. Phys.: Gastrointest. Liver
`Physiol. 243, in the press
`7 Forte, J.G., Forte, T.M. and Saltman, P. (1967) J. Cell Bioi.
`69, 293, 304
`8 Ganser, A.L. and Forte, J.G. (1973) Biochim. Biophys. Acta
`307, 169-180
`9 Forte, J.G., Ganser, A.L., Beesley, R. and Forte, T.M.
`(1974) Gastroenterology 69, 825-852
`10 Saccomani, G., Helander, H.F., Crago, S., Chang, H.H.,
`Dailey, D.W. and Sachs, G. (1979) J. Cell Bioi. 83, 271-283
`I I Schackmann, R., Schwartz, A., Saccomani, G. and Sachs,
`G. (1977) J. Membrane Bioi. 32, 361-381
`12 Chang, H., Saccomani, G., Rabon, E., Schackmann, R. and
`Sachs, G. (1977) Biochim. Biophys. Acta 464, 313-327
`13 Sachs, G., Chang, H.H., Rabon, E., Schackmann, R., Lewin,
`M. and Saccomani, G. (1976) J. Bioi. Chern. 251, 7690-7698
`14 Forte, J.G., Machen, T. and Obrink, K.J. (1980) Annu. Rev.
`Physiol. 42, I 11-126
`15 Saccomani, G., Stewart, H.B., Shaw, D., Lewin, M. and
`Sachs, G. ( 1977) Biochim. Biophys. Acta 465, 3 I 1-330
`16 J~Jgensen, P.L. (1975) Biochim. Biophys. Acta 401, 399-415
`17 Fiske, C.H. and SubbaRow, Y. (1925) J. Bioi. Chern. 66,
`375-400
`18 Torriani, A. (1968) Methods Enzymol. 12B, 212-215
`19 Rabon, E., Chang, H. and Sachs, G. (1978) Biochemistry
`17, 3345
`20 Lowry, O.H., Rosebrough, N.J., Farr, A.L. and Randall,
`R.J. (1951) J. Bioi. Chern. 193, 265-275
`21 Levy, H.M., Leber, P.D. and Ryan, E.M. (1963) J. Bioi.
`Chern. 238, 3654-3659
`22 Stewart, H.B., Wallmark, B. and Sachs, G. (1981) J. Bioi.
`Chern. 256, 2682-2690
`23 Saccomani, G., Shah, G., Spenney, J.G. and Sachs, G.
`(1975) J. Bioi. Chern. 250, 4802-4809
`24 Ray, T.K. and Forte, J.G. (1976) Biochim. Biophys. Acta
`443, 451-467
`25 Wallmark, B. and Mardh, S. (1979) J. Bioi. Chern. 2544,
`I I 899- II 902
`26 Wallmark. B. and Stewart, H.B., Rabon, E., Saccomani, G.
`and Sachs, G. (1980) J. Bioi. Chern. 25, 5313-5319
`27 Kyte, J. (1975) J. Bioi. Chern. 250, 7443-7449
`28 Black, J.W., Duncan, W.A.M., Durant, C.J., Ganellin, C.R.
`and Parsons, E.M. (1972) Nature 236, 385-390
`29 Hirschowitz, B.l. and Sachs, G. (1969) Gastroenterology 56,
`693-702

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket