Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G671-81, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26847387

ABSTRACT

A computer model, constructed for evaluation of integrated functioning of cellular components involved in acid secretion by the gastric parietal cell, has provided new interpretations of older experimental evidence, showing the functional significance of a canalicular space separated from a mucosal bath by a gland lumen and also shedding light on basolateral Cl(-) transport. The model shows 1) changes in levels of parietal cell secretion (with stimulation or H-K-ATPase inhibitors) result mainly from changes in electrochemical driving forces for apical K(+) and Cl(-) efflux, as canalicular [K(+)] ([K(+)]can) increases or decreases with changes in apical H(+)/K(+) exchange rate; 2) H-K-ATPase inhibition in frog gastric mucosa would increase [K(+)]can similarly with low or high mucosal [K(+)], depolarizing apical membrane voltage similarly, so electrogenic H(+) pumping is not indicated by inhibition causing similar increase in transepithelial potential difference (Vt) with 4 and 80 mM mucosal K(+); 3) decreased H(+) secretion during strongly mucosal-positive voltage clamping is consistent with an electroneutral H-K-ATPase being inhibited by greatly decreased [K(+)]can (Michaelis-Menten mechanism); 4) slow initial change ("long time-constant transient") in current or Vt with clamping of Vt or current involves slow change in [K(+)]can; 5) the Na(+)-K(+)-2Cl(-) symporter (NKCC) is likely to have a significant role in Cl(-) influx, despite evidence that it is not necessary for acid secretion; and 6) relative contributions of Cl(-)/HCO3 (-) exchanger (AE2) and NKCC to Cl(-) influx would differ greatly between resting and stimulated states, possibly explaining reported differences in physiological characteristics of stimulated open-circuit Cl(-) secretion (≈H(+)) and resting short-circuit Cl(-) secretion (>>H(+)).


Subject(s)
Computer Simulation , Parietal Cells, Gastric/metabolism , Potassium/metabolism , Animals , Bicarbonates/metabolism , Chlorides/metabolism , Humans , Ion Pumps/metabolism
2.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G699-710, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24578340

ABSTRACT

Selective inhibitors of myosin or actin function and confocal microscopy were used to test the role of an actomyosin complex in controlling morphology, trafficking, and fusion of tubulovesicles (TV) containing H-K-ATPase with the apical secretory canaliculus (ASC) of primary-cultured rabbit gastric parietal cells. In resting cells, myosin IIB and IIC, ezrin, and F-actin were associated with ASC, whereas H-K-ATPase localized to intracellular TV. Histamine caused fusion of TV with ASC and subsequent expansion resulting from HCl and water secretion; F-actin and ezrin remained associated with ASC whereas myosin IIB and IIC appeared to dissociate from ASC and relocalize to the cytoplasm. ML-7 (inhibits myosin light chain kinase) caused ASC of resting cells to collapse and most myosin IIB, F-actin, and ezrin to dissociate from ASC. TV were unaffected by ML-7. Jasplakinolide (stabilizes F-actin) caused ASC to develop large blebs to which actin, myosin II, and ezrin, as well as tubulin, were prominently localized. When added prior to stimulation, ML-7 and jasplakinolide prevented normal histamine-stimulated transformations of ASC/TV and the cytoskeleton, but they did not affect cells that had been previously stimulated with histamine. These results indicate that dynamic pools of actomyosin are required for maintenance of ASC structure in resting cells and for trafficking of TV to ASC during histamine stimulation. However, the dynamic pools of actomyosin are not required once the histamine-stimulated transformation of TV/ASC and cytoskeleton has occurred. These results also show that vesicle trafficking in parietal cells shares mechanisms with similar processes in renal collecting duct cells, neuronal synapses, and skeletal muscle.


Subject(s)
Actins/metabolism , Nonmuscle Myosin Type IIB/metabolism , Parietal Cells, Gastric , Transport Vesicles , Animals , Azepines/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Cell Physiological Phenomena/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , H(+)-K(+)-Exchanging ATPase/metabolism , Naphthalenes/pharmacology , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/pathology , Rabbits , Transport Vesicles/drug effects , Transport Vesicles/physiology
3.
Annu Rev Physiol ; 72: 273-96, 2010.
Article in English | MEDLINE | ID: mdl-20148676

ABSTRACT

The gastric parietal cell was the first system where a regulated membrane recycling hypothesis was proposed as the principal means for moving molecular transporters between cellular compartments. Upon stimulation, massive membrane flow from an endosomal compartment of tubulovesicle membranes to the apical secretory surface places the ATP-driven pumps in position to secrete a solution of strong acid in collaboration with several other membrane transporters. This review focuses on the membrane recycling pathway and proteins that support the recruitment and redistribution of H,K-ATPase-rich membranes, including those involved in signal transduction, membrane targeting, docking, and fusing, in addition to the integral role of the actin cytoskeleton and its associated proteins in the process of membrane recycling. Although much of the evidence discussed here comes from parietal cell studies, other physiological transport systems, as well as less complex cellular and in vitro models, are examined and cited for generality of principle.


Subject(s)
H(+)-K(+)-Exchanging ATPase/metabolism , Parietal Cells, Gastric/enzymology , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , Endocytosis/physiology , Gastric Acid/metabolism , Gene Expression Regulation, Enzymologic/physiology , Humans , SNARE Proteins/metabolism , SNARE Proteins/physiology , Signal Transduction/physiology , rab GTP-Binding Proteins/physiology
4.
J Pharmacol Sci ; 122(3): 213-22, 2013.
Article in English | MEDLINE | ID: mdl-23877017

ABSTRACT

The proposed mechanism for proton pump inhibitors (PPIs) is that PPIs are activated at low pH to the sulfenamide form, which reacts with the sulfhydryl group of cysteine(s) at the active site of the proton pump, to produce reducible disulfide-bonded PPI-proton pump conjugates. However, this mechanism cannot explain the observations that some PPI-protein conjugates are irreducible. This study was designed to investigate the chemistry of the irreducible conjugates by mass spectrometry, using three PPIs and 17 cysteine-containing peptides. While some peptides favored the formation of reducible PPI-peptide adduct, the other peptides mainly produced irreducible adducts. Characterization of the irreducible adduct revealed that the irreducible bonding required the participation of both a sulfhydryl group and a nearby primary amino group. High resolution mass spectrometry suggested a molecular structure of the irreducible adduct. These results suggested a reaction mechanism in which the PPI pyridone form reacted with an amino group and a sulfhydryl group to form an irreducible adduct. The irreducible adduct becomes the dominant product over time because of the irreversible nature of the pyridone-mediated reaction. These findings may explain the irreducible inhibition of H/K-ATPase by PPIs and their relatively slow biological turnover in vivo. [Supplementary materials: available only at http://dx.doi.org/10.1254/jphs.13058FP].


Subject(s)
Cysteine/chemistry , Peptides/chemistry , Proton Pump Inhibitors/chemistry , Proton Pump Inhibitors/pharmacology , 2-Pyridinylmethylsulfinylbenzimidazoles/chemistry , 2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Catalytic Domain , Enzyme Inhibitors , H(+)-K(+)-Exchanging ATPase , Hydrogen-Ion Concentration , Lansoprazole/chemistry , Lansoprazole/pharmacology , Mass Spectrometry , Molecular Structure , Omeprazole/chemistry , Omeprazole/pharmacology , Pantoprazole , Pyridones , Sulfamerazine
5.
Am J Physiol Cell Physiol ; 303(12): C1301-11, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23099641

ABSTRACT

In primary culture, the gastric parietal cell's deeply invaginated apical membrane, seen in microscopy by phalloidin binding to F-actin (concentrated in microvilli and a subapical web), is engulfed into the cell, separated from the basolateral membrane (which then becomes the complete plasma membrane), and converted, from a lacy interconnected system of canaliculi, into several separate vacuoles. In this study, vacuolar morphology was achieved by 71% of parietal cells 8 h after typical collagenase digestion of rabbit gastric mucosa, but the tight-junctional protein zonula occludens-1 (ZO-1) was completely delocalized after ∼2 h, when cells were ready for culturing. Use of low-Ca(2+) medium (4 mM EGTA) to release cells quickly from gastric glands yielded parietal cells in which ZO-1 was seen in a small spot or ring, a localization quickly lost if these cells were then cultured in normal Ca(2+) but remaining up to 20 h if they were cultured in low Ca(2+). The cells in low Ca(2+) mostly retained, at 20 h, an intermediate morphology of many bulbous canalicular expansions ("prevacuoles"), seemingly with narrow interconnections. Histamine stimulation of 20-h cells with intermediate morphology caused colocalization of proton-pumping H-K-ATPase with canaliculi and prevacuoles but little swelling of those structures, consistent with a remaining apical pore through which secreted acid could escape. Apparent canalicular interconnections, lack of stimulated swelling, and lingering ZO-1 staining indicate inhibition of membrane fission processes that separate apical from basolateral membrane and vacuoles from each other, suggesting an important role for extracellular Ca(2+) in these, and possibly other, endocytotic processes.


Subject(s)
Calcium/pharmacology , Parietal Cells, Gastric/cytology , Vacuoles/metabolism , Animals , Cells, Cultured , H(+)-K(+)-Exchanging ATPase/metabolism , Histamine/pharmacology , Microvilli/metabolism , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/metabolism , Rabbits , Vacuoles/drug effects , Zonula Occludens-1 Protein/metabolism
6.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G591-600, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21719736

ABSTRACT

Potassium ions are required for gastric acid secretion. Several potassium channels have been implicated in providing K(+) at the apical membrane of parietal cells. In examining the mRNA expression levels between gastric mucosa and liver tissue, KCNJ15 stood out as the most highly specific K(+) channel in the gastric mucosa. Western blot analysis confirmed that KCNJ15 is abundant in the stomach. Immunofluorescence staining of isolated gastric glands indicated that KCNJ15 was expressed in parietal cells and chief cells, but not in mucous neck cells. In resting parietal cells, KCNJ15 was mainly found in puncta throughout the cytoplasm but was distinct from H(+)-K(+)-ATPase. Upon stimulation, KCNJ15 and H(+)-K(+)-ATPase become colocalized on the apical membranes, as suggested by immunofluorescence staining. Western blot analysis of the resting and the stimulated membrane fractions confirmed this observation. From nonsecreting preparations, KCNJ15-containing vesicles sedimented after a 4-h centrifugation at 100,000 g, but not after a 30-min spin, which did sediment most of the H(+)-K(+)-ATPase-containing tubulovesicles. Most of the KCNJ15 containing small vesicle population was depleted upon stimulation of parietal cells, as indicated by the fact that the KCNJ15 signal was shifted to a large membrane fraction that sedimented at 4,000 g. Our results demonstrate that, in nonsecreting parietal cells, KCNJ15 is stored in vesicles distinct from the H(+)-K(+)-ATPase-enriched tubulovesicles. Furthermore, upon stimulation, KCNJ15 and H(+)-K(+)-ATPase both translocate to the apical membrane for active acid secretion. Thus KCNJ15 can be added to the family of apical K(+) channels in gastric parietal cells.


Subject(s)
H(+)-K(+)-Exchanging ATPase/metabolism , Parietal Cells, Gastric/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Aminopyrine/metabolism , Animals , HEK293 Cells , Humans , Mice , Potassium Channels, Inwardly Rectifying/biosynthesis , RNA, Messenger/metabolism , Rabbits
7.
Proc Natl Acad Sci U S A ; 105(46): 17955-60, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-19004773

ABSTRACT

Slc26a9 is a recently identified anion transporter that is abundantly expressed in gastric epithelial cells. To study its role in stomach physiology, gene targeting was used to prepare mice lacking Slc26a9. Homozygous mutant (Slc26a9(-/-)) mice appeared healthy and displayed normal growth. Slc26a9 deletion resulted in the loss of gastric acid secretion and a moderate reduction in the number of parietal cells in mutant mice at 5 weeks of age. Immunofluorescence labeling detected the H-K-ATPase exclusively on the apical pole of gastric parietal cells in Slc26a9(-/-) mice, in contrast to the predominant cytoplasmic localization in Slc26a9(+/+) mice. Light microscopy indicated that gastric glands were dilated, and electron micrographs displayed a distinct and striking absence of tubulovesicles in parietal cells and reductions in the numbers of parietal and zymogen cells in Slc26a9(-/-) stomach. Expression studies indicated that Slc26a9 can function as a chloride conductive pathway in oocytes as well as a Cl(-)/HCO(3)(-) exchanger in cultured cells, and localization studies in parietal cells detected its presence in tubulovesicles. We propose that Slc26a9 plays an essential role in gastric acid secretion via effects on the viability of tubulovesicles/secretory canaliculi and by regulating chloride secretion in parietal cells.


Subject(s)
Antiporters/deficiency , Cell Membrane/pathology , Gastric Acid/metabolism , Gene Deletion , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/pathology , Animals , Animals, Newborn , Anion Transport Proteins/metabolism , Antiporters/metabolism , Biomarkers/metabolism , COS Cells , Chlorocebus aethiops , Fluorescent Antibody Technique , H(+)-K(+)-Exchanging ATPase/metabolism , Hydrogen-Ion Concentration , Immunoblotting , Mice , Mice, Knockout , Parietal Cells, Gastric/enzymology , Parietal Cells, Gastric/ultrastructure , SLC4A Proteins , Sulfate Transporters , Titrimetry , Xenopus
8.
Am J Physiol Cell Physiol ; 299(2): C431-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20505040

ABSTRACT

Ezrin is an important membrane/actin cytoskeleton linker protein, especially in epithelia. Ezrin has two important binding domains: an NH(2)-terminal region that binds to plasma membrane and a COOH-terminal region that binds to F-actin only after a conformational activation by phosphorylation at Thr567 of ezrin. The present experiments were undertaken to investigate the detailed cellular changes in the time course of expression of ezrin-T567 mutants (nonphosphorylatable T567A and permanent phospho-mimic T567D) in parietal cells and to assess ezrin distribution and its influence on the elaborate membrane recruitment processes of these cells. T567A mutant and wild-type (WT) ezrin were consistently localized to the apical plasma membrane, even with overexpression. On the other hand, T567D went first to apical membrane at early times and low expression levels, then accumulated mainly at the basal surface after 24 h. Overexpression of WT or T567A led to incorporation of internal membranes to apical vacuoles, while overexpression of T567D led to large incorporation of apical and intracellular membranes (including H-K-ATPase) to the basal surface. Differences in polar distribution of ezrin suggest a role for the linker protein in promoting formation and plasticity of membrane surface projections, forming the basis for a novel theory for ezrin as an organizer and regulator of membrane recruitment. A model simulating the cellular distribution of ezrin and its associated membrane- and F-actin-binding forms is given to predict redistributions observed with phosphorylation and mutant overexpression, and it can easily be modified as more specific information regarding binding constants and specific sites becomes available.


Subject(s)
Cell Communication/physiology , Cell Polarity/physiology , Cytoskeletal Proteins/physiology , Actins/metabolism , Actins/physiology , Animals , Cell Line , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Cytoskeleton/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gastric Mucosa/physiology , Humans , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/physiology , Rabbits , Time Factors
9.
FEBS Lett ; 581(22): 4318-24, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17716669

ABSTRACT

Syntaxin and Munc18 are essential for regulated exocytosis in all eukaryotes. It was shown that Munc18 inhibition of neuronal syntaxin 1 can be overcome by CDK5 phosphorylation, indicating that structural change disrupts the syntaxin-Munc18 interaction. Here, we show that this phosphorylation promotes the assembly of Munc18b-syntaxin 3-SNAP25 tripartite complex and membrane fusion machinery SNARE. Using siRNAs to screen for genes required for regulated epithelial secretion, we identified the requirements of CDK5 and Munc18b in cAMP-dependent gastric acid secretion. Biochemical characterization revealed that Munc18b bears a syntaxin 3-selective binding site located at its most C-terminal 53 amino acids. Significantly, the phosphorylation of Thr572 by CDK5 attenuates Munc18b-syntaxin 3 interaction and promotes formation of Munc18b-syntaxin 3-SNAP25 tripartite complex, leading to an assembly of functional Munc18b-syntaxin 3-SNAP25-VAMP2 membrane fusion machinery. Thus, our studies suggest a novel regulatory mechanism in which phosphorylation of Munc18b operates vesicle docking and fusion in regulated exocytosis.


Subject(s)
Epithelium/metabolism , Munc18 Proteins/metabolism , Parietal Cells, Gastric/metabolism , Qa-SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Epithelium/enzymology , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/enzymology , Phosphorylation , Protein Binding , RNA, Small Interfering/metabolism , Rabbits
10.
J Histochem Cytochem ; 52(1): 77-86, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688219

ABSTRACT

High-pressure freezing (HPF) is currently the most reliable method to obtain an adequately frozen sample for high-resolution morphological evaluation. Here we applied the HPF technique to isolated rabbit gastric glands to reveal structural evidence that may be correlated with functional activity of gastric parietal cells. This approach provided well-preserved fine structure and excellent antigenicity of several parietal cell proteins. Microtubules were abundant in the cytoplasm and frequently appeared to be associating with tubulovesicles. Interestingly, many electron-dense coated vesicles were apparent around the intracellular canaliculi (IC) of resting parietal cells, consistent with active membrane retrieval from the apical membranes. Immunolabeling of H+/K+-ATPase was evident on the endocytic components (e.g., multivesicular bodies) and tubulovesicles. After histamine stimulation, the parietal cells characteristically showed expanded IC membranes with varied features of their apical microvilli. The labeling density of H+/K+-ATPase was four-fold higher on the IC membrane of stimulated parietal cells than on that of resting parietal cells. Immunolabeling of ezrin was clearly identified on the IC and basolateral membranes of parietal cells, corresponding to their F-actin-rich sites. The present findings provide a new insight into the correlation of cell structure and function in gastric parietal cells.


Subject(s)
H(+)-K(+)-Exchanging ATPase/metabolism , Parietal Cells, Gastric/enzymology , Parietal Cells, Gastric/ultrastructure , Animals , Cimetidine/pharmacology , Cryopreservation , Cytoskeletal Proteins , Gastric Mucosa , Histamine/pharmacology , Immunohistochemistry , In Vitro Techniques , Organ Preservation , Phosphoproteins/metabolism , Rabbits , Subcellular Fractions/enzymology
11.
J Gastroenterol ; 48(12): 1343-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23478938

ABSTRACT

BACKGROUND: Proton pump inhibitors (PPIs) are pro-drugs requiring an acidic pH for activation. The specificity of PPI toward the proton pump is mainly due to the extremely low pH at the parietal cell canalicular membrane where the pump is located. Reactivity of PPIs was also observed in moderately acidic environments like the renal collecting duct. But no PPI effect on lysosomal enzymes has been observed possibly because the previous studies were performed with liver tissue, where PPIs are metabolized. METHODS: The reactivity of PPIs (omeprazole, lansoprazole and pantoprazole) with a cysteine-containing peptide was analyzed by mass spectrometry, and the impact of PPIs on lysosomal enzymes was evaluated in cultured cells and mice. The effect of PPIs on the immune system was examined with a mouse tumor immunotherapy model. RESULTS: Incubation of a cysteine-containing peptide with PPIs at pH5 led to the conversion of most of the peptide into PPI-peptide adducts. Dose dependent inhibition of lysosomal enzyme activities by PPIs was observed in cultured cells and mouse spleen. Further, PPI counteracted the tumor immunotherapy in a mouse model. CONCLUSIONS: Our data support the hypothesis that many of the PPI adverse effects are caused by systematically compromised immunity, a result of PPI inhibition of the lysosomal enzymes. This novel mechanism complements the existing mechanisms in explaining the increased incidence of tumorigenesis and infectious diseases among PPI users and underlie the ongoing concern about the overuse of PPIs in adult and pediatric populations.


Subject(s)
Anti-Ulcer Agents/pharmacology , Enzymes/drug effects , Lysosomes/drug effects , Proton Pump Inhibitors/pharmacology , 2-Pyridinylmethylsulfinylbenzimidazoles/administration & dosage , 2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Animals , Anti-Ulcer Agents/administration & dosage , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzymes/metabolism , Humans , Hydrogen-Ion Concentration , Immune System/drug effects , Immunotherapy/methods , Lansoprazole/administration & dosage , Lansoprazole/pharmacology , Lysosomes/enzymology , Male , Mass Spectrometry/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Omeprazole/administration & dosage , Omeprazole/pharmacology , Pantoprazole , Prodrugs , Proton Pump Inhibitors/administration & dosage , Spleen/drug effects , Spleen/enzymology
12.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G185-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074636

ABSTRACT

ERM (ezrin, radixin, and moesin) proteins play critical roles in epithelial and endothelial cell polarity, among other functions. In gastric glands, ezrin is mainly expressed in acid-secreting parietal cells, but not in mucous neck cells or zymogenic chief cells. In looking for other ERM proteins, moesin was found lining the lumen of much of the gastric gland, but it was not expressed in parietal cells. No significant radixin expression was detected in the gastric glands. Moesin showed an increased gradient of expression from the neck to the base of the glands. In addition, the staining pattern of moesin revealed a branched morphology for the gastric lumen. This pattern of short branches extending from the glandular lumen was confirmed by using antibody against zonula occludens-1 (ZO-1) to stain tight junctions. With a mucous neck cell probe (lectin GSII, from Griffonia simplicifolia) and a chief cell marker (pepsinogen C), immunohistochemistry revealed that the mucous neck cells at the top of the glands do not express moesin, but, progressing toward the base, mucous cells showing decreased GSII staining had low or moderate level of moesin expression. The level of moesin expression continued to increase toward the base of the glands and reached a plateau in the base where chief cells and parietal cells abound. The level of pepsinogen expression also increased toward the base. Pepsinogen C was located on cytoplasmic granules and/or more generally distributed in chief cells, whereas moesin was exclusively expressed on the apical membrane. This is a clear demonstration of distinctive cellular expression of two ERM family members in the same tissue. The results provide the first evidence that moesin is involved in the cell biology of chief cells. Novel insights on gastric gland morphology revealed by the moesin and ZO-1 staining provide the basis for a model of cell maturation and migration within the gland.


Subject(s)
Chief Cells, Gastric/chemistry , Gastric Mucosa/chemistry , Microfilament Proteins/analysis , Animals , Cell Differentiation , Cell Membrane/chemistry , Chief Cells, Gastric/enzymology , Cytoplasmic Granules/chemistry , Cytoskeletal Proteins/analysis , Fluorescent Antibody Technique , Gastric Mucosa/cytology , Gastric Mucosa/enzymology , Membrane Proteins/analysis , Parietal Cells, Gastric/chemistry , Pepsinogen C/analysis , Phosphoproteins/analysis , Plant Lectins , Rabbits , Tight Junctions/chemistry , Zonula Occludens-1 Protein
13.
Am J Physiol Cell Physiol ; 295(1): C192-202, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18480298

ABSTRACT

In a comparison of three different tissues, the membrane cytoskeleton linker protein ezrin was found to assume high levels of phosphorylation on threonine-567 (T567) in the brush border membranes of renal proximal tubule cells and small intestine enterocytes, in contrast to the apical canalicular membrane of gastric parietal cells. Together with an earlier observation that increased T567 phosphorylation is associated with more elaborate microvilli in parietal cells, this comparative study suggested a higher phosphorylation level requirement for the denser and more uniform distribution of microvilli at brush border surfaces. Using a kinase inhibitor, staurosporin, and metabolic inhibitor, sodium azide, relatively high turnover of ezrin T567 phosphorylation was observed in all three epithelia. Aiming to understand the role of phosphorylation turnover in these tissues, detergent extraction analysis of gastric glands and proximal tubules revealed that an increased phosphorylation on ezrin T567 greatly enhanced its association with F-actin, while ezrin-membrane interaction persisted regardless of the changes of phosphorylation level on ezrin T567. Finally, expression of Thr567Asp mutant ezrin, which mimics the phospho-ezrin state but does not allow turnover, caused aberrant growth of membrane projections in cultured proximal tubule cells, consistent with what had previously been observed in several cell lines and gastric parietal cells. These results fit into a model of surface plasticity, which posits that the turnover of phosphorylation on T567 empowers ezrin to relax and reposition membrane to the underlying cytoskeleton under varying conditions of filament growth or rapid membrane expansion (or depletion).


Subject(s)
Cytoskeletal Proteins/metabolism , Epithelial Cells/metabolism , Actins/metabolism , Animals , Cell Membrane/metabolism , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Enterocytes/metabolism , In Vitro Techniques , Intestinal Mucosa/cytology , Intestine, Small/cytology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Microvilli/metabolism , Mutation , Organ Specificity , Phosphorylation , Rabbits , Threonine/metabolism
14.
Biochemistry ; 46(35): 10145-52, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17696364

ABSTRACT

The gastric H,K-ATPase is related to other cation transport ATPases, for example, Na,K-ATPase and Ca-ATPase, which are called E1-E2 ATPases in recognition of conformational transitions during their respective transport and catalytic cycles. Generally, these ATPases cannot utilize NTPs other than ATP for net ion transport activity. For example, under standard assay conditions, rates of NTP hydrolysis and H+ pumping by the H,K-ATPase for CTP are about 10% of those for ATP and undetectable with GTP, ITP, and UTP. However, we observed that H,K-ATPase will catalyze NTP/ADP phosphate exchange at similar rates for all of these NTPs, suggesting that a common phosphoenzyme intermediate is formed. The present study was undertaken to evaluate the specificity of nucleotides to power the H,K-ATPase and several of its partial reactions, including NTP/ADP exchange, K+-catalyzed phosphatase activity, and proton pumping. Results demonstrate that under conditions that promote the conformational change of the K+ bound form of the enzyme, K.E2, to E1, all NTPs tested support K+-stimulated NTPase activity and H+ pumping up to 30-50% of that with ATP. These conditions include (1) the presence of ADP as well as the NTP energy source and (2) reduced K+ concentration on the cytoplasmic side to approximately 0. These data conform to structural models for E1-E2 ATPases whereby adenosine binding promotes the K.E2 to E1 conformational change and K+ deocclusion.


Subject(s)
Adenosine Triphosphate/metabolism , Gastric Mucosa/enzymology , H(+)-K(+)-Exchanging ATPase/metabolism , Nucleotides/metabolism , Proton-Translocating ATPases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Diphosphate/metabolism , Animals , Biological Transport, Active , Calcium-Transporting ATPases/metabolism , Guanosine Triphosphate/metabolism , H(+)-K(+)-Exchanging ATPase/chemistry , Ion Transport , Potassium/metabolism , Protein Conformation , Proton Pump Inhibitors , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Substrate Specificity , Swine
15.
Am J Physiol Cell Physiol ; 293(3): C874-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17553936

ABSTRACT

In its dormant state, the membrane cytoskeletal linker protein ezrin takes on a NH(2) terminal-to-COOH terminal (N-C) binding conformation. In vitro evidence suggests that eliminating the N-C binding conformation by Thr(567) phosphorylation leads to ezrin activation. Here, we found for resting gastric parietal cells that the levels of ezrin phosphorylation on Thr(567) are low and can be increased to a small extent ( approximately 40%) by stimulating secretion via the cAMP pathway. Treatment of cells with protein phosphatase inhibitors led to a rapid, dramatic increase in Thr(567) phosphorylation by 400% over resting levels, prompting the hypothesis that ezrin activity is regulated by turnover of phosphorylation on Thr(567). In vitro and in vivo fluorescence resonance energy transfer analysis demonstrated that Thr(567) phosphorylation opens the N-C interaction. However, even in the closed conformation, ezrin localizes to membranes by an exposed NH(2) terminal binding site. Importantly, the opened phosphorylated form of ezrin more readily cosediments with F-actin and binds more tightly to membrane than the closed forms. Furthermore, fluorescence recovery after photobleaching analysis in live cells showed that the Thr567Asp mutant had longer recovery times than the wild type or the Thr567Ala mutant, indicating the Thr(567)-phosphorylated form of ezrin is tightly associated with F-actin and the membrane, restricting normal activity. These data demonstrate and emphasize the functional importance of reversible phosphorylation of ezrin on F-actin binding. A novel model is proposed whereby ezrin and closely associated kinase and phosphatase proteins represent a motor complex to maintain a dynamic relationship between the varying membrane surface area and filamentous actin length.


Subject(s)
Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Gastric Mucosa/cytology , Parietal Cells, Gastric/metabolism , Actins/metabolism , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , In Vitro Techniques , Phosphorylation , Protein Conformation , Rabbits
16.
Mol Cell Proteomics ; 5(8): 1437-49, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16737952

ABSTRACT

ARF6 GTPase is a conserved regulator of membrane trafficking and actin-based cytoskeleton dynamics at the leading edge of migrating cells. A key determinant of ARF6 function is the lifetime of the GTP-bound active state, which is orchestrated by GTPase-activating protein (GAP) and GTP-GDP exchanging factor. However, very little is known about the molecular mechanisms underlying ARF6-mediated cell migration. To systematically analyze proteins that regulate ARF6 activity during cell migration, we performed a proteomic analysis of proteins selectively bound to active ARF6 using mass spectrometry and identified a novel ARF6-specific GAP, ACAP4. ACAP4 encodes 903 amino acids and contains two coiled coils, one pleckstrin homology domain, one GAP motif, and two ankyrin repeats. Our biochemical characterization demonstrated that ACAP4 has a phosphatidylinositol 4,5-bisphosphate-dependent GAP activity specific for ARF6. The co-localization of ACAP4 with ARF6 occurred in ruffling membranes formed upon AIF(4) and epidermal growth factor stimulation. ACAP4 overexpression limited the recruitment of ARF6 to the membrane ruffles in the absence of epidermal growth factor stimulation. Expression of GTP hydrolysis-resistant ARF6(Q67L) resulted in accumulations of ACAP4 and ARF6 in the cytoplasmic membrane, suggesting that GTP hydrolysis is required for the ARF6-dependent membrane remodeling. Significantly the depletion of ACAP4 by small interfering RNA or inhibition of ARF6 GTP hydrolysis by overexpressing GAP-deficient ACAP4 suppressed ARF6-dependent cell migration in wound healing, demonstrating the importance of ACAP4 in cell migration. Thus, our study sheds new light on the biological function of ARF6-mediated cell migration.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Movement/physiology , GTPase-Activating Proteins/metabolism , Intracellular Membranes/metabolism , ADP-Ribosylation Factor 6 , Actins/metabolism , Aluminum Compounds/chemistry , Amino Acid Substitution , Cell Membrane/metabolism , Cytoskeleton/physiology , Epidermal Growth Factor/metabolism , Fluorides/chemistry , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Membrane Microdomains/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Proteomics
17.
Am J Physiol Cell Physiol ; 288(6): C1242-54, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15788482

ABSTRACT

Ezrin is a member of ezrin, radixin, moesin (ERM) protein family that links F-actin to membranes. The NH(2)- and COOH-terminal association domains of ERM proteins, known respectively as N-ERMAD and C-ERMAD, participate in interactions with membrane proteins and F-actin, and intramolecular and intermolecular interactions within and among ERM proteins. In gastric parietal cells, ezrin is heavily represented on the apical membrane and is associated with cell activation. Ezrin-ezrin interactions are presumably involved in functional regulation of ezrin and thus became a subject of our study. Fluorescence resonance energy transfer (FRET) was examined with cyan fluorescent protein (CFP)- and yellow fluorescent protein (YFP)-tagged ezrin incorporated into HeLa cells and primary cultures of parietal cells. Constructs included YFP at the NH(2) terminus of ezrin (YFP-Ez), CFP at the COOH terminus of ezrin (Ez-CFP), and double-labeled ezrin (N-YFP-ezrin-CFP-C). FRET was probed using fluorescence microscopy and spectrofluorometry. Evidence of ezrin oligomer formation was found using FRET in cells coexpressing Ez-CFP and YFP-Ez and by performing coimmunoprecipitation of endogenous ezrin with fluorescent protein-tagged ezrin. Thus intermolecular NH(2)- and COOH-terminal association domain (N-C) binding in vivo is consistent with the findings of earlier in vitro studies. After the ezrin oligomers were separated from monomers, FRET was observed in both forms, indicating intramolecular and intermolecular N-C binding. When the distribution of native ezrin as oligomers vs. monomers was examined in resting and maximally stimulated parietal cells, a shift of ezrin oligomers to the monomeric form was correlated with stimulation, suggesting that ezrin oligomers are the membrane-bound dormant form in gastric parietal cells.


Subject(s)
Membrane Proteins/physiology , Parietal Cells, Gastric/physiology , Phosphoproteins/physiology , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , Cytoskeletal Proteins , Fluorescence Resonance Energy Transfer , Gene Expression/physiology , HeLa Cells , Humans , In Vitro Techniques , Membrane Proteins/metabolism , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/ultrastructure , Phosphoproteins/metabolism , Protein Binding , Rabbits
18.
J Cell Sci ; 118(Pt 19): 4381-91, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16144865

ABSTRACT

Phosphorylation of the membrane-cytoskeleton linker protein ezrin has been functionally linked to acid secretion and vesicle recruitment to the apical secretory membrane in gastric parietal cells. Phosphorylation of the conserved T567 residue of ezrin has been shown to alter the N/C oligomerization of ezrin and promote the formation of actin-rich surface projections in other cells. To test the importance of T567 as a regulatory site for ezrin in parietal cell activation, we incorporated wild-type (WT) and mutant forms of ezrin, including the nonphosphorylatable T567A mutation and a mutant mimicking permanent phosphorylation, T567D. All ezrin constructs included C-terminal cyan-fluorescent protein (CFP) and were incorporated into adenoviral constructs for efficient introduction into cultured parietal cells from rabbit stomach. Fluorescence microscopy was used to localize CFP-ezrin and monitor morphological responses. Accumulation of a weak base (aminopyrine) was used to monitor receptor-mediated acid secretory response of the cultured cells. Similar to endogenous ezrin, WT and T567A CFP-ezrin localized heavily to apical membrane vacuoles with considerably lower levels associated with the surrounding basolateral membrane. Interestingly, H,K-ATPase within cytoplasmic tubulovesicles was incorporated into the apical vacuoles along with WT and T567A mutant ezrin. In these parietal cells secretagogue stimulation produced a striking vacuolar expansion associated with HCl secretion and the secretory phenotype. Expression of T567D CFP-ezrin was quite different, being rarely associated with apical vacuoles. T567D was more typically localized to the basolateral membrane, often associated with long spikes and fingerlike projections. Moreover, the cells did not display secretagogue-dependent morphological changes and, to our surprise, H,K-ATPase was recruited to the T567D CFP-ezrin-enriched basolateral projections. We conclude that T567 phosphorylation, which is probably regulated through Rho signaling pathway, may direct ezrin to membrane-cytoskeletal activity at the basolateral membrane and away from apical secretory activity. The large basolateral expansion is predicted to recruit membranes from sources not normally targeted to that surface.


Subject(s)
Cell Polarity , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/metabolism , Phosphoproteins/metabolism , Threonine/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cell Surface Extensions/metabolism , Cells, Cultured , Cytoskeletal Proteins , H(+)-K(+)-Exchanging ATPase/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Phenotype , Phosphoproteins/genetics , Phosphorylation , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vacuoles/metabolism
19.
Traffic ; 6(8): 654-66, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15998321

ABSTRACT

Syntaxins are differentially localized in polarized cells and play an important role in vesicle trafficking and membrane fusion. These soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins are believed to be involved in tubulovesicle trafficking and membrane fusion during the secretory cycle of the gastric parietal cell. We examined the cellular localization and distribution of syntaxin-1 and syntaxin-3 in rabbit parietal cells. Fractionation of gastric epithelial cell membranes showed that syntaxin-1 was more abundant in a fraction enriched in apical plasma membranes, whereas syntaxin-3 was found predominantly in the H,K-ATPase-rich tubulovesicle fraction. We also examined the cellular localization of syntaxins in cultured parietal cells. Parietal cells were infected with CFP-syntaxin-1 and CFP-syntaxin-3 adenoviral constructs. Fluorescence microscopy of live and fixed cells demonstrated that syntaxin-1 was primarily on the apical membrane vacuoles of infected cells, but there was also the expression of syntaxin-1 in a subadjacent cytoplasmic compartment. In resting, non-secreting parietal cells, syntaxin-3 was distributed throughout the cytoplasmic compartment; after stimulation, syntaxin-3 translocated to the apical membrane vacuoles, there co-localizing with H,K-ATPase, syntaxin-1 and F-actin. The differential location of these syntaxin isoforms in gastric parietal cells suggests that these proteins may be critical for maintaining membrane compartment identity and that they may play important, but somewhat different, roles in the membrane recruitment processes associated with secretory activation.


Subject(s)
Antigens, Surface/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Parietal Cells, Gastric/metabolism , Vesicular Transport Proteins/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Actins/metabolism , Adenoviridae/genetics , Animals , Antigens, Surface/genetics , Cell Fractionation , Cells, Cultured , Green Fluorescent Proteins/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Histamine/pharmacology , Membrane Proteins/genetics , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/ultrastructure , Promoter Regions, Genetic , Qa-SNARE Proteins , Rabbits , SNARE Proteins , Syntaxin 1 , Transfection
20.
Am J Physiol Cell Physiol ; 289(2): C493-505, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15800055

ABSTRACT

HCO3- secretion by gastric mucous cells is essential for protection against acidic injury and peptic ulcer. Herein we report the identification of an apical HCO3- transporter in gastric surface epithelial cells. Northern hybridization and RT-PCR demonstrate the expression of this transporter, also known as SLC26A9, in mouse and rat stomach and trachea (but not kidney). In situ hybridization in mouse stomach showed abundant expression of SLC26A9 in surface epithelial cells with apical localization on immunofluorescence labeling. Functional studies in HEK-293 cells demonstrated that SLC26A9 mediates Cl-/HCO3- exchange and is also capable of Cl--independent HCO3- extrusion. Unlike other anion exchangers or transport proteins reported to date, SLC26A9 activity is inhibited by ammonium (NH4+). The inhibitory effect of NH4+ on gastric HCO3- secretion was also indicated by reduced gastric juxtamucosal pH (pHjm) in rat stomach in vivo. This report is the first to describe the inhibition of HCO3- transport in vitro and the reduction of pHjm in stomach in vivo by NH4+. Given its critical localization on the apical membrane of surface epithelial cells, its ability to transport HCO3-, and its inhibition by NH4+, we propose that SLC26A9 mediates HCO3- secretion in surface epithelial cells and is essential for protection against acidic injury in the stomach. Disease states that are associated with increased ammonia (NH3)/NH4+ generation (e.g., Helicobacter pylori) may impair gastric HCO3- secretion and therefore predispose patients to peptic ulcer by inhibiting SLC26A9.


Subject(s)
Chloride-Bicarbonate Antiporters/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Quaternary Ammonium Compounds/metabolism , Animals , Blotting, Northern , Blotting, Western , Chloride-Bicarbonate Antiporters/genetics , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , In Situ Hybridization , Mice , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL