Anda di halaman 1dari 34

14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.

SGM LaTeX2e(2002/01/18) P1: fhd


10.1146/annurev.physiol.65.072302.114200

Annu. Rev. Physiol. 2003. 65:10331


doi: 10.1146/annurev.physiol.65.072302.114200
Copyright c 2003 by Annual Reviews. All rights reserved
First published online as a Review in Advance on December 9, 2002

CELL BIOLOGY OF ACID SECRETION BY


THE PARIETAL CELL

Xuebiao Yao1,2 and John G. Forte1


1
Department of Molecular and Cell Biology University of California, Berkeley, California
94720, and 2Laboratory of Cell Dynamics, University of Science and Technology of China,
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

Hefei, China 230027; email: xbyao@uclink.berkeley.edu; jforte@uclink.berkeley.edu

Key Words membrane recruitment, membrane fusion, gastric secretion,


cytoskeleton, SNARE, HCl secretion
by HINARI on 05/16/07. For personal use only.

Abstract Acid secretion by the gastric parietal cell is regulated by paracrine,


endocrine, and neural pathways. The physiological stimuli include histamine, acetyl-
choline, and gastrin via their receptors located on the basolateral plasma membranes.
Stimulation of acid secretion typically involves an initial elevation of intracellular
calcium and/or cAMP followed by activation of a cAMP-dependent protein kinase
cascade that triggers the translocation and insertion of the proton pump enzyme,
H,K-ATPase, into the apical plasma membrane of parietal cells. Whereas the
H,K-ATPase contains a plasma membrane targeting motif, the stimulation-mediated
relocation of the H,K-ATPase from the cytoplasmic membrane compartment to the
apical plasma membrane is mediated by a SNARE protein complex and its regulatory
proteins. This review summarizes the progress made toward an understanding of the
cell biology of gastric acid secretion. In particular we have reviewed the early signaling
events following histaminergic and cholinergic activation, the identification of multi-
ple factors participating in the trafficking and recycling of the proton pump, and the
role of the cytoskeleton in supporting the apical pole remodeling, which appears to be
necessary for active acid secretion by the parietal cell. Emphasis is placed on identify-
ing protein factors that serve as effectors for the mechanistic changes associated with
cellular activation and the secretory response.

INTRODUCTION

Secretion of HCl into the stomach is mediated by the oxyntic (acid-secreting)


cell, one of several epithelial cell types within gastric glands. In mammalian gas-
tric mucosa, oxyntic cells project peripherally, onto the walls of the gland and
thus are commonly called parietal cells, which is the designation we use in this
review. Gastric acid secretion is a tightly regulated process triggered by ligand-
receptor binding at the basolateral plasma membrane with ultimate output of H+,
Cl and H2O across the apical plasma membrane of the parietal cell. The phys-
iological stimuli include acetylcholine, gastrin, and especially, histamine, which
0066-4278/03/0315-0103$14.00 103
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

104 YAO FORTE

operate via basolateral membrane receptors. Stimulation of acid secretion typically


involves an initial elevation of intracellular calcium and cAMP, followed by acti-
vation of protein kinase cascades, which trigger the translocation and insertion of
H,K-ATPasethe proton pump enzymeinto the apical plasma membrane of the
parietal cell. This review primarily focuses on research over the past 10 years that
has sought to identify and describe specific roles for proteins involved in the activa-
tion, membrane translocation, and recycling processes that underlie acid secretion.
Table 1 provides a summary of the various proteins that have been implicated in the
functional activity of parietal cell secretion. In the course of this review, relevant
earlier studies are briefly discussed and references given to related review articles.
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

AN ELABORATE STRUCTURAL BASIS


FOR ACID SECRETION
by HINARI on 05/16/07. For personal use only.

The parietal cell is a highly specialized epithelial cell with several distinctive
morphological characteristics that directly bear on its functional activities (1).
The apical plasma membrane has the unusual structural form of a series of small
canals (canaliculi) that invaginate from the surface and project throughout the
entire cell interior, with frequent interconnections among canaliculi. In the non-
secreting or resting parietal cell, these apical canalicular surfaces are lined with
short, stubby microvilli that are supported by prominent actin microfilaments in-
cluding accessory actin-binding proteins. Throughout the cytoplasmic space there
is an abundance of membranous structures, rich in H,K-ATPase, that take the mor-
phological form of vesicles, tubules, and cisternal sacs; these are commonly called
H,K-ATPase-rich tubulovesicles.
Parietal cells undergo a profound morphological transition upon activation of
acid secretion. Although limited by the optics of light microscopy, careful histo-
logical examination by Golgi in the late 19th century noted the enlargement of
canaliculi that occurred in secreting parietal cells. Electron micrographs of maxi-
mally stimulated cells have since revealed dilated canalicular spaces, a dramatically
expanded apical membrane surface with elongated microvilli, and diminution of
cytoplasmic tubulovesicles (24). Mounting evidence has contributed to a general
consensus for the membrane recycling hypothesis which holds that activation of
acid secretion results in a fusion-based recruitment of H,K-ATPase-rich cytoplas-
mic membranes into the apical plasma membrane (5). However, an alternate view
has argued that the H,K-ATPase-rich membranes are contiguous with the apical
plasma membrane in both resting and stimulated states, thus negating the need
for a fusion-based recruitment process (68). Recent evidence has clarified the
morphological argument through the use of high-pressure, rapid-freeze fixation to
prepare gastric parietal cells for electron microscopy (EM) and analysis of three-
dimensional structure (9). Models constructed from ultra-thin serial sections, as
well as tomographic reconstruction of thick tissue sections using high voltage EM,
clearly demonstrate the separation of tubulovesicular membranes from the plasma
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 105

TABLE 1 Summary of parietal cell proteins implicated in the cell activation process
Mass Cellular Presumed
Name (kDa) location function Reference

Proton pump
H,K-ATPase = 96 Tubulovesicle (resting) H+ pumping (86)
-, -subunits = 6080 Apical PM (secreting) stabilize -subunit (115)
Trafficking and membrane recycling proteins
Syntaxin 1 34 Apical PM (?) Docking protein (92, 93)
Syntaxin 3 34 Cytoplasm (resting) H,K-ATPase translocation (92, 94)
Apical PM (secreting) (94)
SNAP-25 25 Apical PM SNARE pairing (92)
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

VAMP-2 18 Cytoplasm (resting) H,K-ATPase translocation (92, 93)


Apical PM (secreting)
Rab11 23 Cytoplasm (resting) H,K-ATPase translocation (94, 101)
Apical PM (secreting) (94, 101)
Rab25 23 ? H,K-ATPase translocation (100)
Rab11-interacting Tubulovesicle (resting) Rab11-myosin Vb binding (103)
proteins (FIP1 and FIP2)
by HINARI on 05/16/07. For personal use only.

pp75/Rip11 75 Tubulovesicle (resting) Rab11 binding (103)


SCAMPs 37 Tubulovesicle (resting) Vesicle trafficking (93)
Clathrin (heavy and 160; 35 Apical PM, tubulovesicles Endocytosis (104, 105)
light chains)
Dynamin 110 Apical PM Endocytosis (105)
-, -, -adaptins Tubulovesicle (resting) Endocytosis (104, 105)
Regulatory kinases
PKA cat. = 43 Cytoplasm Activation (17)
reg. = 52 Basolateral PM Anchoring
PKC (multiple isozymes) 80 Cytosol; membranes Protein phosphorylation (52, 53)
PI3 kinase cat. = 110 ? PI phosphorylation
reg. = 85
CAM kinase 50 Cytosol Protein phosphorylation (54)
MAP kinase 34 Cytosol Protein phosphorylation (59, 60)
MLCK 110 Cytosol Myosin lc phosphorylation (146)
Cytoskeleton
Actin
-cytoplasmic 43 Apical microvilli Microvillar formation (134)
-cytoplasmic 43 Basolateral PM Polarity formation (?) (134)
Tubulins 55 Cytoplasm TV trafficking ? (150)
Ezrin 80 Apical microvilli PKA-regulated membrane- (22, 24)
actin-filament linker
Coroninse 66 Apical microvilli PKC-dependent membrane- (28)
actin linker
Myosin V 110 Endosome Rab11a-binding; recycling (113)
Lasp-1 28 Apical PM Membrane-cytoskeleton (39, 41)
linking protein
Ankyrin 110 Apical PM H,K-ATPase binding (125)
linking protein
Spectrin 220/240 Apical PM Membrane-cytoskeleton (125)
Other proteins
AKAP120 120 ? PKA anchoring (146)
Calpain I cat. = 80 Cytosol Hydrolysis of ezrin (147)
reg. = 30
Parchorin 65 Cytosol (resting) Cl channel regulation (26, 42)
PM (stimulated)

PM = plasma membrane; TV = tubulovesicles.


14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

106 YAO FORTE

membrane and generally confirm early studies using chemical fixation techniques.
This revisiting of parietal cell ultrastructure, together with the requirement for
recruitment/fusion-based proteins in parietal cell activation (see below) reinforces
the recruitment-recycling model of parietal cell activation. In addition, this new
fixation method has revealed that the abundant parietal cell mitochondria form an
extensive reticular network throughout the cytoplasm, as has been reported for
several other cell types. Because of the close coupling between acid secretion and
oxidative metabolism, it will be of interest to evaluate whether, and to what extent,
dynamics of mitochondria are related to the cell activation.
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

SIGNAL TRANSDUCTION UNDERLYING GASTRIC


ACID SECRETION
Activation of acid secretion by parietal cells is triggered by paracrine, endocrine,
by HINARI on 05/16/07. For personal use only.

and neural stimuli in vivo. Study of the cell biology of acid secretion has been
greatly facilitated by the technique of gastric gland isolation, developed by
Berglindh & Obrink, along with the method to monitor acid secretion in vitro
by accumulation of the weak base, aminopyrine (10).
From the pioneering work of Chew and her colleagues (11, 12), primary cul-
tures of parietal cells have also been of special value in localizing functionally
relevant proteins and defining events in the secretory cascade. Many intracellular
signaling pathways have now been identified with their contributing roles in pari-
etal cell activation, including protein kinase A (PKA), protein kinase C (PKC),
Ca2+-calmodulin (CaM) kinase II, phosphatidylinositol 3-kinase (PI3 kinase), and
several other downstream kinases. Histaminergic stimulation is by far the most
potent activation pathway observed for the stimulation of gastric acid secretion
in vitro. Although cholinergic and gastrinergic stimulation can be seen in vitro,
the magnitude of stimulation for many species is much reduced compared with
stimulation in vivo, most likely because parietal cells are in close contact with
histamine-releasing enterochromaffin-like (ECL) cells in vivo (13). Isolated ca-
nine parietal cells appear to be an exception, tending to be more responsive to
cholinergic stimuli than to histamine (1). In this review we first focus on signaling
events underlying cAMP-mediated stimulation and the PKA stimulation pathway,
and then move to a discussion of cholinergic pathways and an overview of other
kinase pathways.

cAMP-Mediated PKA Activation and Protein Phosphorylation


Evidence implicating cAMP as an intracellular messenger mediating HCl secre-
tion was presented by Roth & Ivy more than 50 years ago (14). Using isolated
gastric glands, Chew et al. (15) directly showed that histamine elevates intracel-
lular levels of cAMP leading to activation of PKA (16), and specifically type I
cAMP-dependent protein kinase (17). Activation of PKA initiates a cascade of
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 107

phosphorylation events through the activation of other downstream effectors. Col-


lectively, these events trigger membrane and cytoskeletal rearrangements within
the parietal cell, as well as increase electrical conductance across the gastric epithe-
lium (1). In the 1980s, several groups reported on stimulus-associated phosphory-
lation of parietal cell proteins, most of which were simply identified by molecular
size (1821). By tracing the phosphorylation of proteins that occurred concomi-
tant with stimulation of gastric glands by histamine, Urushidani and his colleagues
(19, 22) identified an 80-kDa peripheral membrane protein, later characterized as
the cytoskeletal-associated protein ezrin (23, 24), and a 120-kDa protein, now
called parchorin and recently implicated in Cl and water transport (25, 26). In
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

addition, several stimulation-related phosphoproteins earlier identified by Chew


and her colleagues on the basis of molecular size (18, 27) are now recognized as
40-kDa lasp-1, 66-kDa coroninse (28), and CSPP-28, the latter being a calcium-
sensitive phosphoprotein of 28-kDa (29). Possible functional roles for each of
these phosphoproteins are discussed below.
The early studies of signaling processes related to acid secretion were carried
by HINARI on 05/16/07. For personal use only.

out on intact gastric glands. Because the plasma membrane provides a barrier
to separate intracellular environment from extracellular bath, the development of
permeable parietal cell models was important for a more direct manipulation of
cytosol. Several permeabilized models, in which electroporation (30) and digitonin
(3033) were used to render gastric glandular cells permeable, have provided useful
information. However, none of these earlier permeabilized preparations could be
transformed from the resting to secreting state by the addition of secretagogues or
cAMP. This problem was eliminated by the use of -toxin, isolated from Staphylo-
coccus aureus, as a permeabilizing agent. The -toxin permeabilized gastric gland
model can be triggered by cAMP to effect the resting-to-secreting transition that
is correlated with the phosphorylation, from 32P-ATP, of a dozen phosphoproteins
(34), several of which are similar to those mentioned above from the intact gland
studies.
The narrow pore size generated by -toxin (3 nm in diameter) allows passage
of only small molecules such as nucleotides. Thus the -toxin model has been
useful in studies of metabolism and protein phosphorylation associated with stim-
ulation (34, 35), but access to large macromolecules is required for biochemical
reconstitution of parietal cell activation. To this end, several permeabilized sys-
tems have been developed to allow the introduction of large peptides and proteins
while retaining the resting-to-secreting transition in response to the addition of
cAMP. One system employing the detergent -escin tested a variety of inhibitory
peptides that block the activities of several protein kinases, including PKA, PKC,
myosin light chain kinase (MLCK), and CaM kinase (36). These peptide inhibi-
tion experiments confirmed the roles of PKA and MLCK, but not CaM kinase and
PKC, in parietal cell secretion. In addition, the inclusion of peptides that inhibit
Arf (adenosine ribosylation factor) into -escin-permeabilized glands was shown
to attenuate the cAMP-stimulated parietal cell activation, suggesting a functional
role for Arf protein in parietal cell secretion.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

108 YAO FORTE

An alternative model system has used the Streptococcal toxin streptolysin O


(SLO) to permeabilize gastric glands. With this model, Ammar et al. (37) were
able to show the incorporation of fluorescent-labeled actin into the pool of endoge-
nous F-actin in SLO-permeabilized glands. They also tested the action of several
syntaxin isoforms on the cAMP-activated secretory response (as discussed below
in the section on SNARE proteins).
Yet another approach for permeabilized models is to use a depleted system,
such as the digitonin model, and probe for cytoplasmic constituents that restore
functional activity. Searching for players underlying the cAMP-mediated parietal
cell activation, Akagi et al. (38) carried out a reconstitution experiment in which
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

they added brain cytosol and purified fractions into digitonin-permeabilized gas-
tric glands. They demonstrated a stimulatory activity by brain cytosol as judged by
aminopyrine uptake. The stimulatory activity was further characterized as phos-
phatidylinositol transfer protein (PITP). Although PITP is a stimulatory factor for
acid secretion, it is not associated with the cAMP-dependent pathway. This re-
constitution model promises to be of importance for the further identification and
by HINARI on 05/16/07. For personal use only.

function of critical components necessary for parietal cell activation, including


other PKA effectors; it will also address questions of how known proteins such as
ezrin and lasp integrate the cAMP signal to membrane cytoskeleton remodeling.
Lasp-1 was initially identified in the parietal cell as a 40-kDa phosphoprotein
related to cAMP-mediated activation of acid secretion (18). Lasp-1 has been iden-
tified as a signaling molecule that is phosphorylated upon elevation of [cAMP]i in
pancreas and intestine, as well as gastric mucosa, and is selectively expressed in
ion-transporting cells within epithelial tissues, such as cortical regions of pancreatic
and salivary duct cells and in certain F-actin-rich cells in the distal tubule/collecting
duct (34, 35). Lasp-1 has been characterized as a new LIM protein subfamily mem-
ber containing an N-terminal LIM domain, a C-terminal SH3 domain, and two
internal nebulin repeats, with speculation that lasp-1 may function as an adaptor
molecule to relay the upstream signaling to downstream effectors (39). Recent
biochemical evidence revealed that lasp-1 binds to nonmuscle filamentous (F)
actin, but not monomeric (G) actin, in a phosphorylation-dependent manner (40).
The apparent Kd of bacterially expressed his-tagged lasp-1 binding to F-actin was
2 M, with a saturation stoichiometry of 1:7. Phosphorylation of recombinant
lasp-1 with PKA increased the Kd for lasp-1 binding to F-actin and decreased the
Bmax. The major PKA-dependent phosphorylation sites were localized in rabbit
lasp-1 to S99 and S146 (40).
Immunostaining of gastric glands stimulated by the cAMP-dependent pathway
showed that lasp-1 was redistributed from the basolateral membrane to the api-
cal canalicular membrane of parietal cells upon stimulation (41). Alternatively,
exposure of gastric mucosal fibroblasts to the protein kinase C activator, PMA, in-
duced the formation of lamellipodial extensions and nascent focal complexes that
contained both lasp-1 and F-actin (40). The selective phosphorylation-dependent
regulation of lasp-1 in F-actin-rich epithelial cells and the recruitment of lasp-1 to
cellular regions associated with dynamic actin turnover suggest that this protein
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 109

may play an integral and specific role in the regulation of cytoskeletal/membrane-


based cellular activities.
Parchorin was first identified by SDS-PAGE as a 120-kDa phosphoprotein as-
sociated with histamine/cAMP stimulation of acid secretion (19); the cDNA se-
quence of parchorin has recently been cloned, having an actual molecular mass of
65 kDa (26). Its anomalous migration on gels is due to an unusually high content
of acidic amino acids. Parchorin is a novel protein that has significant homology
to the family of chloride intracellular channels (CLIC) and has been localized to
a variety of epithelial tissues that are active in the transport of salt and water,
especially choroid plexus, salivary glands, and lacrimal glands, in addition to the
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

parietal cell (25, 26). Endogenous parietal cell parchorin is present in the cytosol
and relocated to the apical plasma membrane upon histamine stimulation, very
reminiscent of the translocation of H,K-ATPase in response to stimulation. When
parchorin, linked to green fluorescent protein (GFP), was exogenously expressed
in kidney cells, GFP-parchorin also appeared in the cytosol and was relocated to
the plasma membrane when Clefflux from the cells was triggered. Interestingly,
by HINARI on 05/16/07. For personal use only.

parchorin was co-purified with a new type of kinase activity (42), suggesting that
there may be some functional link between that kinase and the phosphorylated
form of parchorin.

Cholinergic Pathways of Activation


Activation of parietal cells by cholinergic agents acting on muscarinic M3 receptors
clearly involves an elevation of intracellular Ca2+ ([Ca2+]i). Studies on isolated
gastric glands with Ca2+-sensitive fluorescent probes revealed a rapid spike in
parietal cell [Ca2+]i, up to 0.150.5 M within 46 s after addition of the M3
agonist carbachol, followed by a sustained lower-level elevation of [Ca2+]i, that
persisted during the resulting stimulation of acid secretion by the glands (43, 44).
The secretagogue-dependent changes in [Ca2+]i occur from an intracellular store
of bound Ca2+ and are independent of extracellular Ca2+, unless the stores are first
depleted (45, 46). Buffering of the rise in [Ca2+]i by the chelating agent BAPTA
prevents the acid secretory response to carbachol but permits subsequent stimula-
tion by histamine or forskolin (47). Intracellular Ca2+ stores are relatively easily
depleted by cholinergic stimulation in a Ca2+-free medium, although the stores can
be replenished from extracellular sources through a La3+-sensitive pathway (45).
The carbachol-induced rise in [Ca2+]i has been correlated with a rise in parietal
cell inositol trisphosphate (IP3) levels, thus implicating IP3 as the messenger from
the M3 receptor to the internal Ca2+ stores (43). The role of PKC in cholinergic
activation has been more controversial, particularly through the use of PKC activa-
tors such as phorbol esters (e.g., TPA), which have been shown to inhibit or activate
both carbachol- and histamine-stimulated acid secretion (4851). Chew et al. (52)
measured the PKC expression in parietal cells, as well as the response of gastric
glands to a variety of PKC inhibitors. Their data showed several isozymes of PKC
present with some, particularly PKC-, being localized by immunocytochemistry
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

110 YAO FORTE

to a parietal cell compartment similar to filamentous actin (52). Inhibitors of


PKC activity, such as Ro 31-8220, were found to potentiate both carbachol- and
histamine-mediated acid secretion, also causing a dose-dependent decrease in the
phosphorylation of the cytoskeletal-related proteins ezrin and pp66, later charac-
terized as coroninse (28, 52). These data suggest a negative modulatory role for
PKC in acid secretion and were confirmed in principle by an independent study
(53), although there was some disagreement regarding responsiveness of specific
PKC isozymes.
A role for CaM kinase in parietal cell activation stimulated by the cholinergic
pathway was suggested by Tsunoda et al. (54) who found that KN-62, a pharma-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

cological inhibitor of CaM kinase II, inhibited carbachol-mediated acid secretion,


but did not alter the rise in cytosolic Ca2+ caused by carbachol. KN-62 was without
effect on histamine- or forskolin-mediated responses (55). The presence of CaM
kinase II in gastric cells was verified by enzymatic assay (54).
The calcium-sensitive phosphoprotein of 28 kDa known as CSPP28 was orig-
inally discovered based on its phosphorylation associated with cholinergic stim-
by HINARI on 05/16/07. For personal use only.

ulation (27). CSPP28 is rapidly phosphorylated in intact parietal cells in re-


sponse to both the cholinergic agonist, carbachol, and the calcium ionophore,
ionomycin (29). Subsequent studies showed that recombinant CSPP28 was phos-
phorylated by both crude parietal cell homogenate and purified CaM kinase II in
a calcium/calmodulin-dependent manner (29). Whereas CSPP28 is closely tied to
a calcium-mediated phosphorylation, it would be of great interest to ascertain the
nature of CaM kinase-mediated phosphorylation on CSPP28 and whether such
modification regulates the function of CSPP28 in the activation process.
Carbachol has been shown to induce IB kinase activity in canine parietal
cells via Ca2+- and PKC-dependent pathways (56). IB kinase appears to be a
key element in the signaling cascade that activates NF-B (57, 58), which is an
important transcription factor and regulator of numerous genes modulating the
inflammatory response. Carbachol is also a potent inducer of several families of
protein kinases known as mitogen-activated protein kinases (MAPKs) or extra-
cellular signal-regulated protein kinases (ERKs) and the c-Jun N-terminal protein
kinases (JNKs) in the canine parietal cell system (5961). Generally, these kinases
are known to be fundamental signaling elements in cellular functions of growth,
differentiation, and secretion (6265). However, it is uncertain as to what specific
role these kinases play in the acid secretory activation pathway because presumed
specific inhibitors of the various kinases reportedly produce both inhibitory and
enhancing effects. For example, a pronounced up-regulation of p38 kinase, another
member of the MAPK family of protein kinases, occurred in response to carba-
chol and could be blocked by the p38 kinase-specific inhibitor SB-203580 (66).
Interestingly, carbachol-induced aminopyrine uptake by intact canine parietal cells
was potentiated several-fold by SB-203580, suggesting that active p38 kinase was
inhibitory to acid secretion (66). Collectively these data demonstrate the extensive
interrelationship of signaling elements that are activated via M3-muscarinic stim-
ulation of gastric parietal cells. It remains to be established which elements are
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 111

directly involved in the activation of parietal cell secretion, which are modulatory
or inhibitory to the activation, and which are true downstream effector proteins
rather than secondary and tertiary reporters of changes in metabolic load.

Modulation by EGF and TGF-


To reveal the mechanisms underlying the actions of EGF and TGF- on acid se-
cretion, Chew et al. (67) studied the effects of these agents on isolated parietal cells
and primary cultures. They categorized two apparently opposed responses: (a) an
acute inhibition of histamine- and carbachol-mediated acid secretion and (b) an en-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

hancement of acid secretory function with chronic exposure to the growth factors.
Because of the observed differential effects of the acute and chronic responses to
tyrosine kinase inhibitors, they proposed that EGF and TGF- modulate parietal
cell function by multiple signaling pathways. They reasoned that a soluble tyrosine
kinase was involved in mediating the chronic effects of EGF, whereas the acute
potentiation of histamine-stimulated secretion by certain tyrosine kinase inhibitors
by HINARI on 05/16/07. For personal use only.

was probably not mediated by receptor-associated tyrosine kinases. In a further


study of these growth factors on parietal cell activation, these authors showed that
EGF biphasically activated extracellular signal-regulated protein kinases, known
as ERK-1 and ERK-2, with a peak response occurring at approximately 5 min,
followed by a sustained activation for at least 2 h (68). In contrast to EGF, the phor-
bol ester PKC activator TPA induced a sustained activation of ERK-1 and ERK-2
for at least 2 h. Carbachol also activated ERK-1 and ERK-2, but the response was
weaker and monophasic. However, the ERK-signaling cascade was not modulated
by elevated intracellular free Ca2+ or cAMP.
Takeuchi et al. (59, 60) confirmed the biphasic effects of EGF on isolated canine
parietal cells and tested the role of ERKs as possible signal modulators for gastric
secretion. In their studies, carbachol caused a pronounced increase in the kinase
activity of ERK2 in extracts prepared from canine parietal cells (gastrin and EGF
had modest effects, and histamine had no effect), but unlike Chews group, these
authors found that the stimulatory response to carbachol appeared to be dependent
on elevated [Ca2+]i and that a specific inhibitor of ERK, PD-98059, abolished the
stimulatory response. Addition of the ERK inhibitor to intact parietal cells led to a
modest elevation, at best, of aminopyrine uptakes stimulated by carbachol or other
secretagogues, thus the ERK pathway has a minimal role in the direct activation
of gastric secretion. This conclusion is supported by later work suggesting that
the increased ERK activity stimulated by EGF appears coupled to H,K-ATPase
-subunit expression (69) which may be the reason for the slightly enhanced
secretory activity associated with ERK activation and may even form the basis for
the long-term stimulatory effects of EGF. The serine-threonine protein kinase Akt
appears to be another element in the EGF activation cascade leading to increased
H,K-ATPase expression in canine parietal cells (70).
Tsunoda et al. (71) employed chemical inhibitors of tyrosine kinases, genis-
tein and erbstatin, to test for their ability to release the parietal cell from the
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

112 YAO FORTE

inhibitory action of EGF/TGF- on acid secretion. Whereas these two inhibitors


totally abolished the TGF--mediated inhibition, genistein but not erbstatin, also
potentiated histamine- and forskolin-triggered acid secretion. Because genistein
is a relatively selective inhibitor for cytosolic tyrosine kinases such as c-src, it is
likely that some cytosolic tyrosine kinase negatively regulates parietal cell secre-
tion. It has been reported that activation of tyrosine kinase such as c-src stimulates
the dynamin-dependent endocytosis of renal outer medullary potassium channels
(72). It is possible that EGF/TGF- exert their inhibition on histamine-stimulated
acid secretion by promoting the retrieval of H,K-ATPase from the apical plasma
membrane. Although a tyrosine residue in the cytoplasmic tail of the -subunit
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

of H,K-ATPase has been implicated in proton pump recycling (73), it would be


of great interest to test whether tyrosine kinases such as c-src accelerate the re-
cycling of H,K-ATPase and to ascertain the molecular mechanism underlying the
regulation of H,K-ATPase recycling by tyrosine phosphorylation.
by HINARI on 05/16/07. For personal use only.

Modulation by Duodenal and Neural Peptides


Physiological modulation of gastric secretion by a variety of peptides, including
the duodenal hormone cholecystokinin (CCK) and the neuropeptide vasoactive
intestinal peptide (VIP), has been well established. Of prime importance for the
action of CCK and VIP is the paracrine release of somatostatin by D cells in the
antrum and fundus (74, 75). Some of the effects of somatostatin are peripheral to
parietal cells, e.g., inhibit release of gastrin from antral G cells (75, 76) and inhibit
release of histamine from fundic ECL cells (77, 78), but somatostatin also has
direct inhibitory effects on parietal cells (79, 80). Somatostatin receptors on the
parietal cell have been characterized as sstR2-type receptors (84), which operate
via an inhibitory guanine nucleotide-binding protein (Gi) to attenuate the activity
of adenylate cyclase. Inhibition of Gi by pertussis toxin reversed the ability of
somatostatin to inhibit acid secretion and restored the adenylate cyclase-mediated
production of cAMP by isolated canine parietal cells stimulated with histamine and
forskolin, but did not alter the stimulatory effects of dbcAMP or Ca2+ pathway
(79, 82). In addition somatostatin may have long-term effects on parietal cells
because it has been shown to inhibit expression of early response genes, such as
c-fos, via a pertussis toxin-sensitive inhibitory pathway in isolated canine parietal
cells (83, 84).

TRAFFICKING OF H,K-ATPASE ASSOCIATED WITH


ACID SECRETION
In the resting parietal cell the proton pump resides in cytoplasmic tubulovesicles in
an inactive form, presumably because of low permeability of these membranes to
K+ (85). Upon stimulation by secretagogues, H,K-ATPase translocates and inserts
into the apical plasma membrane which has, or acquires, K+ and Cl conductance
21 Jan 2003 11:26 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 113

pathways that lead to active proton pumping. H,K-ATPase is a P-type ATPase


composed of a catalytic subunit (-subunit) and an accessory subunit (-subunit).
The H,K-ATPase catalyzes the electroneutral exchange of intracellular protons
for extracellular potassium ions, thus generating the enormous proton gradients
associated with gastric HCl secretion (1, 86). Early EM studies support a model
in which H,K-ATPase-containing vesicles fuse with the apical plasma membrane
upon stimulation, thus recruiting the pump enzyme into the plasma membrane
(87). In the following section we discuss those protein elements that have been
identified in parietal cells and have been shown (or implicated) to have a role in
directing the trafficking and recycling of H,K-ATPase-rich membranes underlying
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

HCl secretion. Figure 1 provides a cartoon summary of the membrane recycling


scheme, indicating the location of putative participating proteins.

SNARE Proteins
by HINARI on 05/16/07. For personal use only.

It is widely accepted that the fundamental components of the machinery required


for membrane fusion are highly conserved in both constitutive and regulated mem-
brane trafficking pathways (88, 89). These components include N-ethylmaleimide-
sensitive factor (NSF), the soluble NSF attachment proteins (-, -, and -SNAPs),
and SNAP receptors (SNAREs). Membrane-associated SNAREs were first identi-
fied in detergent extracts of brain membranes (90). In these extracts, the SNAREs
formed a tightly bound complex consisting of SNAP-25, syntaxin, and a protein
known as vesicle-associated membrane protein (VAMP). The specific cleavage
of each of these proteins by specific neurotoxins causes a blockade in neuro-
transmitter release, indicating that SNAP-25, syntaxin, and VAMP are essential
for exocytosis in synaptic termini (91). Peng et al. (92) showed that a syntaxin
isoform known as syntaxin 3 and the VAMP-2 isoform (also known as synapto-
brevin) are specifically associated with H,K-ATPase-containing tubulovesicles in
gastric parietal cells, suggesting their possible involvement in secretory activation.
Calhoun & Goldenring (93) and Calhoun et al. (94) confirmed the localization of
syntaxin 3, VAMP-2, and another secretory carrier membrane protein known as
SCAMP to the tubulovesicle fraction, and more importantly demonstrated their re-
location to the apical plasma membrane after stimulation. Because these data were
consistent with a role for syntaxin 3 in apical trafficking of H,K-ATPase, Ammar
et al. (37) carried out experiments in which recombinant syntaxin 3 was added
into SLO-permeabilized gastric glands as a potential competitor for endogenous
SNARE complex formation. Significantly, exogenous syntaxin 3, but not the syn-
taxin 5 isoform, blocked cAMP/ATP-mediated HCl secretion in a dose-dependent
manner. The authors reasoned that the soluble exogenous syntaxin 3 reacted with
endogenous cognate SNAREs to prevent the recruitment of secretory pumps, thus
providing direct evidence for the involvement of the syntaxin 3 isoform in parietal
cell activation.
Primary cultures of parietal cells have been particularly useful in defining the
trafficking and recycling of membrane components associated with stimulation of
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

114 YAO FORTE

acid secretion (95). Karvar et al. (96) used an adenovirus-based GFP reporter to
show that GFP-VAMP-2 is co-localized with H,K-ATPase to cytoplasmic mem-
branes of resting parietal cell cultures and that GFP-VAMP-2 is translocated along
with the pump enzyme to the apical plasma membrane after stimulation by his-
tamine. To test the involvement of VAMP-2 in parietal cell activation, they treated
SLO-permeabilized glands with tetanus toxin, which is a highly specific pro-
tease for VAMP proteins. They demonstrated a correlation between the cleavage
of VAMP-2 and the inhibition of glandular acid secretion, confirming an earlier
suggestion that the proteolytic cleavage of VAMP-2 inhibited acid secretion in
isolated parietal cells that were permeabilized by SLO and stimulated with cAMP
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

(97). Further support for the participation of SNARE proteins in the recruitment of
H,K-ATPase is offered by recent studies of Karvar et al., who doubly infected pari-
etal cell cultures with genes constructed with different fluorescent proteins linked
to VAMP-2 and SNAP-25 (98). In resting cells, the two proteins were localized
to different membrane compartments: VAMP-2 to H,K-ATPase-rich cytoplasmic
vesicles, and SNAP-25 to apical membrane vacuoles. Upon stimulation with his-
by HINARI on 05/16/07. For personal use only.

tamine, the two signals became coincident in the expanded plasma membrane
consistent with SNARE complex formation associated with membrane recruit-
ment. Furthermore, introduction of a SNAP-25 construct lacking the functional C
terminus inhibited acid secretion by the isolated cells.

Rab Proteins
In addition to SNARE proteins, small GTPase proteins have long been implicated
in vesicular membrane trafficking (99). Studies in several systems, primarily in
neuronal synapses, have demonstrated that Rab3 family members redistribute from
a membrane-bound location to the soluble cytosolic fraction upon fusion of secre-
tory granules with target plasma membranes. Rab proteins are then recycled back
onto mature secretory vesicles after reinternalization of the membrane. Although
this cycle is well established for Rab3, far less is known about the functional activ-
ity of other Rab proteins during vesicle fusion and recycling. In the gastric parietal
cell, there are several Rab proteins, including Rab11 and Rab25 (93, 100, 101). In
nonsecreting parietal cells, the Rab11a isoform is associated with H,K-ATPase-
containing tubulovesicles that fuse with the apical plasma membrane in response
to secretory agonists such as histamine (94).
Using matrix-assisted laser desorption mass spectrometry, Duman et al. (102)
confirmed that Rab11 is associated with H,K-ATPase-enriched gastric micro-
somes, and their data provided a stoichiometric estimate of one Rab11 per six
copies of H,K-ATPase. Furthermore, Rab11 exists in at least three forms on rabbit
gastric microsomes: The two most prominent resemble Rab11a, whereas the third
resembles Rab11b. Using an adenoviral expression system, Duman et al. expressed
the dominant-negative mutant Rab11a N124I, in primary cultures of rabbit pari-
etal cells. Rab11a N124I is a mutant of Rab11a in which aspargine (N) at position
124 has been replaced with isoleucine (I). The mutant was well expressed with a
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 115

distribution similar to that of H,K-ATPase in resting parietal cells; however, mutant


Rab11a arrested the stimulus-dependent morphological transition of the parietal
cells and acid secretion measured by aminopyrine uptake. Specificity was verified
by addition of tetracycline, which blocked mutant Rab expression and restored the
morphological transition and acid secretory function. These studies demonstrate
that Rab11a is a prominent GTPase associated with gastric tubulovesicles and
suggest that it plays a direct role in regulating parietal cell activation.
In a search for downstream effectors, a number of Rab11-interacting proteins
have been identified as co-enriched with Rab11a and H,K-ATPase on parietal cell
tubulovesicle membranes: rab11-family interacting protein 1 (rab11-FIP1), rab11-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

family interacting protein 2 (rab11-FIP2), and pp75/rip11 (103). The binding of


rab11-FIP1 and rab11-FIP2 to Rab11a was dependent upon a conserved C-terminal
amphipathic -helix. Moreover, these Rab11-interacting proteins were found to
translocate with Rab11a and the H,K-ATPase upon stimulation of parietal cells with
histamine. These results suggest that the function of Rab11a in plasma membrane
recycling is dependent upon a number of protein effectors.
by HINARI on 05/16/07. For personal use only.

Clathrin Coat and Other Adaptor Proteins


An important, though somewhat ignored, aspect of regulating gastric secretion
has to do with the uptake and internalization of the extensive apical membrane
when the stimulus is withdrawn and the parietal cell returns to the resting state.
Given the similarities of regulated H,K-ATPase trafficking with recycling of cargo
through the apical recycling endosomes of many epithelial cells, it seems reason-
able to speculate that clathrin and its adaptor proteins regulate some part of an
apical recycling pathway as in other epithelial cells. Using immunological probes,
Okamoto et al. (104) identified -adaptin and clathrin heavy chain on tubulovesi-
cles isolated from gastric parietal cells. Further biochemical characterization of
the tubulovesicular clathrin adaptor complex suggested a subunit composition in-
cluding -, - and -adaptins, as well as other subunits with molecular masses
of 50 and 19 kDa, all of which could polymerize in vitro into basket-like struc-
tures of 120 nm diameter (105). For parietal cells in the nonsecreting state,
immuno-electron microscopy revealed that clathrin is relatively abundant at or
near the apical canalicular membrane specifically in association with coated pits
and coated vesicles, as well as accumulated at the ends of tubulovesicular pro-
files where the membranes are acutely curved. These results indicate that, even in
resting cells, active membrane trafficking between the apical membrane and the
tubulovesicular compartment is an ongoing process. They are also consistent with
an hypothesis that the secretory cycle is regulated with respect to the relative rates
of exocytic (stimulation) and endocytic (recycling) processes, rather than a binary
on or off system. It must be pointed out that, although clathrin and its adaptors are
well represented and appear actively involved in some aspect of apical membrane
recycling in the parietal cell, there is only presumptive evidence that H,K-ATPase
is the cargo.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

116 YAO FORTE

Dynamin
Another component of clathrin-dependent trafficking pathways is the large
GTPase dynamin, which is essential for receptor-mediated endocytosis and is
clearly known to interact with clathrin and coat-forming proteins, but its precise
function in vesicle formation remains controversial (106, 107). Given the distri-
bution of clathrin and clathrin adaptors in the parietal cell, it is not surprising that
a dynamin-like molecule should be associated with the apical membrane. In fact,
dynamin II has been immunolocalized to the apical membrane of both resting
and stimulated parietal cells (94, 105). Interestingly, among the various cell types
in gastric glands, dynamin appears to be expressed predominantly, if not exclu-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

sively, in the parietal cell (105). Although it has been proposed that dynamin acts
as a pinchase for constriction of endocytic membrane separation from plasma
membrane, this remains to be established (107).
Using a GST-dynamin II fusion protein as an affinity matrix, Okamoto et al.
identified two proteins that bind to dynamin II, the nonreceptor tyrosine kinase
by HINARI on 05/16/07. For personal use only.

c-src and lasp-1 (108). Moreover, the binding of c-src and lasp-1 was specific for the
proline-rich domain of dynamin II. Their study also showed the presence of c-src
on tubulovesicular membranes, with an active tyrosine kinase that phosphorylated
tubulovesicular proteins in vitro, one of which may be the 100-kDa H,K-ATPase.
A potential trafficking role for c-src on tubulovesicular membranes is of inter-
est because c-src has been found on endosomal membranes (109), as a regulator
of other apical membrane trafficking pathways (110, 111), and shown to regu-
late membrane transporters directly by phosphorylation (112). The interaction of
lasp-1 with dynamin II is also of interest, suggesting the possibility of some regula-
tory role for lasp-1 in coordinating the actin cytoskeleton and vesicular trafficking
machinery via dynamin (108). As with c-src, the functional consequences of lasp-1
dynamin II interaction remain to be characterized.
Although these interactive data provide interesting possibilities for connection
between signaling molecules such as c-src and the downstream effectors, such
as dynamin, lasp-1 and even H,K-ATPase, the role of the in vitro protein-protein
interactions in parietal cell physiology and their regulation during cell activation
remain to be characterized.

Myosin Vb
Given the importance of Rab11a in tubulovesicle trafficking, Lapierre et al. (113)
set up a yeast two-hybrid screen for proteins interacting with active Rab11a.
Among three Rab11-interacting proteins discovered, one is myosin Vb. These
authors further characterized myosin Vb in polarized MDCK cells. They found
that myosin Vb is associated with apical endosomes. Furthermore, this associa-
tion depends on the integrity of microtubules, providing the interesting suggestion
of a role for myosin Vb in plasma membrane recycling in connection with the
microtubule cytoskeleton. Expression of the C-terminal tail of myosin Vb dis-
persed the distribution profile of transferrin receptor and retarded recycling of the
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 117

receptor back to the plasma membrane. By analogy with a proposed function of


myosin Va in melanosome trafficking (114), it is also possible that the mutant
disrupted the association of the myosin Vb-endosomal complex with the actin-
based cytoskeleton, which resulted in an inhibition of apical plasma membrane
recycling.

Structure and Function of H,K-ATPase


H,K-ATPase shares several structural homologies with another P-type ATPase,
Na,K-ATPase (86, 115). They are both composed of an -subunit, predicted to
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

span the membrane 10 times, and a glycosylated -subunit of type II integral


membrane protein. Early in the biosynthetic pathway, the -subunits assemble with
-subunits in the endoplasmic reticulum. Assembly of the holoenzyme appears to
involve interactions between cytoplasmic, extracytoplasmic and transmembrane
domains of both subunits (116, 117) and requires insertion of the / complex
by HINARI on 05/16/07. For personal use only.

into the plasma membrane. Although cross-assembly of H,K-ATPase and Na,K-


ATPase subunits can occur under contrived in vitro conditions, the -subunit of
each ATPase preferentially assembles with its respective -subunit (118).
Whereas the amino acid sequence between the H,K-ATPase and Na,K-ATPase is
highly conserved, these enzymes differ in several important attributes, including the
cations they respectively transport and their targeted locations. The Na,K-ATPase
is concentrated at the basolateral membrane of most epithelial cells, whereas, the
gastric H,K-ATPase is ultimately targeted to the apical membrane of secreting
parietal cells. Also, the H,K-ATPase represents the major cargo protein for the
regulated recycling associated with acid secretion.
The recruitment of vesicular coat proteins has been shown to depend upon both
the presence of membrane protein cargo and the lipid composition of the target
membranes (119). Thus cargo can regulate the formation of vesicular carriers.
Complete deletion of either the -subunit (120) or the -subunit (121) in knockout
mice was shown to have predictably profound effects on parietal cell structure and
function. In both cases, the mice were achlorhydric. Although parietal cells were
histologically identifiable, both knockout mice exhibited a dramatic diminution
of tubulovesicular membranes. The residual amount of -subunit expressed in the
-subunit knockout mice was not sufficient to produce a tubulovesicular network
(120), suggesting that the assembled holoenzyme is required as cargo for the
biogenesis of the tubulovesicular compartment.
Expression studies in heterologous cell lines have shown that the -subunit
of the gastric H,K-ATPase encodes sorting and targeting information responsible
for the apical distribution of the pumps. By analyzing the sorting behavior of a
number of chimeric pumps composed of complementary portions of -subunits
from the H,K-ATPase and Na,K-ATPase, Dunbar et al. (122) identified a portion
of the gastric H,K-ATPase, which is sufficient to redirect the normally basolateral
Na,K-ATPase to the apical surface in transfected LLC-PK cells. This motif resides
within the fourth of the ten predicted transmembrane domains of the -subunit.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

118 YAO FORTE

There are seven N-glycosylation sites on the -subunit of the H,K-ATPase


(123). To examine the role of the carbohydrate chains on holoenzyme activity and
stability, Asano et al. (124) carried out site-directed mutagenesis on the -subunit
in which one, several, or all of the asparagine residues in the N-glycosylation sites
were replaced by glutamine. Removing any one of seven carbohydrate chains from
the -subunit did not have a significant effect on the activity of H,K-ATPase or
its delivery to the apical surface. However, removal of all the carbohydrate chains
prevented assembly of the holoenzyme and all associated functional activity. In
contrast, enzyme activity and /-subunit assembly were retained when three
carbohydrate chains were removed, but surface delivery of the -subunit and its
associated -subunit was arrested, indicating that the surface delivery mechanism
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

is more dependent on the carbohydrate chains than is the expression of H,K-ATPase


activity and holoenzyme assembly.
In addition to stabilizing its complementary -subunit, the -subunit of
H,K-ATPase may contain a tyrosine-based endocytotic motif that is essential for
recycling of the pump after withdrawal of stimulation. Courtois-Coutry et al. (73)
by HINARI on 05/16/07. For personal use only.

made transgenic mice that expressed a mutant H,K-ATPase -subunit, in which a


critical tyrosine residue in the cytoplasmic tail was mutated to alanine. These mice
developed a pathology resembling that in idiopathic hypersecretion of acid into
the stomach. Immuno-electron microscopy data suggest that the -subunit, and
presumably the -subunit, in these transgenic mice are constitutively expressed at
the apical membrane and fail to re-internalize. The authors concluded that that the
mutated -subunit, and therefore the pump enzyme, could not interact with the
endocytotic machinery to allow re-sequestration of the stimulated apical surface
in reversion to the resting state.
Biochemical studies suggest that the cytoskeletal proteins ankyrin and spectrin
interact with the H,K-ATPase in the microsomal fraction of resting parietal cells
and appear to relocate with H,K-ATPase to the apical plasma membrane of the
secreting parietal cells (125). Whereas these authors speculated that ankyrin and
spectrin form a functional complex to stabilize H,K-ATPase at the apical membrane
upon the stimulation, it would be of great interest to ascertain whether ankyrin and
spectrin directly bind to H,K-ATPase and how such interactions are regulated.

ACTIN-BASED CYTOSKELETON IS ESSENTIAL


FOR PARIETAL CELL ACTIVATION
Actin
Interactions between the plasma membrane and the cytoskeleton play a central
role in a variety of physiological processes including mobilization of membrane
receptors, endocytosis, exocytosis (e.g., hormone secretion and transmitter re-
lease), cell polarity, and morphology (126, 127). An essential role for the cortical
actin cytoskeleton in parietal cell secretion has been implied from morphological
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 119

analyses (13) and from studies with microfilament disrupters such as cytocha-
lasin (128, 129). Highly organized microfilaments are a characteristic feature of
microvilli within the canaliculus of the gastric parietal cell. However, the radial
arrangement of the actin filaments in proximity to the microvillar membrane in
parietal cells (3) is distinctly different from the central core localization of actin
filaments of intestinal microvilli (130).
The actin gene family encodes a number of structurally related, but functionally
distinct, protein isoforms that modulate contraction in muscle tissues and control
shape and motility of nonmuscle cells (131). In mammals, there are at least six
different actin isoforms, each encoded by a separate gene, and they differ by
<10% of the amino acid sequence, primarily in the N-terminal region (132).
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

The development of cytoplasmic - and -actin isoform-specific antibodies has


provided powerful tools for localizing actin isoforms in situ (133).
Using actin isoform-specific antibodies and isolated gastric glands as an ep-
ithelial model, Yao et al. (134) demonstrated that actin isoforms, in particular
the cytoplasmic - and -actin nonmuscle isoforms, are differentially polarized
by HINARI on 05/16/07. For personal use only.

in epithelial cells. They found that -actin was predominantly localized to the
apical plasma membrane of all glandular cells, including the tortuous microvilli-
enriched canalicular surface of parietal cells, as well as the entire gland lumen,
whereas -actin was predominantly localized to the basolateral membrane.
The canaliculi of resting parietal cells are replete with short microvilli that
include radially organized actin microfilament bundles projecting along the mi-
crovillus and extending one or more micrometers into the cytoplasm (3). The
radical membrane transformations of cell activation result in elongation of mi-
crovilli, which suggests a commensurate shift in the ratio of monomeric G-actin
to filamentous F-actin. However, measurements on the state of actin in gastric
glands revealed the following: (a) Parietal cell actin is primarily organized in the
F-actin form (90%); (b) the microfilament disrupter cytochalasin D destabilizes
the F-actin and inhibits acid secretion; and (c) phalloidin, which stabilizes F-actin
filaments, does not inhibit acid secretion (135). Surprisingly, the authors could find
no significant change in the ratio of F- to G-actin when the cells were transformed
from rest to maximal secretion: F-actin remained about 90% of the total. These
data are consistent with an hypothesis that microfilamentous actin is necessary for
membrane recruitment underlying parietal cell secretion; however, they suggest
that rapid exchange between G- and F-actin is not essential for the secretory pro-
cess and that the parietal cell maintains actin in a highly polymerized state. This
conclusion was underscored by the work of Ammar et al. (136) who tested the actin
monomer-sequestering agent latrunculin B (Lat B) on parietal cell structure and
function. Lat B inhibited acid secretion and increased the extractable monomeric
actin but only at relatively high doses (1070 M). Because the authors observed
high sensitivity of other parietal cell functions to Lat B (e.g., formation of lamel-
lipodia was inhibited at 0.1 M), they reasoned that there were distinct pools of
exchangeable actin in parietal cells, with microvillar microfilaments being partic-
ularly stable owing to the presence of stabilizing, capping, and bundling proteins.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

120 YAO FORTE

They proposed that the resistance of acid secretory function to Lat B is the result
of stable actin filament-turnover pathways that minimize the accessibility of the
actin monomer to Lat B.
The actin cytoskeleton has been linked to the mechanism of potentiation of
cAMP-mediated acid secretion by carbachol in rabbit gastric glands (137). The
observed potentiation was dependent upon release of Ca2+ from intracellular stores
regulated by the type 3 IP3 receptor (the major subtype in the parietal cell), but
it was unaffected by changes in extracellular Ca2+ or inhibitors of either PKC or
CAM kinase II. On the other hand, the disrupter of actin filaments, cytochala-
sin D, preferentially blocked the secretory effect of carbachol and its synergism
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

with cAMP, as well as the release of Ca2+ from stores. Treatment of glands
with cytochalasin D also caused redistribution of the IP3 receptor from a plasma
membrane-like fraction to the microsomal fraction, suggesting a dissociation of
the stores from the plasma membrane and a functional coupling between actin
filaments and the receptor-mediated Ca2+ store.
by HINARI on 05/16/07. For personal use only.

Ezrin-Actin Interactions
An actin-binding/regulatory protein that may have a primary role in parietal cell
function was mentioned above as the stimulation-dependent 80-kDa phospho-
protein called ezrin. Ezrin was independently identified by various investigators
for their special interests (138140) and implicated as a cytoskeleton-membrane
linker protein. Ezrin is the best studied member of the ERM (ezrin-radixin-moesin)
family (141). These proteins share approximately 75% primary sequence identity
and contain an N-terminal globular domain followed by an -helical region and
a C-terminal tail domain (142). Although ezrin was originally purified with the
microfilament bundles from intestinal microvilli, the native, full-length protein
possessed no convincing F-actin binding activity in tests that used -actin from
skeletal muscle, a commonly used rich source of pure actin. However, C-terminal
fragments of ezrin were shown to bind filamentous -actin (143); consequently,
Bretscher et al. (142) developed a model of intra- and intermolecular folding of
full-length ezrin to account for the discrepancy.
Ezrin was first identified in parietal cells on the basis of PKA-dependent phos-
phorylation concomitant with stimulation of gastric glands (19, 22). Double im-
munostaining for ezrin and for -actin revealed that these two proteins are abundant
and primarily co-localized to the same regions within parietal cells, characteris-
tic of the tortuous apical canalicular surface wending through most of the cell
(23, 24, 134). Interestingly, ezrin is almost exclusively localized to parietal cells,
whereas staining for the -actin isoform is intense along the apical borders of all
cell types lining the gland lumen as well as within apical canaliculi of parietal
cells. Because the -actin isoform is primarily distributed near the basolateral
membrane of parietal cells (134), it appears that ezrin is co-localized with -actin,
but not -actin, as a subset of the F-actin microfilaments.
On the basis of actin-ezrin cyto-localization studies and the knowledge that
-actin is not expressed in parietal cells, Yao et al. (144) hypothesized that ezrin
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 121

preferentially binds to nonmuscle actin isoforms rather than to skeletal muscle


-actin. Using a co-sedimentation assay that compared purified -actin with skele-
tal muscle -actin, ezrin appeared in the pellet only when -actin was polymer-
ized, whereas a known actin-binding protein, the S1 tryptic fragment of myosin II,
showed no selectivity between these actin isoforms. The calculated data suggest a
stoichiometry of 1 ezrin bound per 12 actin molecules. These studies indicate that
gastric ezrin possesses F-actin binding in an isoform-specific manner. In fact, the
binding of full-length ezrin to actin was lately demonstrated using a novel solid
phase actin-binding assay (145).
In their search for a novel class of pharmacological antiulcer agents, Urushidani
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

et al. (146) reported that a compound designated ME3407, a potent acid secretion
inhibitor, arrested the morphological transition of cells stimulated by histamine.
Because ME3407 was found to liberate the phosphoprotein ezrin from the api-
cal plasma membrane, as well as to inhibit histamine-triggered translocation of
H,K-ATPase, these authors hypothesized that ME3407 may exert its inhibitory
action on the modulation of protein phosphorylation required for recruitment of
by HINARI on 05/16/07. For personal use only.

H,K-ATPase. Dephosphorylation of ezrin has been correlated with its dissociation


from the plasma membrane-cytoskeleton (147). ME3407 was also observed to in-
hibit MLCK and PI3 kinase activities measured in vitro (44). Because wortmannin
is a well known inhibitor of these latter kinases, it was tested and found to inhibit
glandular acid secretion, similar to the effect of ME3407. Recent studies, however,
have differentiated the effects of wortmannin and ME3407 (37). Both compounds
were found to inhibit acid secretion by intact gastric glands, but only ME3407
e inhibited acid secretion in SLO-permeabilized glands and liberated ezrin from
its membrane-cytoskeletal locus at the canalicular surface. Thus it appears that
the targets of these two inhibitors are not identical. Future studies may attempt to
define whether and to what extent phosphorylation of ezrin might be altered by
ME3407 and relate it to the modulation of parietal cell secretion.

Ezrin Interacts with Other Cellular Proteins


Using an overlay assay, Dransfield et al. (148) showed that ezrin binds to the
regulatory RII subunit of PKA in parietal cells, suggesting it might be involved in
the functional localization of PKA as an anchoring protein. In addition, the authors
showed that the RII subunit of PKA is capable of binding to full-length ezrin in
cell extracts, arguing that inter- or intramolecular interaction of ezrin masks ezrin
activity in binding to other cellular proteins. It was also reported that the N-terminal
domain of ezrin binds to EBP50, which mediates ezrin association with the plasma
membrane, although the physiological relevance of such interaction remains to be
characterized (144).
In a search for Ca2+-mediated signaling events in parietal cell activation, Yao
et al. (149) discovered the presence of calpain I (-calpain) in gastric parietal cells
and its interaction with ezrin. Further studies showed that parietal cell calpain
I can be activated by an ionomycin-mediated elevation of intracellular calcium;
furthermore, the activation of calpain triggers the release and hydrolysis of ezrin
21 Jan 2003 11:27 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

122 YAO FORTE

(149), followed by the collapse of the actin cytoskeleton in parietal cells. These
studies suggest the importance of ezrin in the integrity of the actin cytoskeleton of
parietal cells. Significantly, the activation of calpain, as evidenced by the hydrolysis
of ezrin, was correlated with the inhibition of parietal cell secretion, presumably
through the loss of ezrin. This ezrin-calpain interaction is unique because the
calpain cleavage site is located to the C-terminal 200 amino acids of ezrin where
sequences of the ERM family are divergent. To explore the possible regulation
by calcium and calpain I of the interaction between -actin and ezrin, Herman
and his associates overexpressed calpastatin, an endogenous inhibitor of calpain,
in fibroblast cells. The overexpression resulted in a diminished calpain activity
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

and increased ezrin protein level, which was experimentally correlated with an
inhibition of filopodial formation (150), suggesting that spatial activation of calpain
may be necessary for actin dynamics associated with spreading at the leading edge.
It will be of interest to evaluate whether the hydrolysis of ezrin, or the liberation of
ezrin from actin cytoskeleton, is essential for formation of cytoplasmic extensions
such as filopodia or microvilli.
by HINARI on 05/16/07. For personal use only.

A NOTE ON THE FUSION REACTION


Thus far we have discussed the signal transduction events of parietal cell activation
in terms of (a) the structural elements (cytoskeleton) that support and promote the
translocation of H,K-ATPase-rich membranes from their cytoplasmic location to
the apical membrane and (b) the cognitive elements (SNAREs) that assure mem-
branes are directed to and associate with highly specific docking sites. (c) A third
and equally important element has generally been given less attention, the fusion
mechanism itself, i.e, the means by which two distinct apposing lipid bilayers
interact and flow into one. A schematic view of these three cooperative processes
contributing to membrane recruitment is shown in Figure 2. The proposal that
phospholipid surface chemistry and fusion processes play a role in membrane
recruitment and secretion extends back to 1963, when morphological transforma-
tions of acid-secreting cells were correlated with phospholipid turnover in bullfrog
gastric mucosa (87). Although this was an important step in formulating the mem-
brane recycling hypothesis, little work was done on the fusion process per se until
recently. Using separate assays of fluorescence dequenching and intervesicular
protein transfer, Duman et al. (151) have demonstrated that gastric tubulovesicular
membranes will fuse with each other (homotypic fusion) or with liposomes of
the appropriate composition. The fusion reaction is highly specific for a trigger-
ing system that must include either Ca2+ or ATP (EC50 is 0.15 M and 1 mM,
respectively) and is dependent on the presence of protein in at least one of the
two vesicular pools, although the specific protein has not been identified. Whereas
these in vitro fusion data are preliminary, they do offer a system by which the
interfacial reactions of fusion might be studied, with the potential to identify new
classes of proteins that facilitate the lipid mixing processes.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 123

PERSPECTIVES
Although great progress has been made over the past 10 years in characterizing
the mechanisms and participants in parietal cell secretion, the challenge ahead is
to define the precise function and the physiological regulation of the implicated
proteins and identify new players (152). By analyzing both the cell biology and
physiology of phenotypic changes in cultured parietal cells and even transgenic
animals expressing mutant regulatory proteins, it will be possible to determine
whether and how a particular molecule operates upon the signaling cascade and
downstream effector reactions of parietal cell activation. Molecular engineering
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

of fluorescent reporters on a desired molecule has emerged as an exciting way to


correlate the cytological changes to molecular function of individual regulatory
proteins in real-time imaging of live parietal cells. Such studies will consolidate
protein-protein interactions that have been inferred from indirect techniques into
a physiological model for parietal cell activation and relate these data to molec-
by HINARI on 05/16/07. For personal use only.

ular medicine of associated disorders such as peptic ulcer disease and gastric
carcinoma.

The Annual Review of Physiology is online at http://physiol.annualreviews.org

LITERATURE CITED
1. Forte JG, Soll A. 1989. Cell biology of etal cell transformation. J. Cell Sci. 108:
hydrochloric acid secretion. See Ref. 153, 112741
pp. 20728 7. Berglindh T, Dibona DR, Ito S, Sachs
2. Ito S. 1987. Functional gastric morphol- G. 1980. Probes of parietal cell function.
ogy. In Physiology of the Gastrointestinal Am. J. Physiol. Gastrointest. Liver Phys-
Tract, ed. LR Johnson, pp. 81751. New iol. 238:G165G76
York: Raven 8. Pettitt JM, van Driel IR, Toh BH, Gleeson
3. Forte TM, Machen TE, Forte JG. 1977. PA. 1996. From coiled tubules to a secre-
Ultrastructural changes in oxyntic cells tory canaliculus: a new model for mem-
associated with secretory function: a brane transformation and acid secretion
membrane-recycling hypothesis. Gastro- by gastric parietal cells. Trends Cell Biol.
enterology 73:94155 6:4952
4. Helander HF. 1981. The cells of the gas- 9. Duman JG, Pathak NJ, Ladinsky MS,
tric mucosa. Int. Rev. Cytol. 70:21789 McDonald KL, Forte JG. 2002. Three-
5. Forte JG, Yao X. 1996. The membrane- dimensional reconstruction of cytoplas-
recruitment-and-recycling hypothesis of mic membrane networks in parietal cells.
gastric HCl secretion. Trends Cell Biol. J. Cell Sci. 115:125158
6:4548 10. Berglindh T, Obrink KJ. 1976. A method
6. Pettitt JM, Humphris DC, Barrett SP, Toh for preparing isolated glands from the rab-
BH, van Driel IR, Gleeson PA. 1995. bit gastric mucosa. Acta Physiol. Scand.
Fast freeze-fixation/freeze-substitution 96:15059
reveals the secretory membranes of the 11. Chew CS. 1994. Parietal cell culture: new
gastric parietal cell as a network of heli- models and directions. Annu. Rev. Phys-
cally coiled tubule. A new model for pari- iol. 56:44561
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

124 YAO FORTE

12. Chew CS, Ljungstrom M, Smolka A, protein involved in parietal cell stimula-
Brown MR. 1989. Primary culture of sec- tion. Am. J. Physiol. Gastrointest. Liver
retagogue-responsive parietal cells from Physiol. 256:G1070G81
rabbit gastric mucosa. Am. J. Physiol. 23. Hanzel DK, Urushidani T, Usinger WR,
Gastrointest. Liver Physiol. 256:G254 Smolka A, Forte JG. 1989. Immunolog-
G63 ical localization of an 80-kDa phospho-
13. Prinz C, Zanner R, Gerhard M, Mahr S, protein to the apical membrane of gastric
Neumayer N, et al. 1999. The mecha- parietal cells. Am. J. Physiol. Gastrointest.
nism of histamine secretion from gastric Liver Physiol. 256:G1082G89
enterochromaffin-like cells. Am. J. Phys- 24. Hanzel D, Reggio H, Bretscher A, Forte
iol. Cell Physiol. 277:C845C55 JG, Mangeat P. 1991. The secretion-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

14. Roth JA, Ivy AC. 1944. The synergistic stimulated 80 K phosphoprotein of pari-
effect of caffeine upon histamine in rela- etal cells is ezrin, and has properties of
tion to gastric secretion. Am. J. Physiol. a membrane cytoskeletal linker in the
142:10713 induced apical microvilli. EMBO J. 10:
15. Chew CS, Hersey SJ, Sachs G, Berglindh 236373
T. 1980. Histamine responsiveness of iso- 25. Mizukawa Y, Nishizawa T, Nagao T, Kita-
by HINARI on 05/16/07. For personal use only.

lated gastric glands. Am. J. Physiol. Gas- mura K, Urushidani T. 2002. Cellular dis-
trointest. Liver Physiol. 238:G312G20 tribution of parchorin, a chloride intracel-
16. Chew CS. 1983. Forskolin stimulation of lular channel-related protein, in various
acid and pepsinogen secretion in isolated tissues. Am. J. Physiol. Cell Physiol. 282:
gastric glands. Am. J. Physiol. Cell Phys- C786C95
iol. 245:C371C80 26. Nishizawa T, Nagao T, Iwatsubo T,
17. Chew CS. 1985. Parietal cell protein Forte JG, Urushidani T. 2000. Molecu-
kinases. Selective activation of type I lar cloning and characterization of a novel
cAMP-dependent protein kinase by his- chloride intracellular channel-related pro-
tamine. J. Biol. Chem. 260:754050 tein, parchorin, expressed in water-secre-
18. Chew CS, Brown MR. 1987. Histamine ting cells. J. Biol. Chem. 275:1116473
increases phosphorylation of 27- and 27. Brown MR, Chew CS. 1989. Carbachol-
40-kDa parietal cell proteins. Am. J. induced protein phosphorylation in pari-
Physiol. Gastrointest. Liver Physiol. 253: etal cells: regulation by [Ca2+]i. Am. J.
G823G29 Physiol. Gastrointest. Liver Physiol. 257:
19. Urushidani T, Hanzel DK, Forte JG. G99G110
1987. Protein phosphorylation associated 28. Parente JA Jr, Chen X, Zhou C, Petro-
with stimulation of rabbit gastric glands. poulos AC, Chew CS. 1999. Isola-
Biochim. Biophys. Acta. 930:20919 tion, cloning, and characterization of a
20. Modlin IM, Oddsdottir M, Adrian TE, new mammalian coronin family member,
Zdon MJ, Zucker KA, Goldenring JR. coroninse, which is regulated within the
1987. A specific histamine-stimulated protein kinase C signaling pathway. J.
phosphoprotein in isolated parietal cells. Biol. Chem. 274:301725
J. Surg. Res. 42:34853 29. Parente JA, Goldenring JR, Petropoulos
21. Malinowska DH, Sachs G, Cuppoletti J. AC, Hellman U, Chew CS. 1996. Purifi-
1988. Gastric H+ secretion: histamine cation, cloning, and expression of a
(cAMP-mediated) activation of protein novel, endogenous, calcium-sensitive,
phosphorylation. Biochim. Biophys. Acta 28-kDa phosphoprotein. J. Biol. Chem.
972:95109 271:20096101
22. Urushidani T, Hanzel DK, Forte JG. 1989. 30. Malinowska DH, Koelz HR, Hersey SJ,
Characterization of an 80-kDa phospho- Sachs G. 1981. Properties of the gastric
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 125

proton pump in unstimulated permeable phosphoprotein within the cAMP signal-


gastric glands. Proc. Natl. Acad. Sci. USA ing pathway in the gastric parietal cell.
78:590812 Am. J. Physiol. Cell Physiol. 275:C56
31. Hersey SJ, Steiner L. 1985. Acid for- C67
mation by permeable gastric glands: en- 40. Chew C, Chen X, Parente JA Jr, Tarrer
hancement by prestimulation. Am. J. S, Okamoto C, Qin H-Y. 2002. Lasp-1
Physiol. Gastrointest. Liver Physiol. 248: binds to non-muscle F-actin and is local-
G561G68 ized within multiple sites of dynamic actin
32. Hersey SJ, Steiner L. 1988. Stimulation assembly. J. Cell Sci. In press
of acid formation in permeable gastric 41. Chew CS, Parente JA Jr, Chen X, Chapon-
glands by valinomycin. Am. J. Physiol. nier C, Cameron RS. 2000. The LIM and
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

Gastrointest. Liver Physiol. 255:G313 SH3 domain-containing protein, lasp-1,


G18 may link the cAMP signaling pathway
33. Malinowska DH. 1990. Permeabilizing with dynamic membrane restructuring ac-
parietal cells. Methods Enzymol. 192: tivities in ion transporting epithelia. J. Cell
10824 Sci. 113:203545
34. Yao X, Karam SM, Ramilo M, Rong Q, 42. Urushidani T, Chow D, Forte JG. 1999.
by HINARI on 05/16/07. For personal use only.

Thibodeau A, Forte JG. 1996. Stimula- Redistribution of a 120 kDa phosphopro-


tion of gastric acid secretion by cAMP tein in the parietal cell associated with
in a novel -toxin-permeabilized gland stimulation. J. Membr. Biol. 168:20920
model. Am. J. Physiol. Cell Physiol. 271: 43. Chew CS, Brown MR. 1986. Release of
C61C73 intracellular Ca2+ and elevation of inosi-
35. Rong Q, Utevskaya O, Ramilo M, tol trisphosphate by secretagogues in pari-
Chow DC, Forte JG. 1998. Nucleotide etal and chief cells isolated from rabbit
metabolism by gastric glands and H+- gastric mucosa. Biochim. Biophys. Acta
K+-ATPase-enriched membranes. Am. J. 888:11625
Physiol. Gastrointest. Liver Physiol. 274: 44. Chew CS. 1986. Cholecystokinin, car-
G103G10 bachol, gastrin, histamine, and forskolin
36. Akagi K, Nagao T, Urushidani T. 1999. increase [Ca2+]i in gastric glands. Am.
Responsiveness of -escin-permeabilized J. Physiol. Gastrointest. Liver Physiol.
rabbit gastric gland model: effects of func- 250:G814G23
tional peptide fragments. Am. J. Physiol. 45. Negulescu PA, Machen TE. 1988. Release
Gastrointest. Liver Physiol. 277:G736 and reloading of intracellular Ca stores
G44 after cholinergic stimulation of the pari-
37. Ammar DA, Zhou R, Forte JG, Yao X. etal cell. Am. J. Physiol. Cell Physiol.
2002. Syntaxin 3 is required for cAMP- 254:C498C504
induced acid secretion: streptolysin O- 46. Negulescu PA, Machen TE. 1988. In-
permeabilized gastric gland model. Am. J. tracellular Ca regulation during secreta-
Physiol. Gastrointest. Liver Physiol. 282: gogue stimulation of the parietal cell. Am.
G23G33 J. Physiol. Cell Physiol. 254:C130C40
38. Akagi K, Nagao T, Urushidani T. 47. Negulescu PA, Reenstra WW, Machen
2001. Reconstitution of acid secretion TE. 1989. Intracellular Ca requirements
in digitonin-permeabilized rabbit gastric for stimulus-secretion coupling in parietal
glands. Identification of cytosolic regula- cell. Am. J. Physiol. Cell Physiol. 256:
tory factors. J. Biol. Chem. 276:28171 C241C51
78 48. Hanson PJ, Hatt JF. 1989. Intracellular
39. Chew CS, Parente JA Jr, Zhou C, Baranco signalling and regulation of gastric acid
E, Chen X. 1998. Lasp-1 is a regulated secretion. Q. J. Exp. Physiol. 74:60734
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

126 YAO FORTE

49. Anderson NG, Hanson PJ. 1985. Involve- IKK-1 and IKK-2: cytokine-activated IkB
ment of calcium-sensitive phospholipid- kinases essential for NF-kB activation.
dependent protein kinase in control of Science 278:86066
acid secretion by isolated rat parietal cells. 59. Takeuchi Y, Yamada J, Yamada T, Todisco
Biochem. J. 232:60911 A. 1997. Functional role of extracellular
50. Beil W, Mannschedel W, Sewing KF. signal-regulated protein kinases in gastric
1987. Protein kinase C and parietal cell acid secretion. Am. J. Physiol. Gastroin-
function. Biochem. Biophys. Res. Com- test. Liver Physiol. 273:G1263G72
mun. 149:72028 60. Takeuchi Y, Pausawasdi N, Todisco A.
51. Brown MR, Chew CS. 1987. Multiple ef- 1999. Carbachol activates ERK2 in iso-
fects of phorbol ester on secretory activity lated gastric parietal cells via multiple
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

in rabbit gastric glands and parietal cells. signaling pathways. Am. J. Physiol. Gas-
Can. J. Physiol. Pharmacol. 65:184047 trointest. Liver Physiol. 276:G1484G92
52. Chew CS, Zhou CJ, Parente JA Jr. 1997. 61. Nagahara A, Wang L, Del Valle J, Todisco
Ca2+-independent protein kinase C iso- A. 1998. Regulation of c-Jun NH2-
forms may modulate parietal cell HCl terminal kinases in isolated canine gastric
secretion. Am. J. Physiol. Gastrointest. parietal cells. Am. J. Physiol. Gastrointest.
by HINARI on 05/16/07. For personal use only.

Liver Physiol. 272:G246G56 Liver Physiol. 275:G740G48


53. Nandi J, Loo A, Kim SW, Levine RA. 62. Cobb MH, Goldsmith EJ. 1995. How
1999. Expression and characterization of MAP kinases are regulated. J. Biol. Chem.
protein kinase C in isolated rabbit parietal 270:1484346
cells. Int. J. Mol. Med. 3:52126 63. Davis RJ. 1993. The mitogen-activated
54. Tsunoda Y, Funasaka M, Modlin IM, protein kinase signal transduction path-
Hidaka H, Fox LM, Goldenring JR. way. J. Biol. Chem. 268:1455356
1992. An inhibitor of Ca2+/calmodulin- 64. Treisman R. 1995. Journey to the surface
dependent protein kinase II, KN-62, in- of the cell: Fos regulation and the SRE.
hibits cholinergic-stimulated parietal cell EMBO J. 14:490513
secretion. Am. J. Physiol. Gastrointest. 65. Widmann C, Gibson S, Jarpe MB, John-
Liver Physiol. 262:G118G22 son GL. 1999. Mitogen-activated pro-
55. Mamiya N, Goldenring JR, Tsunoda tein kinase: conservation of a three-kinase
Y, Modlin IM, Yasui K, et al. 1993. module from yeast to human. Physiol.
Inhibition of acid secretion in gastric Rev. 79:14380
parietal cells by the Ca2+/calmodulin- 66. Pausawasdi N, Ramamoorthy S, Stepan
dependent protein kinase II inhibitor KN- V, Del Valle J, Todisco A. 2000. Regula-
93. Biochem. Biophys. Res. Commun. tion and function of p38 protein kinase
195:60815 in isolated canine gastric parietal cells.
56. Todisco A, Ramamoorthy S, Pausawasdi Am. J. Physiol. Gastrointest. Liver Phys-
N, Tacey K. 1999. Carbachol activates iol. 278:G24G31
IkappaB kinase in isolated canine gas- 67. Chew CS, Nakamura K, Petropoulos
tric parietal cells. Biochem. Biophys. Res. AC. 1994. Multiple actions of epidermal
Commun. 261:87784 growth factor and TGF- on rabbit gastric
57. DiDonato JA, Hayakawa M, Rothwarf parietal cell function. Am. J. Physiol. Gas-
DM, Zandi E, Karin M. 1997. A cytokine- trointest. Liver Physiol. 267:G818G26
responsive IkB kinase that activates the 68. Nakamura K, Zhou CJ, Parente J, Chew
transcription factor NF-kB. Nature 388: CS. 1996. Parietal cell MAP kinases: mul-
54854 tiple activation pathways. Am. J. Physiol.
58. Mercurio F, Zhu H, Murray BW, Gastrointest. Liver Physiol. 271:G640
Shevchenko A, Bennett BL, et al. 1997. G49
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 127

69. Kusayanagi S, Takeuchi Y, Todisco A, update of biology and physiology. Gen.


Mitamura K. 2002. Extracellular signal- Pharmacol. 32:41322
regulated protein kinases mediate H+,K+- 79. Park J, Chiba T, Yamada T. 1987. Mecha-
ATPase -subunit gene expression. Bio- nisms for direct inhibition of canine gas-
chem. Biophys. Res. Commun. 290:1289 tric parietal cells by somatostatin. J. Biol.
94 Chem. 262:1419096
70. Todisco A, Pausawasdi N, Ramamoorthy 80. Del Valle J, Chiba T, Park J, Yamada
S, Del Valle J, Van Dyke RW, Askari FK. T. 1993. Distinct receptors for chole-
2001. Functional role of protein kinase cystokinin and gastrin on canine fundic
B/Akt in gastric acid secretion. J. Biol. D-cells. Am. J. Physiol. Gastrointest.
Chem. 276:4643644 Liver Physiol. 264:G811G15
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

71. Tsunoda Y, Modlin IM, Goldenring JR. 81. Wyatt MA, Jarvie E, Feniuk W, Humphrey
1993. Tyrosine kinase activities in the PP. 1996. Somatostatin sst2 receptor-
modulation of stimulated parietal cell acid mediated inhibition of parietal cell func-
secretion. Am. J. Physiol. Gastrointest. tion in rat isolated gastric mucosa. Br. J.
Liver Physiol. 264:G351G56 Pharmacol. 119:90510
72. Sterling H, Lin DH, Gu RM, Dong K, 82. Schmidtler J, Rosenthal W, Offermanns
by HINARI on 05/16/07. For personal use only.

Hebert SC, Wang WH. 2002. Inhibition S, Schusdziarra V, Classen M, Schepp


of protein-tyrosine phosphatase stimu- W. 1992. Pertussis toxin reverses prost-
lates the dynamin-dependent endocyto- aglandin E2- and somatostatin-induced
sis of ROMK1. J. Biol. Chem. 277:4317 inhibition of rat parietal cell H(+)-
23 production. Cell Signal 4:32129
73. Courtois-Coutry N, Roush D, Rajendran 83. Todisco A, Campbell V, Dickinson CJ,
V, McCarthy JB, Geibel J, et al. 1997. A Del Valle J, Yamada T. 1994. Molecu-
tyrosine-based signal targets H/K-ATPase lar basis for somatostatin action: inhibi-
to a regulated compartment and is re- tion of c-fos expression and AP-1 binding.
quired for the cessation of gastric acid se- Am. J. Physiol. Gastrointest. Liver Phys-
cretion. Cell 90:50110 iol. 267:G245G53
74. Bengtsson P, Lundqvist G, Nilsson G. 84. Todisco A, Takeuchi Y, Yamada J, Sa-
1989. Inhibition of acid formation and doshima JI, Yamada T. 1997. Molecular
stimulation of somatostatin release by mechanisms for somatostatin inhibition
cholecystokinin-related peptides in rabbit of c-fos gene expression. Am. J. Physiol.
gastric glands. J. Physiol. 419:76574 Gastrointest. Liver Physiol. 272:G721
75. Walsh JH. 1988. Peptides as regulators of G26
gastric acid secretion. Annu. Rev. Physiol. 85. Lee HC, Forte JG. 1978. A study of H+
50:4163 transport in gastric microsomal vesicles
76. Beglinger C, Hildebrand P, Meier R, using fluorescent probes. Biochim. Bio-
Bauerfeind P, Hasslocher H, et al. 1992. A phys. Acta 508:33956
physiological role for cholecystokinin as 86. Sachs G, Kaunitz J, Mendlein J, Wallmark
a regulator of gastrin secretion. Gastroen- B. 1989. Biochemistry of gastric acid se-
terology 103:49095 cretion: H+-K+-ATPase. See Ref. 153, pp.
77. Athmann C, Zeng N, Scott DR, Sachs G. 22953
2000. Regulation of parietal cell calcium 87. Kasbekar DK, Forte GM, Forte JG. 1968.
signaling in gastric glands. Am. J. Physiol. Phospholipid turnover and ultrastructural
Gastrointest. Liver Physiol. 279:G1048 changes in resting and secreting bullfrog
G58 gastric mucosa. Biochim. Biophys. Acta
78. Chen D, Zhao CM, Lindstrom E, Hakan- 163:113
son R. 1999. Rat stomach ECL cells 88. Rothman JE, Warren G. 1994.
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

128 YAO FORTE

Implications of the SNARE hypoth- 97. Lehnardt S, Ahnert-Hilger G, Bigalke H,


esis for intracellular membrane topology Jons T. 2000. Acid secretion of pari-
and dynamics. Curr. Biol. 4:22033 etal cells is paralleled by a redistribu-
89. Bennett MK, Scheller RH. 1994. A molec- tion of NSF and ,-SNAPs and inhibited
ular description of synaptic vesicle mem- by tetanus toxin. Histochem. Cell Biol.
brane trafficking. Annu. Rev. Biochem. 114:38791
63:63100 98. Karvar S, Yao X, Duman JG, Zhou R, Liu
90. Bennett MK, Calakos N, Scheller RH. Y, Forte JG. 2002. Inhibiton of acid se-
1992. Syntaxin: a synaptic protein im- cretion by C-terminal mutant of SNAP-25
plicated in docking of synaptic vesicles in gastric parietal cells. Gastroenterology
at presynaptic active zones. Science 257: 122:A253. (Abstr. M1017)
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

25559 99. Zerial M, McBride H. 2001. Rab proteins


91. Schiavo G, Shone CC, Bennett MK, as membrane organizers. Nat. Rev. Mol.
Scheller RH, Montecucco C. 1995. Bo- Cell Biol. 2:10717
tulinum neurotoxin type C cleaves a sin- 100. Goldenring JR, Shen KR, Vaughan HD,
gle Lys-Ala bond within the carboxyl- Modlin IM. 1993. Identification of a small
terminal region of syntaxins. J. Biol. GTP-binding protein, Rab25, expressed
by HINARI on 05/16/07. For personal use only.

Chem. 270:1056670 in the gastrointestinal mucosa, kidney, and


92. Peng XR, Yao X, Chow DC, Forte JG, lung. J. Biol. Chem. 268:1841922
Bennett MK. 1997. Association of syn- 101. Goldenring JR, Smith J, Vaughan HD,
taxin 3 and vesicle-associated membrane Cameron P, Hawkins W, Navarre J. 1996.
protein (VAMP) with H+/K+-ATPase- Rab11 is an apically located small GTP-
containing tubulovesicles in gastric pari- binding protein in epithelial tissues. Am.
etal cells. Mol. Biol. Cell 8:399407 J. Physiol. Gastrointest. Liver Physiol.
93. Calhoun BC, Goldenring JR. 1997. Two 270:G515G25
Rab proteins, vesicle-associated mem- 102. Duman JG, Tyagarajan K, Kolsi MS,
brane protein 2 (VAMP-2) and secretory Moore HP, Forte JG. 1999. Expression of
carrier membrane proteins (SCAMPs), rab11a N124I in gastric parietal cells in-
are present on immunoisolated parietal hibits stimulatory recruitment of the H+-
cell tubulovesicles. Biochem. J. 325:559 K+-ATPase. Am. J. Physiol. Cell Physiol.
64 277:C361C72
94. Calhoun BC, Lapierre LA, Chew CS, 103. Hales CM, Griner R, Hobdy-Henderson
Goldenring JR. 1998. Rab11a redis- KC, Dorn MC, Hardy D, et al. 2001. Iden-
tributes to apical secretory canaliculus tification and characterization of a fam-
during stimulation of gastric parietal cells. ily of Rab11-interacting proteins. J. Biol.
Am. J. Physiol. Cell Physiol. 275:C163 Chem. 276:3906775
C70 104. Okamoto CT, Karam SM, Jeng YY,
95. Agnew BJ, Duman JG, Watson CL, Col- Forte JG, Goldenring JR. 1998. Identi-
ing DE, Forte JG. 1999. Cytological trans- fication of clathrin and clathrin adaptors
formations associated with parietal cell on tubulovesicles of gastric acid secretory
stimulation: critical steps in the activation (oxyntic) cells. Am. J. Physiol. Cell Phys-
cascade. J. Cell Sci. 112:263946 iol. 274:C1017C29
96. Karvar S, Yao X, Duman JG, Hybiske 105. Okamoto CT, Duman JG, Tyagarajan K,
K, Liu Y, Forte JG. 2002. Intracellu- McDonald KL, Jeng YY, et al. 2000.
lar distribution and functional importance Clathrin in gastric acid secretory (pari-
of vesicle-associated membrane protein-2 etal) cells: biochemical characterization
in gastric parietal cells. Gastroenterology and subcellular localization. Am. J. Phys-
123:28190 iol. Cell Physiol. 279:C833C51
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 129

106. Schmid SL, McNiven MA, De Camilli sion, intracellular routing, and functional
P. 1998. Dynamin and its partners: a properties of Na,K-ATPase. J. Biol. Chem.
progress report. Curr. Opin. Cell Biol. 10: 273:3082635
50412 117. Geering K. 2001. The functional role of
107. Sever S, Damke H, Schmid SL. 2000. Gar- subunits in oligomeric P-type ATPases. J.
rotes, springs, ratchets, and whips: putting Bioenerg. Biomembr. 33:42538
dynamin models to the test. Traffic 1:385 118. Koenderink JB, Hermsen HP, Swarts HG,
92 Willems PH, De Pont JJ. 2000. High-
108. Okamoto CT, Li R, Zhang Z, Jeng YY, affinity ouabain binding by a chimeric
Chew CS. 2002. Regulation of protein gastric H+,K+-ATPase containing trans-
and vesicle trafficking at the apical mem- membrane hairpins M3-M4 and M5-M6
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

brane of epithelial cells. J. Control Re- of the 1-subunit of rat Na+,K+-ATPase.


lease 78:3541 Proc. Natl. Acad. Sci. USA 97:1120914
109. Kaplan KB, Swedlow JR, Varmus HE, 119. Kirchhausen T. 1999. Adaptors for
Morgan DO. 1992. Association of p60c- clathrin-mediated traffic. Annu. Rev. Cell
src with endosomal membranes in mam- Dev. Biol. 15:70532
malian fibroblasts. J. Cell Biol. 118:321 120. Spicer Z, Miller ML, Andringa A, Riddle
by HINARI on 05/16/07. For personal use only.

33 TM, Duffy JJ, et al. 2000. Stomachs of


110. Luton F, Verges M, Vaerman JP, Sudol M, mice lacking the gastric H,K-ATPase -
Mostov KE. 1999. The SRC family pro- subunit have achlorhydria, abnormal pari-
tein tyrosine kinase p62yes controls poly- etal cells, and ciliated metaplasia. J. Biol.
meric IgA transcytosis in vivo. Mol. Cell Chem. 275:2155565
4:62732 121. Scarff KL, Judd LM, Toh BH, Gleeson
111. Freedman SD, Katz MH, Parker EM, PA, Van Driel IR. 1999. Gastric H+,K+-
Gelrud A. 1999. Endocytosis at the api- adenosine triphosphatase beta subunit is
cal plasma membrane of pancreatic aci- required for normal function, develop-
nar cells is regulated by tyrosine kinases. ment, and membrane structure of mouse
Am. J. Physiol. Cell Physiol. 276:C306 parietal cells. Gastroenterology 117:605
C11 18
112. Thomas SM, Brugge JS. 1997. Cellular 122. Dunbar LA, Caplan MJ. 2001. Ion pumps
functions regulated by Src family kinases. in polarized cells: sorting and regulation
Annu. Rev. Cell Dev. Biol. 13:513609 of the Na+, K+- and H+, K+-ATPases. J.
113. Lapierre LA, Kumar R, Hales CM, Biol. Chem. 276:2961720
Navarre J, Bhartur SG, et al. 2001. Myo- 123. Tyagarajan K, Lipniunas PH, Townsend
sin Vb is associated with plasma mem- RR, Forte JG. 1997. The N-linked
brane recycling systems. Mol. Biol. Cell oligosaccharides of the -subunit of rab-
12:184357 bit gastric H,K-ATPase: site localization
114. Wu XS, Rao K, Zhang H, Wang F, Sell- and identification of novel structures. Bio-
ers JR, et al. 2002. Identification of an or- chemistry 36:1020012
ganelle receptor for myosin-Va. Nat. Cell 124. Asano S, Hoshina S, Nakaie Y, Watan-
Biol. 4:27178 abe T, Sato M, et al. 1998. Functional ex-
115. Chow DC, Forte JG. 1995. Functional pression of putative H+-K+-ATPase from
significance of the -subunit for het- guinea pig distal colon. Am. J. Physiol.
erodimeric P-type ATPases. J. Exp. Biol. Cell Physiol. 275:C669C74
198(Pt 1):117 125. Smith PR, Bradford AL, Joe EH, An-
116. Hasler U, Wang X, Crambert G, Beguin gelides KJ, Benos DJ, Saccomani G.
P, Jaisser F, et al. 1998. Role of -subunit 1993. Gastric parietal cell H+-K+-ATPase
domains in the assembly, stable expres- microsomes are associated with isoforms
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

130 YAO FORTE

of ankyrin and spectrin. Am. J. Physiol. Functionally distinct pools of actin in se-
Cell Physiol. 264:C63C70 cretory cells. Am. J. Physiol. Cell Physiol.
126. Carraway KL, Carraway CA. 1989. 281:C407C17
Membrane-cytoskeleton interactions in 137. Muto Y, Nagao T, Yamada M, Mikoshiba
animal cells. Biochim. Biophys. Acta 988: K, Urushidani T. 2001. A proposed mech-
14771 anism for the potentiation of cAMP-
127. Yao X, Forte JG. 1996. Membrane- mediated acid secretion by carbachol. Am.
cytoskeletal interaction in regulated exo- J. Physiol. Cell Physiol. 280:C155C65
cytosis and apical insertion of vesicles in 138. Hunter T, Cooper JA. 1981. Epidermal
epithelial cells. In Current Topics in Mem- growth factor induces rapid tyrosine phos-
branes, ed. WJ Nelson, pp. 7396. San phorylation of proteins in A431 human tu-
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

Diego, CA: Academic mor cells. Cell 24:74152


128. Rosenfeld GC, McAllister E, Thompson 139. Bretscher A. 1983. Purification of an
WJ. 1981. Cytochalasin inhibition of iso- 80,000-dalton protein that is a component
lated rat gastric parietal cell function. J. of the isolated microvillus cytoskeleton,
Cell. Physiol. 109:5357 and its localization in nonmuscle cells. J.
129. Black JA, Forte TM, Forte JG. 1982. The Cell Biol. 97:42532
by HINARI on 05/16/07. For personal use only.

effects of microfilament disrupting agents 140. Vaheri A, Carpen O, Heiska L, Helander


on HCl secretion and ultrastructure of TS, Jaaskelainen J, et al. 1997. The
piglet gastric oxyntic cells. Gastroenterol- ezrin protein family: membrane-cytoske-
ogy 83:595604 leton interactions and disease associa-
130. Mooseker MS, Tilney LG. 1975. Orga- tions. Curr. Opin. Cell Biol. 9:65966
nization of an actin filament-membrane 141. Gautreau A, Louvard D, Arpin M. 2002.
complex. Filament polarity and mem- ERM proteins and NF2 tumor suppressor:
brane attachment in the microvilli of in- the Yin and Yang of cortical actin orga-
testinal epithelial cells. J. Cell Biol. 67: nization and cell growth signaling. Curr.
72543 Opin. Cell Biol. 14:1049
131. Herman IM. 1993. Actin isoforms. Curr. 142. Bretscher A, Chambers D, Nguyen R,
Opin. Cell Biol. 5:4855 Reczek D. 2000. ERM-Merlin and EBP50
132. Cleveland DW, Lopata MA, MacDonald protein families in plasma membrane or-
RJ, Cowan NJ, Rutter WJ, Kirschner MW. ganization and function. Annu. Rev. Cell
1980. Number and evolutionary conserva- Dev. Biol. 16:11343
tion of - and -tubulin and cytoplasmic 143. Algrain M, Turunen O, Vaheri A, Lou-
- and -actin genes using specific cloned vard D, Arpin M. 1993. Ezrin contains
cDNA probes. Cell 20:95105 cytoskeleton and membrane binding do-
133. Chaponnier C, Gabbiani G. 1989. Gel- mains accounting for its proposed role
solin modulation in epithelial and stro- as a membrane-cytoskeletal linker. J. Cell
mal cells of mammary carcinoma. Am. J. Biol. 120:12939
Pathol. 134:597603 144. Yao X, Cheng L, Forte JG. 1996. Bio-
134. Yao X, Chaponnier C, Gabbiani G, Forte chemical characterization of ezrin-actin
JG. 1995. Polarized distribution of actin interaction. J. Biol. Chem. 271:722429
isoforms in gastric parietal cells. Mol. 145. Roy C, Martin M, Mangeat P. 1997. A
Biol. Cell 6:54157 dual involvement of the amino-terminal
135. Forte JG, Ly B, Rong Q, Ogihara S, domain of ezrin in F- and G-actin bind-
Ramilo M, et al. 1998. State of actin in ing. J. Biol. Chem. 272:2008895
gastric parietal cells. Am. J. Physiol. Cell 146. Urushidani T, Muto Y, Nagao T, Yao
Physiol. 274:C97C104 X, Forte JG. 1997. ME-3407, a new an-
136. Ammar DA, Nguyen PN, Forte JG. 2001. tiulcer agent, inhibits acid secretion by
14 Jan 2003 13:55 AR AR177-PH65-05.tex AR177-PH65-05.SGM LaTeX2e(2002/01/18) P1: fhd

CELL BIOLOGY OF ACID SECRETION 131

interfering with redistribution of H+-K+- 150. Potter DA, Tirnauer JS, Janssen R, Croall
ATPase. Am. J. Physiol. Gastrointest. DE, Hughes CN, et al. 1998. Calpain regu-
Liver Physiol. 272:G1122G34 lates actin remodeling during cell spread-
147. Chen J, Cohn JA, Mandel LJ. 1995. ing. J. Cell Biol. 141:64762
Dephosphorylation of ezrin as an early 151. Duman JG, Singh G, Lee GY, Machen TE,
event in renal microvillar breakdown and Forte JG. 2002. Ca2+ and Mg2+/ATP in-
anoxic injury. Proc. Natl. Acad. Sci. USA dependently trigger homotypic membrane
92:749599 fusion in gastric secretory membranes.
148. Dransfield DT, Bradford AJ, Smith J, Traffic 3:20317
Martin M, Roy C, et al. 1997. Ezrin is a 152. Soroka CJ, Chew CS, Hanzel DK, Smolka
cyclic AMP-dependent protein kinase an- A, Modlin IM, Goldenring JR. 1993.
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

choring protein. EMBO J. 16:3543 Characterization of membrane and cy-


149. Yao X, Thibodeau A, Forte JG. 1993. toskeletal compartments in cultured pari-
Ezrin-calpain I interactions in gastric pari- etal cells: immunofluorescence and con-
etal cells. Am. J. Physiol. Cell Physiol. focal microscopic examination. Eur. J.
265:C36C46 Cell Biol. 60:7687
by HINARI on 05/16/07. For personal use only.
22 Jan 2003 20:43 AR AR177-05-COLOR.tex AR177-05-COLOR.SGM LaTeX2e(2002/01/18) P1: GDL

Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org


by HINARI on 05/16/07. For personal use only.

Figure 1 Schematic model for regulated tubulovesicle cycling in the parietal cell.
In resting cells, the proton pump H,K-ATPase is sequestered in inactive cytoplasmic
tubulovesicles (TVs). Secretagogue stimulation triggers the translocation of TVs ulti-
mately leading to activation of proton pumping (solid arrows). Once the stimulus has
decayed, the proton pump is internalized to terminate acid secretion (dotted arrows).
The regulated TV cycle can be divided into following steps: (a) Docking. TVs that
contains H,K-ATPase and intrinsic factors dock at the active zone of the target mem-
brane. H,K-ATPase is inactive owing to low permeability to K+. Docking is defined as
the initial contact/interaction between vesicle membrane and target membrane medi-
ated by specific proteins, such as VAMP-2, syntaxins, SNAP-25, and exocyst protein
complex SEC6/8. Docking can occur between vesicles and apical plasma membrane
(heterotypic) and among vesicle membranes themselves (homotypic). (b) Priming.
After docking, TVs go through a maturation process that enables competency for
membrane fusion, possibly mediated by PKA-dependent phosphorylation and Ca2+-
dependent activation. (c) Fusion/proton pumping. Lipids of primed TVs are competent
to mix with those of the apical membrane allowing H,K-ATPase to partition into the
membrane. The proton pump begins active H+ transport while TV contents such as
intrinsic factor exocytose into the gland lumen. (d ) Endocytosis. After the stimulus
removal, the H,K-ATPase-rich regions of apical membrane are retrieved into the cy-
toplasm. The initial internalization is facilitated via clathrin-coated pits and dynamin.
(e) Endosome fusion. Coated TVs fuse with apical early endosome. This may be facil-
itated by small GTPases and their accessory proteins. ( f ) Budding. TVs are reformed
chiefly by budding from recycling endosomes. The newly formed TVs are either com-
petent for translocation and subsequent docking, or committed to a homotypic fusion
to gain competence. (g) Translocation. H,K-ATPase-containing TVs translocate back
to the active zone either by diffusion or by motor proteins.
22 Jan 2003 20:43 AR AR177-05-COLOR.tex AR177-05-COLOR.SGM LaTeX2e(2002/01/18) P1: GDL

Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org


by HINARI on 05/16/07. For personal use only.

Figure 2 Schematic model representing three stages in the membrane recruitment


process. (a) The trafficking and migration of cytoplasmic membranes containing cargo
transport proteins is facilitated by the cytoskeleton (microfilaments and microtubules)
possibly through resident motor proteins. (b) Effective recruitment can occur only
where cargo vesicles can recognize and dock on target plasma membranes via highly
specific interactive proteins (e.g., SNARE proteins), possibly regulated by small GT-
Pases such as the Rab proteins. (c) After the membranes are tightly docked, fusion can
occur only when microscopic regions of the two highly distinctive membrane interfaces
flow together to form the fusion pore. For biological membranes, the reaction probably
involves activation of hydrophobic protein(s) that spans the hydrophilic interface and
permits phospholipids to flow into a fusion pore. Phospholipases may also participate.
All of these processes are carefully regulated by receptor-mediated signaling events
involving protein kinases and phosphatases.
P1: FDS
January 17, 2003 11:23 Annual Reviews AR177-FM

Annual Review of Physiology,


Volume 65, 2003

CONTENTS
FrontispieceJean D. Wilson xiv
PERSPECTIVES, Joseph F. Hoffman, Editor
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

A Double Life: Academic Physician and Androgen Physiologist,


Jean D. Wilson 1
CARDIOVASCULAR PHYSIOLOGY, Jeffrey Robbins, Section Editor
Lipid Receptors in Cardiovascular Development, Nick Osborne
by HINARI on 05/16/07. For personal use only.

and Didier Y.R. Stainier 23


Cardiac Hypertrophy: The Good, the Bad, and the Ugly, N. Frey
and E.N. Olson 45
Stress-Activated Cytokines and the Heart: From Adaptation to
Maladaptation, Douglas L. Mann 81
CELL PHYSIOLOGY, Paul De Weer, Section Editor
Cell Biology of Acid Secretion by the Parietal Cell, Xuebiao Yao
and John G. Forte 103
Permeation and Selectivity in Calcium Channels, William A. Sather
and Edwin W. McCleskey 133
Processive and Nonprocessive Models of Kinesin Movement,
Sharyn A. Endow and Douglas S. Barker 161
COMPARATIVE PHYSIOLOGY, George N. Somero, Section Editor
Origin and Consequences of Mitochondrial Variation in Vertebrate
Muscle, Christopher D. Moyes and David A. Hood 177
Functional Genomics and the Comparative Physiology of Hypoxia,
Frank L. Powell 203
Application of Microarray Technology in Environmental
and Comparative Physiology, Andrew Y. Gracey and
Andrew R. Cossins 231
ENDOCRINOLOGY, Bert W. OMalley, Section Editor
Nuclear Receptors and the Control of Metabolism,
Gordon A. Francis, Elisabeth Fayard, Frederic Picard, and
Johan Auwerx 261

vii
P1: FDS
January 17, 2003 11:23 Annual Reviews AR177-FM

viii CONTENTS

Insulin Receptor Knockout Mice, Tadahiro Kitamura, C. Ronald Kahn,


and Domenico Accili 313
The Physiology of Cellular Liporegulation, Roger H. Unger 333
GASTROINTESTINAL PHYSIOLOGY, John Williams, Section Editor
The Gastric Biology of Helicobacter pylori, George Sachs,
David L. Weeks, Klaus Melchers, and David R. Scott 349
Physiology of Gastric Enterochromaffin-Like Cells, Christian Prinz,
Robert Zanner, and Manfred Gratzl 371
Insights into the Regulation of Gastric Acid Secretion Through Analysis of
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

Genetically Engineered Mice, Linda C. Samuelson and Karen L. Hinkle 383


NEUROPHYSIOLOGY, Richard Aldrich, Section Editor
In Vivo NMR Studies of the Glutamate Neurotransmitter Flux
and Neuroenergetics: Implications for Brain Function,
by HINARI on 05/16/07. For personal use only.

Douglas L. Rothman, Kevin L. Behar, Fahmeed Hyder,


and Robert G. Shulman 401
Transducing Touch in Caenorhabditis elegans, Miriam B. Goodman
and Erich M. Schwarz 429
Hyperpolarization-Activated Cation Currents: From Molecules
to Physiological Function, Richard B. Robinson and
Steven A. Siegelbaum 453
RENAL AND ELECTROLYTE PHYSIOLOGY, Steven C. Hebert, Section Editor
Macula Densa Cell Signaling, P. Darwin Bell, Jean Yves Lapointe,
and Janos Peti-Peterdi 481
Paracrine Factors in Tubuloglomerular Feedback: Adenosine, ATP,
and Nitric Oxide, Jurgen Schnermann and David Z. Levine 501
Regulation of Na/Pi Transporter in the Proximal Tubule,
Heini Murer, Nati Hernando, Ian Forster, and Jurg Biber 531
Mammalian Urea Transporters, Jeff M. Sands 543
Terminal Differentiation of Intercalated Cells: The Role of Hensin,
Qais Al-Awqati 567
RESPIRATORY PHYSIOLOGY, Carole R. Mendelson, Section Editor
Current Status of Gene Therapy for Inherited Lung Diseases,
Ryan R. Driskell and John F. Engelhardt 585
The Role of Exogenous Surfactant in the Treatment of Acute Lung
Injury, James F. Lewis and Ruud Veldhuizen 613
Second Messenger Pathways in Pulmonary Host Defense,
Martha M. Monick and Gary W. Hunninghake 643
P1: FDS
January 17, 2003 11:23 Annual Reviews AR177-FM

CONTENTS ix

Alveolar Type I Cells: Molecular Phenotype and Development,


Mary C. Williams 669
SPECIAL TOPIC: LIPID RECEPTOR PROCESSES, Donald W. Hilgemann,
Special Topic Editor
Getting Ready for the Decade of the Lipids, Donald W. Hilgemann 697
Aminophospholipid Asymmetry: A Matter of Life and Death,
Krishnakumar Balasubramanian and Alan J. Schroit 701
Regulation of TRP Channels Via Lipid Second Messengers,
Roger C. Hardie 735
Annu. Rev. Physiol. 2003.65:103-131. Downloaded from arjournals.annualreviews.org

Phosphoinositide Regulation of the Actin Cytoskeleton,


Helen L. Yin and Paul A. Janmey 761
Dynamics of Phosphoinositides in Membrane Retrieval and
Insertion, Michael P. Czech 791
by HINARI on 05/16/07. For personal use only.

SPECIAL TOPIC: MEMBRANE PROTEIN STRUCTURE, H. Ronald Kaback,


Special Topic Editor
Structure and Mechanism of Na,K-ATPase: Functional Sites
and Their Interactions, Peter L. Jorgensen, Kjell O. Hakansson,
and Steven J. Karlish 817
G Protein-Coupled Receptor Rhodopsin: A Prospectus,
Slawomir Filipek, Ronald E. Stenkamp, David C. Teller, and
Krzysztof Palczewski 851

INDEXES
Subject Index 881
Cumulative Index of Contributing Authors, Volumes 6165 921
Cumulative Index of Chapter Titles, Volumes 6165 925

ERRATA
An online log of corrections to Annual Review of Physiology chapters
may be found at http://physiol.annualreviews.org/errata.shtml

Anda mungkin juga menyukai