Sie sind auf Seite 1von 81

Physiol Rev 93: 189 268, 2013

doi:10.1152/physrev.00015.2012

GASTRIC ACID, CALCIUM ABSORPTION, AND


THEIR IMPACT ON BONE HEALTH
Sascha Kopic and John P. Geibel
Departments of Surgery and Cellular and Molecular Physiology, Yale School of Medicine,
New Haven, Connecticut
Kopic S, Geibel JP. Gastric Acid, Calcium Absorption, and Their Impact on Bone
Health. Physiol Rev 93:189 268, 2013; doi:10.1152/physrev.00015.2012.Calcium balance is essential for a multitude of physiological processes, ranging from cell
signaling to maintenance of bone health. Adequate intestinal absorption of calcium is a
major factor for maintaining systemic calcium homeostasis. Recent observations indicate that a reduction of gastric acidity may impair effective calcium uptake through the intestine.
This article reviews the physiology of gastric acid secretion, intestinal calcium absorption, and their
respective neuroendocrine regulation and explores the physiological basis of a potential link between these individual systems.

INTRODUCTION
GASTRIC ACID SECRETION
INTESTINAL CALCIUM ABSORPTION
REGULATION OF CALCIUM HOMEOSTASIS
THE STOMACH AND CALCIUM
CONCLUSIONS

189
189
203
212
231
238

I. INTRODUCTION
The average adult human body contains 1.6% calcium,
which relates to 1,120 g in a 70-kg individual (743). Ninetynine percent of the calcium is stored in bone and teeth and
is therefore inaccessible to most physiological processes
(743). Although the amount of the immediately accessible
11 g (1%) of calcium may seem miniscule, this fraction
represents a pivotal constituent of our body. It serves a
broad diversity of roles, which range from intracellular signaling and maintenance of membrane integrity to muscle
contraction and neuronal transmission.
To allow for these calcium-dependent processes to function, our body undertakes extensive measures to keep the
intracellular and extracellular calcium concentrations and
the gradient between these two compartments stable. The
extracellular calcium concentration is typically clamped at
1.1 mM, whereas the intracellular environment is kept at
a 10,000 times lower concentration. In consequence, relatively small disturbances in calcium homeostasis can lead to
severe symptoms, such as cardiac arrhythmias or cognitive
dysfunctions. To maintain eucalcemia, our body is therefore tightly regulating the balance between calcium absorption by the intestine and calcium excretion by the kidney. In
addition, calcium is deposited in or extracted from bone,
which serves as a dynamic calcium reservoir. These three
organ systems, i.e., the intestine, the kidney, and bone, are
precisely controlled by a complex endocrine network,

which primarily consists of the calcitropic hormones: 1,25dihydroxyvitamin D [1,25(OH)2-vitamin D], parathyroid
hormone (PTH), and calcitonin.
This review mainly focuses on the question as to how calcium
enters the body through the intestine and how this mechanism
is regulated via the endocrine system. Furthermore, the process
of gastric acid secretion as related to calcium homeostasis will
be reviewed in detail. This may seem surprising, as gastric acid
secretion and intestinal calcium absorption are two distinct
physiological processes, which on first examination may not
seem to be interdependent. However, recent clinical studies
suggest that there may be a relationship between reduced gastric acid secretion and increased risk for sustaining bone fractures, which asks the question whether we need gastric acid to
absorb calcium efficiently through the intestine, or whether the
stomach exerts endocrine functions that impact bone health.
Indeed, it has been put forward several decades ago that gastric acid solubilizes calcium that is then complexed with other
dietary constituents, thereby allowing for a more efficient absorption in the intestine (18, 520, 699, 797). Furthermore, it is
long known that a partial or complete resection of the stomach
results in decreased bone density, also leading to fractures (58,
305, 732, 876). The stomach, the intestine, and bone are therefore functionally more intertwined than one may initially assume. This review will independently analyze the processes of
gastric acid secretion, intestinal calcium absorption, and their
respective neuroendocrine control and will conclude with a
critical attempt at illustrating where these two seemingly independent organ systems intersect in terms of calcium homeostasis and bone health.

II. GASTRIC ACID SECRETION


The stomach is a unique organ that fulfills multiple roles.
The main function of the gastric mucosa is to secrete con-

0031-9333/13 Copyright 2013 the American Physiological Society

189

Downloaded from on December 8, 2014

I.
II.
III.
IV.
V.
VI.

SASCHA KOPIC AND JOHN P. GEIBEL


centrated hydrochloric acid, which provides a chemical barrier against ingested pathogens and aids in the digestion of
foodstuffs. To achieve these functions, the gastric gland
contains specialized cells that pump protons into the gastric
lumen in an effort to acidify the contents of the stomach.
These cells are known as parietal cells, or oxyntic cells.
Since concentrated acid is a noxious substance, the gastric
mucosa has to undertake extensive measures to protect itself from tissue injury. The protection is accomplished by
secreting mucus from mucus neck cells, but also by tightly
regulating the secretion of acid (see sect. IIB). A variety of
specialized endocrine cells in the gastric mucosa are involved in the regulation of gastric acid secretion. A perturbation of either protective mechanism can lead to severe
tissue damage, resulting in gastric ulcers. This section discusses the process of how gastric acid is secreted by reviewing the molecular mechanism underlying acid secretion in
the parietal cell and its neuroendocrine regulation.

A. Apical Ion Transport in the Parietal Cell

1. H-K-ATPase
The gastric H-K-ATPase belongs to the
family of P2-type ATPases, which also includes the ubiquitous Na-K-ATPase and the sarcoplasmic reticulum
Ca2-ATPase (SERCA). As the name implies, it exchanges
one intracellular hydrogen ion for one extracellular potassium ion at the expense of ATP. ATP is provided to the
pump by a large network of mitochondria, which occupy
up to 40% of the cell volume, making the parietal cell one of
the most mitochondria-rich cells in the body (292). In the
A) STRUCTURE.

Apical

Basolateral
PPIs
APAs

H+

SSTR

SST

H2

Hist

CCK2

Gast

M3

ACh

K+

cAMP

K+
KCNQ1
Kir

Ca2+
Cl
CFTR
CIC-2?
SLC26A9
Parietal cell

FIGURE 1. Parietal cell model. The gastric parietal cell is equipped with apical ion transport mechanisms that
allow for the secretion of concentrated hydrochloric acid. Activation of basolateral secretagogue receptors
mainly leads to an increase in either cAMP (histamine) or calcium (acetylcholine, gastrin), causing apical
insertion and activation of the H-K-ATPase. Somatostatin reduces intracellular cAMP levels. ACh, acetylcholine; APAs, acid pump antagonists; Gast, gastrin; Hist, histamine; PPIs, proton pump inhibitors; SST,
somatostatin.

190

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

The gastric parietal cell is responsible for acidifying the


stomach by secreting concentrated acid. Gastric acid secre-

tion depends on the apical extrusion of three ions. Protons


are pumped into the gastric lumen by a proton pump, the
gastric H-K-ATPase, to acidify the gastric content to a
pH of as low as 1. Chloride is secreted via apical chloride
channels to ensure formation of HCl and to provide the
counter-ion conductance to protons. Lastly, potassium
leaves the parietal cell apically in a recycling mechanism,
thereby fueling reciprocal proton transport by the H-KATPase (FIGURE 1). It has been demonstrated in numerous
investigations that disruption of one of these ion transport
mechanism renders the parietal cell incapable of secreting
gastric acid (705, 820, 1013, 1029).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH

In the resting parietal cell, H-K-ATPase is


stored in tubulovesicles throughout the cell (292). Following neuronal or hormonal stimulation (see sect. IIB), these
vesicles are postulated to fuse with the apical pole, which is
characterized by multiple microvilli-lined membrane invaginations, the so-called secretory canaliculi (292). This
distinct apical morphology of the parietal cells maximizes
cell surface and thereby allows for insertion of a high number of proton pumps per cell following stimulation. The
changes in membrane morphology and insertion of H-KATPase are extremely dynamic to ensure fine regulation of
gastric acid secretion (973). H-K-ATPase containing
tubulovesicle fusion relies on SNARE complex formation. In particular, the SNARE proteins syntaxin 3/7/12/
13, VAMP2/8, and SNAP-25 were implicated to be candidates mediating this process (548 550, 624). The functional significance of these proteins was, for example,
demonstrated in primary rabbit parietal cell cultures expressing a SNAP-25 mutation, which was shown to reduce
their capacity to secrete gastric acid (548).
B) TRAFFICKING.

Apart from SNARE proteins, the small GTPases of the rab


family (rab2/11a/25/27b) are involved in the regulation of
H-K-ATPase vesicle trafficking (147, 293, 386, 387,
1049, 1070). Functional data especially substantiate the
importance of rab11a and rab27b. In parallel to SNAP-25
defective cells, parietal cells transfected with a rab11a and
rab27b mutant secrete acid less effectively (293, 1049).
After stimulation, in the off-phase of gastric acid secretion,
H-K-ATPase has to be retrieved from the plasma mem-

brane for recycling (336). It is plausible that the initial step


of this process relies on the formation of clathrin-coated pits
and subsequent vesicle budding. Indeed, clathrin was identified fairly early on H-K-ATPase containing tubulovesicles, although a functional role was not demonstrated
(813). One of the multiple clathrin binding proteins is Huntingtin interacting protein 1 related (Hip1r) which aids in
vesicle formation and membrane trafficking (309). It is
strongly expressed in parietal cells, especially in the vicinity
of secretory canaliculi (522). Functionally, Hip1r-deficient
animals present with a decreased number of parietal cells,
loss of tubulovesicles, and decreased acid output (522,
561).
2. Chloride secretion
Apical chloride secretion provides the second component
for the formation of concentrated HCl and maintains overall electroneutrality during acid secretion. The importance
of chloride efflux for the process of gastric acid secretion
has been established in the 1980s. Patch-clamp measurements demonstrated the presence of chloride conductance
on the apical pole of the parietal cell in Necturus, the human
parietal cell line HGT-1, and rabbit parietal cells (259, 935,
940). All reports demonstrated a sensitivity of the chloride
current to cAMP or histamine, which is a common second
messenger promoting acid secretion or a direct acid secretagogue, respectively (259, 935, 940). Simple flux measurements in isolated parietal cell vesicles had indicated the
presence of a chloride conductance pathway even earlier
(232, 895, 1169). In these early experiments, inhibition of
chloride flux with chloride channel blockers also abolished
proton transport which underlines the necessity of intact
chloride secretion for acid secretion to take place (232, 895,
1169). However, the molecular identity of the chloride
pathway remained elusive. Today, at least three candidates
have been put forward as potential mediators of apical chloride secretion in the parietal cell: the cystic fibrosis conductance regulator (CFTR), chloride channel protein 2 (ClC-2),
and solute carrier 26 A 9 (SLC26A9) (FIGURE 1).
A) CFTR. CFTR represents a common apical chloride conduc-

tance pathway in a broad variety of epithelia, such as the


airways, intestine, and pancreas. Its mutation is responsible
for the most widespread inherited disease, namely, cystic
fibrosis (CF), which results in increased mortality due to
secretory defects and concomitant infections. The presence
of CFTR has been confirmed in gastric mucosa by in situ
hybridization, albeit at low quantities (1044). Nevertheless,
functional measurements in isolated gastric glands demonstrated a decreased acid secretory capacity in animals carrying the most common mutation responsible for CF
(F508) (1013). Furthermore, acid secretion was reduced
in wild-type animals when a specific CFTR inhibitor was
applied (1013). Although these observations may suggest a
direct involvement of CFTR in the process of chloride secretion, it is plausible that CFTR rather has a regulatory

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

191

Downloaded from on December 8, 2014

process of proton extrusion, the H-K-ATPase can overcome a massive acid gradient of 6 pH units, which is necessary to achieve sufficient gastric acidification. The pump
itself is a heterodimer, consisting of a subunit and a
subunit, while the individual pumps assemble as ()4 tetramers on the parietal cell surface (1). The subunit consists of 10 transmembrane domains and contains the catalytic site, which mediates ion exchange. The subunit stabilizes the subunit and is heavily glycosylated (41, 1105).
Mutational analysis of the glycosylated asparagine residues
suggests that these sites are critical for adequate membrane
delivery of the entire pump (41, 1105). Furthermore, the
subunit prevents a reversal of ion transport by a ratchetlike mechanism, which allows H-K-ATPase to pump
against the imposed high proton gradient (4, 294). Both
subunits share a significant degree of homology to Na-KATPase (697, 1012). This close relationship to other P2type ATPase has historically been exploited for homology
modeling of H-K-ATPase based on the crystal structure
of SERCA, which had been acquired in several conformational states (762, 815, 1092, 1093). Recently, however,
direct structural information on H-K-ATPase has been
obtained by electron crystallography, also in the presence of
the acid pump antagonist SCH28080 (2 4).

SASCHA KOPIC AND JOHN P. GEIBEL


effect on H-K-ATPase (1013). In other tissues, CFTR
can interact with a variety of ion transport proteins, such as
NHE, forming regulatory complexes, making an interaction with H-K-ATPase plausible (1013).
B) CLC-2. ClC-2 has been proposed as an alternative chloride
secretion pathway to CFTR in other epithelia, such as the
lung and intestine (207, 404, 675, 766). ClC-2 has been
cloned from rabbit gastric mucosa, which led to the hypothesis that the channel may also be involved in acid secretion
(706). However, follow-up investigations revealed that the
role of ClC-2 is much less clear. The studies revealed controversial results regarding the channels expression in the
gastric mucosa (488, 706, 1001). While the initial observations reported mRNA and cDNA expression in rabbit gastric mucosa, no protein could be detected in human and rat
gastric glands (488, 706, 1001). The importance of ClC-2 in
the stomach has further been severely challenged by the
creation of a ClC-2 (/) animal model. Although ClC-2deficient animals present with a distinct phenotype characterized by testicular and retinal abnormalities, no defect in
acid secretion was observed (118).

3. Potassium recycling
Even before the identification of H-K-ATPase, it has
been observed that potassium is necessary for acid secretion
to take place (335). To prevent the luminal depletion of
potassium, which would impair proton pumping by H-

192

A) KCNQ1. KCNQ1 is a typical shaker-like six transmembrane spanning domain voltage-gated potassium channel
(1144). It was initially identified in the heart, where its
mutation can be responsible for cardiac arrhythmias
(1144). Yet, studies in KCNQ1 (/) animals revealed no
electrocardiographical abnormalities (641). Rather than
suffering from cardiac abnormalities, these animals surprisingly exhibited a distinct gastric phenotype with gastric hyperplasia, dilated gastric glands, vacuolated parietal cells,
hypochlorhydria, and hypergastrinemia (641). This observation led to the speculation that KCNQ1 may be the channel responsible for potassium recycling. Subsequently, immunohistochemical studies confirmed a colocalization of
the channel with H-K-ATPase, and acid secretion was
shown to be inhibited by pharmacological blockade (253,
391). Direct measurement of acid secretion in KCNQ1
(/) mice with modified Ussing chambers (pH stat) later
confirmed the initially observed hypochlorhydria (1029).
Interestingly, luminal substitution of potassium could rescue the acid secretory deficit, indicating that hypochlorhydria ensued from a true lack of apical potassium secretion
rather than a general morphological defect of the KCNQ1
(/) parietal cell (1029).

KCNQ1 is a peculiar channel in that it has a low conductance in acidic environments. In the context of the extreme
acidic milieu surrounding the parietal cell, this would impede its function as a potassium recycling pathway. To
circumvent this limitation, KCNQ1 attaches to a regulatory
subunit (KCNE2), which modulates the channels gating
properties and current amplitude (253, 391, 1087). Coassembly with KCNE2 activates KCNQ1 at acidic pH values
and thus facilitates the process of potassium secretion into
the gland lumen (391, 436). The importance of KCNE2 for
proper channel function is underlined by the observation
that KNCE2 (/) animals display a phenotype similar to
KCNQ1 (/) mice, i.e., hypochlorhydria, altered parietal
cell morphology, and hypergastrinemia (917).
B) KIR CHANNELS.

Apart from KCNQ1, several members of


the inward-rectifier potassium channel (Kir) family have
been proposed to be involved in gastric acid secretion, albeit

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

C) SLC26A9. Lastly, evidence suggests that chloride may leave


the apical pole via SLC26A9, a chloride-bicarbonate antiporter. Both SLC26A9 and an antiporter from the same
anion exchanger family (SLC26A6) have been detected in
the tubulovesicles of parietal cells (845, 1179, 1180). Concerning the functional involvement, the authors speculate
about two potential roles SLC26A9 may play in parietal cell
physiology. Being a chloride-bicarbonate exchanger, its activation would entail alkalinization of the gastric lumen
by bicarbonate efflux and simultaneous chloride uptake
(1180). Since this would neutralize H -K -ATPasemediated proton extrusion, it has been suggested that
SLC26A9 activates in the off-phase of acid secretion to
neutralize tubulovesicular pH during vesicle retrieval
(1180). Alternatively, SLC26A9 may function as a chloride
secretion pathway that contributes to acid secretion. This
hypothesis is based on the observation that SLC26A9 can
also exhibit the behavior of a bona fide chloride channel,
rather than an anion antiporter (88, 281). Undoubtedly,
further functional investigations are needed to delineate its
exact role in the parietal cell. Its genetic disruption, however, leads to a severely altered parietal cell morphology
that is characterized by dilation of gastric glands, loss of
tubulovesicles, and decreased acid output (1180). Although
these results do not answer whether SLC26A9 serves as an
apical chloride efflux pathway, they indicate that it may be
necessary for normal parietal cell function.

K-ATPase, potassium has to leak through potassium


channels or transporters into the gland lumen to ensure
adequate supply to H-K-ATPase (FIGURE 1). This process is referred to as potassium recycling. Early flux measurements in isolated H-K-ATPase containing parietal
cell vesicles had already indicated the presence of a large
potassium conductance during H-K-ATPase activity
(1169). The exact molecular identity of the potassium efflux
pathway is, however, under debate. The list of candidates
that have been put forward to be responsible for potassium
recycling during acid secretion is long and includes KCNQ1
(Kv7.1), KCNJ10 (Kir4.1), KCNJ15 (Kir4.2), KCNJ2
(Kir2.1.), and KCC4.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


the amount of functional evidence supporting a role of these
channels is smaller and the field is divided about the relative
contribution of each channel. Kir 2.1, 4.1, 4.2, and 7.1 were
all confirmed on an mRNA level in gastric mucosa (353,
431, 707). On a protein level, immunohistochemistry demonstrated colocalization of Kir 2.1, 4.1, and 4.2 with HK-ATPase (353, 431, 556, 707). Cell fractionation experiments further indicated trafficking of Kir 4.1 and 4.2 to the
cell surface, following parietal cell stimulation (431, 556).
A most recent observation monitored acid secretion in Kir
4.1 (/) mice (1028). Surprisingly, loss of Kir 4.1 results in
augmented rather than impaired acid secretion, accompanied by upregulated H-K-ATPase expression (1028).
This makes a contribution of Kir 4.1 to potassium recycling
highly unlikely. Instead, it has been proposed that the channel may balance excessive potassium loss through KCNQ1
and may be involved in membrane recycling (1028). In summary, more investigations will be necessary to clarify the
roles of the individual Kir channels.
C) KCC4.

B. Control of Acid Secretion


Gastric acid secretion is subjected to precise regulation. The
complex regulatory machinery that orchestrates the secretion
of gastric acid consists of hormonal (gastrin, somatostatin),
paracrine (histamine, somatostatin), and neuronal components (FIGURE 2). The need for this tight regulation is highlighted by conditions that lead to a hypersecretion of gastric
acid, such as Zollinger-Ellisson syndrome (ZES; gastrinoma).
Gastric hypersecretion can overcome the measures our body
undertakes to protect itself from the acid and thereby lead to
peptic ulcers. A fine on-demand regulation of acid secretion is
thus pivotal to ensure the balance between an adequately low
intragastric pH and tissue protection.
According to the well-established model of acid secretion,
the parietal cell is activated by neuronal input from the
vagus nerve, endocrine input from gastrin-producing G
cells, and paracrine input from histamine-producing enterochromaffin-like (ECL) cells (FIGURES 1 AND 2). The
distinct substances released by these cells, i.e., acetylcho-

1. Cholinergic stimulation/vagus nerve


Since the seminal experiments conducted by Pavlov on
dogs, we know that the mere prospect of food ingestion or
sham-feeding is sufficient to trigger the secretion of gastric
acid (833). This first of three phases of acid secretion is
called the cephalic phase and is mostly mediated through
the vagus nerve (595, 725, 910). Hence, before the advent
of pharmacological inhibitors, vagotomy has been an effective surgical procedure to control acid-related disorders
(301).
The parietal cell receives neuronal input from the vagus nerve
that is relayed via cholinergic postganglionic enteric fibers in
the enteric nervous system (ENS) (FIGURES 1 AND 2). In addition, the vagus nerve activates G-cells to release gastrin,
resulting in an indirect stimulation of the parietal cell. Direct cholinergic activation occurs mostly via muscarinic M3
receptors, which have been identified on the surface of the
parietal cell (507, 541, 846). The M3 receptor is a classic
seven-transmembrane domain GPCR. Predictably, knockout of M3 receptors leads to an impairment of gastric acid
secretion and compensatory hypergastrinemia due to negative feedback (9). Following acetylcholine binding, M3 receptor activation mostly causes an increase in intracellular
calcium concentrations (44, 1163). Calcium rises in response to PLC-mediated IP3 generation and subsequent mobilization from intracellular stores (190). The primary kinases activated by the M3 receptor are protein kinase C
(PKC) and calcium/calmodulin-dependent protein kinase II
(CaMKII) (136, 196, 314 316, 773, 774, 1095). While
activation of CaMKII has a clear stimulatory effect on acid
secretion, PKC has been reported to have dual effects, although reports of an inhibitory role predominate numerically (23, 73, 136, 196, 313, 314, 316, 597, 755, 773,
1095). It has been postulated that the expression of different PKC isoforms may account for this dichotomy (313,
314). Current evidence suggests that the PKC- isoform has
a suppressing effect by trans-inhibiting CaMKII activity,
whereas PKC- increases the baseline levels of intracellular
calcium, thereby sensitizing the parietal cell to subsequent

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

193

Downloaded from on December 8, 2014

Apart from being secreted through channels, potassium and chloride may exit the parietal cell through transporters. This alternative hypothesis is corroborated by a
recent observation of Fuji et al. (352). The group reported
that the K-2Cl cotransporter KCC4 coimmunoprecipitates
with H-K-ATPase in apical membrane fractions of parietal cells (352). Furthermore, flux measurements in H-KATPase containing vesicles showed decreased chloride and
proton transport under pharmacological blockade of
KCC4, suggesting a functional coupling of KCC4 to HK-ATPase (352). Although the hypothesis that both potassium and chloride leave the cell via a transporter is intriguing, the observation is, as of now, solitary and needs
further experimental validation.

line, histamine, and gastrin, directly or indirectly stimulate


the parietal cell by inducing insertion of H-K-ATPase at
the apical membrane and are thus commonly referred to as
acid secretagogues. The main inhibitor of parietal cell acid
secretion is somatostatin, which is secreted by the D-cells
of the gastric mucosa (FIGURES 1 AND 2). Because of the
complexity of the network that controls the release of
acid into the stomach, it has been historically challenging
to dissect the relative role of each individual regulatory
component. Without a doubt, knockout models have
greatly aided us in the last years to gain a more profound
understanding of this process, despite their limitations of
chronic compensation. The subsequent chapter aims to
summarize the key players in our canonical model of acid
regulation.

SASCHA KOPIC AND JOHN P. GEIBEL


Oxyntic mucosa
Lumenal

Antrum
Basolateral

D-cell

Lumenal

Somatostatin

Basolateral

D-cell

VPAC

PACAP
VIP

CCK1

CCK

ENS

Somatostatin

Low
lumenal
pH
ECL-cell
SSTR

SST
G-cell

PAC1

PACAP

CCK2

Gast

SSTR
ENS

CaSR
CaSR

ENS

Histamine
Calcium
Amino acids
Polyamines

PPIs
APAs

SST

Gastrin

ACh
GRP

CaSR

H,K-ATPase

H2

Hist
Circulation

K+
CCK2

Gast

M3

ACh
ENS

SSTR

SST

Parietal cell

Somatostatin
D-cell
FIGURE 2. Neuroendocrine regulation of gastric acid secretion. In addition to direct neuronal regulation, the
parietal cell receives paracrine signals from neighboring ECL- and D-cells. Gastrin is produced in the antral
mucosa of the stomach and reaches the oxyntic mucosa via the circulation (endocrine regulation). Gastrinmediated histamine release represents one of the major stimulatory pathways leading to the secretion of
gastric acid (gastrin-histamine axis). The secretion of gastrin is closely tied to intragastric pH (via somatostatin), thereby creating a negative-feedback loop. ACh, acetylcholine; APAs, acid pump antagonists; ENS, enteric
nervous system; Gast, gastrin; Hist, histamine; PPIs, proton pump inhibitors; SST, somatostatin.

stimulation (313, 314). Apart from PKC and CaMKII activation, cholinergic signaling activates parietal cell MAPKs,
which is partially a downstream effect of PKC activation
(771, 1039, 1062, 1063). MAPK activation seems to have a
biphasic effect on acid secretion (acute inhibition and
chronic augmentation) and also serves as a mediator of
trophic responses in the parietal cell. For example, pro-

194

longed MAPK activation (72h) has been shown to serve as


a maturation and differentiation signal leading to a transformation of parietal cell morphology in vitro (1039). The
change in morphology is accompanied by a downregulation
of H-K-ATPase gene expression (1039). As of now, it is
challenging to put these findings into a physiological perspective.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

H+

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


In addition to M3 receptors, M1 receptors have also been
implicated to play a role in the process of acid secretion.
This hypothesis was derived from the observation that the
M1 receptor is expressed in gastric mucosa and that its
blocker pirenzepine can inhibit gastric acid secretion (29,
466). Most evidence pointed to an expression of M1 on
ECL-cells, where it was speculated to regulate the release of
histamine (437, 507). More recent findings somewhat surprisingly report that pirenzepine also suppresses acid secretion in M1-deficient animals. Furthermore, these animals
show a normal phenotype in terms of acid output (10).
These observations question both the involvement of M1
receptors in acid secretion and the specificity of pirenzepine.
Lastly, knockout studies point towards a contribution of the
M5 receptor to the regulation of acid secretion, as its deletion
correlates with decreased acid output (10). Yet, M5 receptor
mRNA could only be detected in whole stomach homogenates, but not in gastric mucosa per se, making its localization
to the submucosal enteric plexus more likely (10).
2. Gastrin/G-cell

A) SYNTHESIS.

The gastrin cDNA encodes a 101-amino acid


pre-pro-hormone that undergoes extensive posttranslational processing (113, 519, 551, 552, 1161). In brief, the
pre-pro-hormone is first cleaved NH2-terminally to create
progastrin and then truncated to two main core proteins,
G17 and G34, which can exist in glycine extended (G17Gly; G34-Gly) or terminally amidated (G17-NH2, G34NH2) forms. Furthermore, a fraction of progastrin (47%
in humans) is sulfated at Tyr66 in the course of its passage
through the Golgi apparatus, thereby giving rise to sulfated
and nonsulfated isoforms of gastrin (22). Sulfation has no
influence on the acid secretory response, as the affinity to
the gastrin receptor remains unchanged (399, 596). G17NH2 is the main circulating form that mediates the secretory effects of gastrin. Although the glycine-extended forms
have a low affinity towards the gastrin receptor (CCK2) and
thus play no role in gastric acid secretion (they are four to
five orders of magnitude less potent in inducing acid secretion), it is still important to acknowledge their existence
(178, 722). First, they serve as substrates for the synthesis of
amidated gastrin and are cosecreted with gastrin by the
G-cells (1040, 1051). Second, they potentiate the acid se-

B) REGULATION OF RELEASE. Gastrin is released by the G-cell in


response to a variety of stimuli of different origin. Direct
neuronal stimulation of the G-cell occurs via ACh and gastrin releasing peptide (GRP), which are released by postganglionic neurons of the enteric nervous system. The postganglionic fibers themselves receive input from the efferent
fraction of the vagus nerve (86, 485). On the other hand,
food-related signals, such as calcium, amino acids, and
amines, can also directly trigger gastrin secretion (FIGURE 2)
(257). The secretory stimuli culminate in an increase in
intracellular calcium concentrations, leading to vesicle
fusion and gastrin secretion. The main inhibitory signal
for gastrin secretion is somatostatin, which reaches the
G-cells in a paracrine fashion from neighboring D-cells
(632, 975).

With regard to the neuronal control of gastrin secretion, it is


generally thought that vagal stimulation increases the release of gastrin, although some conflicting evidence exists
(310, 694). Latest experiments that assessed local gastrin
concentrations utilizing microdialysis, however, clearly
show an increase in gastrin levels following acute electrical
vagal stimulation (310). The vagus nerve then synapses on
neurons of the ENS, which are thought to release either the
neurotransmitter ACh or GRP on a G-cell, leading to secretion of gastrin (295, 635, 694, 960, 978). It should be noted
that a recent investigation failed to observe increased gastrin levels, following exogenous GRP administration in humans (456). Yet, GRP itself serves as a clear acid secretagogue, although potentially not via gastrin (456). Whether
these conflicting observations are attributable to species differences (most earlier observations utilized rodent models)
remains to be elucidated. The ENS is also thought to mediate parietal and G-cell activation in response to mechanical
distension of the stomach (455, 962, 977). The neurohormonal response to gastric stretch is an integral part of the
gastric phase of acid secretion. Closer examination, however, reveals that the reports are very conflicting in that it is
not clear whether a pure mechanical distension stimulates
or inhibits gastrin release (455, 664, 803, 962, 977). A
biphasic model characterized by initial inhibition of gastrin
secretion under low volumes followed by stimulation under
high volumes has been suggested, but awaits further confirmation (977).

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

195

Downloaded from on December 8, 2014

Gastrin has been discovered in 1906 by John S. Edkins, who


injected gastric extracts of pig and cat stomachs into the
jugular vein of cats and observed a subsequent increase in
acid secretion (298). Gastrin is a peptide hormone that is
produced in specialized G-cells, located in the antral section
of the stomach (FIGURE 2) and endocrine cells in the duodenum, small intestine, colon, pancreas, testis, and pituitary. It is the main mediator of the so-called gastric phase of
acid secretion, which initiates when the ingested food enters
the stomach. The gastric phase accounts for the majority of
the acid secretory response of the stomach.

cretory response to amidated gastrin, although they have no


intrinsic ability to induce acid secretion (178). Third, progastrin and glycine extended gastrins were shown to act as
a proliferative signal, especially in the colon (20, 482, 994,
1145). This is also of pathophysiological relevance as both
forms can promote cancer growth by presumably inhibiting
apoptosis and inducing angiogenesis (71, 87, 900). For example, it was shown that overexpression of progastrin in
mice is a predisposing factor for the development of colorectal or bronchoalveolar cancers (587, 1017).

SASCHA KOPIC AND JOHN P. GEIBEL

The plasma levels of gastrin are closely tied to the intragastric pH. Low intraluminal pH is a potent inhibitor of gastrin
release, which serves as a negative-feedback mechanism to
impede an overproduction of acid. Conversely, a more alkali intragastric pH induces the secretion of gastrin, which
accounts for the commonly observed hypergastrinemia in
states of acid suppression, such as during proton pump
inhibitor (PPI) therapy. The pH dependency of serum gastrin levels is mainly relayed via somatostatin, as acid directly stimulates somatostatin release (see sect. IIB4). Somatostatin, released by neighboring antral D-cells, in turn acts
as the main inhibitor of gastrin secretion (FIGURE 2). The
physical proximity to G-cells allows for a fine paracrine
regulation of gastrin release. Although it is generally accepted that intragastric pH mostly modulates local somatostatin levels, the G-cell may also directly sense intragastric
pH via CaSR. CaSR is acid sensitive, and it has been shown
that isolated rat G-cells secrete less gastrin when the extracellular pH is dropped from 7.4 to 5.5 (569). However,
more investigations are needed to substantiate this evidence. Furthermore, gastrin release is also inhibited by neuronal regulation by the ENS. The neurotransmitter galanin
has been demonstrated to exert a direct inhibitory effect on
isolated G-cells (695, 961).
C) CELLULAR EFFECTS. Following secretion, gastrin enters the
bloodstream and acts on its target cells in an endocrine

196

fashion. Its half-life is determined by its rate of elimination


from the plasma which mainly occurs by metabolism in the
kidney, gut, and brain (419, 420). The importance of renal
elimination is corroborated by the observation that patients
with renal failure present with higher plasma gastrin levels
(818, 1075).
The two primary target cells of gastrin are the histaminesecreting ECL cell and the parietal cell. Gastrin exerts its
functions via binding to the cholecystokinin receptor type 2
(CCK2), a seven transmembrane domain G protein-coupled
receptor, which is expressed on mature parietal and ECL
cells, but also on gastric stem cells (560, 596, 608, 769, 772,
904). On the ECL cell, gastrin binding causes the release of
histamine, which in turn stimulates the parietal cell in a
paracrine fashion (FIGURE 2) (see sect. IIB3) (412). This
activation cascade is commonly referred to as the gastrinhistamine axis. Evidence for a direct, i.e., nonhistaminerelayed, activation of H-K-ATPase in the parietal cell by
gastrin exists, but is far less substantiated (459, 1024,
1025). Gastrin may sensitize the parietal cell to subsequent
secretagogue stimulation, rather than acting as a bona fide
secretagogue itself. Canonically it is widely accepted that
gastrin exerts its physiological effects mostly via activation
of ECL cells (24, 1131). Knock-out of gastrin leads to a
severe impairment of basal and stimulated acid secretion
(179, 347). Apart from stimulating acid secretion, gastrin
serves as a pivotal proliferative signal for the gastric mucosa
in general (60, 410, 534, 631, 816). It is commonly observed that elevated plasma gastrin levels lead to substantial
mucosal proliferation (60, 410, 534, 631, 816). This phenomenon has been extensively described in various knockout animals suffering from hypochlorhydria and concomitant hypergastrinemia, but also in patients with ZES (39,
515, 584, 1066). The source of mucosal cell proliferation is
progenitor cells located in the isthmus region of the gastric
gland (545). Expression of the CCK2 has been confirmed on
several gastric progenitor cells (560, 769). Furthermore,
gastrin has been shown to stimulate cell migration from the
progenitor region along the gastric gland axis (578). Mucosal hyperplasia thus ensues most likely via a direct activation of precursor cells by gastrin. Although hypergastrinemia causes a generalized mucosal hyperplasia, ECL cells
seem to be particularly regulated by gastrin, as their relative
fraction compared with other mucosal cells increases under
prolonged gastrin exposure (60, 410, 631). Conversely, the
absence of the CCK2 almost entirely eliminates mature
ECL-cells from the gastric mucosa (177, 622). [Somewhat
surprisingly this does not occur when gastrin itself is
knocked out (179, 347).] It should be noted that the M3
receptor seems to be necessary as a cofactor mediating the
trophic effects of gastrin, as its absence is associated with a
normal mucosal phenotype despite elevated serum gastrin
levels (see above) (9). The mechanism underlying this interdependency between gastrin and the M3 receptor is as of
now elusive. The cholinergic and gastrin systems also seem

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Dietary components, such as amino acids and calcium, can


directly promote the secretion of gastrin and can thus sustain acid secretion in the gastric and intestinal phase as
digestion progresses (652, 1074). A rise in serum calcium
concentrations evokes a similar effect. The correlation between calcium and gastrin is discussed in a separate section
(see sect. VD2). It has been unclear for a long time as to how
these dietary components activate the G-cell. An involvement of the ENS has been proposed as the most likely explanation in the past. More recent observations, however,
strongly indicate that the calcium-sensing receptor (CaSR)
represents the molecular link between luminal dietary constituents and G-cell activation (325). The CaSR and its role
in the stomach are discussed separately and shall only be
summarized at this point (FIGURE 8) (see sect. IVD). First,
the same dietary components, i.e., amino acids, amines, and
calcium, which have all been shown to trigger gastrin release also function as activators of CaSR (652, 1074). Second, CaSR is expressed on the apical and basolateral side of
the G-cell, which allows it to act as nutrient sensor both in
the gastric lumen and the circulation (142, 182, 886).
Third, direct activation of the CaSR is known to stimulate
acid secretion (145, 291, 373). Finally, and most importantly, CaSR (/) animals lack the gastrin secretory response to intraluminal instillation of peptone, calcium, and
phenylalanine (325). In light of this evidence, it is highly
likely that CaSR is the long elusive luminal nutrient sensor
that regulates the secretion of gastrin from the G-cell.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


to be intertwined with regard to acid secretion. In the absence of the CCK2 receptor, the parietal cells acid secretory
response to the secretagogue carbachol (ACh analog) is
abolished, while the response to histamine remains intact
(543). Again, one can only speculate about the molecular
basis of this interaction.
In conclusion, gastrin is the most important activator of
acid secretion in the stomach. The role of gastrin, and especially its glycine extended forms, has evolved beyond being
a mere acid secretagogue to being an important global regulator of cell growth and differentiation. Furthermore, the
regulation of gastrin by the levels of plasma calcium provokes the question as to whether gastrin itself in turn has an
impact on global calcium homeostasis. A subsequent section makes an attempt at addressing this question (see sect.
VD2).
3. Histamine/ECL cell

A) SYNTHESIS AND REGULATION OF RELEASE.

Histamine is the
effector of the gastrin-histamine axis and directly stimulates
the parietal cell to secrete hydrochloric acid (FIGURE 2).
Histamine is derived from the amino acid histidine, which is
enzymatically converted to histamine by L-histidine decarboxylase (HDC) (957). The effects of genetic HDC deletion
are predictably severe: animals lacking HDC have a low
basal acid output that does not respond to exogenous administration of gastrin (1066).
Histamine is stored in secretory granules of the ECL cell and
is released into the surrounding milieu in response to stimulation by gastrin and neuronal signals. Stimulation by gastrin occurs via activation of its GPCR CCK2 (772, 904).
Gastrin affects the ECL cell in multiple ways. First, gastrin
exposure increases the levels of HDC expression by enhancing its transcription and inhibiting its degradation, to allow
for increased synthesis of histamine (268, 331). The molecular mechanism underlying increased HDC transcription is

Apart from gastrin, ECL cells are stimulated by pituitary


adenylate cyclase activating polypeptide (PACAP), which is
a neuropeptide expressed in the ENS of the gastric mucosa
(737, 1054). PACAP has homology to vasoactive intestinal
polypeptide (VIP) and binds to a distinct receptor (PAC-1)
on the ECL cell (1207, 1208). Binding of PACAP to PAC-1
induces release of histamine (672, 798, 944, 1207). Similar
results have been obtained with VIP, which is attributable
to partial agonism at PAC-1 (798, 941). Historically, investigations yielded controversial results with regard to the
effects of exogenously administrated PACAP on acid secretion. Both an inhibition and stimulation of acid secretion
following PACAP injection are reported (760, 862, 944,
1207). This discrepancy is most likely attributable to the
fact that PACAP can also act as an agonist of the VIP receptor (VPAC) on the somatostatin-secreting D-cell, leading to a concomitant suppression of acid secretion by somatostatin release (1207). Indeed, if an anti-somatostatin antibody is injected into rats simultaneously with PACAP,
acid secretion is elevated threefold from baseline (compared
with 1.5-fold in the absence of an anti somatostatin antibody) (1207). Evidence points to the fact that the PACAP-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

197

Downloaded from on December 8, 2014

Histamine has been discovered as early as 1910 by Dale,


Barger and Laidlow in extracts of ergot fungi (63, 237). In
1920, Popielski for the first time described its effect on the
secretion of gastric acid (866). He observed that subcutaneous administration of histamine resulted in increased acid
secretion (866). Furthermore, he concluded that this effect
was independent of the vagus nerve, as secretion still took
place after vagotomy and administration of atropine. This
led Popielski to postulate that histamine exerts its effects
directly on the level of the gastric gland (866). The hypothesis that histamine acts in a paracrine fashion on parietal
cells and that its release is regulated by the levels of gastrin
has been put forward for the first time by Emmelin and
Kahlson in 1944 (306). At this point, the cellular source of
histamine was still obscure. It was only in the late 1960s
that histamine had been histochemically localized to the
ECL cells of the gastric gland (413, 1084).

fairly well understood. Following CCK2 activation, increased transcription of HDC is mediated via a PKC- and
ERK-dependent pathway (470, 472). The HDC gene promoter is then activated by at least three distinct nuclear
factors which bind to gastrin response elements, resulting in
gene transcription (889, 890). Apart from augmenting gene
transcription, gastrin regulates the degradation of HDC,
which further increases intracellular enzyme levels (331,
1214). Second, gastrin enhances the transcription of the vesicular monoamine transporter type 2 (VMAT2; SLC18A2),
which is responsible for accumulating histamine in the secretory vesicles (376). Similarly to HDC, this effect depends
on PKC and ERK activation and binding of a nuclear factor
to a gastrin response element in the VMAT2 promoter region (164, 1154). It should be mentioned that gastrin regulates the transcription of a plethora of other genes which
serve a diverse array of roles, ranging from growth to metabolism (346). Amongst many others these include chromogranin A, which is essential for granule packaging and is
a precursor of pancreastatin (see sect.VD3) (231). Third,
gastrin induces the fusion of secretory granules and the
release of histamine into the gland environment. Secretion
follows a biphasic elevation of intracellular calcium concentrations after activation of CCK2 (1201). The biphasic increase has been proposed to result from initial IP3-mediated
release from intracellular stores, which is followed by subsequent influx of calcium via L-type calcium channels from
the extracellular space (1201). The importance of intracellular store mobilization has been contested by a different
group, which proposed that solely influx trough L-type, and
to a lesser extent N-type, calcium channels triggers the secretory response (673). Lastly, gastrin has a trophic effect
on the ECL cell (see sect. IIB2).

SASCHA KOPIC AND JOHN P. GEIBEL


stimulated release of somatostatin is of particular importance in the mouse, as most studies showing a suppression
of acid secretion after PACAP administration were conducted in murine models.

The ECL cell is inhibited by a variety of substances, the


most prominent of which is somatostatin (204, 590, 798,
941). Somatostatin is produced in D-cells of the oxyntic
mucosa and reaches the ECL cell in a paracrine fashion
where it binds to the somatostatin receptor (SST2 and potentially SST5) (FIGURE 2) (570, 873). Receptor binding
leads to inhibition of histamine exocytosis via blockade of
mostly L-type calcium channels (105). This impedes the
elevation of intracellular calcium concentrations caused by
ECL activators, such as gastrin (see above) (873). In addition, somatostatin also inhibits the proliferation of ECL
cells (570). Somatostatin can thus be seen as the global
hormonal antagonist to gastrin with regard to ECL cell
function and proliferation. The neuronal inhibition of ECL
cells is mainly carried out by the neuropeptide galanin (105,
672, 798, 1209). Galanin is localized to neurons of the ENS
and demonstrated an inhibitory effect on histamine secretion in in vitro and in vivo models (105, 302, 672, 731, 798,
1209). Similarly to somatostatin, the molecular mechanism
underlying its inhibitory effect is an interference with calcium signaling via closure of L-type calcium channels (105).
Lastly, prostaglandin E and nitric oxide also act as inhibitors of histamine release (105, 554, 798, 1002). Although
neuropeptide YY (PYY) and calcitonin gene-related peptide
(CGRP) have also been implicated in playing a role in ECL
cell regulation, a detailed discussion is omitted in light of
contradictory results which range from stimulation to inhibition of secretion (672, 674, 798, 1210).
B) CELLULAR EFFECTS. As mentioned earlier, stimulation of the
ECL cell is translated into an elevation in intracellular calcium concentrations, leading to exocytosis of preformed

198

Very small amounts of histamine are sufficient to induce


acid secretion. Histamine acts via the H2 receptor on the
parietal cell, which has been discovered by Sir J. W. Black in
1972 (106). For this seminal discovery, he was later
awarded the Nobel Prize in Physiology and Medicine. The
H2 receptor belongs to the family of seven-transmembrane
domain GPCRs. Its activation predominantly leads to increases in the intracellular levels of cAMP, but also of calcium, which serve as stimulatory signals for H-K-ATPase
trafficking (67, 189, 738, 840, 1026, 1143). In analogy,
pharmacological agents that elevate cAMP, such as IBMX
or forskolin, induce acid secretion (1026, 1191). The increase in cAMP is due to activation of adenylate cyclase via
Gs. The role of calcium in the process of histamine secretion
remains a controversial matter. First, the mechanism leading to histamine-induced increases in intracellular calcium
has been subject of discussion. Evidence exists that histamine can, apart from adenylate cyclase, also activate PLC,
leading to calcium release from intracellular stores (607,
1142, 1143). Conversely, it has been suggested that the
observed increases in intracellular calcium are a byproduct
of cAMP-mediated PKA activation, which in turn can regulate the opening of calcium channels (144, 189, 840). Second, it is questionable to what degree the calcium signal is
an integral and necessary part of the acid secretory response
to histamine (738, 840). Chelation of the transitory calcium
increases with BAPTA abolishes only the secretory response
of isolated gastric glands to histamine by 40%, while it
completely eliminates the response to cholinergic stimulation (738). Also, live fluorescence imaging in isolated glands
showed no spatiotemporal correlation between the histamine-induced increases in calcium and the onset of acid
secretion, thereby questioning an involvement of calcium in
the secretory response (840).
H2 receptor knockout animals effectively illustrate the significance of the histamine-gastrin axis in gastric physiology.
Lack of the H2 receptor leads to a complete failure of gastrin or histamine to induce acid secretion (584). The secretory response to carbachol, however, remains intact (584).
Hypergastrinemia develops as a feedback mechanism with
the aim of reestablishing acid secretion, leading to mucosal

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

PACAP has very similar effects on the ECL cell as gastrin.


Similarly to gastrin, PACAP causes histamine release by
increasing intracellular calcium concentrations via calcium
influx through L-type, but also ligand-gated calcium channels (673). In further analogy to gastrin, PACAP upregulates the expression of HDC and exerts trophic effects on
the ECL cell (590, 729, 810). Contradictory results with
regard to the effects of acetylcholine on histamine release
exist. It has been reported that acetylcholine can either stimulate or has no effect on the secretion of histamine in in vitro
experiments on isolated ECL cells (481, 672, 674, 941,
946). In vivo application of muscarinic agonists, followed
by measurement of histamine concentrations using microdialysis, also yielded no evidence for cholinergic stimulation
(798). Conversely, it is well accepted that adrenergic stimulation leads to an increase in histamine release; however,
the physiological relevance of adrenergic activation of ECL
cells is not entirely clear (636, 672, 674, 798, 871, 941).

histamine-containing secretory vesicles. The molecular


mechanism of vesicle fusion with the apical membrane relies on the formation of the core SNARE complex, consisting of syntaxin, synaptobrevin, and SNAP-25. Synaptotagmin presumably acts as a calcium sensor relaying the
intracellular calcium signal to the vesicle fusion protein
apparatus. The expression of all SNARE complex proteins has been confirmed in the ECL cell (471, 477,
1215). In accordance with these findings, introduction of
the neurotoxins tetanus toxin light chain and botulinum
toxin, which cleave constituents of the SNARE complex
apparatus and thereby render it nonfunctional, result in
inhibition of histamine secretion (477).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


hypertrophy (584). In light of the central role of the H2
receptor in parietal cell physiology, it has been successfully
used as a pharmacological target with the aim of suppressing gastric acid output (see sect. IIC2).
4. Somatostatin/D-cell
Somatostatin was isolated for the first time in 1973 from
ovine hypothalamus and characterized as an inhibitor of
growth hormone release from the pituitary gland (124). A
few years later somatostatin was identified in endocrine
cells of the stomach, which we now know as D-cells (638).

Somatostatin is the global antagonist of the acid secretagogues. It is produced by intestinal and gastric D-cells, the
latter of which exist in two populations in the stomach (61):
an antral population locally inhibits the release of gastrin
from G-cells, whereas a population localized to the acidproducing oxyntic mucosa directly regulates the parietal
cell and inhibits histamine release from the ECL cell (FIGURE 2) (19). The morphology of the D-cell is characteristic
in that it possesses long cytoplasmic processes, which allow
it to communicate with and regulate neighboring cells in a
paracrine fashion (620, 632). It is worthwhile to distinguish
the two populations of gastric D-cells, as each population
possesses unique physiological properties (1202).
The antral D-cell is mostly regulated by the local concentrations of gastrin, cholecystokinin, and intraluminal pH.
Gastrin induces somatostatin secretion from D-cells, which
causes reciprocal inhibition of gastrin release from neighboring G-cells, thereby creating a local negative-feedback
loop (976, 1011, 1202). The molecular mechanism underlying this loop is, however, less clear. CCK2 receptor is, if at
all, only expressed at very low levels in the antral mucosa
(749, 905, 967). It has been proposed that gastrin stimulates somatostatin release in the antrum in a receptor-independent mechanism (1202). This may be accomplished via
direct cell-cell contacts between the G- and the D-cell,
which have been demonstrated with electron microscopy
(620). Conversely, evidence for cholecystokinin and its
stimulatory role for somatostatin release via CCK1 is more

One of the most important stimulators of D-cell secretion is


the intragastric pH. A seminal observation demonstrating a
correlation between gastric acidity and the amount of secreted somatostatin was made in dogs in the 1970s. It has
been shown that the amount of somatostatin directly increases in antral venous blood following gastric HCl infusion, while somatostatin levels were unaffected in venous
blood from the oxyntic mucosa (982). Similar observations
were later made in isolated mouse stomach, however without topographic discrimination (975). Two main hypotheses as to how somatostatin is regulated by intragastric pH
exist. The first states that the D-cell can directly act as a pH
sensor, and the second postulates that the pH sensing is
mediated by neurons, which in turn act on D-cells. To accomplish putative direct pH sensing, several antral D-cells
are equipped with a distinct morphological feature. They
possess apical projections that are in contact with the glandular lumen, potentially allowing them to constantly monitor the intraluminal milieu (620). These D-cells have been
termed open type. Conversely, the D-cells of the oxyntic
mucosa are mostly of the closed type, meaning that they are
embedded in the mucosa without luminal contact. The molecular identity of the putative apical pH sensor remains
elusive. However, the presence of CaSR, which has pH
sensing properties, was recently confirmed in preliminary
studies on the D-cell and may represent a possible candidate
for this mechanism (770). Apart from directly acting on
D-cells, the effect of pH on somatostatin secretion may be
mediated via afferent spinal neurons. Over 80% of the spinal afferent neurons contain the neuropeptide CGRP (397,
758, 1047, 1116). Perfusion models of antral sleeves have
shown that the acid-induced rise in somatostatin is accompanied by a concomitant increase in the concentrations of
the neuropeptide CGRP (708). Furthermore, application of
a CGRP receptor blocker inhibited the release of somatostatin following acid exposure (708). As D-cells are known to

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

199

Downloaded from on December 8, 2014

A) SYNTHESIS AND REGULATION OF RELEASE. Somatostatin is a


peptide hormone that exists in two primary forms that differ in their respective peptide length. The most abundant
form in the gastric mucosa is somatostatin-14 (consisting of
14 amino acids), whereas somatostatin-28 only constitutes
a minute fraction of the total gastric somatostatin content
(198, 1125). The two forms of somatostatin are cleavage
products of a larger 116-amino acid pre-prohormone (preprosomatostatin), which in turn is processed to the 92amino acid-long prosomatostatin (1000). It should be mentioned that other cleavage products, such as antrin or somatostatin-28(112) exist and are secreted together with
somatostatin (77, 885). Their physiological significance is,
however, less well understood.

substantiated (749, 905, 967, 1202). Cholecystokinin is


structurally closely related to gastrin (both share an identical 5-amino acid COOH terminus) and also exists in various peptide lengths (767). It is secreted by I-cells of the small
intestine following protein and fat-rich chyme entering the
duodenum, and thus represents a classical mediator of the
intestinal phase of acid secretion (594). As its name implies,
cholecystokinin has originally been described as a stimulator of gallbladder contraction; however, its inhibitory influence on gastric acid secretion is now well accepted and
extensively described (593, 1203). Cholecystokinin can
bind to both the CCK1 and CCK2 receptor with almost
equal affinity, whereas the actions of gastrin are almost
exclusively mediated by the CCK2 receptor. The dual affinity of cholecystokinin would imply a possible stimulatory
effect on acid secretion via activation of CCK2 on ECL cells;
however, in vivo the inhibitory effect mediated by activation of CCK1 and CCK2 on D-cells prevails (593, 966,
1202, 1203).

SASCHA KOPIC AND JOHN P. GEIBEL


express the CGRP receptor, an involvement of CGRP in
acid sensing is plausible (558). Again, this provokes the
question of how CGRP-containing neurons may sense acidity on a molecular basis. The acid-sensitive channels transient receptor potential vanilloid channel (TRPV1) and the
acid-sensing ion channel 3 (ASIC3) had been proposed as
molecular acid sensors; however, latest experiments have
shown that the increase in CGRP still occurs in the genetic
absence of the channels (47, 96, 163).

B) CELLULAR EFFECTS.

The effects of somatostatin on its target


cells are mediated by the SST2 receptor. Knockout of the receptor causes a 10-fold increase in basal acid output, which
exemplifies the pivotal role somatostatin plays as a global suppressant of acid secretion (715). Somatostatin acts on all main
cell types that are involved in the process of acid secretion, i.e.,
parietal cells, ECL cells, and G-cells (FIGURE 2). The inhibition
of the G- and ECL cells has been discussed in the respective
sections. In the parietal cell, somatostatin has a clear direct
inhibitory effect on secretagogue-induced acid secretion (827,
1177). This effect is partially attributable to activation of Gi,
leading to inhibition of adenylate cyclase and a subsequent
decrease in intracellular cAMP levels (827).

A) SECRETIN. Secretin is a 27-amino acid peptide hormone


that is synthesized in duodenal S-cells and secreted into the
circulation in response to a low duodenal pH or passage of
digestive products, such as fat (195, 955, 1153). A subpopulation of secretin-producing cells is also present in the
gastric mucosa, where it may influence acid secretion in a
paracrine manner (191193). Given its secretory stimulus,
it is regarded as a classic effector of the intestinal phase of
acid secretion. When it was first discovered in 1902 by
Bayliss and Starling (interestingly secretin was the first hormone ever to be discovered), it was noted that secretin induces pancreatic bicarbonate secretion, which leads to a
buffering of the gastric acid entering the duodenum (68). In
the stomach, secretin acts as an inhibitor of gastric motility
and acid secretion (141, 194, 269, 374, 532, 564, 656, 677,
1107). The exact mechanism as to how secretin attenuates
the secretion of acid is not exactly known, and several hypotheses have been put forward. For example, it has been
shown that secretin induces the secretion of somatostatin
from isolated D-cells (141). Increases in somatostatin levels
were also observed in isolated perfused stomach models (205,
374). Others have proposed that secretin activates vagal primary afferent neurons, which in turn leads to neuronal modulation of acid secretion (656, 659). In opposition to this theory, it has also been demonstrated that the inhibitory effects of
secretin are independent of vagotomy (677).
B) OXYNTOMODULIN. Oxyntomodulin is a peptide hormone
produced in the mammalian intestine. It is closely related to
glucagon and contains its entire amino acid sequence, extended by a COOH-terminal octapeptide (66). In isolated
parietal cells, oxyntomodulin acts as an activator of acid
secretion (959). The integrated response to oxyntomodulin
is, however, opposite. Systemic injection decreases gastric
acid secretion in rat, cat, and human test subjects (53, 157,
285, 524, 525, 965). The inhibitory effect is most likely
mediated via somatostatin release (53).

In conclusion, somatostatin acts as the global brake on acid


secretion. By acting on G-cells, ECL cells, and parietal cells,
it exerts its inhibitory action on every link in the regulatory
chain leading to the secretion of gastric acid.

It was recognized in the early 1950s that serotonin was present in the antral mucosa of dog stomachs
(323). Serotonin is stored in granules of enterochromaffin
cells of the antrum (1099). It is released into the circulation
and the gastric lumen in response to vagal stimulation (107,
649). Intraluminal acidification serves as another stimulus
for serotonin release (1196). Serotonin has an inhibitory
effect on the secretion of gastric acid (107, 153, 521, 650,
720, 903). It is still poorly understood where serotonin
interferes with acid secretion.

5. Other substances

D) NEUROTENSIN.

A variety of other substances have been shown to have


either direct or indirect effects on acid secretion. In the

200

C) SEROTONIN.

Neurotensin is a 13-amino acid neuropeptide that was originally isolated from calf hypothalamus
(161). In the periphery, it is also produced and secreted
postprandially by specialized endocrine cells (N-cells) of the

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Neuropeptides of the gastric ENS that stimulate the secretion of somatostatin include PACAP and VIP, which both
bind to the VPAC receptor expressed on D-cells (199, 657,
1207). The presence of VIP and PACAP containing neurons, which integrate signals from the vagus nerve, has been
demonstrated in the gastric mucosa (302, 737). Furthermore, cholinergic signals can act on the antral D-cell via the
M3 receptor to promote secretion of somatostatin (140).
This is in sharp contrast to D-cells from the oxyntic mucosa
that are inhibited by cholinergic signals (197, 200, 1182).
As mentioned earlier, the D-cells in the oxyntic mucosa also
differ in their morphology. D-cells in the oxyntic mucosa
are of the closed type and have thus not been implicated to
participate in acid sensing. They exert their acid-suppressive effects by the paracrine regulation of ECL and parietal
cells. Further functional divergence between antrum and the
oxyntic mucosa has been demonstrated in the regulation of the
somatostatin mRNA. For example, suppression of acid secretion with omeprazole in fasted animals markedly decreased
somatostatin mRNA levels in the antrum, whereas the levels in
the oxyntic mucosa were affected to a much lesser extent,
which further corroborates the hypothesis that the antral cells
are involved in luminal chemosensation (945).

interest of conciseness, their physiological effects will only


be discussed briefly at this point.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


small intestine (863). Various investigations have demonstrated that neurotensin suppresses the secretion of gastric
acid and delays gastric emptying (25, 108, 486, 985). This
has been shown by direct systemic injection, but also by
immunoneutralization of endogenous neurotensin in a reverse approach (25, 108, 486, 985). In disagreement with
these findings, other investigators could only inhibit acid
secretion at unphysiologically high serum concentrations of
750 pmol (747). Of note, physiological postprandial neurotensin levels were measured to be 15 pmol by the same
investigators, questioning the role of neurotensin as a physiological endocrine inhibitor of acid secretion (747). Neurotensin is also located to nerve fibers of the enteric nervous
system in the stomach, indicating that it may act as a local
neuronal rather than an endocrine regulator. It has been
proposed that neurotensin may induce the secretion of somatostatin and thereby exert its inhibitory action on acid
secretion (53, 414). Most recently, however, the low-affinity neurotenstin type 2 receptor (NTS2) has been identified
on the parietal cell, suggesting a direct influence.

F) NITRIC OXIDE.

Nitric oxide (NO) is an important signaling


molecule that plays a role in multiple physiological processes, such as vasodilation or the immune response. Indeed, NO has been shown to mediate the hyperemic response of the gastric mucosa that occurs during acid secretion (553). However, NO also directly influences the
production of acid. The effect of NO on acid secretion is
most likely inhibitory (81, 82, 555, 1002; opposed by Ref.
426). NO is produced by various forms of NO synthases,
one of which has been localized at high concentrations in
cells in the vicinity of parietal cells, allowing for a putative
paracrine regulation (80). NO has been proposed to exert
its inhibitory action by either directly inhibiting the parietal
cell or by suppressing the release of histamine from ECL
cells (81, 82, 555, 1002). Intracellular increases in cGMP

G) INTERLEUKINS. Interleukins (IL) are cytokines that mainly


coordinate immune responses. In particular, IL-1 has been
shown to impact gastric acid secretion. IL-1 is a general
proinflammatory cytokine that plays an important role in
the stomach in the context of Helicobacter pylori infection.
H. pylori infection triggers an elevation of IL-1 levels as
part of the hosts immune response (65). Peripheral injection of IL-1 can profoundly suppress gastric acid secretion
(912, 947, 1059, 1101, 1132). Multiple explanations for
this observation have been put forward. It has been suggested the IL-1 acts in the CNS, as intrathecal injection
also has an acid-suppressive effect (948, 949). Others have
suggested that IL-1 promotes formation of prostaglandins
or NO, which in turn inhibit acid secretion (312, 947,
1101). Yet, a direct effect on parietal cells and ECL cells is
the most likely explanation, as both cell types express the
IL-1 receptor and have been shown to be inhibited in their
function in isolated cell models (69, 70, 872, 958).

C. The Pharmacological Suppression


of Acid Secretion
Decreasing gastric acidity is indicated in many pathological
contexts, including gastric reflux disease or peptic ulcer disease. This target can be achieved by two main pharmacological approaches: 1) the inhibition of gastric acid secretion or 2) the intraluminal neutralization of already secreted
gastric acid (antacids). Gastric acid secretion can be attenuated by either directly blocking its final molecular effector,
namely, H-K-ATPase (PPIs and acid pump antagonists),
or by interfering with the neurohormonal signaling pathway leading to its secretion (H2 antagonists). The following
section attempts to discuss the four most common substance classes employed to increase intragastric pH.
1. Direct pharmacological inhibition of H-K-ATPase
The inhibition of H-K-ATPase-mediated proton transport represents the main contemporary pharmacological
strategy for reducing gastric acidity. An increase of gastric
pH is the main factor ameliorating acid-related disorders
and has been show to directly correlate with healing rates of,
for example, GERD (74). Two main substance groups exert
their acid-reducing effect via inhibiting H-K-ATPase function: PPIs and acid pump antagonists (APAs). Both substances
achieve this aim by distinct mechanisms.
Omeprazole was the first clinically available PPI (324). The
first patent on omeprazole was filed in 1979 by the Swedish
company Astra AB (today AstraZeneca). The introduction
as a prescription PPI followed in 1989. Today, the omeprazole enantiomer esomeprazole (S-omeprazole) generates the

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

201

Downloaded from on December 8, 2014

E) GHRELIN. Ghrelin is a recently discovered 28-amino acid


peptide hormone that is synthesized in P/D1 cells of the
fundus (242). Since its discovery, multiple functions have
been ascribed to it, ranging from being a regulator of appetite to being a modulator of bone remodeling. Its effects on
bone are described in a separate section of this review (see
sect. VD1). Apart from these functions, ghrelin also has
been implicated to affect gastric acid secretion, although it
remains a matter of discussion in which direction, as peripherally administered ghrelin has been reported to stimulate,
inhibit, or not affect acid secretion (278, 355, 654, 719).
The reason for these dichotomic results is largely unclear.
The fact that ghrelin circulates in acylated and desacylated
forms adds further complexity to the subject (491). Indeed,
acylated ghrelin has been shown to stimulate acid secretion
following peripheral injection, whereas desacylated ghrelin
remained without effect (938). Further investigations are
needed to clarify the controversy surrounding ghrelin and
its influence on gastric acid secretion.

concentrations have been observed in both cell types after


NO exposure, suggesting that guanylate cyclase is an intracellular target for NO (82, 1002).

SASCHA KOPIC AND JOHN P. GEIBEL


second highest revenue of all pharmaceuticals in the United
States and is only surpassed by the statin atorvastatin (512).
Furthermore, in the United States, PPIs are available as
over-the-counter formulations, making them accessible for
the broad public. This is partially made possible by the high
safety profile of PPIs with a low incidence of unspecific
adverse effects. Recently, however, concerns about the
long-term effects of chronic acid suppression have emerged
with regard to its impact on bone health (see sect. VA).

Suppression of acid secretion can never be complete, as


H-K-ATPase is subjected to a constant turnover (half-life
50 h) and needs to be stimulated for the conversion of the
PPI to take place (936). Nevertheless, PPIs are highly effective in reducing gastric acidity. Depending on the PPI and
the regimen, overall intragastric pH can be elevated by several pH units, up to a pH of 6 (compared with 12 at
baseline) (137, 425, 1036). For an excellent summary of PPI
efficacy, please refer to Reference 1036.
APAs represent the second class of H-K-ATPase inhibitors. Unlike PPIs, they do not undergo irreversible binding,
but rather act as potassium competitive antagonists. The

202

2. H2 antagonists
The development of H2 blockers is inseparably intertwined
with Sir Blacks discovery of the H2 receptor on the gastric
parietal cell at the Smith Kline and French Laboratories
(now GlaxoSmithKline) (106). In his original publication,
Sir Black also describes burimamide as a competitive H2
antagonist that can effectively inhibit pentagastrin-stimulated gastric acid output in human volunteers (106). Further
development of the antagonist led to the synthesis of cimetidine, which was first commercially introduced in 1976 in
the United Kingdom, followed by the United States in 1977.
Other commonly used members of H2-antagonist family
now include ranitidine, famotidine, and nizatidine.
H2 antagonists prevent histamine-mediated stimulation of
the parietal cell by competitively interfering with its receptor. Although this effectively terminates the gastrin-histamine axis, the parietal cell is still susceptible to cholinergic
stimulation via the M3 receptor. This partial inhibition
mainly accounts for the lower clinical efficacy of H2 antagonists compared with PPIs, which directly target H-KATPase as the final target of all parietal cell stimuli (384).
For example, a meta-analysis concluded that patients
treated for bleeding peptic ulcers are about twice as likely to
suffer from persistent or recurrent bleeding if treated with
H2 antagonists compared with PPIs (384). Another metaanalysis also demonstrated a higher efficacy of PPIs in treating esophagitis (83% healing rate with PPIs compared with
52% with H2 antagonists)(567). Today, H2 antagonists are
largely superseded by PPIs due to their higher clinical efficacy. Furthermore, with the exception of famotidine, H2
antagonists are extensively metabolized in the liver by the
CYP-450 system, leading to substantial drug-drug interaction profile (for review, see Ref. 506).
3. Antacids
Antacids directly neutralize gastric acid allowing immediate
short-term control of heartburn. They exist in various salt
formulations, the most common of which are carbonate
salts, such as CaCO3, MgCO3, or NaHCO3. The use of
calcium carbonate as a dietary calcium supplement is dis-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

PPIs are delivered as pro-drugs through the bloodstream to


the parietal cell. They are weak bases (pKa 4), which can
easily pass the cell membrane and accumulate in acidic compartments, such as the secretory canaliculus of the parietal
cell. The pro-drug is then converted to the pharmacologically active cyclic sulphenamide by the acidic pH in the
secretory canaliculus (670, 1007, 1135). Their specific accumulation in acidic milieus and their pH-catalyzed conversion to active substances confers specificity and thus a high
safety profile to PPIs. Once activated, PPIs bind covalently
via disulfide bonds to H-K-ATPase, thereby inhibiting its
capacity to pump protons (89, 90, 1005, 1006, 1008). The
pattern of the cysteine residues, which are involved in PPI
binding, differ among the respective members of the PPI
family: cysteine-813 reacts with all PPIs. In addition,
omeprazole reacts with cysteine-892, lansoprazole with
cysteine-321, and pantoprazole and tenatoprazole, respectively, with cysteine-822 (89, 90, 1005, 1006, 1008). Since
the binding is covalent and irreversible, the inhibitory effect
of PPIs lasts long beyond their plasma half-life, which usually ranges between 0.5 and 2 h depending on the specific
PPI (581, 1036). PPIs are generally metabolized by the hepatic cytochrome P-450 system, in particular CYP2C19
and CYP3A4. This is of particular clinical importance, as
CYP2C19 polymorphisms are known to exist. These polymorphisms can impact the pharmacokinetics of PPIs by
affecting the metabolic rate of CYP2C19, which may have
consequences for the optimal therapeutic regimen (582).
Esomeprazole and rabeprazole seem to be less dependent on
CYP2C19 metabolism (516, 983). Apart from the pattern
of cysteine reactivity, half-life and metabolism, PPIs also
vary in oral bioavailability (581).

duration of inhibition is thus directly dependent on the


plasma concentration of the inhibitor. As predicted by homology modeling, mutational analysis, but also recent
structural data, APAs bind in the luminal cavity of H-KATPase in the vicinity of the potassium entry site where they
exert their inhibitory action (2, 42, 761, 1104, 1106). Although the inhibition of acid secretion has been shown to be
very effective, these substances are generally not in clinical use
(577). For example, clinical trials of the APA AZD08650 have
shown no additional therapeutic effect compared with the PPI
gold-standard, which resulted in abandonment of the drug in a
clinical setting (262, 539).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


cussed in a separate section (see sect. VC). Although each
formulation possesses its own spectrum of side effects, a
notable condition in the context of this review is the milkalkali syndrome, which is the result of a concomitant overingestion of calcium and alkali, such as CaCO3. Although
the syndrome became less prevalent with the introduction
of modern ulcer therapies, it still poses a significant risk for
patients that may ingest CaCO3 as a calcium supplement
for the prevention of osteoporosis or as an antacid on a
regular basis. Milk-alkali syndrome presents with the triad
of metabolic alkalosis (carbonate), hpercalcemia (calcium),
and renal insufficiency. The above average ingestion of calcium leads to increased plasma calcium levels due to excess
absorption and impaired renal secretion. PTH levels are low
due to negative feedback (6). Hypercalcemia further causes
renal vasoconstriction, which decreases the glomerular filtration rate, and renal fluid loss because of activation of the
CaSR, which in turn has loop-diuretic-like effects (see sect.
IVD3). The activation of the CaSR is further potentiated by
the metabolic alkalosis, which increases its sensitivity to
calcium. All abnormalities are usually reversible after withdrawal of the offending agent and adequate treatment.

The intestine is responsible for the absorption of dietary


calcium into our systemic circulation. Although the kidney
also plays a pivotal role in calcium homeostasis by retaining
and balancing systemic calcium via regulating its excretion
into the urine, renal calcium handling will not be the subject
of this review.
Current recommendations suggest that an average 40-yrold adult should ingest 1,000 mg of calcium on a daily
basis (218). In the United States, this requirement is mostly
met (57). Up to 72% of the dietary calcium intake is attributable to dairy products (218). Typically, the intestine absorbs between 25 and 35% of the ingested calcium (1193).
This occurs via two distinct pathways: 1) a paracellular
pathway and 2) a transcellular pathway.

A. Transcellular Calcium Absorption


1. Calcium entry
Evidence for active transport of calcium across the intestinal
epithelium was established very early. With the use of a calcium radioisotope, various groups demonstrated 1,25(OH)2vitamin D-dependent calcium transport against an imposed
concentration gradient in the small intestine of the rat (952,
954, 1150). It has also been observed that active transport
can be induced by a low-calcium diet, which we now know
to stimulate the production of 1,25(OH)2-vitamin D (1133,
1134). The degree of transport was highest in the duodenum and decreased in the more distal segments (952). The
duodenum is still considered the primary site where the bulk
of transcellular transport occurs. To conduct active transcellular calcium absorption, the enterocyte has to be equipped
with an apical calcium entry pathway, a mechanism for cytosolic calcium shuttling, and a basolateral calcium exit pathway
(FIGURE 3). As the cytosolic concentration of free calcium
is kept at a constant low level with typical concentrations
of 100 nM, apical calcium influx follows its electrochemical gradient into the cell. In contrast, the extrusion
of calcium on the basolateral membrane against an uphill
gradient either directly requires ATP or energy stored in
the sodium gradient.
A) THE SEARCH FOR THE APICAL CALCIUM ENTRY PATHWAY.

Calcium absorption via the paracellular route is tied to a


downhill concentration gradient between the luminal and
the extracellular compartment and occurs throughout the
entire intestine (although solvent drag induced paracellular
flux may also play a role at low luminal calcium concentrations; Refs. 266, 1071, 1098). Conversely, transcellular absorption can also take place against an uphill gradient, but
requires molecular machinery in the form of distinct calcium transport proteins which are expressed on the apical
and basolateral membranes of the enterocyte. This process
directly requires energy in the form of hydrolyzable ATP
and is alternatively termed active transport (versus passive paracellular transport). The proximal small intestine,
i.e., the duodenum and the jejunum, is the main site for
transcellular calcium absorption (825).

The molecular identity of the apical calcium entry pathway was unclear for a long time. Early experiments in isolated duodenal
brush-border vesicles revealed that calcium uptake was passive, saturable, sensitive to ruthenium red, 1,25(OH)2-vitamin
D dependent, and functionally optimal at a pH of 7.5 (740).
This black box characterization suggested that a specific
carrier was responsible for calcium absorption and in retrospect already provided us with accurate key characteristics of the transient receptor potential vanilloid channel
type 6 (TRPV6), which was later established as the primary
apical calcium uptake channel (740). In subsequent attempts to further unravel the nature of the calcium uptake
mechanism, various voltage-gated L-type calcium channel
blockers were used (474, 717, 838). Although isolated duodenal cells accumulated less calcium following application

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

203

Downloaded from on December 8, 2014

III. INTESTINAL CALCIUM ABSORPTION

Since a concentration gradient is not a prerequisite for this


process, transcellular transport allows us to absorb calcium
even when the calcium concentration in the chyme is fairly
low. The relative importance of each respective absorption
pathway thus alternates with the amount of ingested calcium (46, 824, 1224). The rate of paracellular calcium uptake is canonically thought to remain constant, while transcellular transport can be upregulated under conditions of
dietary calcium restriction (46, 824, 1224). This regulation
occurs via the active metabolite of vitamin D [1,25(OH)2vitamin D], which serves as a stimulator for transcellular
calcium uptake to prevent systemic calcium depletion.

SASCHA KOPIC AND JOHN P. GEIBEL

Enterocyte
D
D
VDR
D
RXR VDR

Transcription
Ca2+
PMCA

Transcription

Ca2+
Ca2+

Na+
Ca2+

Calbindin-D 9k
NCX

TRPV6

D
FIGURE 3. Transcellular and paracellular calcium absorption in the intestine. The transcellular intestinal
absorption of calcium relies on apical calcium entry through TRPV6, intracellular calcium transport by calbindin-D9k, and basolateral calcium extrusion via either NCX or PMCA. 1,25(OH)2-vitamin D regulates most of
these ion transport proteins on a transcriptional level. 1,25(OH)2-vitamin D passes the plasma membrane of
the enterocyte and binds to its receptor (VDR), which then heterodimerizes with RXR to initiate transcription.
Evidence also suggests that 1,25(OH)2-vitamin D regulates the permeability of tight junctions, which gate the
paracellular absorption of calcium. D, 1,25(OH)2-vitamin D.

of the inhibitors and 1,25(OH)2-vitamin D stimulation


(717), in vivo calcium entry proved to be fairly insensitive to
their effects (with the exception of verapamil, which demonstrated some degree of inhibition if applied at very high
concentration in the millimolar range) (474, 838). The conflicting reports may be attributable to the different experimental models that were used, as isolated single cells do not
allow discrimination between apical and basolateral transport mechanisms. Furthermore, it has been argued that Ltype calcium channels may play a role in the stimulatory
pathway of vitamin D, rather than in calcium uptake per se
(717). In conclusion, an involvement of L-type calcium
channels seemed rather inconclusive and the identity of the
calcium entry channel remained elusive.
B) TRPV6. The cloning of the calcium transport protein subtype 1 (CaT1) in 1999 finally marked a turning point in the
search for the elusive intestinal calcium entry channels
(838). The work was pioneered by Hoenderop and colleagues who had identified the main calcium entry protein
in the kidney (epithelial calcium channel type 1, ECaC) via
an expression cloning strategy a few months earlier (474).

204

CaT1 was identified by a similar approach. A rat duodenal


cDNA library was functionally screened using a calcium
uptake assay in a Xenopus oocyte expression system (838).
This screening process yielded the 727-amino acid protein
CaT1, which showed a 75% sequence homology to rabbit
ECaC. Homology analysis also demonstrated a relationship
to the vanilloid reptor type 1 (VR1), a nonspecific cation
channel that is activated by capsaicin, the pungent ingredient in chili peppers, and mostly mediates pain signaling
through afferent sensory neurons (838). The nomenclature
changed over time as new channel proteins were identified,
and today we consider CaT1, ECaC, and VR1 to be members of the same transient potential receptor vanilloid
(TRPV) ion channel family. Literature now refers to ECaC
as TRPV5, to CaT1 as TRPV6, and to VR1 as TRPV1.
The structure of TRPV6 was predicted to have six transmembrane domains and four ankyrin repeat domains,
which serve as cytoskeletal linking sites (838). Channel conductance was not dependent on other ions and was inhibitable by a low extracellular pH (838). Calcium uptake was
reduced by as much as 70% at a pH of 5.5, which con-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Ca2+

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


firmed the early black box data observations made in intestinal brush-border vesicles (838). This behavior seemed
counterintuitive to the investigators, as TRPV6 expression
was highest in the duodenum, which is exposed to an acid
load from the stomach (838). Duodenal pH has been reported to be as low as 6.1 6.6 but is even lower acutely
after gastric emptying (5.4), which would entail significant inhibition of TRPV6-mediated calcium uptake (284,
290, 838). Conductance was also modestly sensitive (10
15% inhibition) to L-type calcium channel blockers at high
concentrations, which partially clarified the preceding ambiguous observations made by other groups with these inhibitors (318, 337, 717, 838).

TRPV6 displays some unique biophysical properties. As


mentioned earlier, TRPV6 consists of six transmembrane
segments, like many voltage-gated cation channels. Unlike
these channels, TRPV6 lacks the voltage sensor domain in
the fourth -helix and is in a constitutively open state at
resting membrane potential. Analysis of the quarterny
structure shows that it assembles in tetramers (476). The
ankyrin domains were implicated to be responsible for tetramer formation; however, more recent structural data obtained by crystallography question this hypothesis (311,
848). It is thought that heterotetramers can also be formed
with subunits of closely related TRPV5 (476). In contrast to
most other members of the TRP channel family, TRPV6 is a
very selective channel for calcium (475, 1058, 1197). The
relative permeability of TRPV6 for calcium over sodium
(PCa/PNa) is 100, whereas TRPV1, for example, has a
selectivity of PCa/PNa 10 (although this has recently been
shown to be variable) (206, 475, 1197). This high selectivity is crucial for the maintenance of a constant membrane
potential in the enterocyte during calcium absorption.
TRPV6 is strongly inward rectifying, which has been attributed to magnesium ions plugging the channel pore for outward ion movement during states of depolarization (1126).
Magnesium also exerts a voltage-independent inhibitory effect on TRPV6 current; however, the mechanism underlying this observation is unclear (475, 1126). Furthermore,

In 2001 it was demonstrated that ATP modulates TRPV6


activity by preventing channel rundown (475). Recent work
by Al-Ansary et al. (15) suggests that ATP directly binds to
the channel, thereby inhibiting inactivation by locking the
channel in the open conformation. Binding of ATP may be
antagonized by channel phosphorylation through PKC
(15). In conclusion, TRPV6 is precisely regulated by its own
microenvironment. Calcium, magnesium, and protons have
an inhibitory effect on the channel, whereas ATP prevents
channel inactivation.
It is important to remember that apart from being a nutrient, calcium is a crucial intracellular messaging molecule.
Therefore, the enterocyte has to tightly control its intracellular concentration and adapt uptake to energy status and
basolateral extrusion. To ensure this delicate intracellular
homeostasis, TRPV6 is associated with and regulated by a
variety of auxiliary proteins. The first protein that has been
identified to interact with TRPV6 was the S100A10-annexin2 complex (1112). The S100A10-annexin2 complex
has been implicated to play a role in protein trafficking and
membrane anchoring. Its association with TRPV6 is essential for channel trafficking to occur, as a disruption of the
interaction leads to cytosolic scattering of the channel
(1112). Formation of the S100A10-annexin2 complex and
its association with TRPV6 involves activation of PKA and
calcineurinA (CnA) (117). Another protein that is required
for TRPV6 membrane insertion is rab11a (1111). In analogy to S100A10-annexin2, perturbation of rab11a binding
results in decreased surface expression of TRPV6 and channel retention in the cytosol (1111). Furthermore, the protein
kinases SGK1 and WNK3 are known to promote membrane insertion of TRPV6; however, their exact site of action is still unclear (114, 1031). Once trafficked to the membrane, physical channel stability is maintained by a variety
of auxiliary proteins. The COOH-terminal tail of TRPV6
contains a PDZ protein binding motif. PDZ proteins serve
as membrane anchors and protein scaffolds and mediate the
assembly of multiprotein complexes, thereby regulating
channel activity. The PDZ protein sodium hydrogen exchanger regulating factor (NHERF4) (aka PDZK2) has recently been identified to interact with TRPV6 (574, 1113).
It has been speculated that NHERF4 serves as a scaffold for
TRPV6 at the apical pole, which is underlined by the observation that knockdown of NHERF4 by RNAi leads to decreased TRPV6 current in HEK293 cells (574, 1113). Apart

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

205

Downloaded from on December 8, 2014

Subsequently, the human analog of rat TRPV6 was cloned


(it had 97% sequence homology and was rather confusingly
named ECaC2), and its expression was confirmed in human
duodenum (64). The generation of a TRPV6 antibody allowed for first localization studies, which confirmed an apical localization of TRPV6 and thus reaffirmed its role as the
primary apical calcium entry pathway in the intestine
(1220). Tissue expression of TRPV6 varies among species
(461). In humans, TRPV6 was identified in the duodenum,
jejunum, stomach, esophagus, kidney, placenta, mammary
gland, pancreas, prostate, testis, and salivary gland, but was
also found to be upregulated in a series of malignancies,
including prostate, breast, colon, and ovarian cancer (461,
792, 836, 839, 1168, 1220). The role of TRPV6 in these
tissues mostly remains to be elucidated.

TRPV6 gating is sensitive to intracellular calcium. Increases


in calcium were shown to inhibit the channel, resembling a
negative-feedback mechanism (475). Although global calcium concentrations in the cell largely remain constant, the
channel microenvironment is exposed to fluctuations in local calcium. This feedback loop may be important for finely
tuning the amount of calcium influx and preventing calcium
overload of the enterocyte. The putative mechanism of this
regulation will be discussed later in this section.

SASCHA KOPIC AND JOHN P. GEIBEL


from trafficking to the apical membrane, channel number
can be regulated by internalization and degradation. One
possibility of decreasing functional channels at the cell surface is protein ubiquitination. The process of ubiquitination
involves enzymatic tagging of target proteins, thus directing
them to the proteasome or lysosome for rapid degradation.
The degradation tag is transferred to the target protein by
E3 ubiquitin protein ligases, such as Nedd4 2. It is already
well understood how Nedd4 2-mediated ubiquitination
can decrease the number, but also directly modulate the
activity, of the epithelial sodium channel (ENaC) in the
kidney (825). A recent study demonstrated that a similar
mechanism applies to TRPV6 (1212). Coexpression of
Nedd4 2 and TRPV6 in oocytes resulted in decreased calcium flux and channel numbers (1212). Nedd4 2-mediated
TRPV6 ubiquitination could thus serve as a mechanism of
rapidly regulating channel retrieval from the plasma membrane (1212).

As previously discussed, the activity of TRPV6 is finely


regulated by intracellular calcium concentrations. Elevations in intracellular calcium levels exert an autoinhibitory effect on channel opening (475). Shortly after the
identification of TRPV6, calmodulin (CaM) was shown
to bind to the channel in a calcium-dependent manner,
and it was speculated that it mediates the calcium feedback response (461, 791). Subsequent investigations demonstrated that CaM may indeed represent the molecular
calcium sensor (263, 619). Elegant FRET studies reported
that CaM dynamically associates with TRPV6 in the presence of calcium and that this association is terminated when
intracellular calcium is depleted (263). The association between TRPV6 and CaM also correlates with decreased current flux through the channel (263). Although the ankyrin
repeat domains of the channel were thought to possibly
serve as binding sites for CaM, more recent structural insights provide rebutting evidence for this hypothesis (848).
Instead, the COOH-terminal tail of TRPV6 has been suggested as the site where CaM binding occurs (263, 619,
791). Hence, CaM can tune ion flux through TRPV6 and

206

Phosphorylation by kinases and dephosphorylation by


phosphatases represent a common cellular strategy for rapidly modulating channel gating properties. The non-receptor tyrosine kinase Src has emerged as a candidate for the
direct phosphorylation of TRPV6, thereby increasing channel conductance (1042). Src was previously shown to modulate TRPV4 activity (1178). The effects of Src are antagonized by the phosphatase PTP1B. Both enzymes act on the
tyrosine residues Y161/162, which are located in the NH2terminal tail of the channel (1041).
An interesting interaction without direct relevance for the
intestine has been reported between renal TRPV5 and
klotho (170). Klotho is a -glucuronidase with a transmembrane anchor that can be cleaved, resulting in shedding of
the enzymatically active domain of the protein into the
urine (170, 612). Klotho can then increase TRPV5-mediated calcium uptake by hydrolyzing N-linked oligosaccharides on extracellular channel domains, thereby preventing
channel retrieval from the plasma membrane (170). Although a recent report indicates that klotho can also affect
TRPV6 activity in vitro, it is not expressed in the intestine
and thus may only interact with renal TRPV6 (612, 683).
Although this finding has no implications for intestinal calcium uptake, various bacteria and neutrophils produce
-glucuronidase, which may affect TRPV6 activity in the
intestine (359, 1004).
Recently, Stumpf et al. (1046) described an association between cyclophilin B (CyB) and TRPV6 in the placenta. Coexpression of both proteins in oocytes increased calcium
uptake (1046). CyB was also detected by Western blot analysis in the small intestine and colon; however, colocalization and functional studies in intestinal tissue remain to be
performed (1046).
C) THE TRPV6 KNOCKOUT CONUNDRUM.

As will be discussed in
more detail later, 1,25(OH)2-vitamin D is one of the key
hormonal regulators of systemic calcium homeostasis (see
sect. IVA). Increased levels of 1,25(OH)2-vitamin D lead to
enhanced intestinal calcium absorption. Shortly after cloning of TRPV6, it was recognized that the channel is positively regulated at the mRNA level by 1,25(OH)2-vitamin D
(614, 1030, 1109, 1110). The TRPV6 promoter has multiple binding sites for the 1,25(OH)2-vitamin D receptor
(VDR) and is thus directly sensitive to increases in
1,25(OH)2-vitamin D levels (736). Consequently, VDR-deficient animals display a marked decrease in TRPV6 mRNA
levels, whereas administration of 1,25(OH)2-vitamin D in
wild-type animals results in an increase in mRNA transcription (1030, 1109, 1110). In conjunction with the rapidly expanding characterization of the channel itself, these observations supported the dogma that TRPV6 is the essential player

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Several other proteins have been identified to associate with


TRPV6 that do not affect channel numbers, but rather directly regulate channel activity. The nisnap1 gene was identified in the late 1990s; however, its function remained elusive (992). Recently, TRPV6 was shown to interact with
nisnap1 (971). It is expressed in the intestine, but not the
kidney, which is unusual as TRPV6 and TRPV5 are generally regulated by similar auxiliary proteins. Electrophysiological measurements showed that nisnap1 inhibits TRPV6
without affecting its surface expression (971). Very similar
functional properties were attributed to RGS2, a protein
that is mainly known to alter the GTPase activity of G
proteins. In analogy to nisnap1, RGS2 can inhibit TRPV6
at the plasma membrane without affecting its trafficking
dynamics (970).

most likely mediates the channels sensitivity to intracellular calcium.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


in transcellular calcium uptake and that this pathway is
strongly regulated by 1,25(OH)2-vitamin D.

These controversial findings allow several conclusions and


speculations. 1) As with any model of targeted gene disruption, a compensatory mechanism may be in place that
masks and distorts the physiological importance of TRPV6.
The animals may upregulate other, yet unidentified, calcium transport mechanisms to compensate for the loss of
TRPV6 function. 2) Rather than being a constitutively
active pathway, transcellular calcium absorption through
TRPV6 only occurs in states of low dietary calcium intake.
This hypothesis has been put forward very early and has been
reaffirmed by a recent study that observed an increase in bone
turnover in TRPV6 (/) animals on a low-calcium diet
(46, 666, 824, 1224). Mobilization of bone calcium may be
necessary in these animals to maintain systemic calcium
concentrations, which are double challenged by the lack of
TRPV6 and the insufficient calcium intake (666). Furthermore, it has been extensively described that a low-calcium
diet induces gene expression of TRPV6 [presumably
through an increase in 1,25(OH)2-vitamin D levels], suggesting a role of the channel during states of insufficient
dietary calcium supply (76, 132, 639). 3) 1,25(OH)2-vitamin D may have many more targets in the intestinal mucosa
than previously anticipated and may also regulate calcium

D) GENETIC POLYMORPHISMS OF TRPV6.

Single nucleotide polymorphisms (SNPs) are variations in the genomic sequence


that occur in one single base. If the frequency of a SNP or a
specific set of SNPs (haplotype) increases in a population
over time compared with other SNPs, it can be concluded
that this set of SNPs is associated with an evolutionary
advantage, meaning that this gene locus was under selection. Recent reports from various groups indicate that
TRPV6 was subjected to strong selection (12, 502, 1023,
1050). Traces for selection can be found in any non-African
population (12, 502, 1023). The selective event in Europeans has presumably occurred 7,000 years ago and
coincides with the development of agriculture (502). This
correlation also holds true for other populations (502). It
has been speculated that a change in diet or resistance to
emerging disease led to selection and fixation of the now
conserved haplotype (502). Interestingly, selection took
place in parallel in many populations, pointing towards a
strong selective stress that developed independently in
each region (502).
The electrophysiological properties of ancestral and derived
TRPV6 were investigated by two groups (502, 1050).
Hughes et al. (502) observed no statistically significant divergence in channel behavior. The derived channel only
displayed a tendency towards decreased sensitivity to the
autoinhibition by calcium, albeit not significant (P
0.094) (502). The authors speculated that differences in
protein-protein interactions may have led to selection
(502). Conversely, Sudo et al. (1050) reported increased
calcium influx through the derived channel, which may constitute an evolutionary advantage by facilitating dietary calcium uptake. Regardless of the controversial functional
data, these insights indirectly underline the importance of
TRPV6 and provide a counterweight to the conclusions
drawn from the TRPV6 (/) animal model. It is unquestionable that TRPV6 was under parallel independent selection in many regions across the planet. This provokes the
question of how strong selection can occur for a derived
channel whose physiological difference to the ancestral
form seems to be very subtle, yet complete disruption of the
channel in the mouse model only causes a very mild phenotype. This inconsistency adequately exemplifies the caveat
that underlies the conclusions drawn from (/) animals
and reminds us of the fact that we are far from understanding the exact physiology of transcellular calcium uptake.

E) CAV1.3. TRPV6 may not be the only channel mediating


apical calcium absorption in the intestine. A recent investi-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

207

Downloaded from on December 8, 2014

With the introduction of novel genetic techniques, a TRPV6


(/) mouse was created in 2007 by Bianco et al. (94). The
animals presented with decreased intestinal calcium uptake,
as measured by serum concentrations of a calcium radioisotope following gavage, decreased femoral bone density
(9.3%) and increased 1,25(OH)2-vitamin D levels, as a result of feedback regulation (94). These findings were in
accordance with the postulated role of TRPV6 as the primary 1,25(OH)2-vitamin D-sensitive calcium uptake mechanism in the intestine. However, the generation of another
TRPV6 (/) animal line by Benn et al. (76) a year later
spawned a controversy in the field. In these animals, baseline calcium uptake was identical between wild-type and
(/) groups. Surprisingly, calcium uptake could be increased in the (/) group by a low calcium diet and, more
importantly, by 1,25(OH)2-vitamin D (an observation that
was also confirmed by another group; Ref. 615) (76). These
findings profoundly questioned the role of TRPV6 in calcium absorption and suggested that a different molecular
target of 1,25(OH)2-vitamin D mediates the increase in calcium uptake after exposure. All these observations were in
sharp contrast to the initial report by Bianco et al. (94).
However, Benn et al. (76) also reported that compared with
wild-type animals calcium uptake was reduced in TRPV6deficient animals that were fed a low-calcium diet. This
suggests that the animals may not be able to adequately
increase absorption, when confronted with a low dietary
availability of calcium, which in turn suggests a role of
TRPV6 during states of dietary calcium insufficiency.

uptake through the paracellular pathway (76, 615). In conclusion, the generation of TRPV6 (/) animals has sustainably challenged our model of transcellular calcium absorption by questioning the relative importance of TRPV6
and by introducing new potential targets of 1,25(OH)2vitamin D regulation.

SASCHA KOPIC AND JOHN P. GEIBEL

2. Calbindin-D 9k
The following section will address the question of what
happens to dietary calcium after it enters the enterocyte. It
should be considered that transcellular calcium uptake and
cytosolic calcium homeostasis are two contradicting requirements for the enterocyte. A prominent transcellular
flux of free calcium will inevitably interfere with housekeeping functions of the cell, such as intracellular signaling.
Furthermore, it has been calculated from in vivo data that if
calcium were to diffuse freely (simple diffusion) through the
cytosol, uptake rates would only be 1/70th of the actually
measured values (129). Simple diffusion would constitute a
bottleneck in the process of transcellular calcium absorption if cytosolic calcium concentrations should be kept low.
A partial solution to this problem was found as early
as 1966, when Wasserman et al. (1151) identified a
1,25(OH)2-vitamin D-inducible calcium binding protein in
the chick intestine. The authors observed that calcium radioisotopes traveled faster across a cellophane membrane if
suspended in intestinal homogenates from rachitic, i.e.,
1,25(OH)2-vitamin D deficient, chicks than if suspended in
the homogenates from rachitic animals treated with
1,25(OH)2-vitamin D (1151). This indicated that calcium
was bound to a protein in the enterocyte and that expression of this protein was controlled by 1,25(OH)2-vitamin D
(1151, 1152). Subsequently, the calcium binding protein
was further characterized (1073, 1149, 1152). Expression
levels were shown to be highest in the duodenum and to
gradually decrease in more distal segments, which correlated with the degree of calcium uptake that has been attributed to each intestinal segment respectively in prior
functional investigations (1073). The mammalian isoform,

208

which had a lower molecular mass of 9 kDa (hence the


name calbindin-D 9k), compared with the 28 kDa of the
avian isoform, was later identified (138, 280, 283, 358).
Concerning the functional role of calbindin-D 9k, it had
already been speculated very early after its discovery that it
may serve as a calcium shuttling protein (951). Indeed, initial calculations and later very basic experimental data confirmed that calbindin-D 9k may mediate facilitated diffusion of calcium between the two poles of the enterocyte, in
analogy to the transport of oxygen by myoglobin in the
muscle (321, 602). In a very fundamental investigation,
calcium flux was measured between chambers that were
separated by dialysis membranes. A 51% increase in transchamber calcium flux occurred in the presence of calbindin-D 9k (321). However, it is hard to relate this number to
physiological values, given the simplification underlying the
experimental model. It has subsequently been calculated
that calbindin-D 9k may facilitate diffusion of calcium by a
factor of up to 60 (129). Rather than envisioning calbindin-D 9k as a protein that individually shuttles calcium ions
across the cell, one should imagine calbindin-D 9k as a
calcium gradient amplifier (129, 602). As the intracellular
concentration of free calcium is in the nanomolar range, the
intracellular gradient of free calcium between the apical and
basolateral pole can also only be in the nanomolar range.
Diffusional flux, however, directly correlates with the concentration gradient and can in consequence only be very
small. Calbindin-D 9k is expressed in the enterocyte in the
micromolar range and dynamically binds and releases calcium (714). Since the association between calbindin-D 9k
and calcium is dynamic, the local concentration of calbindin-D 9k-bound calcium will directly correlate with the
concentration of free calcium. It is thus the concentration
gradient of calbindin-D 9k-bound calcium throughout the
cell that determines the flux rate of calcium. As this gradient
can be in the micromolar range, calbindin-D 9k serves as a
calcium gradient amplifier (129, 602). It should be noted
that the real experimental data on calbindin-D 9k are fairly
scarce and that the bulk of scientific effort has gone into
mathematical modeling of facilitated diffusion and calcium
binding kinetics (129, 321, 322, 602, 1021).
Functionally, several correlations between calbindin-D 9k
and calcium uptake were identified. The calbindin-D 9k
content of each intestinal segment correlates linearly with
its ability for calcium uptake (129, 824, 1021). Although
this relation holds true on the experimental and the mathematical modeling level, it does not prove causality (129,
824, 1021). Furthermore, 1,25(OH)2-vitamin D and a lowcalcium diet induce calbindin-D 9k on both the mRNA and
protein level, suggesting that it is involved in the process of
regulated calcium absorption (289, 604). In accordance
with these findings, VDR-deficient animals have a decreased calbindin-D 9k mRNA content that cannot be rescued by exogenous 1,25(OH)2-vitamin D administration
(663, 1194). Although a 1,25(OH)2-vitamin D responsive

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

gation provides another explanation for the initially observed partial sensitivity of transcellular calcium uptake to
L-type calcium channel blockers. The voltage-gated calcium channel Cav1.3, a member of the L-type calcium channel family, was recently identified in the apical membranes
of the distal jejunum and proximal ileum (751). Previous
observations were emulated, as the investigators again demonstrated a decrease in calcium absorption following application of L-type calcium channel inhibitors in the corresponding segments (751). The authors argued that uptake
through Cav1.3 may have previously been misinterpreted as
paracellular calcium uptake (751). However, it should be
noted that the calcium uptake assay used in this report did
not discriminate between transcellular and paracellular calcium movement. Subsequently, it has been observed that
L-type inhibitor-sensitive calcium flux is linked to stimulation of glucose uptake through the glucose transporter type
2 (GLUT2) (692, 750). Cav1.3 may serve as an alternative
calcium entry pathway that is active in states of luminal
calcium abundance and that coregulates glucose absorption
(692, 750). Further investigations will be needed to determine the contribution of Cav1.3 to dietary calcium absorption.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


element (VDRE) had been identified in the 5=-flanking region of the calbindin-D 9k gene, later mutational analysis
demonstrated that this site was not essential for transcriptional regulation of calbindin-D 9k by 1,25(OH)2-vitamin
D (217, 240). Hence, it is not clear how 1,25(OH)2-vitamin
D exactly regulates calbindin-D 9k on a molecular level.
Interestingly, intestinal calbindin-D 9k mRNA levels can be
rescued in VDR (/) animals by a high-calcium/phosphorus/lactose diet, while extraintestinal calbindin-D 9k
mRNA levels are unaffected (663). This suggests that, apart
from endocrine regulation through 1,25(OH)2-vitamin D,
intestinal calbindin-D 9k is also regulated by local factors
(663). A direct short-term stimulatory effect of oral calcium
intake on calbindin-D 9k levels had been observed before in
a wild-type background (647). The mechanisms underlying
the effect of diet on calbindin-D 9k levels are, however, still
obscure.

In light of the insights gained through knockout animals,


the role of calbindin-D 9k remains somewhat unclear.
Again, it is difficult to dissect the physiological function of
calbindin-D 9k in these animal models, given the assumption that the organism will pursue compensation for the loss
of a gene product. In conclusion, it is undisputed that calbindin-D 9k is regulated by 1,25(OH)2-vitamin D, that it
can bind calcium and that theoretical modeling and very
fundamental experimental models verify that it can facilitate diffusion of calcium across the enterocyte. The (/)
models suggest that animals can compensate for the loss of
calbindin-D 9k and maintain normal calcium homeostasis
(13, 76, 613, 639). Furthermore, TRPV6, calbindin-D 9k
and double (/) animals can still increase their calcium
uptake in response to 1,25(OH)2-vitamin D or a low-calcium diet, respectively, albeit not to the same extent as
wild-type animals (13, 76, 615). One may conclude that
neither of these proteins is necessary for transcellular uptake; however, it seems more likely that 1,25(OH)2-vitamin
D has more targets than previously postulated and that
compensatory mechanisms are in place.
3. Basolateral calcium extrusion
Once calcium reaches the basolateral membrane, it is extruded into the extracellular space, which completes the
process of intestinal absorption. As extracellular calcium
concentrations are higher than cytosolic concentrations,
this process requires energy. Two proteins are responsible
for this task. 1) The PMCA extrudes calcium at the direct
expense of ATP, whereas 2) the sodium-calcium exchanger
(NCX) utilizes the energy stored in the sodium gradient to
transport calcium out of the cell (FIGURE 3). It is this basolateral exit process that requires energy during transcellular
calcium uptake and that allows us to absorb calcium against
an uphill gradient when dietary concentrations are low.
A) THE PLASMA MEMBRANE CALCIUM ATPASE. The plasma membrane calcium ATPase (PMCA) is a virtually ubiquitous
protein that is responsible for intracellular calcium homeostasis by pumping calcium into the extracellular milieu,
thereby keeping intracellular concentrations low. The
pump belongs to the family of P-type primary ion transport
ATPases, which among others also includes gastric H-KATPase (see sect. IIA1). Four isoforms of the protein exist;
however, variety is increased by splicing (562, 1043). The
PMCA1b splice variant is most predominant in the small
intestine and the duodenum in particular, which suggest an
involvement of this isoform in the process of transcellular
calcium absorption (341, 492). Given the ubiquitous expression of PMCA, a detailed review of its structure and

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

209

Downloaded from on December 8, 2014

Despite the correlation between calbindin-D 9k levels and


1,25(OH)2-vitamin D and the mathematical and experimental models, which support its role as a calcium shuttling
protein, no clear evidence for the involvement of calbindin-D 9k in transcellular calcium absorption existed. This
had sparked controversy among investigators. In fact, a
very early study concluded that although 1,25(OH)2-vitamin D induced both calbindin-D 9k and calcium absorption, a discrepancy existed in the time course of both effects
(424). Later, synthetic 1,25(OH)2-vitamin D derivatives,
designed for a maximized effect on cell differentiation and
suppressed calciotropic activity, failed to increase serum
calcium levels while causing a sevenfold increase in calbindin-D 9k mRNA [native 1,25(OH)2-vitamin D increases
both serum calcium and calbindin-D 9k mRNA](604). In
2006, a calbindin-D 9k (/) mouse was created by Kutuzova et al. (613). The animals presented with no apparent
phenotype abnormalities and had normal serum calcium
concentrations (613). Subsequent analysis of these animals
showed that they responded equally well to 1,25(OH)2vitamin D with regard to calcium absorption as wild-type
animals (13). Of note, calcium absorption was assessed by
measuring serum calcium following gavage of a calcium
radioisotope and did not discriminate between transcellular
and paracellular absorption (13). A different group replicated the findings made in the calbindin-D 9k (/) mouse
and also did not find any appreciable variation in phenotype or serum calcium concentrations (639). However, the
investigators observed that during preweaning, when both
calcium demand and absorption are at their peak, gene
expression of TRPV6 and the basolateral calcium extruder,
plasma membrane calcium ATPase isoform 1b (PMCA1b),
were highly induced in the calbindin-D 9k (/) animals
(128, 639). This has been interpreted as a compensatory
upregulation of calcium transport proteins to ensure sufficient uptake in a state of high metabolic demand for calcium (639). Subsequently, a TRPV6 and calbindin-D 9k
double (/) animal was created. The mature animals

show no disturbances in calcium homeostasis, although,


similarly to TRPV 6 (/) animals, they cannot increase
calcium uptake in response to a dietary calcium challenge to
the same extent as wild-type animals (76).

SASCHA KOPIC AND JOHN P. GEIBEL


function will be omitted. For a current and detailed review,
please refer to Reference 1043.
PMCA was first characterized in the 1960s in the membrane
of erythrocytes (956). Very early experiments in basolateral
membranes isolated from enterocytes identified a calciumdependent enzyme with phosphatase activity, which served
as first evidence for PMCA in the intestine (100, 742). Subsequent investigations in basolateral vesicles from rat intestine concluded that two distinct calcium transport mechanisms existed in their membranes: an ATP-dependent
(PMCA) and a sodium-dependent (NCX) mechanism (382,
457, 573). Furthermore, it was shown that inhibition of
calmodulin halved the amount of ATP-dependent calcium
transport (573). Today, we know that PMCA is highly regulated by CaM, which can increase both the affinity of the
pump to calcium and its turnover speed by a factor of up to
10 (526, 627, 628, 756).

B) THE NCX.

Long before the molecular identities of PMCA


and NCX were known, it had been observed that calcium
uptake in the intestine was dependent on extracellular sodium (713). The authors concluded that a sodium, calciumexchange diffusion carrier may exist at the basal membrane
of the cell, which proved to be a remarkably accurate
prediction (713). As mentioned before, later investigations
delineated between an ATP and a sodium-dependent transport of calcium on the basolateral enterocyte membrane
(382, 457, 573).

210

In addition, NCX is not subjected to regulation by 1,25(OH)2vitamin D, which further contributes to the ambiguity concerning its importance in the process of calcium absorption.
This has been observed very early, when exogenous administration of 1,25(OH)2-vitamin D to vitamin D-deficient
animals did not increase sodium-dependent calcium transport, while doubling transport through PMCA (382). Furthermore, it was recently shown that a calcium-depleted
diet decreases duodenal NCX1 mRNA levels (583).
Of note, two members of the potassium-dependent sodium
calcium exchanger (NCKX) family, namely, NCKX 3 and
4, were also identified in the small intestine (658, 687, 688).
However, further functional investigations will be needed
to clarify their role.

B. Paracellular Calcium Absorption


If we plot the amount of duodenal calcium absorption as a
function of the luminal calcium concentration, we can observe that the absorption curve is comprised of two distinct
kinetic components: a saturable/exponential component
and a nonsaturable/linear component (824, 825, 1134).
The saturable component represents transcellular calcium
uptake through the enterocyte, as both the number of calcium transport proteins and their turnover rate is limited.
The nonsaturable component reflects calcium uptake
through the paracellular pathway. The saturable compo-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Interestingly, it has also been suggested that calbindin-D 9k


can directly stimulate calcium transport via PMCA (1136,
1137). In addition to intracellular proteins, PMCA is also
quantitatively regulated by endocrine factors. 1,25(OH)2vitamin D can increase both the PMCA mRNA and protein
content in enterocytes (35, 36, 62, 146, 382, 489, 614, 823,
1109; contested by Ref. 1138). The data on the effects of a
low-calcium diet on PMCA transcription are more controversial, as one study suggests induction whereas two other studies
observed reduced mRNA levels (146, 583, 639). Of note,
these observations were made in different species (chick vs.
mouse). The sensitivity of the tissue to 1,25(OH)2-vitamin D is
also reported to decrease with age, which may represent
another confounding factor underlying these observations
(35, 36). In conclusion, the transcriptional regulation of
PMCA1b by 1,25(OH)2-vitamin D and its decreasing expression along the length of the small intestine correlate
well with the canonical model of transcellular calcium absorption. It is challenging to investigate the functional
contribution of PMCA1b in the process of calcium absorption, given its role as a housekeeping protein.
PMCA1 is crucial during development, which results in
embryonic death if knocked out (814, 1198). Conversely,
heterozygote (/) animals present with no apparent
phenotype, albeit parameters linked to calcium homeostasis were not assessed (814).

For a recent review on the structure and function of NCX,


please refer to Reference 687. In brief, NCX exists in three
isoforms (NCX13), which are expressed in a broad variety
of cell types (687). The function of NCX has mainly been
investigated in excitatory tissues, given its role as a highcapacity calcium extrusion mechanism following excitation. To transport calcium against its strong electrochemical gradient, NCX has to utilize three sodium ions to accomplish extrusion of one calcium ion (85, 460). NCX1 is
the predominant isoform in the small intestine, where it has
been detected on the mRNA and the protein levels (275,
688). However, NCX had been postulated to play a more
important role during calcium absorption in the kidney
than in the enterocyte, hence not much data on intestinal
NCX are available to us (473). Furthermore, genetic disruption of NCX1 is embryonically lethal, which imposes
some limitations on our experimental methodology (600).
A more recent study provides some functional evidence for
NCX in the intestine. By measuring intracellular calcium
concentrations with a fluorescent indicator dye, Dong et al.
(275) demonstrated that calcium uptake in a sodium-free
environment (to run NCX in its reversed configuration) was
significantly decreased when NCX was pharmacologically
inhibited. Despite our knowledge that NCX exists in enterocytes and that it can extrude calcium under experimental conditions, it is difficult to assess its relative contribution
to transcellular calcium absorption.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH

Epithelial tight junctions are adhesion points between two


neighboring cells that seal their intercellular space against a
lumen, thereby restricting ion and water movement between the two compartments. As water cannot be directly
transported, its movement is tied to ion fluxes, which in
turn are determined by their respective concentration gradients across the epithelium. Each epithelial cell expresses
an annulus of tight junction proteins at the apical end of the
lateral membrane. Tight junctions are protein complexes
that consist of a variety of intra- and transcellular proteins.
A detailed review of their structure would exceed the scope
of this article (for a recent review, please refer to Ref. 999).
Briefly, the composition of the involved proteins determines
the pore size, ion selectivity, and thereby the relative leakiness of each tight junction and the whole epithelium in
general. Furthermore, tight junctions create a lateral diffusion barrier for membrane proteins and help to maintain the
functional polarity of the epithelial cell. It is important to
recognize that tight junctions are not static complexes, but
can be rather dynamically regulated (999). This allows the
epithelium to change its ion and water permeability in response to various stimuli (999).
As discussed previously, the rate of the nonsaturable
component of calcium absorption is documented to be
fairly constant and insensitive to 1,25(OH)2-vitamin D,
which indicated that 1,25(OH)2-vitamin D may have no
effect on paracellular transport (824, 825, 1224). However, in the late 1990s, Chirayath et al. (201) postulated
that 1,25(OH)2-vitamin D can increase paracellular calcium flux in confluent Caco-2 cell cultures, which form

an epithelia-like monolayer with tight junctions. This


conclusion is based on the observations that 1,25(OH)2vitamin D decreased the transepithelial electric resistance, which is often used as a measure of tight junction
permeability, and induced bidirectional, i.e., also serosal
to mucosal, calcium flux in the cultured monolayers
(201). A more recent investigation further substantiated
this hypothesis. It has been shown that 1,25(OH)2-vitamin
D regulates the mRNA levels of some tight junction proteins
in rat duodenum, which may alter their gating characteristics (354, 614). For example, the mRNA levels of claudin-3,
a protein that directly determines tight junction permeability, were decreased 2.2-fold following 1,25(OH)2-vitamin
D administration (614). Conversely, claudin-2 and -12
mRNA and protein levels were increased (354). Functional
observations in Caco-2 monolayers further demonstrate
that overexpression of claudin-2 and -12 increases electrical
conductivity and facilitates paracellular calcium flux (354).
All of these events are tied to a 1,25(OH)2-vitamin D-mediated promotion of transcriptional events. However, it is
known that 1,25(OH)2-vitamin D can also exert short-term
nongenomic effects (1122) (see sect. IVA6B). A recent investigation presents evidence for augmented solvent-drag
paracellular calcium flux after acute administration of
1,25(OH)2-vitamin D (1098). Solvent-drag mediated flux
may occur at low calcium concentrations in the absence of
a calcium gradient. In this model, calcium is dragged
through the paracellular space by water flux that is fueled
by the hyperosmolar milieu in the paracellular space (266,
626). 1,25(OH)2-vitamin D was shown to induce this calcium flux in the presence of initially equimolar calcium
concentrations between the mucosal and serosal compartments (1098).
Over the last years, evidence for the regulation of the paracellular pathway by 1,25(OH)2-vitamin D has slowly accumulated. It is apparent that the canonical dogma which
postulates that 1,25(OH)2-vitamin D only targets the transcellular pathway has to be revisited. Still, more experimental investigations will be needed to ultimately clarify the
effects of 1,25(OH)2-vitamin D on paracellular calcium
transport. Should tight junctions indeed be subjected to regulation by 1,25(OH)2-vitamin D, this mechanism may partially
explain the sensitivity of calcium uptake in TRPV6 and calbindin-D 9k (/) animals to 1,25(OH)2-vitamin D.

C. Alternative Models of Transcellular


Calcium Absorption
Two alternative models of transcellular calcium absorption
have been put forward. One suggests that calcium is transported through the enterocyte by vesicles (vesicular transport model) rather than facilitated diffusion, and the other
postulates that 1,25(OH)2-vitamin D can induce intestinal calcium absorption in a rapid, nongenomic fashion via a putative
1,25(OH)2-vitamin D surface receptor (transcaltachia).

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

211

Downloaded from on December 8, 2014

nent is less pronounced in the jejunum and disappears completely in the ileum, indicating that transcellular calcium
absorption is restricted to the proximal segments of the
intestine, as discussed previously (825) [this has been contested more recently, when transcellular flux was also noted
in the ileum (46)]. Compared with the transcellular pathway, the paracellular route has not received much scientific
attention. It has been put forward that the rate of paracellular calcium absorption is constant across the length of the
intestine and is neither sensitive to 1,25(OH)2-vitamin D
nor a low-calcium diet (824, 825, 1224). However, provided that enough dietary calcium is available to saturate
the transcellular pathway, observations indicate that net
calcium absorption is highest in the ileum, which has been
attributed to the sojourn time, rather than alterations in
paracellular permeability (290, 710). In the rat, the chyme
spends some 74% of its transit time in the ileum, which
allows for a long exchange period between lumen and
plasma (290). The diffusion rate itself is fairly low and only
amounts to 2% of the rate if calcium were to diffuse freely
between intestinal lumen and plasma (290). This effect is a
consequence of the diffusion barrier that tight junctions,
which act as functional gating molecules of the paracellular
pathway, impose on calcium flux.

SASCHA KOPIC AND JOHN P. GEIBEL


The assumption that calcium is transported through the cell
in vesicles is corroborated by a handful of observations. It
has been demonstrated that 1,25(OH)2-vitamin D treatment increased the number of supranuclear lysosomes in
rachitic chicks, compared with nontreated animals (247). It
was later shown that the calcium content of lysosomes can
more than double following treatment with 1,25(OH)2-vitamin D, suggesting that they may play a role in the process
of transcellular calcium movement (780). Compared with
the cumulative evidence that is in accordance with our canonical model of calcium absorption, the role of vesicular
transport of calcium is still fairly obscure.

IV. REGULATION OF
CALCIUM HOMEOSTASIS
Eucalcemia is maintained by the concerted effort of vitamin
D, PTH, and, to a lesser extent, calcitonin. All three hormones can influence serum calcium concentrations by acting on the intestine, the kidney, or bone. 1,25(OH)2-vitamin D, the active vitamin D metabolite, primarily modulates the intestinal absorption of calcium and will therefore
be discussed in most detail (FIGURE 4). Apart from hormonal regulators that influence the absorption, excretion,
and deposition of calcium, our body needs a mechanism
that allows it to sense the current levels of plasma calcium.
This task is fulfilled by the CaSR. It oversees the precise
regulation of the calcitropic hormones.

A. Vitamin D
As discussed previously, vitamin D is one of the key regulators of calcium homeostasis. Our body has two sources
for vitamin D, namely, a dietary source (vitamin D23) and
an endogenous source that relies on ultraviolet (UV) light
catalyzed synthesis in the skin (vitamin D3) (FIGURE 5).

212

1. Historical perspective
From a historical perspective, the identification of vitamin
D is closely intertwined with attempts to understand the
pathophysiology of rickets. Rickets is characterized by
childhood skeletal deformities resulting from inadequate
osteoid mineralization and calcification of cartilage due to
decreased serum calcium levels during development. The
adult form equivalent of rickets is termed osteomalacia.
With the onset of industrialization, rickets became a prevalent problem in the 18th, 19th, and the beginning of the
20th century. In fact, the disease was so widespread at the
beginning of the 20th century that an investigation conducted by the German pathologist Schmorl on 386 children,
who had died before the age of 4 years, concluded that 90%
of them had had rickets (968). Even in present times nutritional rickets still remains a major public health concern in
developing countries (303, 757, 809). The seminal observations that led to the identification of vitamin D were provided by Mellanby in 1919 (733). He observed that dog
pups who were fed a severely restricted diet consisting of
porridge or bread were consistently developing rickets
(733). Development of rickets could be averted if their diet
was supplemented with cod liver oil, which we now know
to contain a high concentration of vitamin D (733). Mellanby (733) concluded that rickets is a deficiency disease
which develops in consequence of the absence of some accessory food factor or factors. Vitamin A had been discovered shortly before, and it was subsequently speculated that
it may represent the factor promoting bone formation
(733). This hypothesis was later rebutted by American biochemist McCollum who concluded that a substance which
is distinct from fat-soluble [vitamin] A must be responsible
for preventing rickets (727). Furthermore, he stated that his
experiments demonstrate the existence of a fourth Vitamin [vitamin D] whose specific property [. . .] is to regulate
the metabolism of bones (727). In parallel to the unraveling of the dietary component of rickets, scientists were independently discovering the importance of sunlight for disease prevention. The Polish pediatrician Raczynski was most
likely the first to demonstrate evidence for this hypothesis experimentally (881). He kept one dog pup in the shade while a
littermate was kept in the sunlight. Both dogs were breastfed
by their mother. After 6 wk, the bones of the dog that was kept
in the shade contained 36% less calcium (881). These observations were followed up by the German pediatrician Huldschinsky, who healed rachitic children after exposing them
intermittently for 2 months to the UV rays generated by a
mercury vapor quartz lamp (504). Hess and Unger (447) rep-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

The model of transcaltachia mainly relies on the observation that 1,25(OH)2-vitamin D can exert effects that are too
acute to be attributable to transcriptional events. For example, intestinal calcium absorption was increased in chicks
within 14 min of 1,25(OH)2-vitamin D exposure, an onset
which is too rapid as to be of a genomic nature (785). It
should be noted that rapid effects of 1,25(OH)2-vitamin D
have also been suggested to influence the paracellular pathway (see above). Undoubtedly a careful evaluation of the
route of calcium flux, i.e., transcellular versus paracellular,
is necessary. At least, isolated intestinal cells respond with
increased uptake of radiolabeled calcium to acute 1,25(OH)2vitamin D exposure (568). It has been postulated that apart
from the VDR, a membrane-bound 1,25(OH)2-vitamin D exposure receptor, the 1,25(OH)2-vitamin D-MARRS (membrane-associated, rapid response, steroid binding) protein, mediates the acute effects of 1,25(OH)2-vitamin D (568, 779,
785).

Nomenclature in this case is fairly misleading, as vitamins are


per definition substances that cannot be generated by our body
and have to be ingested from an external source. The fact that
we can synthesize vitamin D3 in our skin classifies the substance as a prohormone rather than a vitamin. As we will see,
our current nomenclature is a byproduct of the historic events
leading to the discovery of vitamin D.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


Hypocalcemia
Stimulates PTH secretion

Parathyroid glands

Increases bone resorption

Bone

PTH

Increases renal calcium absorption


Stimulates 1,2
1,25(OH)2-D synthesis

Downloaded from on December 8, 2014

Kidney

Intestine

eases renal
Increases
calcium absorption
Increases intestinal
calcium absorption
1,25(OH)2-D

FIGURE 4. Endocrine regulation of serum calcium levels. Calcium homeostasis is mainly regulated by PTH and
1,25(OH)2-vitamin D. Both hormones act at their respective target organs to increase serum calcium levels.

licated these findings by using sunlight treatment. Hess (446)


later summed up the impact of these revolutionary observations, which now seem intuitive to us: We have known that a
growing plant cannot thrive in the dark, but have failed to
realize that the same laws apply to growing animals.
It was later recognized that it was sufficient to irradiate the
food administered to animals rather than the whole animal
to prevent development of or heal rickets. Thus initially
inert dietary substances with no antirachitic properties
could be activated by UV light (385, 448 450, 505, 1037).
It was concluded that irradiation caused the conversion of a
biological precursor to an active form and that the same
mechanism was physiologically taking place in the skin.
Initially, it was speculated that cholesterol may serve as this

pro-vitamin. It was Windaus and Hess (in collaboration


with Rosenheim) who were the first to uncover its exact
molecular identity. They stated: We conclude from our
experiments with complete certainty that ergosterol [. . .]
represents the anti-rachitic provitamin (1166). In 1928,
Windaus received the Nobel Prize in Chemistry for the
services rendered through his research into the constitution
of the sterols and their connection with the vitamins. The
irradiation product of ergosterol was later purified and
named vitamin D2 (ergocalciferol) (43, 896, 1164, 1167).
Although these findings solved the question as to how UV
irradiation generates Vitamin D2 from ergosterol, which has
antirachitic properties if ingested, the molecular mechanisms
underlying the antirachitic effects of cutaneous sunlight exposure still remained obscure. Since ergosterol is an exclusive

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

213

SASCHA KOPIC AND JOHN P. GEIBEL


Keratinocyte
Dietary vitamin D

7-dehydrocholesterol
UV-B (290-315nm)

Intestinal uptake

Previtamin D
Isomerization

Vitamin D

Vitamin D

Vitamin D

DBP

DBP
Hepatocyte

1,25(OH)2-vitamin D

Vitamin D

Mitochondria:
CYP27A1

ER:
CYP2R1

Downloaded from on December 8, 2014

25(OH)-vitamin D

25(OH)-vitamin D

DBP

Proximal tubule

Glomerular
filtration

25(OH)-vitamin D
PTH

25(OH)-vitamin D

DBP

Mitochondria:
CYP27B1

Megalin
Calcium

1,25(OH)2-vitamin D

Target organs

FIGURE 5. Vitamin D metabolism. Vitamin D can either be synthesized in the skin or absorbed from our diet.
It is then transported to the liver where it undergoes 25-hydroxylation by one of two hepatic enzymes (CYP27A1
or CYP2R1). During transport through the circulation, vitamin D is bound to a carrier protein (DBP). The
25(OH)-vitamin-D-DBP complex passes the glomerular filter and is scavenged from the primary urine by the
apical megalin receptor of the proximal tubule. Here, 25(OH)-vitamin D is converted to the active vitamin D
metabolite 1,25(OH)2-vitamin D. DBP, vitamin D binding protein.

component of yeast and fungal membranes, a different precursor substance had to exist in animal skin. Again, it was
Windaus and colleagues who identified 7-dehydrocholesterol
as the provitamin in porcine skin, which is converted to vitamin D3 (cholecalciferol) under irradiation (1165). After these
discoveries, industrially produced vitamin D has rapidly been
used in medical applications and as a food fortification. To-

214

day, the main portion of dietary vitamin D ingestion in the


United States stems from fortified dairy products (150).
2. Intestinal vitamin D absorption
To exert its antirachitic effects, dietary vitamin D has to be
absorbed into our circulation. Early everted gut sack exper-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH

Following uptake into the enterocyte, vitamin D is packed


into chylomicrons and secreted into the lymph (288, 953). It
has been demonstrated that after intestinal administration
of radioactive labeled vitamin D3, up to 90% of the recovered radioactive tracer was associated with the chylomicron
fraction of the collected intestinal lymph (288). Furthermore, patients suffering from the autosomal recessive chylomicron retention disease (Anderson disease; OMIM
246700), which causes impairment of chylomicron processing and secretion in the enterocyte, can present with insufficient levels of fat-soluble vitamins, such as vitamins D and
E (535). The chylomicron remnants, which include vitamin
D, are then scavenged by the liver after the lymph has entered the circulation through the thoracic duct (287, 407).
However, it has been demonstrated in vitro and in vivo in
hepatectomized and normal rats that vitamin D can directly
transfer from chylomicrons to a vitamin D binding protein
(DBP; see sect. IVA5) in the blood plasma (286, 288). It has
therefore been suggested that at least a fraction of hepatic
vitamin D uptake is mediated through DBP rather than
chylomicrons (286; contested by Ref. 407). In the liver,
vitamin D is then hydroxylated at its 25 position to
25(OH)-vitamin D. The metabolism of vitamin D will be
the subject of a later section.
The intestinal absorption of 25(OH)-vitamin D has been
investigated by many groups, with the aim of optimizing
vitamin D administration in a therapeutic context by bypassing the first metabolic step in the liver (219, 245, 287,
445, 645, 721, 1018, 1019, 1127). In general, enteric up-

take of 25(OH)-vitamin D is more effective than that of


vitamin D, which is partially attributable to a comparably
lower dependency on bile acid secretion (219, 245, 287,
645, 1018, 1019). The observation that patients with
cholestasis still absorb 25(OH)-vitamin D effectively,
whereas vitamin D absorption is impaired, corroborates
this hypothesis (1018). There is some controversy with regard to the transport of 25(OH)-vitamin D following its
absorption (287, 701, 1019). It has been argued that it may
be transported predominantly in the protein fraction of the
lymph (287), i.e., not in chylomicrons, or that it is directly
absorbed into the portal blood (701, 1019).
3. Cutaneous vitamin D synthesis
Cutaneous synthesis is our second source of vitamin D.
Cutaneous production depends on exposure to UVB (290
315 nm) light (FIGURE 5). The UVB photons convert 7-dehydrocholesterol, which is located in the plasma membrane
of keratinocytes, to previtamin D3 (11171120). This dependency on sunlight causes a seasonal variation in vitamin
D3 production, with synthesis being low during the winter
months when the radiation angle of the sun flattens (187,
478, 538, 1035, 1156). In consequence, changes in latitude
result in similar variability in production. Further factors
that decrease production include pigmentation of the skin
and application of sunscreen (209, 723), whereas an increase in altitude promotes production (478). Following
conversion from 7-dehydrocholesterol, previtamin D3
isomerizes to vitamin D3 (479). The isomerization process
is temperature dependent and fairly slow (479, 1117). It has
been calculated that the half-life for the formation of vitamin D3 is 2.5 h (1085). Interestingly, in vitro experiments
conducted in isotropic medium demonstrated that the
isomerization rate was 10 times slower than in in vivo experiments (1085). This was later attributed to the fact that
amphiphatic interactions with phospholipids of the cell
membrane stabilize the previtamin D3 conformer which
then isomerizes to vitamin D3 (1086). The cellular microenvironment of the reaction thus greatly optimizes the isomerization to vitamin D3. After its synthesis, vitamin D3 is
bound to DBP and carried though the bloodstream to its
target organs. Observations made in patients indicate that
the vitamin D3 plasma levels peak 2 days after sunlight
exposure, which is due to the slow isomerization rate in the
skin (7).
4. Vitamin D metabolism and its regulation
Irrespective of the source (endogenous or exogenous), vitamin D is metabolized in the liver to 25(OH)-vitamin D
(FIGURE 5). Evidence for the existence of biologically active
vitamin D metabolites emerged in the 1960s (686, 799).
25(OH)-vitamin D was identified by means of injecting rats
with radiolabeled vitamin D and subsequent silicic acid column chromatography of lipid extracts from serum and var-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

215

Downloaded from on December 8, 2014

iments demonstrated that this process has linear, nonsaturable, and passive kinetics, suggesting that no specific carrier mechanism for vitamin D is in place (484). These observations were later replicated in in vivo models (483).
Absorption is highest in the proximal and mid small intestine (484). Since vitamin D is fat soluble, its absorption
mechanism is similar to that of dietary lipids. In an aqueous
media, vitamin D aggregates in micelle-like structures
(735). Its absorption is aided by the secretion of bile acids,
which is underscored by the observation that patients suffering from cholestasis can present with vitamin D deficiency and develop bone disease, such as osteomalacia or
osteoporosis (245, 445, 563, 721, 894, 953, 1018, 1080).
Apart from bile salts, formation of mixed micelles containing monoglycerides and free fatty acids represents another
factor that aids in vitamin D absorption (884, 1082; contested by Ref. 483). These substances increase micelle size,
which promotes the solubilization of vitamin D, thereby
increasing uptake (884). Clinically, pancreatic insufficiency, causing an impairment of triglyceride breakdown
through insufficient lipase secretion, leads to decreased vitamin D absorption (1080, 1127). This is a particular problem in cystic fibrosis patients, who often develop pancreatic
and concomitant vitamin D insufficiencies (33, 230, 625,
678, 924).

SASCHA KOPIC AND JOHN P. GEIBEL

The mitochondrial 25-hydroxylase (CYP27A1) was first


purified to homogeneity from rabbit liver mitochondria
(235). It was demonstrated that this cytochrome P-450 was
not specific to vitamin D and could also hydroxylate other
substrates, most notably cholesterol (27-hydroxylation),
which represents an important step in the formation of bile
acids (235). In retrospect, CYP27A1 had been purified 4
years earlier; however, only the 27-hydroxylation of cholesterol had been investigated and its effects on vitamin D
had remained obscure (1162). Subsequently, the enzymes
cDNA was cloned from rabbit, rat, and human, and its dual
role in vitamin D and steroid conversion was confirmed (26,
149, 1048, 1103). The full gene structure was identified a
few years later (646). Although no crystal structure of the
enzyme is available to us, a homology model based on other
CYP family members has been proposed (874). In the liver,
CYP27A1 is expressed on the mRNA level in hepatocytes,
endothelial, stellate, and Kupffer cells (1078). Furthermore,
CYP27A1 expression was confirmed in a variety of other
tissues, including duodenum, adrenal glands, kidney, lung,
vascular endothelium, brain, retina, skin, muscle, and osteoblasts; however, their potential contribution to vitamin
D metabolism remains unclear (26, 51, 135, 184, 370, 383,
508, 640, 898, 979). Interestingly, an extrahepatic conversion of vitamin D had been suggested previously to occur in
the kidney and the intestine (1097). Marked differences in
CYP27A1 activity can be observed between males and females. For example, CYP27A1 enzyme activity and mRNA
expression were demonstrated to be increased in female rats

216

(933, 1078). Higher expression in females was also confirmed in biopsy samples from human subjects (370). A
regulation via sex hormones may underlie this phenomenon, as injection of estradiol was shown to induce
CYP27A1 activity (933). Interestingly, seasonal variations
in expression were also observed, which may represent a
confounding factor for decreased 25(OH)-vitamin D levels
during the winter months (370).
It should be noted that CYP27A1 can also hydroxylate
vitamin D3 at other positions (402, 950). These include
positions 27 and 26; however, the ratio for 25-:27-:26hydroxylation has been estimated to be only 100:15:3,
which demonstrates that 25-hydroxylation of vitamin D3 is
the most essential reaction catalyzed by the enzyme (950).
More importantly, CYP27A1 can also use its own product
25(OH)-vitamin D as a substrate to further act as a 1hydroxylase and produce the hormonally active form of
vitamin D, namely, 1,25(OH)2-vitamin D (50, 51, 950). As
will be discussed later, this reaction is normally catalyzed in
the kidney by another CYP family member (CYP27B1). At
the moment, it is not clear what the physiological significance of 1-hydroxylation by CYP27A1 is.
Mutations in CYP27A1 cause the autosomal recessive disorder cerebrotendinous xanthomatosis (CTX; OMIM
213700). The disease was first described in 1937 and is
characterized by cholestanol deposits that are most prominent in tendons, especially the Achilles tendon, the brain,
and the lung. Patients present with progressive neurologic
defects, atherosclerosis, and cataracts and commonly suffer
from diarrhea. The inadequate bile acid synthesis was first
noted in 1974 by Setoguchi et al. (993). Shortly after the
cDNA of CYP27A1 was cloned, it was demonstrated that
mutations of this enzyme were responsible for CTX (148).
In agreement with the dual role of CYP27A1, CTX patients
also suffer from osteoporosis, low 25(OH)-vitamin D levels, and impaired intestinal calcium absorption (83, 320).
Three CYP27A1 mutations that are known to cause CTX
and still lead to protein expression were recreated in vitro,
and enzymatic activity was assayed. Depending on the expression system, these mutants showed lower or higher 25hydroxylation activity than the wt enzyme, which led the
authors to questions the enzymes role in vitamin D metabolism (403). It should be considered that 1) many more
(38) mutations underlying CTX exist, 2) by far not all
patients exhibit disturbances in bone or vitamin D homeostasis, and 3) the investigated mutant enzymes may only
cause disturbances in cholesterol, rather than vitamin D
metabolism (83, 320, 403). In the light of these limitations,
it should be questioned whether CTX represents an apt
model system to evaluate CYP27A1 in the context of vitamin D metabolism.
With the introduction of novel genetic tools, a cyp27a1
(/) mouse was created in 1998 (918). However, the phe-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

ious tissues (686). Chromatography revealed the presence


of a vitamin D metabolite in serum, liver, bone, and feces in
rats and in human serum (256, 686). Furthermore, it was
shown that the vitamin D metabolite was biologically active
by reverting rickets in diseased animals and by increasing
intestinal calcium transport and bone metabolism (686,
752). The metabolite was characterized as 25(OH)-vitamin
D and subsequently successfully synthesized (111, 112). It
was soon recognized that hepatectomized rats were not effectively converting vitamin D to 25(OH)-vitamin D, suggesting that the liver was the main organ responsible for
25-hydroxylation (864, 865). The ability of the liver to
metabolize vitamin D was also confirmed in perfused livers
and tissue homogenates (490). Historically, a long controversy existed with regard to the subcellular localization of
the enzyme that hydroxylates vitamin D (25-hydroxylase)
in the liver, as enzyme activity was observed in both mitochondrial and microsomal fractions of liver homogenates
(93, 102, 103, 258, 696, 1053). Extensive investigations
indicated that both the microsomal and mitochondrial 25hydroxylase are members of the cytochrome P-450 family
(102104, 696, 835). Both fractions demonstrated distinct
enzymatic kinetics. The mitochondrial enzyme was characterized as a low-affinity high-capacity enzyme, whereas the
microsomal fraction displayed high-affinity low-capacity
characteristics (103, 104, 356, 696).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


notype of CTX could not be reproduced, albeit fecal bile
acid content was markedly decreased (918). Rather surprisingly, serum 25(OH)-vitamin D levels were increased in
(/) animals, which either pointed to compensatory upregulation of other, maybe microsomal, enzymes or a noninvolvement of cyp27a1 in vitamin D metabolism in the
mouse (918). The (/) mouse model was later characterized in more detail, but no new conclusions with regard to
vitamin D were drawn (902).
Another caveat that needs to be considered when assessing
the physiological importance of CYP27A1 for vitamin D
metabolism is that the enzyme cannot 25-hydroxylate dietary vitamin D2 (402). This observation underlines the
necessity for another 25-hydroxylase which physiologically
metabolizes vitamin D2. The microsomal 25-hydroxylase
was a clear candidate for this process; however, it has only
recently received more extensive scientific attention.

The regulation of 25-hydroxylation has been the subject of


controversy in the past, which is partially due to the fact
that multiple enzymes may metabolize vitamin D, and that
it was challenging to experimentally discriminate between
these enzyme entities. Thus evidence which supports (91,
92, 741, 1078) and questions (258, 1097) the existence of
25-hydroxylase regulation can be found. A detailed analysis

Following its synthesis, 25(OH)-vitamin D binds to DBP


and is transported to the kidney, where it undergoes further
conversion to 1,25(OH)2-vitamin D, the hormonally active
form of vitamin D (FIGURE 5). Historically, 1,25(OH)2vitamin D was first identified in the nuclei of intestinal cells
as an uncharacterized vitamin D metabolite (429, 634). The
biological activity of the metabolite was determined to be
much higher than that of vitamin D, and it was eventually
isolated and identified as 1,25(OH)2-vitamin D (480, 586,
633, 800). 1,25(OH)2-vitamin D is up to 10 times more
potent than vitamin D. The importance of the kidney for the
synthesis of 1,25(OH)2-vitamin D was soon discovered, as
nephrectomized rats were neither able to convert 25(OH)vitamin D, nor absorb calcium effectively (339, 458).
Briefly thereafter, patients with chronic renal insufficiency
were shown to lack the capability to metabolize 25(OH)vitamin D, which further established the role of the kidney
as the major conversion site (724). The impairment of
1,25(OH)2-vitamin D synthesis in the course of chronic
renal deficiency is a cofactor in the development of renal
osteodystrophy, a bone mineralization deficiency due to
deranged mineral balance. Of note, the kidney is not the
exclusive site of CYP27B1 expression. 1-Hydroxylase has
been detected in a variety of other tissues, including the
placenta, decidua, skin, brain, vascular endothelium, pancreas, colon, but also in monocytes and dendritic cells (451
453, 603, 1204 1206). The role of extrarenal 1,25(OH)2vitamin D synthesis is not entirely clear. Beyond modulating
calcium homeostasis, vitamin D has been shown to have immunomodulatory and antiproliferative effects (see sect. IVA6).
Given these observations, it has been speculated that extrarenal local 1,25(OH)2-vitamin D production and paracrine secretion may represent important factors for the maintenance
of the barrier function in these tissues (451, 453).
In the kidney, 1-hydroxylation of 25(OH)-vitamin D
takes place in the inner mitochondrial membrane of the
epithelial cells of the proximal tubule (393).
The cellular uptake mechanism of 25(OH)-vitamin D by the
tubule cells will be subject of a later section. The 1-hydroxylase responsible for the conversion is also a member of
the cytochrome P-450 family (CYP27B1) and was first
cloned in 1997 by St-Arnaud et al. from rat cDNA (1034).

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

217

Downloaded from on December 8, 2014

The molecular identity of the microsomal 25-hydroxylase


was unclear, until it was unmasked by Cheng et al. (184) in
2003 as CYP2R1. CYP2R1 is expressed on the mRNA level
in a plethora of tissues, but most prominently in the liver
and testes (184). As illustrated by a recent investigation, the
testes may play a role in calcium homeostasis. Patients with
testiculopathy (and concomitant lower CYP2R1 expression) were shown to have decreased 25(OH)-vitamin D levels and osteoporosis (333). The proteins crystal structure
has recently been resolved in complex with vitamin D3
(1045). Unlike CYP27A1, CYP2R1 has been shown to 25hydroxylate both vitamin D2 and vitamin D3, which may be
a solution to the enigma of vitamin D2 metabolism (184).
The physiological relevance of CYP2R1 was underscored
by the characterization of a patient who presented with low
25(OH)-vitamin D levels and was shown to have a transition mutation in the CYP2R1 gene (183). Furthermore, a
remarkable large-scale study has recently tried to establish a
correlation between 25(OH)-vitamin D status and the genotype of 33,996 individuals. It was found that lower
25(OH)-vitamin D levels correlated with variants in the
CYP2R1 gene (1146) (a smaller study came to similar conclusions, Ref. 139). This represents an outstanding finding,
as the influence of CYP27A1 mutations on 25(OH)-vitamin
D production is far less conclusive. Given the disproportionate quantity of scientific data, it is at this moment difficult to evaluate the relative contribution of each 25-hydroxylase to vitamin D metabolism, but more and more
evidence for the importance of CYP2R1 is accumulating.

of these observations is beyond the scope of this review, but


the most recent investigation should be considered in more
detail, given the advances in our experimental repertoire: it
has been demonstrated in the rat that 1,25(OH)2-vitamin D
can downregulate hepatic CYP27A1 transcription with a
concomitant reduction in enzyme activity (1078). These
observations strongly corroborate the hypothesis that the
25-hydroxylation step is subjected to negative-feedback
regulation. The exact mechanism of this regulatory mechanism remains elusive, especially since the CYP27A1 gene is
not under control of a VDRE (369, 988, 1078).

SASCHA KOPIC AND JOHN P. GEIBEL

The activity of CYP27B1 is subjected to tight hormonal


regulation. The key regulator of 1,25(OH)2-vitamin D synthesis is PTH, which is secreted by the parathyroid glands in
response to low serum calcium concentrations in an effort
to increase calcium uptake and release bone calcium into
the circulation. The regulation of calcium homeostasis by
the parathyroid glands and the CaSR will be covered in
subsequent sections (see sect. IV, B and D). Apart from this
stimulatory input, CYP27B1 is under negative control by its
own product. 1,25(OH)2-vitamin D represses CYP27B1 on
a transcriptional level (116, 125, 443, 591, 764). Although
studies in (/) animals suggest that the VDR is essential
for autoinhibition to take place, the promoter region of
CYP27B1 does not include a canonical VDRE (125, 591,
764). It is thus most likely that the transcriptional regulation through 1,25(OH)2-vitamin D is indirect (125,
591). Alternatively, so-called E-box-type elements were
recently proposed to act as negative VDREs (575). Moreover, CYP27B1 can be directly regulated by the local
calcium concentrations. High extracellular calcium inhibits 1,25(OH)2-vitamin D synthesis, whereas low calcium
concentrations induce its production (109). It has been proposed that changes in calcium modulate VDR expression
and thereby the sensitivity of cell to the local negative feedback by 1,25(OH)2-vitamin D (702). Some evidence suggests that the CaSR mediates the regulatory effects of calcium on CYP27B1 activity (FIGURE 8) (702). Other factors
that regulate CYP27B1 activity include fibroblast growth
factor 23 (FGF23), calcitonin, prolactin, sex steroids (at
least in avian species), and phosphate (716, 913, 1067,
1068, 1217).

218

Apart from regulating the synthesis of the vitamin D


metabolites, our body also tightly controls their degradation. The first step of vitamin D catabolism is 24-hydroxylation, which is carried out by the mitochondrial
enzyme CYP24A1. The primary site for vitamin D catabolism is the kidney, but CYP24A1 is also strongly expressed
in other extrarenal tissues, such as the intestine, osteoblasts,
keratinocytes, prostate, placenta, brain, and heart (11, 181,
796). CYP24A1 can hydroxylate both 25(OH)-vitamin D
and 1,25(OH)2-vitamin D, thereby creating 24,25(OH)2vitamin D and 1,24,25(OH)3-vitamin D, respectively (14).
24-Hydroxylation is followed by a series of oxidation/reduction reactions which finally yield the excretable product
calcitroic acid (703, 893). At least in the proximal tubule of
the kidney, the regulation of CYP24A1 is reciprocal to that
of CYP27B1. 1,25(OH)2-vitamin D upregulates CYP24A1,
thereby stimulating its own breakdown, whereas PTH inhibits
CYP24A1 (37, 180, 812, 1069, 1222). While 1,25(OH)2-vitamin D increases the transcription of CYP24A1 (the gene has
two upstream VDREs), PTH exerts its inhibitory effects by
decreasing CYP24A1 mRNA stability (1222). In osteoblastic
and distal convoluted tubule cell lines, PTH has synergistic
effects with 1,25(OH)2-vitamin D in inducing CYP24A1 (37,
1189). In analogy to CYP27B1, CYP24A1 is further regulated
by FGF23, calcitonin, and phosphate (361, 513, 1175).
5. Vitamin D transport and cellular uptake
The hypothesis that vitamin D may be bound to a carrier
substance in serum was first expressed in the late 1950s,
when it was demonstrated that the alpha fraction of human
serum had high anti-rachitic properties (1079). It was later
shown that DBP is identical to the group specific component
(Gc) protein, which had been characterized independently
by Hirschfeld and colleagues around the same time (236,
464, 465). DBP (58 kDa; 458 amino acids) is synthesized
in the liver and is closely related to albumin and -fetoprotein, which are all derived from the same ancestral gene
(221, 875, 1158, 1187, 1188). The crystal structure of DBP
has been resolved at a resolution of 2.3 in complex with
25(OH)-vitamin D (1121). DBP can bind vitamin D and all
of its metabolites (408). There is, however, a difference in
the relative affinity for the vitamin D steroids, with the
affinity for 25(OH)-vitamin D being highest (Kd 108 M),
followed by 1,25(OH)2-vitamin D and vitamin D (Kd
107 M) (408). In humans, vitamin D2 and D3 metabolites are bound with equal affinity to DBP (406). Given its
long plasma half-life, 25(OH)-vitamin D is also measured
as the primary clinical parameter to assess the vitamin D
status of patients. Although the plasma concentration of
DBP is 4 8 M, only 5% of the binding sites are occupied by vitamin D sterols (408, 409). DBP has a very fast
turnover rate. It has been estimated that up to 28% of DBP
are replaced every day (557). This turnover entails a high
demand for synthesis output by the liver. In consequence,
patients with liver disease demonstrate lower DBP and total
vitamin D levels than healthy subjects (97, 409). This rela-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

The human homolog was cloned shortly thereafter (349,


748). Interestingly, mapping of the CYP27B1 gene revealed
that the locus was identical to the gene locus of the autosomal recessive disorder pseudo vitamin D deficiency rickets, type 1A (PDDR1A, OMIM 264700) (1034). PDDR is
characterized by low serum calcium, secondary hyperparathyroidism, and low 1,25(OH)2-vitamin D levels (869) (of
note, the original article wrongly suggests an autosomal
dominant inheritance pattern). Patients can exhibit rickets
or osteomalacia due to increased mobilization of calcium.
The gene locus of PDDR1A had been mapped by linkage
analysis; however, before the cloning of CYP27B1, it was
not clear whether a defect in the enzyme itself or disturbances in its regulation were responsible for the disease
(617, 618, 1223). The clear-cut phenotype of PPDR1A effectively illustrates the pivotal role of CYP27B1 and the
lack of redundancy at this essential step of vitamin D metabolism. The characteristic features of PDDR1A can essentially be emulated if cyp27b1 is knocked out in a mouse
model (238, 822). The serum calcium levels and the secondary hyperparathyroidism can be normalized if these animals
are fed a high-calcium, phosphorus, lactose diet, albeit bone
growth remains impaired (239).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


tionship is reversed during pregnancy, when the levels of
DBP and vitamin D sterols are increased (97, 409, 609, 685,
892). Of note, DBP is not the exclusive carrier substance for
vitamin D sterols. Albumin and lipoproteins were shown to
transport a fraction (15%) of vitamin D (1015).
Given its multitude of functions, DBP has been regarded as an
essential protein. Indeed, an analysis of over 80,000 human
serum samples showed that DBP was present in all of them,
which led to the hypothesis that deleterious mutations of
DBP were lethal (213). Rather surprisingly, DBP (/)
mice thrive well with growth curves identical to their
littermates, although their 25(OH)-vitamin D and
1,25(OH)2-vitamin D levels (total) are severely decreased
(937). If challenged with a low vitamin D diet, the DBP
(/) animals develop secondary hyperparathyroidism,
leading to defects in bone mineralization (937). It is unclear, however, to what extent albumin and lipoproteins
compensate for the loss of the primary vitamin D carrier
substance.

6. Cellular effects of vitamin D


The cellular effects of 1,25(OH)2-vitamin D can be categorized into two major pathways, which are defined by their
respective speed of onset: 1) slow genomic responses and
2) rapid nongenomic responses. Both pathways require
binding of 1,25(OH)2-vitamin D to its intracellular receptor, the VDR (503, 787). The VDR (NR1I1, nuclear receptor subfamily 1, group I, member 1) belongs to the superfamily of nuclear receptors, which amongst others also
includes the estrogen, testosterone, or glucocorticoid receptors. VDR was first identified in the late 1960s in the chromatin fraction of chick intestinal mucosa, where
1,25(OH)2-vitamin D increases the rate of intestinal calcium uptake (430). The 427-amino acid protein (molecular
mass 48.3 kDa) was cloned in 1988 by Baker et al. (59). The
crystal structure of the VDR is available to us with
1,25(OH)2-vitamin D bound to the receptors ligand binding domain (914).
A) GENOMIC EFFECTS. Following ligand binding, nuclear receptors typically act as transcription factors and induce or repress the transcription of certain target genes. In the case of
VDR, 1,25(OH)2-vitamin D binds to the receptor, which
subsequently heterodimerizes with the retinoid X receptor
(RXR) (FIGURE 3) (1027, 1083). The VDR-RXR complex
then interacts with a VDRE in the 5= promoter region of the
regulated gene resulting in transactivation. Alternatively, it
has been proposed that VDR can bind to the VDRE before
ligand binding occurs (920).

I) Intestine. The intestine is one of the primary target sites of


1,25(OH)2-vitamin D. 1,25(OH)2-vitamin D upregulates
the expression of intestinal TRPV6, calbindin-D 9k, and
PMCA, which are canonically regarded to mediate the process of transcellular calcium absorption (FIGURE 3) (see sect.
IIIA). Furthermore, 1,25(OH)2-vitamin D may modulate
calcium uptake through the paracellular route (see sect.
IIIB). By increasing the amount of absorbed calcium,
1,25(OH)2-vitamin D directly elevates serum calcium levels. This represents one of the final links in the regulatory
chain of calcium homeostasis which starts with sensing of
low calcium levels by the parathyroid gland and ends with
increased synthesis of 1,25(OH)2-vitamin D by the kidney.
The significance of 1,25(OH)2-vitamin D for intestinal calcium absorption is illustrated by 1,25(OH)2-vitamin D-deficient patients, which absorb up to 80% less calcium from
their meal compared with healthy individuals (996). Although similar results were obtained in animal models,
probably one of the more illustrative observations has been
made in VDR-deficient animals (825, 1110). Deletion of
VDR correlates with a massively impaired capacity to absorb intestinal calcium resulting in low plasma calcium levels and hyperparathyroidism (1110). This phenotype is re-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

219

Downloaded from on December 8, 2014

Not much is known about the cellular uptake of vitamin D.


As all other steroids, free vitamin D can passively diffuse
through the plasma membrane because of its lipohilic nature. However, due to the high concentration of DBP and its
affinity towards vitamin D ligands, only a fraction of vitamin D circulates in the free form. For example, 0.003% of
25(OH)-vitamin D is transported as an unbound sterol in
serum, which raises the question whether passive diffusion
represents a sufficient uptake pathway (98). At least
25(OH)-vitamin D has been proposed to be delivered to
the proximal kidney tubule for further conversion to
1,25(OH)2-vitamin D by a different and remarkable mechanism. Rather than diffusing passively, it has been proposed
that the 25(OH)-vitamin D-DBP complex passes the glomerular filter and is endocytosed by the epithelial cell of the
proximal tubule (FIGURE 5) (805). The endocytotic process
is mediated by megalin (aka gp330), which is a multifunctional clearance receptor on the luminal membrane. Mice
lacking functional megalin were shown to lose DBP and
vitamin D in the urine and develop vitamin D deficiency
(805). A kidney specific megalin (/) animal was recently
created, and the observations made in the global (/)
animal, which had a very low perinatal survival rate of 2%,
could essentially be replicated (643). Furthermore, defects
in cubulin, a membrane protein that colocalizes with megalin, cause a similar phenotype (806). The experimental data
are further supported by clinical observations made in patients suffering from Fanconi syndrome. Fanconi syndrome is a global reabsorption deficiency of the proximal
tubule, which can develop as a result of heavy metal
poisoning and/or drugs or may have inherited causes.
These patients were shown to lose DBP in their urine,
which may reflect the inability of the tubule cell to endocytose DBP (805, 1076, 1100). A similar endocytotic
uptake mechanism has been proposed for mammary

cells, which can also convert 25(OH)-vitamin D to


1.25(OH)2-vitamin D(926).

SASCHA KOPIC AND JOHN P. GEIBEL


versible by intestine-specific expression of VDR on a VDR
(/) background (712, 1181). Thus animals that lack the
VDR, except in the intestine, present with normal serum
calcium and PTH levels (1181). In conclusion, it is unquestionable that 1,25(OH)2-vitamin D and its receptor are the
key regulators of intestinal calcium absorption.

III) Kidney. The kidney is not only the major site of


1,25(OH)2-vitamin D synthesis (see above), but also represents a vitamin D target organ. The kidney acts as key
regulator of calcium homeostasis by changing the amount
of calcium that is reabsorbed from the primary urine. The
majority of the calcium that is filtered through the glomerulus is reabsorbed in the proximal tubule through the paracellular space, with the amount of absorbed calcium gradually decreasing along the nephron. Fine regulation of calcium absorption occurs in the distal tubule and collecting

220

B) NONGENOMIC EFFECTS.

In contrast to the genomic effects of


1,25(OH)2-vitamin D, which have been known for decades,
the rapid cellular responses have only recently received
more scientific attention. While the transcriptional events of
1,25(OH)2-vitamin D take place on a time scale of a few
hours to days, the rapid nongenomic responses occur within
minutes of exposure. One of the first evidences, which in
retrospect can be attributed to a nongenomic response, was
the observation made in 1941 by Selye that intraperitoneal
injection of steroids had an anesthetic effect (990). Interestingly, the rapid responses to 1,25(OH)2-vitamin D also require the presence of the VDR, as these responses cannot be
elicited in VDR deficient animals (503, 787). It should be
noted that other proteins such as 1,25(OH)2-vitamin-DMARRS have also been suggested as candidates for a membrane associated 1,25(OH)2-vitamin D receptor (782). Attempts at identifying the subcellular localization of the
VDR in the rapid response context have yielded that VDR is
also present in plasma membrane invaginations, the socalled caveolae (503, 802). These VDR-containing membrane microdomains have been identified in multiple tissues, including the intestine, kidney, and lung, and are identified by coexpression of caveolin-1, which is used as a
marker protein for caveolae (503, 802). Functionally, not
many rapid response effects of 1,25(OH)2-vitamin D have
been characterized. For example, it has been demonstrated
that 1,25(OH)2-vitamin D can influence ion channel gating
in osteoblasts, modulate the contraction of cardiomyocytes,
lead to insulin secretion in pancreatic -cells via elevating
intracellular calcium, and cause photoprotection in keratinocytes (273, 540, 1088, 1200, 1216). In the intestine, the
phenomenon of transcaltachia (see sect. IIIC) has been attributed to rapid actions of 1,25(OH)2-vitamin D (801).
In addition to 1,25(OH)2-vitamin D, the VDR also binds
the secondary bile acid lithocholic acid (704). Secondary
bile acids are bile acids that have been metabolized by the
intestinal gut flora. Lithocholic acid is toxic and has been
implicated to play a role in intestinal carcinogenesis (601).
It has been suggested that the VDR may serve as a secondary bile acid sensor and induce lithocholic acid breakdown
through CYP3A activation (537). The noncanonical VDR
stimulation by lithocholic acid may thus serve as an autoprotective mechanism (704). Apart from inducing CYP3A,
lithocholic acid has been demonstrated to increase expres-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

II) Bone. Some evidence exists that 1,25(OH)2-vitamin D


may have direct influences on the formation of bone (FIGURE 4). The VDR is expressed in osteoblasts, osteoclasts,
and chondrocytes (115, 533, 623, 730). Most of the direct
effects of 1,25(OH)2-vitamin D are thought to be mediated
by osteoblasts. 1,25(OH)2-vitamin D has been shown to
promote osteoblast differentiation from mesenchymal stem
cells and to regulate the synthesis of various osteoblast proteins, such as osteocalcin, alkaline phosphatase, collagen I,
osteopontin, and RANKL (45, 78, 79, 665, 709, 804, 870,
925). In general, however, the effects of 1,25(OH)2-vitamin
D on osteoblast maturation and protein synthesis are very
pleiotropic and depended on the duration of the exposure
and the differentiation stage at which the osteoblast is exposed (817). Effects of 1,25(OH)2-vitamin D on the mineralization of bone have also been reported. For example, an
increase in the mineralization of extracellular matrix was
observed following concomitant 1,25(OH)2-vitamin D and
vitamin K exposure (599, 745). Yet, the overall direct influence of 1,25(OH)2-vitamin D on bone metabolism is somewhat obscure. This is illustrated by VDR-deficient animals.
Naturally, these animals develop rickets and osteomalacia
due to impaired intestinal calcium absorption. If the animals are, however, maintained normocalcemic, the skeletal
phenotype is completely rescued (21, 661). Although, these
observations question the importance of 1,25(OH)2-vitamin D as a direct regulator of bone metabolism, a subsequent investigation by Panda et al. (821) came to a different
conclusion. The authors reported that osteoblast numbers,
mineral apposition rates, and overall bone volume were
reduced in normocalcemic (rescue diet) cyp27b1/VDR
(double /) animals, suggesting that the 1,25(OH)2-vitamin D system is necessary for intact bone formation (821).
The reason for these discrepant findings is generally unclear; however, differences in the length of exposure to the
rescue diet have been put forward as a possible cause (821).
For a more detailed review of the effects of vitamin D on
bone, please refer to References 1033, 1114.

duct. The mechanism by which the distal tubule conducts


transcellular calcium absorption is highly analogous to the
proximal intestine. The epithelial cells of the distal tubule
express TRPV5 (the sister channel of TRPV6) as the apical calcium entry channel, calbindin-D 28k and NCX1 and
PMCA1b as basolateral calcium extruders (203, 474, 610,
837, 922). In analogy to the intestine, 1,25(OH)2-vitamin D
upregulates the majority of these proteins in an effort to
increase renal calcium reabsorption (203, 474, 610, 837,
922).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


sion of TRPV6 in intestinal cell lines, which corroborates its
role as a physiological VDR agonist (517).
We are far from understanding the full spectrum of the
effects of 1,25(OH)2-vitamin D, and unfortunately, the
scope of this review does not allow a detailed analysis of
these processes. In general, our knowledge concerning the
physiological role of 1,25(OH)2-vitamin D is massively expanding beyond the horizon of calcium homeostasis. Evidence is accumulating that 1,25(OH)2-vitamin D can influence the renin-angiotensin-aldosterone system and may
thereby act as a regulator of blood pressure (660, 662,
1219). Furthermore, low vitamin D status is associated with
an increased incidence of colorectal, ovarian, and breast
cancer(363, 364, 642, 984). Vitamin D also acts as a potent
modulator of the immune response and cell proliferation.

B. PTH

1. Production and secretion


PTH is a 84-amino acid peptide hormone produced in the
parathyroid glands. Its amino acid sequence was first established in 1970 in bovine (126, 788). Cloning of the human
cDNA followed a decade later (440). The PTH gene encodes a 115-amino acid precursor hormone (preproPTH),
which is enzymatically cleaved in a two step process to its
mature 84-amino acid secreted form (565). The NH2-terminal prepro-signaling sequence is necessary for correct
hormone processing and trafficking (340, 546). This
knowledge is mainly founded on truncation studies and
observations made in patients with mutations in the PTH
gene, which can result in familial isolated hypothyroidism,
a disorder characterized by hypocalcemia and low PTH
levels (38, 340, 828, 1055). For example, a well-characterized T-to-C point mutation in the prepro signaling sequence
that causes FIH leads to accumulation of the precursor hormone in the ER (243, 546). The impaired processing triggers ER stress, ultimately apoptosis and PTH insufficiency
(243).
Following its cleavage to mature PTH, the hormone is
stored in secretory vesicles and released into the circulation
in response to low plasma calcium. The regulation of PTH

2. PTH1R
PTH exerts its physiological effects via activation of a membrane-bound GPCR, the parathyroid hormone receptor
type 1 (PTH1R) (PTH2R is mostly expressed in the CNS
and tissues that are not involved in calcium handling and
will thus not be reviewed). Of note, PTH1R is not exclusively located at the plasma membrane but can also localize
to the cell nucleus. The physiological significance of nuclear
PTH1R is currently unclear but may represent a novel signaling paradigm for the actions of PTH (830, 856, 857,
1155).
Full-length PTH is not required to activate PTH1R. The
NH2-terminal domain of PTH mediates most of the physiological effects of the hormone and is responsible for binding in the -- binding fold of PTH1R (861). This is why
clinically PTH(134) is used as a PTH analog with identical
biological activity (753, 867, 919, 1094). Conversely, NH2terminal truncation of PTH(134) to PTH(234) changes
the characteristics to a partial receptor agonist, whereas
further truncation to PTH(334) results in loss of biological
activity (1094).
PTH1R was first cloned from opossum in 1991, which was
followed by identification of the highly homologous human
cDNA shortly thereafter (536, 964). In the nonactivated
state the receptor is expressed as a homodimer at the cell
surface, which dissociates upon PTH binding (860).
PTH1R is a member of the class B (class 2, secretin family)
GPCRs. As many other GPCRs, it undergoes N-linked glycosylation at four asparagine residues (1218). Mutational
analysis revealed that site-specific mutation of all four sites
decreases cell surface expression, whereas impairment of
fewer glycosylation sites does not seem to have significant
effects on trafficking or ligand binding (1218). Furthermore, the extracellular domain of the receptor includes a
characteristic disulfide bond pattern involving six cysteine
residues (392). These residues are thought to be essential for
stabilizing the hydrophobic -- binding pocket for
PTH, which is conserved among all members of class B

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

221

Downloaded from on December 8, 2014

Experiments dating back to the beginning of the 1900s have


demonstrated that surgical removal of the parathyroid
gland results in tetany. It was recognized very early that
administration of calcium could ameliorate or prevent the
manifestation of tetany, and it was subsequently concluded
that the parathyroid glands play an important role in calcium metabolism (690, 826, 939). In 1925, extracts from
the parathyroids were for the first time shown to control
tetany in dogs (216). This finding marked the discovery of
PTH, which is one of the three prime hormones regulating
calcium homeostasis.

secretion is extremely tight, given the bodys need to maintain the calcium concentration within a narrow window
(1.11.3 mM). Small alterations in calcium homeostasis can
have deleterious effects, for example, on the excitability of
neurons and muscles. The low plasma half-life of PTH of 5
min allows for a precise regulation of this balance (95). To
achieve controlled and rapid on-demand secretion of PTH, the
parathyroid is equipped with an ultrasensitive extracellular
CaSR, which constantly monitors the plasma calcium levels
and triggers intracellular signaling events and PTH release
upon imminent drops in calcium levels (FIGURE 6) (see sect.
IVD). PTH is further regulated on a transcriptional level by
1,25(OH)2-vitamin D, creating a negative-feedback loop
(154, 930, 931).

SASCHA KOPIC AND JOHN P. GEIBEL


Ca
Ca
Ca

Calcium

Ca

PLC

Gq

PIP2

DAG
Kinase phosphorylation

Inhibition of
cell growth

IP3

Calcium

1,25(OH)2-D D

PLA2

ER

D
VDR

AA
Increases VDR expression

Nucleus
Inhibits PTH
secretion
Reduces PTH
synthesis
Parathyroid gland

PTH
FIGURE 6. CaSR signaling in the parathyroid gland. Increased serum calcium levels lead to an inhibition of
PTH secretion. Serum calcium levels are measured by the CaSR receptor. Activation of CaSR causes
generation of arachidonic acid (AA) metabolites, which inhibit the release of PTH and increase the expression
of VDR, thereby increasing the cells sensitivity to the negative feedback exerted by 1,25(OH)2-vitamin D.
1,25(OH)2-vitamin D suppresses the synthesis of PTH. Furthermore, CaSR activation inhibits parathyroid gland
growth.

GPCRs and has recently been crystallized in the presence of


PTH (861).
Binding of PTH causes activation of at least two distinct G
proteins. Gq/11 mediates intracellular calcium release via
phospholipase C (PLC) activation and increases in inositol
trisphosphate (IP3), whereas Gs activates adenylyl cyclase
leading to rises in cAMP (5, 188, 808, 859).
PTH1R can be regulated on a variety of levels, ranging from
trafficking and internalization to direct protein interactions
at the cell surface. Desensitization of PTH1R is mediated by
GRK2 binding/phosphorylation and -arrestin binding,
which uncouples the receptor from its associated G proteins
and triggers its internalization (267, 326, 330, 1123). For

222

example, knockout of -arrestin causes increased and sustained levels of the second messenger cAMP in primary
osteoblast cultures upon PTH stimulation (327). PTH1R
also associates with the scaffolding protein NHERF1,
which stabilizes the receptor at the cell membrane and prevents its endocytosis and desensitization (1022, 1139). This
effect of NHERF1 is partially attributable to a prevention of
an interaction between -arrestin and PTH1R (1140). Colocalization of NHERF1, -arrestin, and PTH1R has been
demonstrated and suggests that NHERF1 is constitutively
bound, whereas -arrestin association is more dependent
on receptor activation (580). Interestingly, it has recently
become apparent that -arrestin not only plays a role in
receptor desensitization, but also mediates activation of
downstream signaling cascades, such as MAPKs, in a G

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

D
RXR VDR

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


protein-independent manner (380). Current scientific effort
thus focuses on the development of so-called biased
PTH1R agonists, which can selectively trigger G protein- or
-arrestin-dependent signaling events, allowing for a more
selective therapeutic repertoire (381, 1160). Furthermore,
NHERF may also modulate the cells response to PTH binding. In the presence of NHERF2, the cells calcium response
via PLC is augmented, whereas the cAMP response is dampened, presumably via recruitment of Gi (698). Both
NHERF and -arrestin provide effective examples of how
receptor-associated proteins can modulate canonical signaling events or even, as is the case with -arrestin, initiate
signaling events in their own right.

3. Cellular effects
PTH exerts its effects in two primary target tissues: bone
and the kidney.
In the kidney, PTH causes phostphaturia, increases calcium
absorption, and induces the synthesis of 1,25(OH)2-vitamin D. In-detail analysis of renal phosphate handling is
beyond the scope of this review and has been summarized
previously (334, 765). In brief, phosphaturia mainly results
from downregulation of the Na-Pi transporter type IIa
(NaPi-IIa) at the apical membrane of the proximal tubule,
thereby reducing the amount of reabsorbed phosphate from
the primary urine. Rather than directly modulating the
transporters activity, PTH exposure mainly affects the
number of active cotransporters on the plasma membrane.
Activation of basolateral PTH1R causes retrieval of NaPiIIa and targets it for lysosomal degradation, resulting in
diminished Pi reuptake (566, 682, 847). Apart from NaPiIIa, at least two other apical phosphate transporters are
present in the proximal tubule: NaPi-IIc and PiT-2 (986,
1124). Currently, there is some evidence that PTH can also
regulate NaPi-IIc and PiT-2, but additional studies remain
to be conducted (855, 987). Although a contribution of
these transporters to renal phosphate reabsorption is highly
likely, knockout studies suggest that NaPi-IIa is responsible
for 80% of total phosphate transport, thus representing
the major uptake mechanism (72, 468).
It has been recognized for over 30 years that PTH can
stimulate the synthesis of the active vitamin D metabolite
1,25(OH)2-vitamin D in the kidney (116, 338) (see sect.
IVA4). 1,25(OH)2-vitamin D in consequence enhances the

Apart from inducing the synthesis of 1,25(OH)2-vitamin D,


PTH can directly upregulate the renal reabsorption of calcium. The regulation of calcium absorption by PTH occurs
in distal segments of the nephron, mostly in the distal convoluted tubule and the connecting tubule (99, 379, 1003,
1172). In close analogy to the duodenum, these segments
express calcium transport proteins, which are responsible
for mediating the process of active transcellular calcium
absorption in the kidney, namely, TRPV5, calbindin-D
28k, and NCX1. PTH can regulate all of these protein levels
on a transcriptional level (1108). Interestingly, this seems to
be accomplished independently of 1,25(OH)2-vitamin D,
which also positively regulates most of these transporters
(see sect. IVA6) (1108). It is therefore difficult to dissect the
relative contribution of each of the two hormones in the
physiological regulation of the calcium transport proteins.
In addition to transcriptional activation, PTH was shown to
cause direct phosphorylation of TRPV5, thereby increasing
its opening probability (249). The channel is phosphorylated at threonine-709 in a PKA-dependent fashion (249).
Elevation of intracellular cAMP levels and concomitant
PKA activation are classical downstream effects of PTH1R
activation in the kidney (248, 1172).
In bone, PTH exerts a dichotomous effect depending on the
pattern of exposure. It is well documented that pulsatile
PTH exposure has anabolic effects on bone mass, whereas
continuous release increases plasma calcium by bone catabolism (FIGURE 7) (27, 348, 401, 469, 1065). The observation that intermittent PTH administration increases bone
mass has led to the use of PTH as a treatment strategy for
osteoporosis (621, 671, 897). The enhanced bone formation mainly results from an increase in osteoblast numbers.
This phenomenon has been partially attributed to a PTHmediated induction of osteoblast differentiation and an inhibition of their apoptosis (75, 274, 518, 530, 531, 684,
969, 1032). Multiple mechanisms underlying the anti-apoptotic effects of intermittent PTH on osteoblasts have been
suggested. Among others, these include runt-related transcription factor 2 (Runx2)-mediated transcription of survival genes and increased DNA repair (75, 969). It has
further been shown that fibroblast growth factor 2 (FGF2)
is partially needed as an endogenous cofactor for the anabolic effects of PTH to take place (934).

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

223

Downloaded from on December 8, 2014

PTH1R can also be regulated by its external environment.


The extracellular receptor domain can be cleaved by metalloproteases, resulting in receptor degradation (579). PTH
binding prevents this proteolytic cleavage, thus stabilizing
PTH1R at the cell surface. The physiological importance of
this mechanism is not yet fully understood. However,
MMPs are involved in bone remodeling and may in consequence locally regulate the sensitivity of osteoblasts to PTH
(579).

intestinal and renal uptake of calcium in an effort to counteract hypocalcemia, which initially led to secretion of PTH.
The PTH-stimulated increase in 1,25(OH)2-vitamin D levels is achieved on a transcriptional level. PTH upregulates
the transcription of CYP27B1, the mitochondrial enzyme
which is responsible for the conversion from 25(OH)-vitamin D to 1,25(OH)2-vitamin D. Transcriptional upregulation occurs via PTH binding to PTH1R, leading to increases
in the second messenger cAMP and activation of PKA (125,
442, 763, 764, 921).

SASCHA KOPIC AND JOHN P. GEIBEL


Intermittent

Chronic
PTH
PTH1R
OPG

RANKL

Osteoblast
proliferation

RANK

Osteoclast
differentiation
H+

Osteoblast

Osteoclast

V-ATPase
FIGURE 7. The effects of PTH on bone. PTH has a dual effect on bone. Intermittent PTH exposure causes
osteoblast proliferation, leading to an increase in bone mass. Continuous PTH exposure results in RANKL
upregulation and concomitant OPG suppression (OPG serves as a decoy receptor for RANKL and prevents its
interaction with osteoclast RANK). The stimulated RANKL-RANK interaction leads to osteoclast proliferation
and increased bone turnover.

In the intestine, several observations suggest that PTH may


have a direct, i.e., non-1,25(OH)2-vitamin-D mediated, effect on intestinal calcium absorption. Both isolated enterocytes and intestinal loops demonstrated an increase in calcium uptake following acute PTH exposure (781, 783,
784). However, more investigations are needed to clearly
establish a direct regulatory role of PTH in the context of
intestinal calcium uptake.

224

4. PTH fragments
It should be noted that full-length PTH(1 84) is not the
only circulating form of the hormone in the body. Various
PTH fragments can be found in the circulation, which partially originate directly from the parathyroid gland and partially represent products of peripheral cleavage. The parathyroid itself releases COOH- and NH2-terminal hormone
fragments, which are generated by cysteine proteases (cathepsin B and H) in distinct secretory vesicles of the gland
(418, 427, 693). Interestingly, the fraction of secreted hormone fragments changes with extracellular calcium conditions. It has been reported that more fragments are released
under conditions of hypercalcemia, when secretion of fulllength PTH is suppressed (417, 418, 605, 726). Peripheral
proteolysis represents the second source of PTH fragments.
This process occurs predominantly in liver and the kidney
(127, 234, 989). The group of fragments that have received
the most amount of scientific attention is the large NH2terminally truncated non-PTH(1 84) fragments. PTH(7
84) is the quantitatively major member of this group, which
is secreted by the parathyroids (233). The group of nonPTH(1 84) fragments can represent up to 20% of circulating PTH, but can increase in patients with renal failure
dramatically to up to 50% because of impaired renal clearance (130, 131, 444, 648). This is of particular interest, as it
has become recently apparent that the non-PTH(1 84)
fragments exert biological activity. In general, non-PTH(1
84) fragments antagonize the effects of PTH in its primary
target tissues, bone and the kidney but also the parathyroid
gland directly. It has been shown that PTH(7 84) can inhibit PTH release from the parathyroid, presumably in an
autocrine fashion, despite low serum calcium concentra-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Continuous PTH exposure, on the other hand, mainly affects osteoclast numbers and activation, thereby increasing
bone turnover. Since osteoclasts are canonically thought to
not express PTH1R (although this view has recently been
challenged, Ref. 260), the catabolic effects of PTH are relayed through osteoblast signaling. The PTH-induced
crosstalk between osteoblasts and osteoclast is mainly mediated by receptor activator of nuclear factor B (RANK),
osteoprotegrin (OPG), and RANK ligand (RANKL). Both
RANKL and OPG are expressed by osteoblasts and exert
opposing actions on osteoclasts. RANKL promotes osteoclastogenesis by binding to RANK on osteoclasts. Conversely, OPG serves as a soluble decoy receptor for RANKL
and thus inhibits its interaction with RANK, thereby suppressing osteoclastogenesis. In accordance with this model,
RANKL-deficient animals develop osteopetrosis because of
insufficient osteoclasts activation (592). Sustained PTH exposure affects RANK-RANKL signaling by downregulating antiresorptive OPG, while simultaneously stimulating production of RANKL by osteoblasts (FIGURE 7)
(350, 497, 679, 689). The enhanced RANK-RANKL signaling induces formation of osteoclasts, which in turn
leads to enhanced bone resorption and elevates serum
calcium levels.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


tions (493). In bone, PTH(7 84) blocks the effects of PTH
on calcium mobilization in thyroparathyroidectomized rats
(786). More detailed investigations revealed that PTH(7
84) reduces calcium release from bone in vivo and inhibits
the formation of osteoclast-like cells in murine primary
marrow cultures (272). In the kidney, PTH(7 84) can inhibit the formation of 1,25(OH)2-vitamin D, presumably
via a posttranscriptional mechanism (768, 1102).
Since activation of PTH1R requires an intact NH2-terminal
domain of PTH, it has been speculated that non-PTH(1
84) fragments exert their function through a pathway distinct of PTH1R. The existence of a COOH-terminal PTH
receptor has thus been postulated (270 272, 786). The molecular identity of this receptor is, however, not yet resolved. Another mechanism through which non-PTH(1
84) fragments may exert their biological activity is downregulation of PTH1R. It has been demonstrated that
PTH(7 84) causes internalization of PTH1R, thus offsetting the effects of PTH by decreasing the number of available receptors at the cell surface (1022).

Calcitonin exerts its physiological functions via activation


of the calcitonin receptor. The calcitonin receptor is a seventransmembrane domain GPCR. In particular, it is a member
of the family B subfamily of GPCRs (669). It shares significant homology with other receptors of the family, which
include the PTH, GHRH, PACAP, VIP, secretin, glucagon,
and glucagon-like peptide receptors.
The calcitonin receptor is expressed in the two most extensively described calcitonin target tissues, i.e., osteoclasts
and the kidney, but also in other adult tissues, such as the
prostate, CNS, skeletal muscle, and placenta (17, 329, 428,
790, 878, 1174). Multiple isoforms of the calcitonin receptor occur in the body, which results from different RNA
splicing directed by tissue-specific promoters (17, 30, 389,
606). Activation of the calcitonin receptor mostly translates
into a rise of intracellular cAMP levels via Gs-dependent
activation of adenylate cyclase (167, 332, 435, 669). Although the cAMP/PKA pathway appears to be dominant,
activation of both PLC and PLD have also been reported
(167, 332, 776).
3. Cellular effects
A) OSTEOCLASTS.

1. Production and secretion

Shortly after the discovery of calcitonin and


its hypocalcemic effects, investigators set out to identify its
physiological site of action. First evidence for an effect of
calcitonin on bone metabolism came from experiments on
rat embryonic bone in tissue culture. It was observed that
calcitonin caused a decreased basal release of calcium from
these preparations (343). These observations served as the
first evidence of how calcitonin lowers serum calcium levels.
Furthermore, calcitonin blocked the resorptive actions of
PTH on bone, albeit only temporarily (342, 343). After 4 6
days of combined treatment (calcitonin PTH), calcium
release rose again (342). This desensitization to the effects
of calcitonin has been coined as the calcitonin escape
phenomenon (342). Today we know that the transient effect of calcitonin on osteoclasts is attributable to a downregulation of surface calcitonin receptors and their synthesis (882, 1061, 1129, 1130).

Calcitonin is encoded by the CALCA gene and is initially


synthesized as a 141-amino acid precursor (preprocalcitonin), which is later processed to the mature 32-amino acid
hormone (637). The same gene also encodes the neuropeptide CGRP. Production of either peptide is dependent on
tissue-specific RNA splicing. Although encoded by a different gene, amylin also belongs to the calcitonin peptide family. All three peptides, i.e., calcitonin, CGRP, and amylin,
share some overlapping functions with regard to osteoclast
suppression (16, 229, 1199). Calcitonin is released from the
C-cells in response to rising concentrations of plasma calcium. The CaSR is responsible for the molecular process of
calcium sensing on the parafollicular cells (351, 367, 728).

As alluded to before, calcitonin directly inhibits the action


of osteoclasts, causing the balance between bone absorption and formation to shift towards anabolism. Calcitonin
exerts its inhibitory effects on osteoclasts via activation of
its receptor, which is expressed in abundance on their surface (428, 790, 878). Exposure to calcitonin triggers distinct morphological changes in the osteoclast. Osteoclasts
are highly motile cells that resorb bone via formation of
so-called resorptive pits, which are membrane invaginations that are luminally acidified by active proton secretion.
Calcitonin has been shown to inhibit the formation of these
resorptive bays in vitro (487, 1057). Furthermore, osteoclast motility is markedly decreased, causing the cell to

C. Calcitonin
Calcitonin is a peptide hormone that has been discovered by
Copp et al. in 1962 as a factor that reduces serum calcium
concentrations (160, 227). Calcitonin production was initially falsely ascribed to the parathyroid glands, and it was
only later that the thyroid gland had been established as the
source of calcitonin (463). The primary sites of calcitonin
production are the parafollicular cells (C-cells) of the thyroid gland. Calcitonin exerts its hypocalcemic effects primarily by inhibition of osteoclast activity. It should be
noted that the importance of calcitonin in day-to-day calcium homeostasis in humans is rather negligible (see sect.
IVC4). For this reason, it will only be reviewed concisely.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

225

Downloaded from on December 8, 2014

In conclusion, non-PTH(1 84) fragments act as PTH antagonists and are secreted by the parathyroid in response to
hypercalcemia.

2. The calcitonin receptor

SASCHA KOPIC AND JOHN P. GEIBEL


enter a state of functional quiescence (169). Apart from
directly altering osteoclast function, calcitonin also affects
osteoclast differentiation. Calcitonin inhibits the formation
of multinucleated mature osteoclasts by arresting their differentiation in more immature stages (1060).
B) KIDNEY.

It has also been speculated that calcitonin may act directly on the collecting duct in a similar fashion to antidiuretic hormone (ADH or vasopressin), i.e., to concentrate the urine by increasing the reabsorption of water
from the primary urine (251). Indeed, calcitonin was
shown to increase the apical expression of aquaporin 2
(AQP2) in principal cells of the collecting duct (119).
Apical insertion of AQP2 and subsequent transepithelial
water movement is the primary mechanism by which
ADH causes concentration of the urine to lower plasma
osmolarity.
In conclusion, the direct impact of calcitonin on renal
calcium handling is quite vague and may be of minor
importance. However, calcitonin also has another, indirect effect on calcium homeostasis. Calcitonin was
shown to be an important regulator of the expression of
CYP27B1, the renal enzyme responsible for the conversion of 25(OH)-vitamin D to 1,25(OH)2-vitamin D
(1009, 1217). In normocalcemic rats, CYP27B1 mRNA
levels were inducible by calcitonin administration, leading to an increase in the production of 1,25(OH)2-vitamin D (1009, 1217). Furthermore, CYP24A1 was in-

226

4. The relevance of calcitonin for


calcium homeostasis
The relevance of calcitonin for day-to-day calcium balance
is highly debatable. This is corroborated by fundamental
observations during conditions of decreased or increased
calcitonin levels, neither of which result in an appreciable
phenotype in terms of calcium balance. For example, patients with medullary carcinomas of the thyroid (MTC), a
tumor of the thyroid C-cells resulting in the hypersecretion
of calcitonin, were shown to have normal bone mineral
densities (1176). Animal studies further substantiate this
conundrum. A reduction in serum calcitonin levels by thyroidectomy in rats did not impact serum calcium levels
(223, 1064). In light of this evidence, the question arises as
to what the physiological role of calcitonin is.
It has been suggested that calcitonin is an evolutionary remnant (462). This is substantiated by the fact that calcitonin
from other species is more potent than human calcitonin.
Teleost calcitonin has the highest biological activity in humans, which may be a result of their higher dependence on
the hormone. For example, salmon calcitonin has an approximately sixfold higher affinity to calcitonin receptor
than human calcitonin (28, 328). Furthermore, it is less
effectively eliminated by the kidney, resulting in a longer
plasma half-life (405). The differences in the biological activity between calcitonin forms led to the introduction of
salmon calcitonin as a treatment for skeletal disorders, such
as osteoporosis or Pagets disease (1077).
Given its ambiguous role in regular calcium homeostasis, it
has been postulated that calcitonin may be of importance
during states of high calcium demand, such as during lactation (1170). It has been demonstrated that calcitonin and
CGRP (/) mice show greater loss of skeletal mass during
lactation than wt animals (1170). Since calcitonin and
CGRP are encoded by the same gene, animals were controlled by CGRP substitution, which was without effect
(1170). Another mechanism by which calcitonin may be
osteoprotective during lactation or pregnancy is by inducing the renal synthesis of 1,25(OH)2-vitamin D (1217).

D. The CaSR
The CaSR is a G protein-coupled membrane-bound receptor that is the primary sensor for calcium and is the first link
in the regulatory chain of calcium homeostasis. By regulating the release of PTH from the parathyroid to modulate

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Renal calcium handling is the second organ


function that is influenced by calcitonin. However, it is
not entirely clear whether calcitonin causes calciuria or
enhances calcium reabsorption from the urine. The conflicting results may be to some extent attributable to species differences. In humans, calcitonin likely increases the
excretion calcium through the urine and thereby acts in
concert with its inhibitory action on osteoclasts to lower
serum calcium levels (31, 32, 143, 215, 819, 1016). Conversely, most studies demonstrating an increase in the
renal reabsorption of calcium and magnesium were conducted in rats, rabbits, or mice (84, 158, 159, 265, 304,
877, 883, 1225). A single investigation established a calcium-conserving effect of calcitonin in human (160). The
primary site of action for calcitonin in the rat is the thick
ascending limb (TAL) of the loop of Henle, where calcitonin has been demonstrated to bind its receptor, increase local adenylate cyclase activity, and promote calcium reabsorption (166, 168). Apart from enhancing the
reabsorption of calcium, calcitonin also promotes the
vectorial transport of NaCl in the rat TAL, thereby amplifying the corticomedullary concentration gradient,
which is a prerequisite for the subsequent concentration
of urine in the collecting duct (265, 304). In the rabbit,
the calcium-conserving effects of calcitonin seem to be
mediated by the distal tubule (1225).

duced in HEK-293 cells following calcitonin exposure,


suggesting a regulatory role of calcitonin in vitamin D
catabolism (361). In addition to its putative direct effect
on calcium reabsorption, calcitonin may thus affect normal calcium metabolism indirectly by modulating the
levels of circulating 1,25(OH)2-vitamin D.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


current serum calcium levels, it presides over the subsequent
hierarchical cascade of vitamin D synthesis and calcium
handling in the vitamin D target organs, such as the intestine, kidney, and bone. More recent investigations have
demonstrated that the CaSR is not exclusively expressed in
the parathyroid gland, but is also present locally in the
vitamin D target organs. This diverse expression suggests
that CaSR can modulate organ function locally and outside
of the strict PTH-vitamin D-organ axis. Thus a short and
local feedback loop is created, which allows the organs to
respond rapidly to the local calcium environment. A detailed description of the receptors role in each tissue is
beyond the scope of this article, but an attempt will be made
to identify the key features of CaSR in these local sites in a
subsequent section (FIGURE 8). For excellent in-depth reviews, please refer to Refs. 372, 711, 906.

1. Structure and signaling


The CaSR was first cloned by Brown et al. (134) in 1993
from bovine parathyroid using a Xenopus oocyte expression cloning system. Cloning of the human CaSR followed
2 years later (366). The CaSR is a 1,028-amino acid protein
that belongs to the superfamily of classic 7-transmembrane
domain G protein-coupled receptors (134, 366). It is mainly
expressed as a homodimer on the cell surface (55). The
dimerization process has been shown to take place in the ER
and is mediated by the formation of disulfide bonds between cysteine residues (C129, C131) and noncovalent interactions of leucine residues (L112, L156) in the extracellular domain of the receptor (54, 529, 858, 888, 1213).
Following assembly in the ER, the CaSR undergoes
N-linked glycosylation in the Golgi apparatus, some of
which is pivotal for cell surface expression (887). The trafficking between ER and Golgi apparatus is regulated by the
small GTP-binding protein Rab1 (1221). Knockdown and
mutations of Rab1 in HEK cells results in decreased numbers of CaSR at the cell surface (1221). Conversely, the
internalization of CaSR is thought to be mediated by ubiquitination by the E3 ligase dorfin (498).

In lack of a crystal structure of CaSR, the exact binding sites


of calcium on the extracellular domain remain subject of
speculation. So far, applicable structural data has only been
obtained from the metabotropic glutamate receptor type I
(mGluR1), which belongs to the same family of type C
GPCRs. In this model, glutamate binds to key residues
which are located in a cavity, embedded in between two
lobular domains (LB1 and LB2) of the extracellular tail
(611). This structural hallmark has been aptly coined the
receptors Venus fly trap module. This motif is conserved
among other GPCRs of the same family. Multiple attempts
to identify the calcium binding sites have been undertaken
using computational homology modeling (499, 500, 1014).
These models have postulated between one and five calcium
binding sites (499, 500, 1014). With the employment of
mutational analysis, it has been possible to validate functionality of some of these putative binding sites (500). Monitoring of the intracellular calcium response to increasing
extracellular calcium levels in CaSR transfected HEK cells
had indicated previously that the Hill coefficient for this
response was 3.1, suggesting that multiple calcium binding sites may exist (834).
It should be noted that the CaSR can also be stimulated by
other polyvalent cations (Mg2, Pb2, Cd2, Fe2, Ba2,
Ni2, Co2, or Gd3) and larger polycationic molecules,
such as spermine, spermidine, putricine, protamine, and
neomycin (134, 416, 880). Furthermore, several substances
can allosterically modify the receptor and potentiate its sensitivity to its direct agonists. These include pharmacological
small molecule substances (calcimimetics) that are in clinical use for the treatment of conditions, such as secondary
hyperparathyroidism, and L-type amino acids, which enable the CaSR to act as a nutrient sensor (220). Truncation
studies have demonstrated that the Venus fly trap motif is
necessary for allosteric modification by L-type amino acids
(759). The affinity of calcium to the CaSR can also be modulated by changes in extracellular pH (279, 879). An acidic
extracellular milieu has been shown to decrease the sensitivity of CaSR to its agonists, whereas an increased extracellular pH has converse effects (879).
The intracellular domain of CaSR contains five PKC phosphorylation sites (366). Mutational analysis demonstrated
that PKC-mediated phosphorylation of the CaSR at Thr888 blunts its response to extracellular calcium, as evidenced by inhibited calcium release from intracellular

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

227

Downloaded from on December 8, 2014

The importance of CaSR as a regulator of calcium balance


is underlined by clinical pathologies that are caused by its
mutation. Loss-of-function mutations cause familial hypocalciuric hypercalcaemia (FHH; OMIM 145980) and
neonatal severe hyperparathyroidism (NSHPT; OMIM
239200), whereas activating mutations cause autosomal
dominant hypocalcemia (OMIM 601198). These mutations mostly change the threshold of receptor activation in
either direction. Currently, 300 different mutations are
reported, two-thirds of which represent inactivating mutations (241). Given its broad expression pattern, the deranged sensing of blood calcium levels in these disorders not
only affects the secretion of PTH from the parathyroid
glands, but also causes local dysfunction in other organs,
such as the kidney where calcium absorption is perturbed.

On the cell surface, the CaSR resides in caveolin-1-rich


plasma membrane domains, which also contain associated
signaling proteins (571). These signaling complexes are
formed with the help of scaffolding proteins. The COOHterminal tail of CaSR binds to filamin A, an actin binding
protein (48, 467). Silencing of filamin A with siRNAs results in the attenuation of MAPK signaling by the receptor
(496) (see below).

SASCHA KOPIC AND JOHN P. GEIBEL

Calcium-sensing receptor
Kidney
Proximal tubule

Collecting duct

Distal convoluted tubule

CaSR

CaSR

CaSR

HPO42

CaSR

CaSR
NaPi-IIa

Na+

H+

Ca2+
V-ATPase

TRPV5

25(OH)-vitamin D

CaSR

CaSR
Mitochondria:
CYP27B1

H2O
1,25(OH)2-vitamin D

AQP2

Enterocyte

ROMK
K+

CaSR
CaSR

NKCC2

CaSR

Na+

CFTR

2Cl

Cl

K+
3

Ca2+
H2O

Stomach

Bone
G-cell

PPIs
APAs

CaSR

CaSR

H+
H,K-ATPase

CaSR

CaSR

K+

?
Gastrin
CaSR

ECL-cell +
parietal cell
activation

Differentiation
and function

RANKL

RANK

?
Osteoblast

Circulation

H+

V-ATPase

Parietal cell

FIGURE 8. CaSR in the gastrointestinal tract, kidney, and bone. Kidney: the effects of CaSR activation on ion
transport in various nephron segments are shown. In the proximal tubule, CaSR stimulates phosphate
absorption and 1,25(OH)2-vitamin D synthesis. In the thick ascending limb of the loop of Henle, CaSR inhibits
apical potassium channels (ROMK), thereby inhibiting NKCC2 (potassium recycling). The resulting changes in
the lumen-positive potential inhibit paracellular calcium uptake. In the distal convoluted tubule, CaSR presumably stimulates apical calcium entry through TRPV5. In the collecting duct, CaSR stimulates proton extrusion
through the V-type ATPase and inhibits urine concentration through AQP2. Stomach: in the parietal cell, CaSR
induces acid secretion by activating H-K-ATPase. In the G-cell, CaSR activation results in gastrin secretion.
Of note, CaSR serves as a luminal nutrient and calcium sensor on the G-cell. Bone: CaSR on osteoblasts
presumably regulates their differentiation and RANKL expression. Intestine: in the intestine, CaSR activation
reduces water secretion by inhibiting chloride secretion through CFTR.

228

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Osteoclast

Downloaded from on December 8, 2014

Intestine
Thick ascending limb of
the loop of Henle

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


stores (56, 246). The phosphorylation occurs in response to
receptor activation and thus represents an autoinhibitory
feedback mechanism (246). -Arrestins are most likely involved in the process of PKC-associated desensitization
(681, 853). Conversely, dephosphorylation of the Thr-888
residue is carried out by a calyculin-sensitive phosphatase,
thereby restoring the receptors initial sensitivity (246). Another mechanism of desensitization is mediated by a G protein receptor kinase (GRK), most likely by interfering with
Gq regulated pathways (see below) (681, 853).

2. CaSR in the parathyroid


CaSR regulates parathyroid function at three levels: 1) the
release of PTH from secretory granules, 2) de novo synthesis of PTH, and 3) parathyroid cell growth.
Activation of CaSR by increasing plasma calcium results in
an inhibition of PTH release, thereby lowering calcium levels. It is thought that this response is mediated by the generation of arachidonic acid metabolites via Gq and PLA2
activation (FIGURE 6) (121, 152). Cultured porcine parathyroid cells demonstrated an increase in arachidonic acid production after CaSR stimulation while PTH release was inhibited (121). Furthermore, exogenous administration of
arachidonic acid suppressed PTH release from the parathyroid cells (121). Similar effects were demonstrated for the
arachidonic acid metabolites 12- and 15-hydroxyeicosatetranoic acid, suggesting that they represent the downstream
effectors of arachidonic acid production (120).
Apart from directly controlling PTH release, CaSR also
modulates PTH synthesis. PTH gene transcription is mainly
regulated by 1,25(OH)2-vitamin D. Binding of 1,25(OH)2vitamin D to the VDR causes a decrease in pre-pro-PTH
mRNA levels creating a negative-feedback loop (154, 930,
931). However, it was recognized before the identification
of the CaSR that serum calcium can modulate the actions of
1,25(OH)2-vitamin D on PTH gene transcription (930). It

The molecular mechanisms underlying the trophic effects of


CaSR activation are less clear. Earlier observations had already suggested that hypocalcemia is associated with parathyroid cell proliferation (778). Currently, the CaSR specific calcimimetics provide the most useful insight into the
regulation of parathyroid growth by CaSR. Calcimimetics
administered in the context of both animal models and
clinical studies of hyperparathyroidism demonstrate that
activation of CaSR leads to a reduction in gland size
(510, 589, 746, 1128). Conversely, inactivating mutations of CaSR result in parathyroid enlargement. Parathyroid-selective genetic disruption of Gq was furthermore shown to cause moderate hyperparathyroidism
with increased plasma PTH levels and gland hyperplasia,
suggesting a role of Gq in the regulation of parathyroid
cell growth (849). Similar findings were reported in
Gq/11 double KO animals (1159).
3. CaSR in the kidney
The CaSR acts as an important regulator of ion and water
homeostasis in the kidney. It should be noted that it can
exert its effects on calcium transport independently of other
hormonal regulators, such as PTH and 1,25(OH)2-vitamin
D. The CaSR is expressed along most of the nephron, albeit
in varying subcellular localizations (FIGURE 8) (907, 908).
In the proximal tubule, CaSR is localized apically at the
base of the brush border, where it has been implicated to
play a role in phosphate transport (52, 907, 909). The primary regulator of phosphate transport in the proximal tubule of the kidney is PTH. In brief, increased PTH levels
inhibit phosphate reabsorption from the lumen. Activation
of the apical CaSR can partially reverse the effects of PTH
and restore phosphate absorption (52). Conversely, PTH
and high phosphate levels reduce CaSR expression (909).
Furthermore, it is likely that the CaSR mediates the inhibitory effects of calcium on 1,25(OH)2-vitamin D synthesis in
the proximal tubule (109, 702).
In the thick ascending limb of the loop of Henle, CaSR is
located on the basolateral membrane (907). In this nephron
segment, the receptor acts as a major modulator of monovalent and polyvalent ion absorption. Activation of CaSR
leads to an inhibition of the apical renal outer medullary

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

229

Downloaded from on December 8, 2014

Once stimulated, the CaSR activates a variety of intracellular signaling cascades. Being a GPCR, most of these processes are mediated by G proteins. Specifically, Gq/11, Gi,
and G12/13 have been shown to be coupled to the CaSR
(40, 175, 495, 849). The expression of all subunits was
confirmed in bovine parathyroid (1115). Gi mediates the
suppression of cAMP levels by inhibiting adenylyl cyclase
and activates the ERK/MAPK pathway (175, 250, 375,
572). Activation of Gq/11 results in increased intracellular
calcium concentrations via activation of PLC and IP3 triggered calcium release (133, 1010). As demonstrated in HEK
cells, this cascade can also activate further downstream
phospholipase A2 leading to production of arachidonic acid
metabolites (415). G12/13 is thought to regulate phospholipase D and phosphatidylinositol 4-kinase (PI 4-K); however, this interaction has only been demonstrated in heterologous cell culture system (494, 495).

was shown that increases in calcium can potentiate the inhibitory effects of 1,25(OH)2-vitamin D (930). This effect is
most likely mediated by CaSR, whose activation can decrease
PTH transcription by augmenting the inhibitory effects of
1,25(OH)2-vitamin D. Molecularly this is achieved by upregulating the expression of the VDR (151, 162, 362, 653, 916).
The current working model states that activation of CaSR
causes an increase of arachidonic acid metabolites and activation of the MAPK pathway, which in turn results in increased
VDR mRNA levels (FIGURE 6) (151). This allows the parathyroid to adjust its 1,25(OH)2-vitamin D sensitivity to the current plasma calcium levels.

SASCHA KOPIC AND JOHN P. GEIBEL

In contrast to the medullary section, the cortical portion of


the thick ascending limb has been proposed to have predominantly active calcium uptake properties, which are under the hormonal regulation of PTH and calcitonin (344,
509). PTH increases calcium absorption in this segment,
and it has been shown that, similarly to phosphate absorption in the proximal tubule, activation of CaSR can suppress the effects of PTH (264, 754). The absorption of NaCl
does not seem to be affected by CaSR (264, 754).
The distal convoluted tubule and the connecting tubule are
responsible for the fine-tuning of calcium reabsorption in
the kidney. To achieve this goal, they are equipped with
molecular machinery, similar to that in the duodenum
(TRPV5, calbindin-D 28k, NCX1, and PMCA1b) to absorb calcium against its electrochemical gradient through
the transcellular pathway (680). In analogy to the proximal
small intestine, these transporters are predominantly regulated through 1,25(OH)2-vitamin D, but also PTH. CaSR
colocalizes with TRPV5 in this segment (1090). Its activation causes increase calcium influx through TRPV5 and
may thereby locally and rapidly adapt active absorption to
the urine calcium concentration (FIGURE 8) (1090).
Apart from regulating calcium and phosphate absorption in
the kidney, CaSR modulates proton and water movement in
the collecting duct. In the intercalated cells of the collecting
duct, apical V-ATPase acidifies the urine in an effort to
maintain systemic acid-base homeostasis. It has been shown
that luminal calcium and neomycin can induce V-ATPase
activity via activation of CaSR, thereby causing proton secretion into the urine (FIGURE 8) (901). Since the formation

230

of calcium kidney stones is dependent on luminal pH, it has


been speculated that this may represent an autoprotective
mechanism that prevents nephrolithiasis (901). Furthermore, stimulation of apical CaSR in the principal cells of the
collecting duct leads to decreased ADH (vasopressin)-stimulated water reabsorption through AQP2 (FIGURE 8) (942,
943). Taken together, activation of CaSR has diuretic effects via inhibiting NKCC2 in the thick ascending limb of
the loop of Henle and by inhibiting AQP2-mediated water
reabsorption the collecting duct.
4. CaSR in the gastrointestinal tract
The CaSR is distributed along most of the gastrointestinal
tract, ranging from the stomach to the large intestine (186,
360, 744, 932, 998). We are now only slowly beginning to
unravel its function in this diverse array of tissues. In the
stomach, CaSR localizes to the basolateral membrane of
the acid-secreting parietal cells and to all membranes of the
gastrin-secreting G-cells (FIGURE 8) (142, 182, 886). Primary cultures of G-cells were shown to release gastrin after
stimulation of the CaSR with calcium (142, 886). The release is mediated via calcium influx into the cytosol through
nonselective cation channels opening after CaSR stimulation (142). These findings provide the molecular basis for
the observation that rises in serum calcium can increase
serum gastrin levels (see sect. IIB2). The apical expression of
CaSR in G-cells theoretically enables it to act as a luminal
nutrient sensor modulating gastric acid secretion and other
parameters. In recent studies there is direct evidence showing that gastrin levels increase in mice after calcium and
L-type amino acid ingestion (325). This effect was abolished in CaSR (/) animals (325). In healthy human test
subjects, pharmacological stimulation of CaSR leads to a
concomitant increase in gastrin levels and gastric acid output (165). CaSR on G-cells was thus postulated to play an
important role in the gastric phase of acid secretion by
maintaining acid output by maintaining gastrin secretion
(325).
Apart from being expressed on G-cells, CaSR is also localized on the basolateral membrane of the acid-secreting parietal cell, where it exerts effects that are independent of
gastrin and other secretagogues. Activation of parietal cell
CaSR has been reported to increase H-K-ATPase-mediated proton secretion, thereby acidifying the gastric lumen
(FIGURE 8) (145, 291, 373). This stimulatory effect was
demonstrated for direct activators, such as calcium or
Gd3, but also allosteric modifiers, such as L-type amino
acids (145, 291, 373). In parallel to other tissues, the intracellular activation signal for H-K-ATPase is mediated by
rises in intracellular calcium, PLC, MAPK, and PKC (899).
In conclusion, both rises in luminal and plasma calcium
concentrations can induce gastric acid secretion either indirectly through gastrin release or directly through parietal
cell activation. The physiological significance of this observation remains the subject of speculation, but may be linked

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

potassium (ROMK; Kir1.1) channel, mainly through arachidonic acid metabolites created by PLA2 (1147, 1148).
Apical ROMK releases potassium ions into the lumen,
which in turn are needed to fuel apical ion uptake through
the Na-K-2Cl (NKCC2) cotransporter. By decreasing
apical potassium efflux, CaSR inhibits sodium and chloride
uptake through NKCC2 (906). This correlation is reflected
in much earlier observations, which report that calcium
infusions can decrease tubular sodium clearance (300, 718,
1052). In addition, impairment of NKCC2 has also implications for calcium absorption. Reduced NKCC2 activity
decreases the lumen-positive potential and negatively affects countercurrent multiplication, and in consequence the
nephrons ability to concentrate urine (434). Both mechanisms will lead to impaired calcium absorption (434). Calcium absorption in the medullary portion of the thick ascending limb is thought to occur predominantly as passive
uptake through the paracellular route (995). Similar observations have been made when blocking NKCC2 pharmacologically with the loop diuretic furosemide (299). It has thus
been proposed that activation of basolateral CaSR has
loop diuretic-like effects, reducing NaCl but also calcium
absorption in the kidney (434).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


to facilitating calcium uptake by increasing acid output. As
described in a subsequent section, it has been speculated
that gastric acid increases the bioavailability of ingested
calcium (see sect. V). Alternatively, CaSR may primarily
function as a nutrient sensor in the stomach (amino acid
sensing), which maintains constant acid output in the gastric (apical G-cell sensing) and postprandial (basolateral
parietal cell) phase of digestion, when circulating levels of
amino acids are high (325).

5. CaSR in bone
It is well established that CaSR is expressed in osteoblasts,
osteoclasts, and their respective precursors (172, 174, 542,
11831185, 1192). The functional role of CaSR in these
cells is, however, less clear. Undoubtedly, both cell lines are
exposed to local fluctuations in calcium concentrations
making an adaptive response to the calcium environment
plausible. Indeed, changes in extracellular calcium concentration have been shown to regulate various cell functions,
mostly in in vitro models. Extracellular calcium can stimulate the proliferation, migration, and differentiation of osteoblasts (174, 297, 1183, 1184, 1186). Similarly, calcium
was proposed as a differentiation signal for osteoclasts
(542, 544, 734). Significant doubt about the in vivo importance of CaSR in bone has emerged with the generation of
the CaSR (/) mice. Although CaSR knockout results in
rickets, these animals suffer from severe hyperparathyroidism, which did not allow a discrimination between the effects of high PTH and CaSR on bone turnover (365). Con-

Although these observations have profoundly questioned


the physiological significance of CaSR in bone, closer examination still favors a role of CaSR in bone turnover. With
the recent advances in genetic methods, an osteoblast-specific CaSR (/) model has been created (171). These animals have severely stunted growth and skeletal development, clearly suggesting an involvement of CaSR in normal
osteoblast function (171). The previous conflicting evidence
gained from global CaSR (/) models with survival rescue
by elimination of PTH synthesis have been attributed to the
possible expression of alternate CaSR splice variants, which
may compensate for the deletion of full-length CaSR in
these animals (171, 915). In an attempt to further elucidate
the function of CaSR in osteoblasts, the reverse approach
has been executed by specifically upregulating CaSR in osteoblasts with use of a constitutively active receptor mutant
(296). Upregulation of CaSR results in bone loss, as evidenced
by a decrease in bone volume and density, specifically of trabecular bone (296). These findings are accompanied by an
increased number of osteoclasts, whereas osteoblast parameters were essentially unchanged (296). Activation of CaSR has
been speculated to promote RANKL production by osteoblasts, which serves as an osteoclastogenic signal (296). Osteoblasts may thus recruit osteoclasts and induce their maturation
via CaSR signaling and increased RANKL expression, which
would explain the observed increase in bone turnover and
osteoclast numbers in the setting of constitutive CaSR activation (241).

V. THE STOMACH AND CALCIUM


Preceding parts of this review have independently summarized the physiology of acid secretion, intestinal calcium
absorption, and their respective regulation. The following

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

231

Downloaded from on December 8, 2014

In the intestine, functional investigations on CaSR have


mainly been conducted in colonic epithelia, where CaSR
localizes to both the basolateral and apical membranes of
the colonic crypt (173, 186, 360). Expression patterns vary
slightly in the small intestine, with general basolateral expression and additional weak apical expression in the villus
(173, 360). Furthermore, CaSR is expressed in both the
Meissners and Auberachs plexuses. Early experiments on
single perfused colonic crypts demonstrated that intracellular calcium concentrations could be increased when exposing the crypts to classic CaSR agonists and that forskolinstimulated fluid secretion could be inhibited (186). This and
subsequent investigations indicate that CaSR plays an important role as a modulator of colonic fluid secretion (185,
186). Subsequently, attempts have been made to take advantage of the constipatory effects of CaSR activation in
pathophysiological settings. Activations of CaSR in the
course of diarrheagenic enterotoxin exposure was shown to
decrease fluid secretion via increased breakdown of cyclic
nucleotides (371). Although the potential clinical applications of ameliorating the symptoms of secretory diarrhea
are promising, more efforts will have to be made to fully
unravel the physiological role of CaSR in intestinal ion and
fluid transport. So far it is not clear whether intestinal CaSR
can modulate calcium absorption, as is the case in the kidney.

comitant genetic ablation of the parathyroid gland or PTH


secretion, however, revealed that the skeletal phenotype of
CaSR single mutation (/) could mostly be rescued, suggesting that the skeletal abnormalities were due to high
circulating PTH levels rather than CaSR inactivation (598,
1096). Furthermore, CaSR does not seem to be the exclusive calcium-sensing mechanism in osteoblasts, as changes
in extracellular calcium can still elicit functional responses
in CaSR (/) osteoblasts (852). This observation has been
attributed to another GPCR with calcium-sensing capabilities, namely, GPRC6A (850, 851). Although GPRC6A has
a higher activation threshold for calcium, it also responds to
the CaSR allosteric activator R568 (851). GPRC6A activation may thus represent a confounding factor in most in
vitro studies on osteoblasts and their modulation by CaSR.
Also, GPRC6A knockout leads to osteopenia, further underlining the possibility of an alternate calcium-sensing
pathway in bone (850). Osteoblasts extracted from these
GPRC6A-deficient animals show decreased sensitivity to
extracellular calcium and in vitro mineralization defects
(854).

SASCHA KOPIC AND JOHN P. GEIBEL


section will attempt to illustrate the functional intersections
between these seemingly unrelated fields. In particular, the
question of whether acid is needed to absorb calcium effectively from the gut or whether the stomach contributes to
the regulation of calcium homeostasis by secretion of an
endocrine substance will be investigated.

A. Proton Pump Inhibitors and the Risk


of Fracture
In May 2010, the Food and Drug Administration (FDA)
released the following safety announcement: Healthcare
professionals and users of proton pump inhibitors should
be aware of the possible increased risk of fractures of the
hip, wrist, and spine with the use of proton pump inhibitors, and weigh the known benefits against the potential
risks when deciding to use them (319).

The roots of this controversy may potentially be traced back


to the 1940s and 1950s. Before the advent of PPIs, total and
partial gastrectomies or vagotomies were performed to control acid-related disorders. It was soon apparent that patients who underwent these radical surgical procedures developed osteoporosis/-malacia (58, 305, 732, 876). A study
that assessed the prevalence of osteomalacia in gastrectomized patients concluded that up to 12% of patients (19%
of females) had histologically overt osteomalacia, although
general disturbances in calcium metabolism were estimated
to occur in up to 28% of patients (208, 368). Other inves-

232

Back in the field of PPIs, the seminal epidemiologic investigation by Yang et al. was subsequently followed up by a
number of studies, which also focused on other types of
fractures, other populations, and other drugs reducing gastric acid output, such as H2 receptor antagonists (202, 228,
252, 394, 400, 559, 868, 923). Although their conclusions
were somewhat controversial, a recent meta-analysis supports the initial hypothesis that a correlation between PPI
intake and fracture risk (hip, spine, and any-site fractures)
exists (1195). The meta-analysis considered 11 studies and
identified an overall odds ratio of 1.30 for all fracture types
combined (1195). There was no association between H2
blocker intake and an increase in fracture risk, although
some single studies supported a link (228, 1195). Another
meta-analysis came to a comparable conclusion with regard
to an increased fracture risk under PPI exposure (616).

B. Gastric Acid and Intestinal


Calcium Uptake
A variety of reasons could theoretically account for the
observation that PPIs increase the likelihood of fractures.
The most prominent hypothesis assumes that the reduced
acidity in the stomach impairs the intestinal absorption of
dietary calcium. This assumption is based on both patient
observations and experimental animal data. Alas, the number of animal studies, which in contrast to investigations in
humans per default allow more radical experimental designs and genetic manipulation, is very small.
1. Effects of gastrectomy, vagotomy, and PPIs on
mineral metabolism in humans
Before discussing the reports that try to correlate PPI use
with calcium uptake, it is worthwhile to examine older
literature on patients that had undergone partial or total
gastrectomy. As discussed previously, these procedures are
known to be linked to bone disease. In contrast to PPIs,
which eliminate the singular factor of acid secretion, gastrectomies also influence gastric emptying, the emulsification of food stuffs, and food habits. It is thus more difficult
to draw conclusions on the influence of acid secretion on
calcium absorption from gastrectomized patients than from
individuals on PPI therapy. A further caveat lies in the type
of gastrectomy, as different surgical procedures are and
were in use. Some surgeries bypass the duodenum (Billroth
II, Roux-en-Y, total gastrectomy), whereas some leave the
duodenal passage intact (Billroth I).

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Proton pump inhibitors (PPIs, see sect. IIC1) are in widespread use for the treatment of acid-related disorders, such
as gastroesophageal reflux disease (GERD) or gastric ulcer
disease. They exert their curative effects by inhibiting the
acid output of the stomach. Over the recent years, mostly
epidemiological evidence has accumulated which links the
intake of PPIs to an increased risk of sustaining fractures,
especially in the elderly population. Yang et al. (1190) published one of the earliest and largest studies investigating
this potential correlation in 2006. Examining a population
of over 13,000 hip fracture cases and over 135,000 controls
over the age of 50, the authors concluded that long-term
(over 1 year) PPI use was associated with an increase in hip
fractures (AOR 1.44) (1190). Although the likelihood of
sustaining a fracture following PPI intake may seem fairly
low, the implications for public health are substantial. This
has multiple reasons: PPIs represent the third most commonly prescribed medication in the United States and are
also available as over-the-counter formulations. Furthermore, there is an ongoing debate whether PPIs are overprescribed, putting certain populations at unnecessary risk of
side effects. In combination with the high incidence of osteoporotic fractures, the mean incidence of hip fractures
alone between 1986 and 2005 was 957 per 100,000 women
over the age of 65 per year, a small increase in risk suddenly
has implications for a very large population (123).

tigations came to lower prevalence results of 510%


(1091). The osteomalacia was also shown to translate into
an increased incidence of fractures in these patients (795).
Naturally, gastrectomy represents a radical intervention,
and the reasons for this correlation may be multifactorial,
but reduced acid output may be of significance.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH

The intrusiveness of gastric surgery makes it difficult to


dissect the influence of gastric acid on these parameters.
This is why vagotomized patients are a somewhat more apt
patient population to study the effects of gastric acid on

calcium uptake, albeit the number of studies on this cohort


is very limited. Vagotomy abolishes the parasympathetic
input to the stomach and thereby decreases the amount of
secreted acid. Although the gross anatomy of the stomach
remains intact, other parameters, such as gastrin levels, are
also deranged given the important role of the vagus nerve in
the regulation of gastric acid secretion (see sect. IIB). While
bone disease is generally not reported in these patients, low
25(OH)-vitamin D levels are common (514, 793). Similarly
to gastrectomy, the 1,25(OH)2-vitamin D levels are concomitantly elevated, suggesting adaptive upregulation potentially to compensate for decreased calcium absorption
(793). As discussed previously, the decreased 25(OH)-vitamin D could be indicative of augmented catabolism of the
vitamin (244). Serum calcium is commonly decreased or in
the lower normal range, while intestinal calcium absorption
is increased, presumably in response to elevated 1,25(OH)2vitamin D (110, 974). Secondary hyperparathyroidism does
not manifest (793, 974).
In general, the disturbance in mineral metabolism is more
pronounced in gastrectomized patients than in vagotomized patients, as evidenced by the higher incidence of bone
disease and secondary hyperparathyroidism. It is challenging to draw clear conclusions on the influence of gastric acid
on calcium absorption in either patient group. Yet, it is
apparent that compensatory mechanisms are in place in
these patients, as evidenced by the increased 1,25(OH)2vitamin D and PTH levels. Less efficient calcium uptake due
to decreased acid output may be one explanation for this,
but without further analysis this conclusion remains speculative.
With the advent of PPIs and H2 blockers, the number of
surgical interventions to control acid-related disorders decreased massively. Given their high specificity, PPIs selectively eliminate gastric acid output. Several investigations
that try to tie PPI intake to a disturbance in mineral metabolism exist. Graziani et al. (396) observed in eight healthy
volunteers that postprandial calcium concentrations did
not increase in subjects on a PPI regime (omeprazole 20 mg
3 daily), whereas control subjects demonstrated a clear
spike in serum calcium levels. Urine calcium excretion was
also reduced compared with the control group (396). A
similar effect of PPIs was later observed by two independent
groups in patients undergoing hemodialysis (395, 423). It
should be noted that neither of these studies directly assessed intestinal calcium absorption, but rather measured
serum calcium as an indirectly related parameter. More
recently, intestinal calcium absorption was measured by
OConnell et al. (807) using a radiolabeled calcium isotope.
The investigators reported that 7 days of PPI (omeprazole
20 mg 1 daily) intake significantly reduced calcium absorption in elderly women under fasting conditions compared with the placebo group. Although these studies support a role of PPIs in reducing calcium uptake, conflicting

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

233

Downloaded from on December 8, 2014

A common finding among gastrectomized patients is their


low 25(OH)-vitamin D levels, while levels of 1,25(OH)2vitamin D seem to be increased (101, 244, 378, 511, 794,
972, 1081). The pathophysiological reason for this is not
entirely clear. It has been argued that bone disease and low
25(OH)-vitamin D are a result of impaired vitamin D absorption following surgery; however, the consensus seems
to be that uptake rates of vitamin D is not impaired in these
patients (245, 378; contested by Ref. 1081). The vitamin D
insufficiency may also be a byproduct of improper nutrition
(378). As fat and milk intolerance can develop, especially in
surgeries which exclude the duodenum (Billroth II), a
change of dietary habits with insufficient intake of the fatsoluble vitamin D may be an underlying cause. Indeed,
long-term longitudinal studies suggest that maintaining
body weight reduces the risk of developing bone disease
after gastrectomy, which emphasizes the role of adequate
nutrition (667, 668). On the other hand, Billroth I and II
patients show the same loss in bone density, although Billroth II surgery (bypassing of the duodenum) is associated
with a much higher degree of fat malabsorption (110, 651).
A different investigation even concluded that Billroth I patients have a higher loss in bone density than Billroth II
patients (794). A more recent report suggests that the problem underlying bone disease after gastrectomy may be impaired calcium absorption, rather than dietary vitamin D
deficiency (244). It has been shown that high 1,25(OH)2vitamin D levels accelerate the breakdown of 25(OH)-vitamin D (210 212, 244). The observed low 25(OH)-vitamin
D levels in gastrectomized patients may thus be a byproduct
of increased catabolism, and not insufficient intake,
whereas the high 1,25(OH)2-vitamin D levels may represent
compensatory upregulation due to insufficient calcium absorption or intake (244). Calcium absorption in gastrectomized patients (Billroth I II) has been reported to be in the
low-normal range, while 1,25(OH)2-vitamin D levels are
increased (794). In line with these findings, secondary hyperparathyroidism, an indicator of compensatory upregulation due to low serum calcium levels, is a known finding
after gastrectomy (101, 244, 1171). Other investigations on
intestinal calcium absorption in gastrectomized patients
came to very contradictory results, ranging from increased
to impaired absorption (8, 34, 255, 398, 585, 794). Many
of these reports failed to assess 1,25(OH)2-vitamin D and
PTH levels, which means that adaptive mechanisms may
mask the insufficient baseline uptake of calcium in the intestine (8, 34, 255, 398, 585, 794). In conclusion, the exact
pathogenesis of postgastrectomy osteopenia remains somewhat unclear. The disorder may be attributable to vitamin
D insufficiency, impaired calcium absorption, inappropriate diet, or a combination of all factors.

SASCHA KOPIC AND JOHN P. GEIBEL


evidence exists. Several investigations that assessed calcium
absorption using radioactive tracers and a whole gut lavage
technique found no evidence for a decrease in absorption
under short-term PPI treatment (421, 991, 1173). It is not
clear why these discrepancies in the outcomes of the trials
exist. There is, however, variability in the experimental
technique to measure calcium uptake, the cohorts investigated (young vs. postemenopausal women vs. dialysis patients) and the form of calcium administration (calcium
salts vs. whole meals), which may partially account for the
divergent results. Indeed, different calcium salts are absorbed with different effectiveness in acid suppressed individuals, which will be subject of later discussion (see sect.
VC). Furthermore, different populations may have different
capacities for endocrine compensation.

2. Effects of gastrectomy, vagotomy, and PPIs on


mineral metabolism in the animal model
To further elucidate the problem of osteopenia following
gastrectomy or PPI use, several animal studies tried to replicate and expand the observations made in human test
subjects.
For example, Axelson et al. (49) measured serum calcium
concentrations in rats who had undergone parathyroidectomy and various surgical procedures to reduce gastric acid
output (vagotomy, antrectomy, gastrectomy). While parathyroidectomy alone predictably reduced serum calcium
levels, the gastric operations (with intact parathyroid
glands) had little to no effect on calcium concentrations
(49). Interestingly, intestinal calcium absorption was even
increased in the latter group. The authors attributed this
observation to a compensatory upregulation of PTH secretion and concomitant 1,25(OH)2-vitamin D production. To
eliminate this factor, gastrectomy or fundectomy was conducted after parathyroidectomy, thereby depriving the animals of their compensatory machinery. This intervention
resulted in massive hypocalcemia and death after a few
days, which led the authors to conclude that acid secretion
is important for the maintenance of calcium homeostasis
(49). Another investigation in rats that had undergone
antrectomy (Billroth I) observed a significantly decreased
absorption of calcium (345). Fundectomy did not affect
calcium absorption (927). However, both studies employed
the balance method to calculate calcium absorption, which
is considered less accurate than using radiotracers. In pigs,

234

In addition, several reports of vagotomy in a rat model


are available to us (49, 307, 308, 928). It has been demonstrated that vagotomy alone has no effect on the rate of
intestinal absorption (307, 928). Secondary hyperparathyroidism was observed by one group, while PTH levels
were reported to be unaffected by the other group (307,
928). 1,25(OH)2-vitamin D was not measured, which
would have provided further evidence of compensation
due to decreased calcium bioavailability. However, if vagotomy and parathyroidectomy are performed together,
intestinal calcium absorption is significantly impaired
compared with vagotomy or parathyroidectomy alone
(307). This is in accordance with the low serum calcium
concentrations found in gastrectomized and parathyroidectomized rats (49).
PPIs were also used to relate acid secretion to bone disease in rats. Bone weight did not change in rats that were
treated for 4 wk with omeprazole (841). Alas, calcium
absorption was not measured in these animals, and a
decrease in bone weight represents a very terminal and
long-term outcome. A more recent investigation by
Schinke et al. (963) demonstrates that mice which have
been genetically manipulated to be achlorhydric (CCK2
/) have decreased serum calcium levels as well as develop osteoporosis and secondary hyperparathyroidism
in an effort to maintain calcium balance (963). This study
is especially noteworthy, as acid secretion is knocked out
selectively in this mouse model while the stomach remains intact (stomach morphology). Furthermore, a genetic mutation that has been associated with osteopetrosis (a disease characterized by increased bone density)
due to osteoclast malfunction was also shown to cause
decreased gastric acid secretion. These patients present
with lower serum calcium values. Rather than being a
product of impaired bone resorption (osteoclast defect),
the hypocalcemia may thus be related to impaired intestinal calcium absorption (gastric acid secretory defect)
(963).

C. Calcium Salts
Many investigators have employed calcium salts to determine the efficacy of intestinal calcium absorption. Furthermore, calcium salts are in wide clinical use as a dietary supplement. As will be discussed in this section,
calcium salts differ in their bioavailability, which not
only represents a potential source of error in experimental designs, but more importantly, has extensive clinical
implications.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

In summary, it is experimentally difficult to unmask the


potential correlation between a reduction in gastric acidity
and calcium absorption, given our bodys high capacity for
compensation. In addition, slight alterations in mineral homeostasis may take years to manifest themselves clinically,
for example, in osteopenia or fractures. Without following
up on test subjects on a long-term basis, snapshot measurements which may still lie within clinically normal range can
be misleading.

total gastrectomy causes massively reduced calcium uptake


and secondary hyperparathyroidism (700). In this study,
the duodenum was surgically bypassed by esophagojejunostomy, thereby eliminating the site of maximal active
calcium absorption and limiting the conclusion that can be
drawn (700).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


(520). The group reported that absorption of calcium carbonate was severely impaired in four male patients suffering
from achlorhydria (520). Compared with five control subjects, who absorbed between 9 and 18% of the ingested
calcium carbonate, these patients only absorbed 0 2%. Interestingly, when gastric acid secretion of one of these patients was stimulated by administration of betazol hydrochloride (a histamine analog), calcium carbonate absorption rose from 2 to 10% (520). This investigation somewhat
spawned the entire controversy of whether gastric acid is
necessary to absorb calcium effectively from the intestine. A
similar investigation was conducted later by Recker
(891) in a larger sample of achlorhydric patients. The
investigator concluded that 1) control subjects absorb
calcium carbonate and calcium citrate equally well,
2) but that achlorhydric patients lose their capability to
absorb calcium carbonate, while calcium citrate absorption is increased (presumably through compensatory upregulation of the absorption via vitamin D) (891). It is
important to mention that these absorption assays were
conducted in the fasting state. When the achlorhydric
patients ingested the calcium carbonate salt together with
a meal, their calcium uptake normalized. It cannot be
conclusively answered which factor of the meal was responsible for the increase, as several food components,
such as fiber and protein, are known to affect calcium
uptake. However, the authors speculated that the pH
(5.8) of the meal was sufficiently low to dissolve the
ingested calcium carbonate (891). Furthermore, a previously cited study that demonstrated decreased calcium
absorption under PPI therapy employed calcium carbonate as source of calcium for the conducted measurements
(807). Patients with gastric bypass surgery also absorb
calcium carbonate less effectively than calcium citrate
(1089). The importance of acid for the absorption of
calcium carbonate was also demonstrated in the previously discussed achlorhydric CCK2 (/) mouse model
(963). The osteoporotic phenotype and secondary hyperparathyroidism in these achlorhydric mice could only be
fully rescued by a high-calcium gluconate (2%) diet, but
not by a high-calcium carbonate (2%) diet (963). In the
light of these reports, it is evident that although the bioavailability of calcium salts in the healthy individual may
be equal, an impairment of acid secretion has a negative
effect on the bioavailability of calcium carbonate, presumably because of decreased solubility. Given the fact
that calcium carbonate is the most commonly used formulation for calcium substitution therapy, these findings
may partially account for the statistical correlation between PPI use and the increased risk of sustaining fractures.
Another factor that needs to be taken into consideration
when assessing solubility of calcium salts is the PCO2
(390). In the local milieu of the duodenum, the PCO2 can
reach values of up to 300 mmHg, resulting from the

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

235

Downloaded from on December 8, 2014

Calcium salts represent the most common supplementation


form of calcium for individuals who do not meet their adequate daily intake. The indications for supplementation
can be diverse, but mostly include conditions such as osteoporosis/-penia or preventative intake after menopause, during glucocorticoid intake or if lactose intolerant. In the year
2000, the National Health Interview Survey concluded that
11% of Americans ingest calcium supplements on a daily
basis (739). Females account for 80% of this population,
mostly to ensure supply after menopause (739). Calcium
salts exist in multiple formulations. The most commonly
used salts are calcium carbonate, calcium citrate, calcium
lactate, and calcium gluconate. Calcium carbonate is the
most widely used formulation, because it contains the highest percentage of elemental calcium per weight (40%),
which equates to small tablet size and easier ingestion (997).
In comparison, calcium citrate contains 21% elemental calcium, calcium lactate 14%, and calcium gluconate 9%
(997). However, the calcium salts do not only differ in their
calcium fraction, but are massively divergent with regard to
their solubility in water. Calcium carbonate is the least water-soluble salt at a neutral pH. For example, calcium citrate
dissolves 17 times more readily in water than calcium carbonate (997). Very fundamental in vitro solubility experiments have shown that after 1 h in 500 ml of water only 1%
of the initial 500 mg of calcium carbonate are dissolved at
37C (997). The solubility of calcium carbonate can be
greatly improved by an acidic environment (390, 997). Adjusting the pH to 5.5 in the same experiment dissolves 86%
of the calcium carbonate; further lowering it to 2.5, a value
that can be expected in the stomach, increases the dissolved
fraction to 100%. Given these differences in solubility, a
plethora of studies have investigated the bioavailability of
the various calcium salts, mostly focusing on the difference
between calcium carbonate and calcium citrate. Again, the
conclusions are heterogenic. Several studies suggest that
calcium carbonate is absorbed less effectively than the more
soluble calcium citrate (422, 438, 439, 789), while others
conclude that there is no difference in bioavailability (432,
433, 520, 547, 831, 832, 891, 997, 1038). A detailed analysis of the individual trials is beyond the scope of this review. It suffices to say that there is strong variability in the
experimental methods (direct absorption measurements vs.
measurement of postprandial serum calcium vs. urine excretion) and design of the studies (populations; administration in the fasting state vs. with a meal). A confounding
factor to the results of these studies may be the gastric pH at
the time of the measurement. As discussed earlier, calcium
carbonate is not very soluble at more alkali pH values,
which may have implications for patients using these supplements while on PPI therapy (997). Furthermore, meals
dramatically affect gastric pH and may change the bioavailability of the supplements. Indeed, there seems to be a correlation between gastric pH and the absorbability of calcium carbonate. The first observation indicative of this association was made by Ivanovich et al. in the late 1960s

SASCHA KOPIC AND JOHN P. GEIBEL


pancreatic secretion of bicarbonate (929). Solubility experiments of calcium carbonate have shown that a high
PCO2 negatively affects its solubility, as the carbonate
enters equilibrium with CO2 (390). Compared with other
calcium salts, the particularly low bioavailability of calcium carbonate in acid suppressed patients may thus be a
compounded effect of reduced acid secretion and a high
duodenal PCO2.

D. The Endocrine Stomach and


Calcium Homeostasis

1. Ghrelin
Ghrelin has been discovered fairly recently (1999) by Kojima and colleagues and is mainly implicated in regulating
food intake in the hypothalamus (588, 777). Ghrelin levels
are inversely correlated with body mass and elevated in
conditions of fasting, such as anorexia nervosa (377). Ghrelin is mainly synthesized and secreted in a pulsatile manner
by special neuroendocrine cells (P/D1 cells) in the fundic
region of the stomach (242, 777). The influence of ghrelin
on gastric acid secretion is discussed in a separate section
(see section IIB5E). A few years after its discovery, it was
shown that ghrelin can also directly affect osteoblasts (254,
357, 576, 691). Ghrelin induces osteoblast proliferation
and differentiation and inhibits their apoptosis (357, 576,
691, 1141). It is not entirely clear whether this effect is
mediated via the ghrelin surface receptor, the growth hormone secretagogue receptor 1a (GHS-R1a), or not. While
the receptor is expressed in rat and murine osteoblasts and
its pharmacological inhibition abolishes the effects of ghrelin on differentiation and proliferation, no GHS-R1a
mRNA could be detected in a human osteoblast cell line
(254, 357, 691). It should be noted that this effect is independent of growth hormone (GH). Ghrelin serves as a potent stimulator of GH secretion from the pituitary gland,
which in turn acts as an activator of osteoblasts through the
GH/IGF-I axis. However, the observations that 1) pharmacological inhibition of GHS-R1a attenuates the effects of
ghrelin and that 2) GH-deficient rats are still sensitive to
ghrelin, suggest a direct effect on osteoblasts (357). In vivo,
the activation of osteoblasts translates to an increase in

236

Several studies aimed to identify a link between serum ghrelin levels and BMD in human populations. The most recent,
and one of the largest (n 707 subjects), investigation
assessed BMD with peripheral quantitative computed tomography (pQCT). This technique allows for separate
analysis of trabecular and cortical bone. The results showed
a positive correlation between ghrelin and trabecular BMD
in elderly men and women (775). A different large-scale
study, investigating a similar cohort (n 977) found no
association using dual-energy X-ray and single-photon absorptiometry (1157). These techniques, however, do not
permit a discrimination between cortical and trabecular
bone. Other small-scale studies also came to contradictory
conclusions (388, 811). The reason for these discrepancies
is elusive. Since the formation of trabecular bone represents
a more dynamic process, its direct measurement may be
more sensitive to subtle changes than overall bone density
measurement (775). Baseline plasma ghrelin levels were
also shown to be inversely correlated to type 1 collagen
C-telopeptide (CTX), a marker for bone resorption (501).
The source of ghrelin represents another potential caveat. In
vitro studies suggest that osteoblasts can also synthesize
ghrelin (254, 357). Ghrelin was identified on the mRNA
and protein level by two investigations (254, 357). A different group did not find evidence for ghrelin in osteoblasts
(214). This has important implications, as ghrelin may be
secreted in an auto-/paracrine fashion, which would make
plasma ghrelin levels less significant for osteoblast activation. On the other hand, (partial) gastrectomy significantly
decreases plasma ghrelin concentrations, which could contribute to postgastrectomy osteopenia, although these may
just be two independent factors. Total gastrectomy causes a
drop in plasma ghrelin levels by as much as 70% (528).
Partial gastrectomy also severely decreases plasma ghrelin;
however, levels normalize depending on the type of resection to 48 88% of the preoperative levels due to compensatory production in the remaining gastric mucosa (528).
This recovery already occurs after 7days. It thus remains to
be elucidated if the slightly decreased ghrelin levels after
small gastric resections can account for the long-term phenomenon of postgastrectomy osteopenia. Furthermore, in
mice, the reduction of bone mass after gastrectomy cannot
be rescued by exogenous administration of ghrelin (277).
Ghrelin administration did also not affect markers of bone
resorption in gastrectomized patients, although these parameters were only measured very acutely 4 h after ghrelin
infusion (501). So far, no data on the effect of chronic
ghrelin treatment on BMD are available.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

Apart from being a mere acid secretory organ, the stomach


also plays an important role as an endocrine organ. It
should be noted that all of the aforementioned surgical or
pharmacological interventions, i.e., gastrectomies, vagotomies, or pharmacological acid suppression therapy, will inevitably impact the endocrine functions of the stomach. It is
therefore plausible that not only the changes in intragastric
pH affect the absorption of calcium, but that the dysregulation of the endocrine stomach is responsible for changes in
calcium homeostasis. The following section will address
how hormones that are secreted by the stomach may impact
calcium and bone homeostasis.

bone mineral density (BMD) in rat and murine models (261,


357). Ghrelin also promotes the formation of new bone
following injury (261). For example, mice that received a
standardized bone injury demonstrated 1.6 times more new
bone surface if treated with ghrelin compared with control
animals (261).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


Although decreased ghrelin levels could in theory be a contributing factor to the reduction of bone mass following
gastrectomy, it is unknown if PPIs can directly affect ghrelin
levels. A potential link between PPI-related fractures and
ghrelin levels remains to be investigated. An indirect association may be present in patients with Helicobacter pylori
infections. These infections represent a common indication
for PPI intake and were suggested to coincide with reduced
ghrelin in plasma and the gastric mucosa (527). Given the
recent discovery of ghrelins impact on osteoblast function,
many questions still remain to be answered. It is, however,
clear that our view of the stomach as a mere acid secretory
pouch needs to be expanded to a new level.
2. Gastrin

At least in the rat, the hypothesis of the gastrin-calcitonin


axis has been severely challenged. Although gastrin decreases plasma calcium levels in rats, the same effect occurs
in (para)thyroidectomized animals, suggesting that calcitonin is not involved in this process (980). Furthermore,
cultured rat thyroid cells could not be stimulated to release
calcitonin if incubated with gastrin (225). Fundectomy- and
omeprazole-induced hypergastrinemia also did not affect
calcitonin levels in rats (843, 927). Interestingly, hypocalcemia after gastrin injection could not be induced in rats
that had been (para)thyroidectomized and gastrectomized
(844, 981). This observation led to the conclusion that gastrin may stimulate the release of an unknown substance
from the rat stomach, which in turn exerts calcitropic activity. In accordance with this hypothesis, mucosal extracts
from rat stomachs were shown to have the same hypocalcemic effects as gastrin and to stimulate uptake of radiolabeled calcium into the bone (844). These findings were also

Whatever the exact effector hormone of gastrin may be,


changes in gastrin levels cannot entirely explain the clinical
phenomenon of postgastrectomy osteopenia and PPI-related fractures. Although vagotomy and PPIs undoubtedly
increase serum gastrin levels through a negative-feedback
mechanism, most partial and all total gastrectomies result
in hypogastrinemia. Yet both hyper- and hypogastrinemic
conditions have similar outcomes, i.e., osteopenia and increased risks of fractures. It is, of course, plausible that
different factors contribute to this outcome in each individual group. Gastrin may be involved in certain pathologies,
but given that its true impact on bone metabolism is somewhat elusive, this assumption remains speculative.

Acid suppression

Calcium solubility

Gastrin

Intestinal absorption

Pancreastatin

Calcitonin

PTH

FIGURE 9. Model summarizing the potential impact of acid suppression on calcium homeostasis.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

237

Downloaded from on December 8, 2014

Gastrin represents one of the main acid secretagogues (see


sect. IIB2). It is secreted by specialized G-cells in the antrum
of the stomach and the duodenum. The released gastrin
enters the circulation and induces acid secretion in gastric
parietal cells via the CCK2 receptor. It has been hypothesized fairly early that plasma gastrin may have an impact on
bone metabolism. Injection of gastrin and its synthetic analog pentagastrin was shown to decrease plasma calcium
levels in pigs and rats in the 1970s (222, 980). This effect
was attributed to gastrin-stimulated release of calcitonin
from the thyroid gland. Indeed, pentagastrin was shown to
be a potent stimulator of calcitonin secretion in various
species and is still in clinical use to evaluate thyroid C-cell
hyperplasia and medullary carcinomas (155, 156, 226, 441,
829). Although a clinical correlation between plasma gastrin levels and plasma calcitonin has been demonstrated by
one study in patients with Zollinger-Ellison syndrome (hypergastrinemic patients) and in pigs, it is unclear if native
gastrin, i.e., not pentagastrin, acts as an important secretagogue for calcitonin in humans (224, 1020). In fact, other
investigations found no association between gastrin and
calcitonin levels in other cohorts (122, 454).

replicated in chicken (842). The unknown hormone was


tentatively named gastrocalcin (844). When ghrelin was
discovered, it was speculated that it might represent a candidate hormone for gastrocalcin. However, unlike gastrocalcin, ghrelin is not under gastrin control, making this
proposition unlikely (276). Subsequent investigations suggested that the origin of gastrocalcin were the gastric ECL
cells. ECL extracts can indeed trigger a calcium second messenger response in osteoblast (629, 630). Yet, functional
evidence for gastrocalcin-mediated osteoblast activation is
still lacking. A recent report postulates that parathyroid
hormone-like hormone (PTHLH) may in fact be gastrocalcin (676). PTHLH exerts similar physiological effects as
PTH by sharing a common receptor and is commonly elevated in paraneoplastic syndromes (1056). PTHLH has
been identified in ECL cells, and its transcription has been
shown to be inducible by gastrin in parietal cells (523, 676).
Of note, PTH causes effects opposite to those assigned to
gastrin and gastrocalcin, namely, hypercalcemia. Further
studies will thus be needed to corroborate this hypothesis.

SASCHA KOPIC AND JOHN P. GEIBEL


3. Pancreastatin
Pancreastatin is a cleavage product of chromogranin A that
was initially isolated from porcine pancreas (1072). Gastric
ECL cells are also known to harbor significant amounts of
chromogranin A and pancreastatin. Pancreastatin is secreted together with histamine from ECL cells in response
to their neuroendocrine stimulation (see sect. IIB3) (176). In
rat, it has been shown that the serum pancreastatin levels
correlate with the secretory status of ECL cells. States that
enhance ECL cell secretion, such as gastrin infusion, resulted in elevated serum pancreastatin levels (411). In accordance with this hypothesis, and of special relevance for
the topic of this review, PPI therapy also resulted in increased serum pancreastatin levels (the ECL is stimulated by
gastrin, which in turn is released in response to high gastric
pH) (411). These observations led the investigators to conclude that ECL cells are a major contributor to the serum
levels of pancreastatin in the rat and that these levels change
in parallel with ECL cell secretion (411). This is also corroborated by the observation that gastrectomy reduces pancreastatin levels in rats (644).

review we have focused on the important role calcium plays


as a first and second messenger in the maintenance of bone
health. By relying on a complex series of receptors, channels, and transport proteins, calcium is tightly controlled at
the cellular and tissue level to ensure its bioavailability to
bone. Modulations to any of these pathways by disease,
mutation, or pharmaceutical perturbation can lead to clinical changes in bone health.

ACKNOWLEDGMENTS
S. Kopic is a Howard Hughes Medical Institute International Student Research Fellow. Special thanks to Sashka
Dimitrievska for her untiring support and critical editing of
the manuscript.
Address for reprint requests and other correspondence: J. P.
Geibel, Yale School of Medicine, 310 Cedar St., BML 238,
New Haven, CT 06510 (e-mail: John.geibel@yale.edu).

In summary, it should be noted that the stomach secretes


not only acid, but also hormones that have been shown to
directly alter calcium and/or bone homeostasis. The secretion of these hormones depends on the neuroendocrine machinery that also regulates acid secretion. It is therefore
plausible that the correlation between states of impaired
acid secretion and impaired bone mineralization is multifactorial by depending on intragastric pH and serum levels
of gastric hormones (FIGURE 9).

No conflicts of interest, financial or otherwise, are declared


by the authors.

REFERENCES
1. Abe K, Kaya S, Taniguchi K, Hayashi Y, Imagawa T, Kikumoto M, Oiwa K, Sakaguchi
K. Evidence for a relationship between activity and the tetraprotomeric assembly of
solubilized pig gastric H/K-ATPase. J Biochem 138: 293301, 2005.
2. Abe K, Tani K, Fujiyoshi Y. Conformational rearrangement of gastric H,K-ATPase
induced by an acid suppressant. Nat Commun 2: 155, 2011.
3. Abe K, Tani K, Fujiyoshi Y. Structural and functional characterization of H,KATPase with bound fluorinated phosphate analogs. J Struct Biol 170: 60 68, 2010.
4. Abe K, Tani K, Nishizawa T, Fujiyoshi Y. Inter-subunit interaction of gastric H,KATPase prevents reverse reaction of the transport cycle. EMBO J 28: 16371643,
2009.
5. Abou-Samra AB, Juppner H, Force T, Freeman MW, Kong XF, Schipani E, Urena P,
Richards J, Bonventre JV, Potts JT Jr. Expression cloning of a common receptor for
parathyroid hormone and parathyroid hormone-related peptide from rat osteoblast-like cells: a single receptor stimulates intracellular accumulation of both cAMP
and inositol trisphosphates and increases intracellular free calcium. Proc Natl Acad Sci
USA 89: 27322736, 1992.
6. Abreo K, Adlakha A, Kilpatrick S, Flanagan R, Webb R, Shakamuri S. The milk-alkali
syndrome. A reversible form of acute renal failure. Arch Intern Med 153: 10051010,
1993.
7. Adams JS, Clemens TL, Parrish JA, Holick MF. Vitamin-D synthesis and metabolism
after ultraviolet irradiation of normal and vitamin-D-deficient subjects. N Engl J Med
306: 722725, 1982.

VI. CONCLUSIONS

8. Agnew JE, Holdsworth CD. The effect of fat on calcium absorption from a mixed
meal in normal subjects, patients with malabsorptive disease, and patients with a
partial gastrectomy. Gut 12: 973977, 1971.

We set out in this review to demonstrate that gastric and


intestinal physiology are intertwined to regulate calcium
absorption and secretion to maintain bone health. In this

9. Aihara T, Fujishita T, Kanatani K, Furutani K, Nakamura E, Taketo MM, Matsui M,


Chen D, Okabe S. Impaired gastric secretion and lack of trophic responses to
hypergastrinemia in M3 muscarinic receptor knockout mice. Gastroenterology 125:
1774 1784, 2003.

238

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

DISCLOSURES
Pancreastatin exerts a variety of metabolic effects. Apart
from influencing energy metabolism, pancreasstatin was
shown to affect the secretion of PTH from the parathyroid
gland. In isolated bovine and porcine parathyroid cells,
pancreastatin has a clear inhibitory effect on PTH secretion
(282, 317, 911). The suppression of PTH functions on a
transcriptional level (1211). Reduced PTH secretion in turn
would have a potential impact on calcium and bone metabolism. Whether the same observations are valid for humans
is less clear, as pancreastatin failed to inhibit PTH secretion
from isolated human parathyroid cells (911). Regardless,
the volume of data on pancreastatin and its influence on the
parathyroid gland is very small, and further investigations
would be necessary to establish this intriguing link. Apart
from potential modulation of parathyroid secretion, pancreastatin has also been shown to have an inhibitory effect
on gastric acid secretion (655).

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


10. Aihara T, Nakamura Y, Taketo MM, Matsui M, Okabe S. Cholinergically stimulated
gastric acid secretion is mediated by M3 and M5 but not M1 muscarinic acetylcholine
receptors in mice. Am J Physiol Gastrointest Liver Physiol 288: G1199 G1207, 2005.

30. Anusaksathien O, Laplace C, Li X, Ren Y, Peng L, Goldring SR, Galson DL. Tissuespecific and ubiquitous promoters direct the expression of alternatively spliced transcripts from the calcitonin receptor gene. J Biol Chem 276: 2266322674, 2001.

11. Akeno N, Saikatsu S, Kawane T, Horiuchi N. Mouse vitamin D-24-hydroxylase:


molecular cloning, tissue distribution, transcriptional regulation by 1alpha,25-dihydroxyvitamin D3. Endocrinology 138: 22332240, 1997.

31. Ardaillou R. Kidney and calcitonin. Nephron 15: 250 260, 1975.

12. Akey JM, Swanson WJ, Madeoy J, Eberle M, Shriver MD. TRPV6 exhibits unusual
patterns of polymorphism and divergence in worldwide populations. Hum Mol Genet
15: 2106 2113, 2006.
13. Akhter S, Kutuzova GD, Christakos S, DeLuca HF. Calbindin D9k is not required for
1,25-dihydroxyvitamin D3-mediated Ca2 absorption in small intestine. Arc Biochem
Biophys 460: 227232, 2007.
14. Akiyoshi-Shibata M, Sakaki T, Ohyama Y, Noshiro M, Okuda K, Yabusaki Y. Further
oxidation of hydroxycalcidiol by calcidiol 24-hydroxylase. A study with the mature
enzyme expressed in Escherichia coli. Eur J Biochem 224: 335343, 1994.
15. Al-Ansary D, Bogeski I, Disteldorf BMJ, Becherer U, Niemeyer BA. ATP modulates
Ca2 uptake by TRPV6 and is counteracted by isoform-specific phosphorylation.
FASEB J 24: 425 435, 2010.
16. Alam AS, Moonga BS, Bevis PJ, Huang CL, Zaidi M. Amylin inhibits bone resorption
by a direct effect on the motility of rat osteoclasts. Exp Physiol 78: 183196, 1993.

32. Ardaillou R, Vuagnat P, Milhaud G, Richet G. Effects of thyrocalcitonin on the renal


excretion of phosphates, calcium and hydrogen ions in man. Nephron 4: 298 314,
1967.
33. Aris RM, Lester GE, Dingman S, Ontjes DA. Altered calcium homeostasis in adults
with cystic fibrosis. Osteoporos Int 10: 102108, 1999.
34. Arman E, Nilsson LH, Reizenstein P. Studies in the dumping syndrome. VI. Calcium
deficiency after partial gastrectomy. Am J Dig Dis 15: 455 462, 1970.
35. Armbrecht HJ, Boltz MA, Kumar VB. Intestinal plasma membrane calcium pump
protein and its induction by 1,25(OH)2D3 decrease with age. Am J Physiol Gastrointest Liver Physiol 277: G41G47, 1999.
36. Armbrecht HJ, Boltz MA, Wongsurawat N. Expression of plasma membrane calcium
pump mRNA in rat intestine: effect of age and 1,25-dihydroxyvitamin D. Biochim
Biophys Acta 1195: 110 114, 1994.
37. Armbrecht HJ, Hodam TL, Boltz MA, Partridge NC, Brown AJ, Kumar VB. Induction
of the vitamin D 24-hydroxylase (CYP24) by 1,25-dihydroxyvitamin D3 is regulated
by parathyroid hormone in UMR106 osteoblastic cells. Endocrinology 139: 3375
3381, 1998.
38. Arnold A, Horst SA, Gardella TJ, Baba H, Levine MA, Kronenberg HM. Mutation of
the signal peptide-encoding region of the preproparathyroid hormone gene in familial isolated hypoparathyroidism. J Clin Invest 86: 1084 1087, 1990.

18. Allen LH. Calcium bioavailability and absorption: a review. Am J Clin Nutr 35: 783
808, 1982.

39. Arnold R, Koop H, Schwarting H, Tuch K, Willemer B. Effect of acid inhibition on


gastric endocrine cells. Scand J Gastroenterol Suppl 125: 14 19, 1986.

19. Alumets J, Ekelund M, El Munshid HA, Hakanson R, Loren I, Sundler F. Topography


of somatostatin cells in the stomach of the rat: possible functional significance. Cell
Tissue Res 202: 177188, 1979.

40. Arthur JM, Collinsworth GP, Gettys TW, Quarles LD, Raymond JR. Specific coupling
of a cation-sensing receptor to G protein -subunits in MDCK cells. Am J Physiol
Renal Physiol 273: F129 F135, 1997.

20. Aly A, Shulkes A, Baldwin GS. Short term infusion of glycine-extended gastrin(17)
stimulates both proliferation and formation of aberrant crypt foci in rat colonic
mucosa. Int J Cancer 94: 307313, 2001.

41. Asano S, Kawada K, Kimura T, Grishin AV, Caplan MJ, Takeguchi N. The roles of
carbohydrate chains of the beta-subunit on the functional expression of gastric
H,K-ATPase. J Biol Chem 275: 8324 8330, 2000.

21. Amling M, Priemel M, Holzmann T, Chapin K, Rueger JM, Baron R, Demay MB.
Rescue of the skeletal phenotype of vitamin D receptor-ablated mice in the setting of
normal mineral ion homeostasis: formal histomorphometric and biomechanical analyses. Endocrinology 140: 4982 4987, 1999.

42. Asano S, Yoshida A, Yashiro H, Kobayashi Y, Morisato A, Ogawa H, Takeguchi N,


Morii M. The cavity structure for docking the K-competitive inhibitors in the gastric
proton pump. J Biol Chem 279: 13968 13975, 2004.

22. Andersen BN. Species variation in the tyrosine sulfation of mammalian gastrins. Gen
Comp Endocrinol 58: 44 50, 1985.
23. Anderson NG, Hanson PJ. Involvement of calcium-sensitive phospholipid-dependent protein kinase in control of acid secretion by isolated rat parietal cells. Biochem
J 232: 609 611, 1985.
24. Andersson K, Cabero JL, Mattsson H, Hakanson R. Gastric acid secretion after
depletion of enterochromaffin-like cell histamine. A study with alpha-fluoromethylhistidine in rats. Scand J Gastroenterol 31: 24 30, 1996.
25. Andersson S, Chang D, Folkers K, Rosell S. Inhibition of gastric acid secretion in dogs
by neurotensin. Life Sci 19: 367370, 1976.
26. Andersson S, Davis DL, Dahlback H, Jornvall H, Russell DW. Cloning, structure, and
expression of the mitochondrial cytochrome P-450 sterol 26-hydroxylase, a bile acid
biosynthetic enzyme. J Biol Chem 264: 8222 8229, 1989.
27. Andreassen TT, Ejersted C, Oxlund H. Intermittent parathyroid hormone (134)
treatment increases callus formation and mechanical strength of healing rat fractures. J Bone Miner Res 14: 960 968, 1999.
28. Andreotti G, Mendez BL, Amodeo P, Morelli MA, Nakamuta H, Motta A. Structural
determinants of salmon calcitonin bioactivity: the role of the Leu-based amphipathic
alpha-helix. J Biol Chem 281: 2419324203, 2006.
29. Anisuzzaman AS, Morishima S, Suzuki F, Tanaka T, Muramatsu I. Identification of
M(1) muscarinic receptor subtype in rat stomach using a tissue segment binding
method, the effects of immobilization stress on the muscarinic receptors. Eur J
Pharmacol 599: 146 151, 2008.

43. Askew FA, Bruce HM, Callow RK, Philpot JS, Webster TA. Crystalline vitamin D.
Nature 128: 758, 1931.
44. Athmann C, Zeng N, Scott DR, Sachs G. Regulation of parietal cell calcium signaling
in gastric glands. Am J Physiol Gastrointest Liver Physiol 279: G1048 G1058, 2000.
45. Atkins GJ, Anderson PH, Findlay DM, Welldon KJ, Vincent C, Zannettino AC,
OLoughlin PD, Morris HA. Metabolism of vitamin D3 in human osteoblasts: evidence for autocrine and paracrine activities of 1 alpha,25-dihydroxyvitamin D3. Bone
40: 15171528, 2007.
46. Auchere D, Tardivel S, Gounelle JC, Drueke T, Lacour B. Role of transcellular
pathway in ileal Ca2 absorption: stimulation by low-Ca2 diet. Am J Physiol Gastrointest Liver Physiol 275: G951G956, 1998.
47. Auer J, Reeh PW, Fischer MJ. Acid-induced CGRP release from the stomach does not
depend on TRPV1 or ASIC3. Neurogastroenterol Motil 22: 680 687, 2010.
48. Awata H, Huang C, Handlogten ME, Miller RT. Interaction of the calcium-sensing
receptor and filamin, a potential scaffolding protein. J Biol Chem 276: 3487134879,
2001.
49. Axelson J, Persson P, Gagnemo-Persson R, Hakanson R. Importance of the stomach
in maintaining calcium homoeostasis in the rat. Gut 32: 1298 1302, 1991.
50. Axen E, Postlind H, Sjoberg H, Wikvall K. Liver mitochondrial cytochrome P450
CYP27 and recombinant-expressed human CYP27 catalyze 1-hydroxylation of
25-hydroxyvitamin D3. Proc Natl Acad Sci USA 91: 10014 10018, 1994.
51. Axen E, Postlind H, Wikvall K. Effects on CYP27 mRNA expression in rat kidney and
liver by 1,25-dihydroxyvitamin D3, a suppressor of renal 25-hydroxyvitamin D3
1-hydroxylase activity. Biochem Biophys Res Commun 215: 136 141, 1995.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

239

Downloaded from on December 8, 2014

17. Albrandt K, Brady EM, Moore CX, Mull E, Sierzega ME, Beaumont K. Molecular
cloning and functional expression of a third isoform of the human calcitonin receptor
and partial characterization of the calcitonin receptor gene. Endocrinology 136:
53775384, 1995.

SASCHA KOPIC AND JOHN P. GEIBEL


52. Ba J, Brown D, Friedman PA. Calcium-sensing receptor regulation of PTH-inhibitable proximal tubule phosphate transport. Am J Physiol Renal Physiol 285: F1233
F1243, 2003.

74. Bell NJ, Burget D, Howden CW, Wilkinson J, Hunt RH. Appropriate acid suppression
for the management of gastro-oesophageal reflux disease. Digestion 51 Suppl 1:
59 67, 1992.

53. Bado A, Cloarec D, Moizo L, Laigneau JP, Bataille D, Lewin MJ. Neurotensin and
oxyntomodulin-(30 37) potentiate PYY regulation of gastric acid and somatostatin
secretions. Am J Physiol Gastrointest Liver Physiol 265: G113G117, 1993.

75. Bellido T, Ali AA, Plotkin LI, Fu Q, Gubrij I, Roberson PK, Weinstein RS, OBrien CA,
Manolagas SC, Jilka RL. Proteasomal degradation of Runx2 shortens parathyroid
hormone-induced anti-apoptotic signaling in osteoblasts. A putative explanation for
why intermittent administration is needed for bone anabolism. J Biol Chem 278:
50259 50272, 2003.

54. Bai M, Trivedi S, Brown EM. Dimerization of the extracellular calcium-sensing receptor (CaR) on the cell surface of CaR-transfected HEK293 cells. J Biol Chem 273:
2360523610, 1998.
55. Bai M, Trivedi S, Kifor O, Quinn SJ, Brown EM. Intermolecular interactions between
dimeric calcium-sensing receptor monomers are important for its normal function.
Proc Natl Acad Sci USA 96: 2834 2839, 1999.
56. Bai M, Trivedi S, Lane CR, Yang Y, Quinn SJ, Brown EM. Protein kinase C phosphorylation of threonine at position 888 in Ca2o-sensing receptor (CaR) inhibits coupling to Ca2 store release. J Biol Chem 273: 2126721275, 1998.
57. Bailey RL, Dodd KW, Goldman JA, Gahche JJ, Dwyer JT, Moshfegh AJ, Sempos CT,
Picciano MF. Estimation of total usual calcium and vitamin D intakes in the United
States. J Nutr 140: 817 822, 2010.
58. Baird IM, Oleesky S. Osteomalacia following gastric surgery. Gastroenterology 33:
284 292, 1957.
59. Baker AR, McDonnell DP, Hughes M, Crisp TM, Mangelsdorf DJ, Haussler MR, Pike
JW, Shine J, OMalley BW. Cloning and expression of full-length cDNA encoding
human vitamin D receptor. Proc Natl Acad Sci USA 85: 3294 3298, 1988.

61. Baldissera FG, Nielsen OV, Holst JJ. The intestinal mucosa preferentially releases
somatostatin-28 in pigs. Regul Pept 11: 251262, 1985.
62. Balesaria S, Sangha S, Walters JR. Human duodenum responses to vitamin D metabolites of TRPV6 and other genes involved in calcium absorption. Am J Physiol Gastrointest Liver Physiol 297: G1193G1197, 2009.
63. Barger G, Dale HH. The presence in ergot and physiological activity of beta-imidazolylethylamine. J Physiol 40: xxxviii-xl, 1910.

77. Benoit R, Ling N, Esch F. A new prosomatostatin-derived peptide reveals a pattern


for prohormone cleavage at monobasic sites. Science 238: 1126 1129, 1987.
78. Beresford JN, Bennett JH, Devlin C, Leboy PS, Owen ME. Evidence for an inverse
relationship between the differentiation of adipocytic and osteogenic cells in rat
marrow stromal cell cultures. J Cell Sci 102: 341351, 1992.
79. Beresford JN, Joyner CJ, Devlin C, Triffitt JT. The effects of dexamethasone and
1,25-dihydroxyvitamin D3 on osteogenic differentiation of human marrow stromal
cells in vitro. Arch Oral Biol 39: 941947, 1994.
80. Berg A, Kechagias S, Sjostrand SE, Ericson AC. Morphological support for paracrine
inhibition of gastric acid secretion by nitric oxide in humans. Scand J Gastroenterol 36:
1016 1021, 2001.
81. Berg A, Redeen S, Ericson AC, Sjostrand SE. Nitric oxide-an endogenous inhibitor of gastric acid secretion in isolated human gastric glands. BMC Gastroenterol
4: 16, 2004.
82. Berg A, Redeen S, Grenegard M, Ericson AC, Sjostrand SE. Nitric oxide inhibits
gastric acid secretion by increasing intraparietal cell levels of cGMP in isolated human
gastric glands. Am J Physiol Gastrointest Liver Physiol 289: G1061G1066, 2005.
83. Berginer VM, Shany S, Alkalay D, Berginer J, Dekel S, Salen G, Tint GS, Gazit D.
Osteoporosis and increased bone fractures in cerebrotendinous xanthomatosis.
Metabolism 42: 69 74, 1993.
84. Berndt TJ, Knox FG. Effects of parathyroid hormone and calcitonin on electrolyte
excretion in the rabbit. Kidney Int 17: 473 478, 1980.

64. Barley NF, Howard A, OCallaghan D, Legon S, Walters JR. Epithelial calcium transporter expression in human duodenum. Am J Physiol Gastrointest Liver Physiol 280:
G285G290, 2001.

85. Bers DM, Ginsburg KS. Na:Ca stoichiometry and cytosolic Ca-dependent activation
of NCX in intact cardiomyocytes. Ann NY Acad Sci 1099: 326 338, 2007.

65. Basso D, Scrigner M, Toma A, Navaglia F, Di Mario F, Rugge M, Plebani M. Helicobacter pylori infection enhances mucosal interleukin-1 beta, interleukin-6, the soluble
receptor of interleukin-2. Int J Clin Lab Res 26: 207210, 1996.

86. Berthoud HR. Morphological analysis of vagal input to gastrin releasing peptide and
vasoactive intestinal peptide containing neurons in the rat glandular stomach. J Comp
Neurol 370: 6170, 1996.

66. Bataille D, Blache P, Mercier F, Jarrousse C, Kervran A, Dufour M, Mangeat P,


Dubrasquet M, Mallat A, Lotersztajn S. Glucagon and related peptides Molecular
structure and biological specificity. Ann NY Acad Sci 527: 168 185, 1988.

87. Bertrand C, Kowalski-Chauvel A, Do C, Resa C, Najib S, Daulhac L, Wang TC,


Ferrand A, Seva C. A gastrin precursor, gastrin-gly, upregulates VEGF expression in
colonic epithelial cells through an HIF-1-independent mechanism. Int J Cancer 126:
28472857, 2010.

67. Batzri S, Gardner JD. Cellular cyclic AMP in dispersed mucosal cells from guinea pig
stomach. Biochim Biophys Acta 541: 181189, 1978.
68. Bayliss WM, Starling EH. The mechanism of pancreatic secretion. J Physiol 28: 325
353, 1902.
69. Beales IL, Calam J. Inhibition of carbachol stimulated acid secretion by interleukin
1beta in rabbit parietal cells requires protein kinase C. Gut 48: 782789, 2001.
70. Beales IL, Calam J. Interleukin 1 beta and tumour necrosis factor alpha inhibit acid
secretion in cultured rabbit parietal cells by multiple pathways. Gut 42: 227234,
1998.
71. Beales IL, Ogunwobi OO. Glycine-extended gastrin inhibits apoptosis in Barretts
oesophageal and oesophageal adenocarcinoma cells through JAK2/STAT3 activation. J Mol Endocrinol 42: 305318, 2009.

88. Bertrand CA, Zhang R, Pilewski JM, Frizzell RA. SLC26A9 is a constitutively active,
CFTR-regulated anion conductance in human bronchial epithelia. J Gen Physiol 133:
421 438, 2009.
89. Besancon M, Shin JM, Mercier F, Munson K, Miller M, Hersey S, Sachs G. Membrane
topology and omeprazole labeling of the gastric H,K-adenosinetriphosphatase.
Biochemistry 32: 23452355, 1993.
90. Besancon M, Simon A, Sachs G, Shin JM. Sites of reaction of the gastric H-KATPase with extracytoplasmic thiol reagents. J Biol Chem 272: 22438 22446, 1997.
91. Bhattacharyya MH, DeLuca HF. The regulation of calciferol-25-hydroxylase in the
chick. Biochem Biophys Res Commun 59: 734 741, 1974.
92. Bhattacharyya MH, DeLuca HF. The regulation of rat liver calciferol-25-hydroxylase.
J Biol Chem 248: 2969 2973, 1973.

72. Beck L, Karaplis AC, Amizuka N, Hewson AS, Ozawa H, Tenenhouse HS. Targeted
inactivation of Npt2 in mice leads to severe renal phosphate wasting, hypercalciuria,
and skeletal abnormalities. Proc Natl Acad Sci USA 95: 53725377, 1998.

93. Bhattacharyya MH, DeLuca HF. Subcellular location of rat liver calciferol-25-hydroxylase. Arch Biochem Biophys 160: 58 62, 1974.

73. Beil W, Mannschedel W, Sewing KF. Protein kinase C and parietal cell function.
Biochem Biophys Res Commun 149: 720 728, 1987.

94. Bianco SD, Peng JB, Takanaga H, Suzuki Y, Crescenzi A, Kos CH, Zhuang L, Freeman
MR, Gouveia CH, Wu J, Luo H, Mauro T, Brown EM, Hediger MA. Marked distur-

240

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

60. Bakke I, Qvigstad G, Brenna E, Sandvik AK, Waldum HL. Gastrin has a specific
proliferative effect on the rat enterochromaffin-like cell, but not on the parietal cell:
a study by elutriation centrifugation. Acta Physiol Scand 169: 29 37, 2000.

76. Benn BS, Ajibade D, Porta A, Dhawan P, Hediger M, Peng JB, Jiang Y, Oh GT, Jeung
EB, Lieben L, Bouillon R, Carmeliet G, Christakos S. Active intestinal calcium transport in the absence of transient receptor potential vanilloid type 6 and calbindinD9k. Endocrinology 149: 3196 3205, 2008.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


bance of calcium homeostasis in mice with targeted disruption of the Trpv6 calcium
channel gene. J Bone Miner Res 22: 274 285, 2007.
95. Bieglmayer C, Prager G, Niederle B. Kinetic analyses of parathyroid hormone clearance as measured by three rapid immunoassays during parathyroidectomy. Clin
Chem 48: 17311738, 2002.
96. Bielefeldt K, Davis BM. Differential effects of ASIC3 and TRPV1 deletion on gastroesophageal sensation in mice. Am J Physiol Gastrointest Liver Physiol 294: G130 G138,
2008.
97. Bikle DD, Gee E, Halloran B, Haddad JG. Free 1,25-dihydroxyvitamin D levels in
serum from normal subjects, pregnant subjects, and subjects with liver disease. J Clin
Invest 74: 1966 1971, 1984.
98. Bikle DD, Gee E, Halloran B, Kowalski MA, Ryzen E, Haddad JG. Assessment of the
free fraction of 25-hydroxyvitamin D in serum and its regulation by albumin and the
vitamin D-binding protein. J Clin Endocrinol Metab 63: 954 959, 1986.
99. Bindels RJ, Hartog A, Timmermans J, Van Os CH. Active Ca2 transport in primary
cultures of rabbit kidney CCD: stimulation by 1,25-dihydroxyvitamin D3 and PTH.
Am J Physiol Renal Fluid Electrolyte Physiol 261: F799 F807, 1991.
100. Birge SJ, Gilbert HR. Indentification of an intestinal sodium and calcium-dependent
phosphatase stimulated by parathyroid hormone. J Clin Invest 54: 710 717, 1974.
101. Bisballe S, Eriksen EF, Melsen F, Mosekilde L, Sorensen OH, Hessov I. Osteopenia
and osteomalacia after gastrectomy: interrelations between biochemical markers of
bone remodelling, vitamin D metabolites, and bone histomorphometry. Gut 32:
13031307, 1991.

103. Bjorkhem I, Holmberg I. Assay and properties of a mitochondrial 25-hydroxylase


active on vitamine D3. J Biol Chem 253: 842 849, 1978.
104. Bjorkhem I, Holmberg I, Oftebro H, Pedersen JI. Properties of a reconstituted
vitamin D3 25-hydroxylase from rat liver mitochondria. J Biol Chem 255: 5244 5249,
1980.
105. Bjorkqvist M, Bernsand M, Eliasson L, Hakanson R, Lindstrom E. Somatostatin,
misoprostol and galanin inhibit gastrin- and PACAP-stimulated secretion of histamine and pancreastatin from ECL cells by blocking specific Ca2 channels. Regul Pept
130: 8190, 2005.
106. Black JW, Duncan WA, Durant CJ, Ganellin CR, Parsons EM. Definition and antagonism of histamine H2-receptors. Nature 236: 385390, 1972.
107. Black JW, Fisher EW, Smith AN. The effects of 5-hydroxytryptamine on gastric
secretion in anaesthetized dogs. J Physiol 141: 2734, 1958.
108. Blackburn AM, Fletcher DR, Bloom SR, Christofides ND, Long RG, Fitzpatrick ML,
Baron JH. Effect of neurotensin on gastric function in man. Lancet 1: 987989, 1980.
109. Bland R, Walker EA, Hughes SV, Stewart PM, Hewison M. Constitutive expression of
25-hydroxyvitamin D3-1alpha-hydroxylase in a transformed human proximal tubule
cell line: evidence for direct regulation of vitamin D metabolism by calcium. Endocrinology 140: 20272034, 1999.
110. Blichert-Toft M, Beck A, Christiansen C, Transbol I. Effects of gastric resection and
vagotomy on blood and bone mineral content. World J Surg 3: 99 102, 133105,
1979.
111. Blunt JW, DeLuca HF. The synthesis of 25-hydroxycholecalciferol. A biologically
active metabolite of vitamin D3. Biochemistry 8: 671 675, 1969.

116. Booth BE, Tsai HC, Morris RC Jr. Vitamin D status regulates 25-hydroxyvitamin
D3-1 alpha-hydroxylase and its responsiveness to parathyroid hormone in the chick.
J Clin Invest 75: 155161, 1985.
117. Borthwick LA, Neal A, Hobson L, Gerke V, Robson L, Muimo R. The annexin
2S100A10 complex and its association with TRPV6 is regulated by cAMP/PKA/CnA
in airway and gut epithelia. Cell Calcium 44: 147157, 2008.
118. Bosl MR, Stein V, Hubner C, Zdebik AA, Jordt SE, Mukhopadhyay AK, Davidoff MS,
Holstein AF, Jentsch TJ. Male germ cells and photoreceptors, both dependent on
close cell-cell interactions, degenerate upon ClC-2 Cl channel disruption. EMBO J
20: 1289 1299, 2001.
119. Bouley R, Lu HA, Nunes P, Da Silva N, McLaughlin M, Chen Y, Brown D. Calcitonin
has a vasopressin-like effect on aquaporin-2 trafficking and urinary concentration. J
Am Soc Nephrol 22: 59 72, 2011.
120. Bourdeau A, Moutahir M, Souberbielle JC, Bonnet P, Herviaux P, Sachs C, Lieberherr M. Effects of lipoxygenase products of arachidonate metabolism on parathyroid
hormone secretion. Endocrinology 135: 1109 1112, 1994.
121. Bourdeau A, Souberbielle JC, Bonnet P, Herviaux P, Sachs C, Lieberherr M. Phospholipase-A2 action and arachidonic acid metabolism in calcium-mediated parathyroid hormone secretion. Endocrinology 130: 1339 1344, 1992.
122. Brandsborg M, Nielsen HE, Brandsborg O, Olsen KJ, Lovgreen NA. The role of
serum gastrin in the secretion of calcitonin: studies in patients with pernicious anaemia and in healthy subjects. Scand J Gastroenterol 15: 2328, 1980.
123. Brauer CA, Coca-Perraillon M, Cutler DM, Rosen AB. Incidence and mortality of hip
fractures in the United States. JAMA 302: 15731579, 2009.
124. Brazeau P, Vale W, Burgus R, Ling N, Butcher M, Rivier J, Guillemin R. Hypothalamic
polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science 179: 7779, 1973.
125. Brenza HL, DeLuca HF. Regulation of 25-hydroxyvitamin D3 1alpha-hydroxylase
gene expression by parathyroid hormone and 1,25-dihydroxyvitamin D3. Arch
Biochem Biophys 381: 143152, 2000.
126. Brewer HB Jr, Ronan R. Bovine parathyroid hormone: amino acid sequence. Proc
Natl Acad Sci USA 67: 18621869, 1970.
127. Bringhurst FR, Stern AM, Yotts M, Mizrahi N, Segre GV, Potts JT Jr. Peripheral
metabolism of PTH: fate of biologically active amino terminus in vivo. Am J Physiol
Endocrinol Metab 255: E886 E893, 1988.
128. Brommage R. Measurement of calcium and phosphorus fluxes during lactation in the
rat. J Nutr 119: 428 438, 1989.
129. Bronner F, Pansu D, Stein WD. An analysis of intestinal calcium transport across the
rat intestine. Am J Physiol Gastrointest Liver Physiol 250: G561G569, 1986.
130. Brossard JH, Cloutier M, Roy L, Lepage R, Gascon-Barre M, DAmour P. Accumulation of a non-(1 84) molecular form of parathyroid hormone (PTH) detected by
intact PTH assay in renal failure: importance in the interpretation of PTH values. J
Clin Endocrinol Metab 81: 39233929, 1996.
131. Brossard JH, Whittom S, Lepage R, DAmour P. Carboxyl-terminal fragments of
parathyroid hormone are not secreted preferentially in primary hyperparathyroidism as they are in other hypercalcemic conditions. J Clin Endocrinol Metab 77: 413
419, 1993.

112. Blunt JW, DeLuca HF, Schnoes HK. 25-Hydroxycholecalciferol. A biologically active
metabolite of vitamin D3. Biochemistry 7: 33173322, 1968.

132. Brown AJ, Krits I, Armbrecht HJ. Effect of age, vitamin D, and calcium on the
regulation of rat intestinal epithelial calcium channels. Arch Biochem Biophys 437:
5158, 2005.

113. Boel E, Vuust J, Norris F, Norris K, Wind A, Rehfeld JF, Marcker KA. Molecular
cloning of human gastrin cDNA: evidence for evolution of gastrin by gene duplication. Proc Natl Acad Sci USA 80: 2866 2869, 1983.

133. Brown E, Enyedi P, LeBoff M, Rotberg J, Preston J, Chen C. High extracellular Ca2
and Mg2 stimulate accumulation of inositol phosphates in bovine parathyroid cells.
FEBS Lett 218: 113118, 1987.

114. Bohmer C, Palmada M, Kenngott C, Lindner R, Klaus F, Laufer J, Lang F. Regulation


of the epithelial calcium channel TRPV6 by the serum and glucocorticoid-inducible
kinase isoforms SGK1 and SGK3. FEBS Lett 581: 5586 5590, 2007.

134. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger
MA, Lytton J, Hebert SC. Cloning and characterization of an extracellular Ca2sensing receptor from bovine parathyroid. Nature 366: 575580, 1993.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

241

Downloaded from on December 8, 2014

102. Bjorkhem I, Hansson R, Holmberg I, Wikvall K. 25-Hydroxylation of vitamin D3 by a


reconstituted system from rat liver microsomes. Biochem Biophys Res Commun 90:
615 622, 1979.

115. Boivin G, Mesguich P, Pike JW, Bouillon R, Meunier PJ, Haussler MR, Dubois PM,
Morel G. Ultrastructural immunocytochemical localization of endogenous 1,25-dihydroxyvitamin D3 and its receptors in osteoblasts and osteocytes from neonatal
mouse and rat calvaria. Bone Miner 3: 125136, 1987.

SASCHA KOPIC AND JOHN P. GEIBEL


135. Brown J 3rd, Theisler C, Silberman S, Magnuson D, Gottardi-Littell N, Lee JM, Yager
D, Crowley J, Sambamurti K, Rahman MM, Reiss AB, Eckman CB, Wolozin B.
Differential expression of cholesterol hydroxylases in Alzheimers disease. J Biol
Chem 279: 34674 34681, 2004.
136. Brown MR, Chew CS. Multiple effects of phorbol ester on secretory activity in rabbit
gastric glands and parietal cells. Can J Physiol Pharmacol 65: 1840 1847, 1987.
137. Bruley des Varannes S, Levy P, Lartigue S, Dellatolas F, Lemaire M, Galmiche JP.
Comparison of lansoprazole with omeprazole on 24-hour intragastric pH, acid secretion and serum gastrin in healthy volunteers. Aliment Pharmacol Ther 8: 309 314,
1994.
138. Bruns ME, Fleisher EB, Avioli LV. Control of vitamin D-dependent calcium-binding
protein in rat intestine by growth and fasting. J Biol Chem 252: 4145 4150, 1977.

156. Care AD, Bruce JB, Boelkins J, Kenny AD, Conaway H, Anast CS. Role of pancreozymin-cholecystokinin and structurally related compounds as calcitonin secretogogues. Endocrinology 89: 262271, 1971.
157. Carles-Bonnet C, Jarrousse C, Niel H, Martinez J, Bataille D. Oxyntomodulin and its
(19 37) and (30 37) fragments inhibit histamine-stimulated gastric acid secretion in
the conscious rat. Eur J Pharmacol 203: 245252, 1991.
158. Carney S, Thompson L. Acute effect of calcitonin on rat renal electrolyte transport.
Am J Physiol Renal Fluid Electrolyte Physiol 240: F12F16, 1981.
159. Carney SL. Acute effect of endogenous calcitonin on rat renal function. Miner Electrolyte Metab 21: 411 416, 1995.
160. Carney SL. Calcitonin and human renal calcium and electrolyte transport. Miner
Electrolyte Metab 23: 43 47, 1997.
161. Carraway R, Leeman SE. The isolation of a new hypotensive peptide, neurotensin,
from bovine hypothalami. J Biol Chem 248: 6854 6861, 1973.

140. Buchan AM, MacLeod MD, Meloche RM, Kwok YN. Muscarinic regulation of somatostatin release from primary cultures of human antral epithelial cells. Pharmacology
44: 33 40, 1992.

162. Carrillo-Lopez N, Alvarez-Hernandez D, Gonzalez-Suarez I, Roman-Garcia P, Valdivielso JM, Fernandez-Martin JL, Cannata-Andia JB. Simultaneous changes in the
calcium-sensing receptor and the vitamin D receptor under the influence of calcium
and calcitriol. Nephrol Dial Transplant 23: 3479 3484, 2008.

141. Buchan AM, Meloche RM, Kwok YN, Kofod H. Effect of cholecystokinin and secretin
on somatostatin release from cultured antral cells. Gastroenterology 104: 1414
1419, 1993.

163. Caterina MJ, Rosen TA, Tominaga M, Brake AJ, Julius D. A capsaicin-receptor homologue with a high threshold for noxious heat. Nature 398: 436 441, 1999.

142. Buchan AM, Squires PE, Ring M, Meloche RM. Mechanism of action of the calciumsensing receptor in human antral gastrin cells. Gastroenterology 120: 1128 1139,
2001.

164. Catlow K, Ashurst HL, Varro A, Dimaline R. Identification of a gastrin response


element in the vesicular monoamine transporter type 2 promoter and requirement
of 20 S proteasome subunits for transcriptional activity. J Biol Chem 282: 17069
17077, 2007.

143. Buclin T, Cosma Rochat M, Burckhardt P, Azria M, Attinger M. Bioavailability and


biological efficacy of a new oral formulation of salmon calcitonin in healthy volunteers. J Bone Miner Res 17: 1478 1485, 2002.
144. Bugrim AE. Regulation of Ca2 release by cAMP-dependent protein kinase. A mechanism for agonist-specific calcium signaling? Cell Calcium 25: 219 226, 1999.
145. Busque SM, Kerstetter JE, Geibel JP, Insogna K. L-type amino acids stimulate gastric
acid secretion by activation of the calcium-sensing receptor in parietal cells. Am J
Physiol Gastrointest Liver Physiol 289: G664 G669, 2005.
146. Cai Q, Chandler JS, Wasserman RH, Kumar R, Penniston JT. Vitamin D and adaptation to dietary calcium and phosphate deficiencies increase intestinal plasma membrane calcium pump gene expression. Proc Natl Acad Sci USA 90: 13451349, 1993.
147. Calhoun BC, Lapierre LA, Chew CS, Goldenring JR. Rab11a redistributes to apical
secretory canaliculus during stimulation of gastric parietal cells. Am J Physiol Cell
Physiol 275: C163C170, 1998.
148. Cali JJ, Hsieh CL, Francke U, Russell DW. Mutations in the bile acid biosynthetic
enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol
Chem 266: 7779 7783, 1991.
149. Cali JJ, Russell DW. Characterization of human sterol 27-hydroxylase. A mitochondrial cytochrome P-450 that catalyzes multiple oxidation reaction in bile acid biosynthesis. J Biol Chem 266: 7774 7778, 1991.
150. Calvo MS, Whiting SJ, Barton CN. Vitamin D intake: a global perspective of current
status. J Nutr 135: 310 316, 2005.
151. Canadillas S, Canalejo R, Rodriguez-Ortiz ME, Martinez-Moreno JM, Estepa JC,
Zafra R, Perez J, Munoz-Castaneda JR, Canalejo A, Rodriguez M, Almaden Y. The
up-regulation of the parathyroid VDR expression by extracellular calcium is mediated by the ERK1/2-MAPK signaling pathway. Am J Physiol Renal Physiol 2010.
152. Canalejo A, Canadillas S, Ballesteros E, Rodriguez M, Almaden Y. Importance of
arachidonic acid as a mediator of parathyroid gland response. Kidney Int Suppl S10
S13, 2003.
153. Canfield SP, Spencer JE. The inhibitory effects of 5-hydroxytryptamine on gastric
acid secretion by the rat isolated stomach. Br J Pharmacol 78: 123129, 1983.

165. Ceglia L, Harris SS, Rasmussen HM, Dawson-Hughes B. Activation of the calcium
sensing receptor stimulates gastrin and gastric acid secretion in healthy participants.
Osteoporos Int 20: 7178, 2009.
166. Chabardes D, Gagnan-Brunette M, Imbert-Teboul M, Gontcharevskaia O, Montegut M, Clique A, Morel F. Adenylate cyclase responsiveness to hormones in various portions of the human nephron. J Clin Invest 65: 439 448, 1980.
167. Chabre O, Conklin BR, Lin HY, Lodish HF, Wilson E, Ives HE, Catanzariti L, Hemmings BA, Bourne HR. A recombinant calcitonin receptor independently stimulates
3=,5=-cyclic adenosine monophosphate and Ca2/inositol phosphate signaling pathways. Mol Endocrinol 6: 551556, 1992.
168. Chai SY, Christopoulos G, Cooper ME, Sexton PM. Characterization of binding sites
for amylin, calcitonin, and CGRP in primate kidney. Am J Physiol Renal Physiol 274:
F51F62, 1998.
169. Chambers TJ, Magnus CJ. Calcitonin alters behaviour of isolated osteoclasts. J Pathol
136: 2739, 1982.
170. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, Hoenderop JG. The
beta-glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science 310:
490 493, 2005.
171. Chang W, Tu C, Chen TH, Bikle D, Shoback D. The extracellular calcium-sensing
receptor (CaSR) is a critical modulator of skeletal development. Sci Signal 1: ra1,
2008.
172. Chang W, Tu C, Chen TH, Komuves L, Oda Y, Pratt SA, Miller S, Shoback D.
Expression and signal transduction of calcium-sensing receptors in cartilage and
bone. Endocrinology 140: 58835893, 1999.
173. Chattopadhyay N, Cheng I, Rogers K, Riccardi D, Hall A, Diaz R, Hebert SC, Soybel
DI, Brown EM. Identification and localization of extracellular Ca2-sensing receptor
in rat intestine. Am J Physiol Gastrointest Liver Physiol 274: G122G130, 1998.
174. Chattopadhyay N, Yano S, Tfelt-Hansen J, Rooney P, Kanuparthi D, Bandyopadhyay
S, Ren X, Terwilliger E, Brown EM. Mitogenic action of calcium-sensing receptor on
rat calvarial osteoblasts. Endocrinology 145: 34513462, 2004.

154. Cantley LK, Russell J, Lettieri D, Sherwood LM. 1,25-Dihydroxyvitamin D3 suppresses parathyroid hormone secretion from bovine parathyroid cells in tissue culture. Endocrinology 117: 2114 2119, 1985.

175. Chen CJ, Barnett JV, Congo DA, Brown EM. Divalent cations suppress 3=,5=-adenosine monophosphate accumulation by stimulating a pertussis toxin-sensitive guanine nucleotide-binding protein in cultured bovine parathyroid cells. Endocrinology
124: 233239, 1989.

155. Care AD, Bates RF, Swaminathan R, Ganguli PC. The role of gastrin as a calcitonin
secretagogue. J Endocrinol 51: 735744, 1971.

176. Chen D, Marvik R, Ronning K, Andersson K, Waldum HL, Hakanson R. Gastrinevoked secretion of pancreastatin and histamine from ECL cells and of acid from

242

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

139. Bu FX, Armas L, Lappe J, Zhou Y, Gao G, Wang HW, Recker R, Zhao LJ. Comprehensive association analysis of nine candidate genes with serum 25-hydroxy vitamin
D levels among healthy Caucasian subjects. Hum Genet 128: 549 556, 2010.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


parietal cells in isolated, vascularly perfused rat stomach. Effects of isobutyl methylxanthin and alpha-fluoromethylhistidine. Regul Pept 65: 133138, 1996.
177. Chen D, Zhao CM, Al-Haider W, Hakanson R, Rehfeld JF, Kopin AS. Differentiation
of gastric ECL cells is altered in CCK(2) receptor-deficient mice. Gastroenterology
123: 577585, 2002.
178. Chen D, Zhao CM, Dockray GJ, Varro A, Van Hoek A, Sinclair NF, Wang TC, Koh TJ.
Glycine-extended gastrin synergizes with gastrin 17 to stimulate acid secretion in
gastrin-deficient mice. Gastroenterology 119: 756 765, 2000.

197. Chiba T, Fujita T, Yamada T. Carbachol inhibits stimulant-induced increases in fundic


D-cell cytosolic Ca2 concentration. Am J Physiol Gastrointest Liver Physiol 257:
G308 G312, 1989.
198. Chiba T, Park J, Yamada T. Biosynthesis of somatostatin in canine fundic D cells. J Clin
Invest 81: 282287, 1988.
199. Chiba T, Taminato T, Kadowaki S, Abe H, Chihara K, Seino Y, Matsukura S, Fujita T.
Effects of glucagon, secretin, and vasoactive intestinal polypeptide on gastric somatostatin and gastrin release from isolated perfused rat stomach. Gastroenterology 79:
6771, 1980.
200. Chiba T, Yamada T. Mechanisms for muscarinic inhibition of somatostatin release
from canine fundic D cells. Metabolism 39: 122124, 1990.

180. Chen KS, DeLuca HF. Cloning of the human 1 alpha,25-dihydroxyvitamin D-3 24hydroxylase gene promoter and identification of two vitamin D-responsive elements. Biochim Biophys Acta 1263: 19, 1995.

201. Chirayath MV, Gajdzik L, Hulla W, Graf J, Cross HS, Peterlik M. Vitamin D increases
tight-junction conductance and paracellular Ca2 transport in Caco-2 cell cultures.
Am J Physiol Gastrointest Liver Physiol 274: G389 G396, 1998.

181. Chen S, Glenn DJ, Ni W, Grigsby CL, Olsen K, Nishimoto M, Law CS, Gardner DG.
Expression of the vitamin d receptor is increased in the hypertrophic heart. Hypertension 52: 1106 1112, 2008.

202. Chiu HF, Huang YW, Chang CC, Yang CY. Use of proton pump inhibitors increased
the risk of hip fracture: a population-based case-control study. Pharmacoepidemiol
Drug Safety 19: 11311136, 2010.

182. Cheng I, Qureshi I, Chattopadhyay N, Qureshi A, Butters RR, Hall AE, Cima RR,
Rogers KV, Hebert SC, Geibel JP, Brown EM, Soybel DI. Expression of an extracellular calcium-sensing receptor in rat stomach. Gastroenterology 116: 118 126, 1999.

203. Christakos S, Brunette MG, Norman AW. Localization of immunoreactive vitamin


D-dependent calcium binding protein in chick nephron. Endocrinology 109: 322324,
1981.

183. Cheng JB, Levine MA, Bell NH, Mangelsdorf DJ, Russell DW. Genetic evidence that
the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proc Natl Acad Sci
USA 101: 77117715, 2004.

204. Chuang CN, Tanner M, Lloyd KC, Wong H, Soll AH. Endogenous somatostatin
inhibits histamine release from canine gastric mucosal cells in primary culture. Am J
Physiol Gastrointest Liver Physiol 265: G521G525, 1993.

184. Cheng JB, Motola DL, Mangelsdorf DJ, Russell DW. De-orphanization of cytochrome P450 2R1: a microsomal vitamin D 25-hydroxilase. J Biol Chem 278: 38084
38093, 2003.

205. Chung I, Li P, Lee K, Chang T, Chey WY. Dual inhibitory mechanism of secretin
action on acid secretion in totally isolated, vascularly perfused rat stomach. Gastroenterology 107: 17511758, 1994.

185. Cheng SX, Geibel JP, Hebert SC. Extracellular polyamines regulate fluid secretion in
rat colonic crypts via the extracellular calcium-sensing receptor. Gastroenterology
126: 148 158, 2004.

206. Chung MK, Guler AD, Caterina MJ. TRPV1 shows dynamic ionic selectivity during
agonist stimulation. Nat Neurosci 11: 555564, 2008.

186. Cheng SX, Okuda M, Hall AE, Geibel JP, Hebert SC. Expression of calcium-sensing
receptor in rat colonic epithelium: evidence for modulation of fluid secretion. Am J
Physiol Gastrointest Liver Physiol 283: G240 G250, 2002.
187. Chesney RW, Rosen JF, Hamstra AJ, Smith C, Mahaffey K, DeLuca HF. Absence of
seasonal variation in serum concentrations of 1,25-dihydroxyvitamin D despite a rise
in 25-hydroxyvitamin D in summer. J Clin Endocrinol Metab 53: 139 142, 1981.
188. Cheung R, Erclik MS, Mitchell J. Increased expression of G11alpha in osteoblastic
cells enhances parathyroid hormone activation of phospholipase C and AP-1 regulation of matrix metalloproteinase-13 mRNA. J Cell Physiol 204: 336 343, 2005.
189. Chew CS. Cholecystokinin, carbachol, gastrin, histamine, and forskolin increase
[Ca2]i in gastric glands. Am J Physiol Gastrointest Liver Physiol 250: G814 G823,
1986.
190. Chew CS, Brown MR. Release of intracellular Ca2 and elevation of inositol trisphosphate by secretagogues in parietal and chief cells isolated from rabbit gastric mucosa. Biochim Biophys Acta 888: 116 125, 1986.
191. Chey WY, Chang CH, Pan HJ, Chang C, Kim BM, Park IS, Chang TM. Evidence on
the presence of secretin cells in the gastric antral and oxyntic mucosa. Regul Pept 111:
183190, 2003.
192. Chey WY, Chang TM, Park HJ, Lee KY, Escoffery R. Secretin-like immunoreactivity
and biological activity in the antral mucosa. Endocrinology 113: 651 656, 1983.
193. Chey WY, Escoffery R. Secretion cells in the gastrointestinal tract. Endocrinology 98:
1390 1395, 1976.
194. Chey WY, Hitanant S, Hendricks J, Lorber SH. Effect of secretin and cholecystokinin
on gastric emptying and gastric secretion in man. Gastroenterology 58: 820 827,
1970.

207. Cid LP, Montrose-Rafizadeh C, Smith DI, Guggino WB, Cutting GR. Cloning of a
putative human voltage-gated chloride channel (CIC-2) cDNA widely expressed in
human tissues. Hum Mol Genet 4: 407 413, 1995.
208. Clark CG, Crooks J, Dawson AA, Mitchell PE. Disordered calcium metabolism after
polya partial gastrectomy. Lancet 1: 734 738, 1964.
209. Clemens TL, Adams JS, Henderson SL, Holick MF. Increased skin pigment reduces
the capacity of skin to synthesise vitamin D3. Lancet 1: 74 76, 1982.
210. Clements MR, Davies M, Fraser DR, Lumb GA, Mawer EB, Adams PH. Metabolic
inactivation of vitamin D is enhanced in primary hyperparathyroidism. Clin Sci 73:
659 664, 1987.
211. Clements MR, Davies M, Hayes ME, Hickey CD, Lumb GA, Mawer EB, Adams PH.
The role of 1,25-dihydroxyvitamin D in the mechanism of acquired vitamin D deficiency. Clin Endocrinol 37: 1727, 1992.
212. Clements MR, Johnson L, Fraser DR. A new mechanism for induced vitamin D
deficiency in calcium deprivation. Nature 325: 62 65, 1987.
213. Cleve H. The variants of the group-specific component. A review of their distribution in human populations. Isr J Med Sci 9: 11331146, 1973.
214. Cocchi D, Maccarinelli G, Sibilia V, Tulipano G, Torsello A, Pazzaglia UE, Giustina A,
Netti C. GH-releasing peptides and bone. J Endocrinol Invest 28: 1114, 2005.
215. Cochran M, Peacock M, Sachs G, Nordin BE. Renal effects of calcitonin. Br Med J 1:
135137, 1970.
216. Collip JB. The extraction of a parathyroid hormone which will prevent or control
parathyroid tetany and which regulates the level of blood calcium. J Biol Chem 63:
395 438, 1925.

195. Chey WY, Lee YH, Hendricks JG, Rhodes RA, Tai HH. Plasma secretin concentrations in fasting and postprandial state in man. Am J Dig Dis 23: 981988, 1978.

217. Colnot S, Ovejero C, Romagnolo B, Porteu A, Lacourte P, Thomasset M, Perret C.


Transgenic analysis of the response of the rat calbindin-D 9k gene to vitamin D.
Endocrinology 141: 23012308, 2000.

196. Chiba T, Fisher SK, Agranoff BW, Yamada T. Autoregulation of muscarinic and
gastrin receptors on gastric parietal cells. Am J Physiol Gastrointest Liver Physiol 256:
G356 G363, 1989.

218. Committee to Review Dietary Reference Intakes for Vitamin D and Calcium. Dietary
Reference Intakes for Calcium and Vitamin D. Washington, DC: National Academy
Press, 2010.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

243

Downloaded from on December 8, 2014

179. Chen D, Zhao CM, Hakanson R, Samuelson LC, Rehfeld JF, Friis-Hansen L. Altered
control of gastric acid secretion in gastrin-cholecystokinin double mutant mice. Gastroenterology 126: 476 487, 2004.

SASCHA KOPIC AND JOHN P. GEIBEL


219. Compston JE, Thompson RP. Intestinal absorption of 25-hydroxyvitamin D and
osteomalacia in primary biliary cirrhosis. Lancet 1: 721724, 1977.
220. Conigrave AD, Quinn SJ, Brown EM. L-Amino acid sensing by the extracellular
Ca2-sensing receptor. Proc Natl Acad Sci USA 97: 4814 4819, 2000.
221. Cooke NE, David EV. Serum vitamin D-binding protein is a third member of the
albumin and alpha fetoprotein gene family. J Clin Invest 76: 2420 2424, 1985.
222. Cooper CW, Biggerstaff CR, Wiseman CW, Carlone MF. Hypocalcemic effect of
pentagastrin and related gastrointestinal hormnal peptides in the rat. Endocrinology
91: 14551461, 1972.
223. Cooper CW, Hirsch PF, Munson PL. Importance of endogenous thyrocalcitonin for
protection against hypercalcemia in the rat. Endocrinology 86: 406 415, 1970.

240. Darwish HM, DeLuca HF. Identification of a 1,25-dihydroxyvitamin D3-response


element in the 5=-flanking region of the rat calbindin D-9k gene. Proc Natl Acad Sci
USA 89: 603 607, 1992.
241. Database Csr. http://www.casrdb.mcgill.ca/ [retrieved September 2011].
242. Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS, Suganuma T, Matsukura S,
Kangawa K, Nakazato M. Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats
and humans. Endocrinology 141: 4255 4261, 2000.
243. Datta R, Waheed A, Shah GN, Sly WS. Signal sequence mutation in autosomal
dominant form of hypoparathyroidism induces apoptosis that is corrected by a
chemical chaperone. Proc Natl Acad Sci USA 104: 19989 19994, 2007.

224. Cooper CW, McGuigan JE, Schwesinger WH, Brubaker RL, Munson PL. Correlation
between levels of gastrin and thyrocalcitonin in pig thyroid venous blood. Endocrinology 95: 302307, 1974.

244. Davies M, Heys SE, Selby PL, Berry JL, Mawer EB. Increased catabolism of 25hydroxyvitamin D in patients with partial gastrectomy and elevated 1,25-dihydroxyvitamin D levels. Implications for metabolic bone disease. J Clin Endocrinol
Metab 82: 209 212, 1997.

225. Cooper CW, Ramp WK, Becker DI, Ontjes DA. In vitro secretion of immunoreactive
rat thyrocalcitonin. Endocrinology 101: 304 311, 1977.

245. Davies M, Mawer EB, Krawitt EL. Comparative absorption of vitamin D3 and 25hydroxyvitamin D3 in intestinal disease. Gut 21: 287292, 1980.

226. Cooper CW, Schwesinger WH, Mahgoub AM, Ontjes DA. Thyrocalcitonin: stimulation of secretion by pentagastrin. Science 172: 1238 1240, 1971.
227. Copp DH, Cameron EC, Cheney BA, Davidson AG, Henze KG. Evidence for calcitonin: a new hormone from the parathyroid that lowers blood calcium. Endocrinology
70: 638 649, 1962.

229. Cornish J, Callon KE, Bava U, Kamona SA, Cooper GJ, Reid IR. Effects of calcitonin,
amylin, calcitonin gene-related peptide on osteoclast development. Bone 29: 162
168, 2001.
230. Couper RT, Corey M, Moore DJ, Fisher LJ, Forstner GG, Durie PR. Decline of
exocrine pancreatic function in cystic fibrosis patients with pancreatic sufficiency.
Pediatr Res 32: 179 182, 1992.
231. Cramer T, Juttner S, Plath T, Mergler S, Seufferlein T, Wang TC, Merchant J, Hocker
M. Gastrin transactivates the chromogranin A gene through MEK-1/ERK- and PKCdependent phosphorylation of Sp1 and CREB. Cell Signal 20: 60 72, 2008.
232. Cuppoletti J, Sachs G. Regulation of gastric acid secretion via modulation of a chloride conductance. J Biol Chem 259: 1495214959, 1984.
233. DAmour P, Brossard JH, Rousseau L, Nguyen-Yamamoto L, Nassif E, Lazure C,
Gauthier D, Lavigne JR, Zahradnik RJ. Structure of non-(1 84) PTH fragments secreted by parathyroid glands in primary and secondary hyperparathyroidism. Kidney
Int 68: 998 1007, 2005.
234. DAmour P, Segre GV, Roth SI, Potts JT Jr. Analysis of parathyroid hormone and its
fragments in rat tissues: chemical identification and microscopical localization. J Clin
Invest 63: 89 98, 1979.
235. Dahlback H, Wikvall K. 25-Hydroxylation of vitamin D3 by a cytochrome P-450 from
rabbit liver mitochondria. Biochem J 252: 207213, 1988.
236. Daiger SP, Schanfield MS, Cavalli-Sforza LL. Group-specific component (Gc) proteins bind vitamin D and 25-hydroxyvitamin D. Proc Natl Acad Sci USA 72: 2076
2080, 1975.
237. Dale HH, Laidlow PP. The physiological action of beta-iminazolylethylamine. J Physiol
41: 318 344, 1910.
238. Dardenne O, Prudhomme J, Arabian A, Glorieux FH, St-Arnaud R. Targeted inactivation of the 25-hydroxyvitamin D(3)-1(alpha)-hydroxylase gene (CYP27B1) creates an animal model of pseudovitamin D-deficiency rickets. Endocrinology 142:
31353141, 2001.
239. Dardenne O, Prudhomme J, Hacking SA, Glorieux FH, St-Arnaud R. Correction of
the abnormal mineral ion homeostasis with a high-calcium, high-phosphorus, highlactose diet rescues the PDDR phenotype of mice deficient for the 25-hydroxyvitamin D-1alpha-hydroxylase (CYP27B1). Bone 32: 332340, 2003.

244

247. Davis WL, Jones RG. Lysosomal proliferation in rachitic avian intestinal absorptive
cells following 1,25-dihydroxycholecalciferol. Tissue Cell 14: 585595, 1982.
248. De Groot T, Kovalevskaya NV, Verkaart S, Schilderink N, Felici M, van der Hagen
EA, Bindels RJ, Vuister GW, Hoenderop JG. Molecular mechanisms of calmodulin
action on TRPV5 and modulation by parathyroid hormone. Mol Cell Biol 31: 2845
2853, 2011.
249. De Groot T, Lee K, Langeslag M, Xi Q, Jalink K, Bindels RJ, Hoenderop JG. Parathyroid hormone activates TRPV5 via PKA-dependent phosphorylation. J Am Soc Nephrol 20: 16931704, 2009.
250. De Jesus Ferreira MC, Helies-Toussaint C, Imbert-Teboul M, Bailly C, Verbavatz JM,
Bellanger AC, Chabardes D. Co-expression of a Ca2-inhibitable adenylyl cyclase
and of a Ca2-sensing receptor in the cortical thick ascending limb cell of the rat
kidney. Inhibition of hormone-dependent cAMP accumulation by extracellular
Ca2. J Biol Chem 273: 1519215202, 1998.
251. De Rouffignac C, Elalouf JM. Effects of calcitonin on the renal concentrating mechanism. Am J Physiol Renal Fluid Electrolyte Physiol 245: F506 F511, 1983.
252. De Vries F, Cooper AL, Cockle SM, Van Staa TP, Cooper C. Fracture risk in patients
receiving acid-suppressant medication alone and in combination with bisphosphonates. Osteoporosis Int 20: 1989 1998, 2009.
253. Dedek K, Waldegger S. Colocalization of KCNQ1/KCNE channel subunits in the
mouse gastrointestinal tract. Pflgers Arch 442: 896 902, 2001.
254. Delhanty PJ, van der Eerden BC, van der Velde M, Gauna C, Pols HA, Jahr H, Chiba
H, van der Lely AJ, van Leeuwen JP. Ghrelin and unacylated ghrelin stimulate human
osteoblast growth via mitogen-activated protein kinase (MAPK)/phosphoinositide
3-kinase (PI3K) pathways in the absence of GHS-R1a. J Endocrinol 188: 37 47, 2006.
255. Deller DJ. Radiocalcium absorption after partial gastrectomy. Am J Dig Dis 11: 10 19,
1966.
256. DeLuca HF, Lund J, Rosenbloom A, Lobeck CC. Metabolism of tritiated vitamin D3
in familial vitamin D-resistant rickets with hypophosphatemia. J Pediatr 70: 828 832,
1967.
257. DelValle J, Yamada T. Amino acids and amines stimulate gastrin release from canine
antral G-cells via different pathways. J Clin Invest 85: 139 143, 1990.
258. Delvin EE, Arabian A, Glorieux FH. Kinetics of liver microsomal cholecalciferol
25-hydroxylase in vitamin D-depleted and -repleated rats. Biochem J 172: 417 422,
1978.
259. Demarest JR, Loo DD, Sachs G. Activation of apical chloride channels in the gastric
oxyntic cell. Science 245: 402 404, 1989.
260. Dempster DW, Hughes-Begos CE, Plavetic-Chee K, Brandao-Burch A, Cosman F,
Nieves J, Neubort S, Lu SS, Iida-Klein A, Arnett T, Lindsay R. Normal human oste-

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

228. Corley DA, Kubo A, Zhao W, Quesenberry C. Proton pump inhibitors and histamine-2 receptor antagonists are associated with hip fractures among at-risk patients.
Gastroenterology 139: 93101, 2010.

246. Davies SL, Ozawa A, McCormick WD, Dvorak MM, Ward DT. Protein kinase
C-mediated phosphorylation of the calcium-sensing receptor is stimulated by receptor activation and attenuated by calyculin-sensitive phosphatase activity. J Biol Chem
282: 15048 15056, 2007.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


oclasts formed from peripheral blood monocytes express PTH type 1 receptors and
are stimulated by PTH in the absence of osteoblasts. J Cell Biochem 95: 139 148,
2005.

280. Dorrington KJ, Hui A, Hofmann T, Hitchman AJ, Harrison JE. Porcine intestinal
calcium-binding protein. Molecular properties and the effect of binding calcium ions.
J Biol Chem 249: 199 204, 1974.

261. Deng F, Ling J, Ma J, Liu C, Zhang W. Stimulation of intramembranous bone repair in


rats by ghrelin. Exp Physiol 93: 872 879, 2008.

281. Dorwart MR, Shcheynikov N, Wang Y, Stippec S, Muallem S. SLC26A9 is a Cl


channel regulated by the WNK kinases. J Physiol 584: 333345, 2007.

262. Dent J, Kahrilas PJ, Hatlebakk J, Vakil N, Denison H, Franzen S, Lundborg P. A


randomized, comparative trial of a potassium-competitive acid blocker (AZD0865)
and esomeprazole for the treatment of patients with nonerosive reflux disease. Am
J Gastroenterol 103: 20 26, 2008.

282. Drees BM, Hamilton JW. Pancreastatin and bovine parathyroid cell secretion. Bone
Miner 17: 335346, 1992.

263. Derler I, Hofbauer M, Kahr H, Fritsch R, Muik M, Kepplinger K, Hack ME, Moritz S,
Schindl R, Groschner K, Romanin C. Dynamic but not constitutive association of
calmodulin with rat TRPV6 channels enables fine tuning of Ca2-dependent inactivation. J Physiol 577: 31 44, 2006.
264. Desfleurs E, Wittner M, Simeone S, Pajaud S, Moine G, Rajerison R, Di Stefano A.
Calcium-sensing receptor: regulation of electrolyte transport in the thick ascending
limb of Henles loop. Kidney Blood Press Res 21: 401 412, 1998.

283. Drescher D, DeLuca HF. Vitamin D stimulated calcium binding protein from rat
intestinal mucosa. Purification and some properties. Biochemistry 10: 23022307,
1971.
284. Dressman JB, Berardi RR, Dermentzoglou LC, Russell TL, Schmaltz SP, Barnett JL,
Jarvenpaa KM. Upper gastrointestinal (GI) pH in young, healthy men and women.
Pharm Res 7: 756 761, 1990.
285. Dubrasquet M, Bataille D, Gespach C. Oxyntomodulin (glucagon-37 or bioactive
enteroglucagon): a potent inhibitor of pentagastrin-stimulated acid secretion in rats.
Biosci Rep 2: 391395, 1982.

265. Di Stefano A, Wittner M, Nitschke R, Braitsch R, Greger R, Bailly C, Amiel C, Roinel


N, de Rouffignac C. Effects of parathyroid hormone and calcitonin on Na, Cl, K,
Mg2 and Ca2 transport in cortical and medullary thick ascending limbs of mouse
kidney. Pflgers Arch 417: 161167, 1990.

286. Dueland S, Helgerud P, Pedersen JI, Berg T, Drevon CA. Plasma clearance, transfer,
and distribution of vitamin D3 from intestinal lymph. Am J Physiol Endocrinol Metab
245: E326 E331, 1983.

266. Diamond JM, Bossert WH. Standing-gradient osmotic flow. A mechanism for coupling of water and solute transport in epithelia. J Gen Physiol 50: 20612083, 1967.

287. Dueland S, Pedersen JI, Helgerud P, Drevon CA. Absorption, distribution, transport
of vitamin D3 and 25-hydroxyvitamin D3 in the rat. Am J Physiol Endocrinol Metab 245:
E463E467, 1983.

268. Dimaline R, Sandvik AK. Histidine decarboxylase gene expression in rat fundus is
regulated by gastrin. FEBS Lett 281: 20 22, 1991.
269. Dinoso V, Chey WY, Hendricks J, Lorber SH. Intestinal mucosal hormones and
motor function of the stomach in man. J Appl Physiol 26: 326 329, 1969.
270. Divieti P, Geller AI, Suliman G, Juppner H, Bringhurst FR. Receptors specific for the
carboxyl-terminal region of parathyroid hormone on bone-derived cells: determinants of ligand binding and bioactivity. Endocrinology 146: 18631870, 2005.
271. Divieti P, Inomata N, Chapin K, Singh R, Juppner H, Bringhurst FR. Receptors for the
carboxyl-terminal region of pth(1 84) are highly expressed in osteocytic cells. Endocrinology 142: 916 925, 2001.
272. Divieti P, John MR, Juppner H, Bringhurst FR. Human PTH-(7 84) inhibits bone
resorption in vitro via actions independent of the type 1 PTH/PTHrP receptor.
Endocrinology 143: 171176, 2002.
273. Dixon KM, Deo SS, Norman AW, Bishop JE, Halliday GM, Reeve VE, Mason RS. In
vivo relevance for photoprotection by the vitamin D rapid response pathway. J
Steroid Biochem Mol Biol 103: 451 456, 2007.
274. Dobnig H, Turner RT. The effects of programmed administration of human parathyroid hormone fragment (134) on bone histomorphometry and serum chemistry
in rats. Endocrinology 138: 4607 4612, 1997.
275. Dong H, Sellers ZM, Smith A, Chow JY, Barrett KE. Na/Ca2 exchange regulates
Ca2-dependent duodenal mucosal ion transport and HCO3 secretion in mice. Am
J Physiol Gastrointest Liver Physiol 288: G457G465, 2005.
276. Dornonville de la Cour C, Bjorkqvist M, Sandvik AK, Bakke I, Zhao CM, Chen D,
Hakanson R. A-like cells in the rat stomach contain ghrelin and do not operate under
gastrin control. Regul Pept 99: 141150, 2001.
277. Dornonville de la Cour C, Lindqvist A, Egecioglu E, Tung YC, Surve V, Ohlsson C,
Jansson JO, Erlanson-Albertsson C, Dickson SL, Hakanson R. Ghrelin treatment
reverses the reduction in weight gain and body fat in gastrectomised mice. Gut 54:
907913, 2005.

288. Dueland S, Pedersen JI, Helgerud P, Drevon CA. Transport of vitamin D3 from rat
intestine. Evidence for transfer of vitamin D3 from chylomicrons to alpha-globulins.
J Biol Chem 257: 146 150, 1982.
289. Duflos C, Bellaton C, Baghdassarian N, Gadoux M, Pansu D, Bronner F. 1,25Dihydroxycholecalciferol regulates rat intestinal calbindin D9k posttranscriptionally.
J Nutr 126: 834 841, 1996.
290. Duflos C, Bellaton C, Pansu D, Bronner F. Calcium solubility, intestinal sojourn time
and paracellular permeability codetermine passive calcium absorption in rats. J Nutr
125: 2348 2355, 1995.
291. Dufner MM, Kirchhoff P, Remy C, Hafner P, Muller MK, Cheng SX, Tang LQ, Hebert
SC, Geibel JP, Wagner CA. The calcium-sensing receptor acts as a modulator of
gastric acid secretion in freshly isolated human gastric glands. Am J Physiol Gastrointest Liver Physiol 289: G1084 G1090, 2005.
292. Duman JG, Pathak NJ, Ladinsky MS, McDonald KL, Forte JG. Three-dimensional
reconstruction of cytoplasmic membrane networks in parietal cells. J Cell Sci 115:
12511258, 2002.
293. Duman JG, Tyagarajan K, Kolsi MS, Moore HP, Forte JG. Expression of rab11a N124I
in gastric parietal cells inhibits stimulatory recruitment of the H-K-ATPase. Am J
Physiol Cell Physiol 277: C361C372, 1999.
294. Durr KL, Abe K, Tavraz NN, Friedrich T. E2P state stabilization by the N-terminal
tail of the H-K-ATPase beta-subunit is critical for efficient proton pumping under
in vivo conditions. J Biol Chem 284: 2014720154, 2009.
295. DuVal JW, Saffouri B, Weir GC, Walsh JH, Arimura A, Makhlouf GM. Stimulation of
gastrin and somatostatin secretion from the isolated rat stomach by bombesin. Am J
Physiol Gastrointest Liver Physiol 241: G242G247, 1981.
296. Dvorak MM, Chen TH, Orwoll B, Garvey C, Chang W, Bikle DD, Shoback DM.
Constitutive activity of the osteoblast Ca2-sensing receptor promotes loss of cancellous bone. Endocrinology 148: 3156 3163, 2007.
297. Dvorak MM, Siddiqua A, Ward DT, Carter DH, Dallas SL, Nemeth EF, Riccardi D.
Physiological changes in extracellular calcium concentration directly control osteoblast function in the absence of calciotropic hormones. Proc Natl Acad Sci USA 101:
5140 5145, 2004.
298. Edkins JS. The chemical mechanism of gastric secretion. J Physiol 34: 133144, 1906.

278. Dornonville de la Cour C, Lindstrom E, Norlen P, Hakanson R. Ghrelin stimulates


gastric emptying but is without effect on acid secretion and gastric endocrine cells.
Regul Pept 120: 2332, 2004.
279. Doroszewicz J, Waldegger P, Jeck N, Seyberth H, Waldegger S. pH dependence of
extracellular calcium sensing receptor activity determined by a novel technique.
Kidney Int 67: 187192, 2005.

299. Edwards BR, Baer PG, Sutton RA, Dirks JH. Micropuncture study of diuretic effects
on sodium and calcium reabsorption in the dog nephron. J Clin Invest 52: 2418 2427,
1973.
300. Edwards BR, Sutton RA, Dirks JH. Effect of calcium infusion on renal tubular reabsorption in the dog. Am J Physiol 227: 1318, 1974.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

245

Downloaded from on December 8, 2014

267. Dicker F, Quitterer U, Winstel R, Honold K, Lohse MJ. Phosphorylation-independent inhibition of parathyroid hormone receptor signaling by G protein-coupled
receptor kinases. Proc Natl Acad Sci USA 96: 5476 5481, 1999.

SASCHA KOPIC AND JOHN P. GEIBEL


301. Edwards LW, Herrington JL Jr. Vagotomy and gastro-enterostomy; vagotomy and
conservative gastrectomy; a comparative study. Ann Surg 137: 873 883, 1953.
302. Ekblad E, Ekelund M, Graffner H, Hakanson R, Sundler F. Peptide-containing nerve
fibers in the stomach wall of rat and mouse. Gastroenterology 89: 73 85, 1985.
303. El Hag AI, Karrar ZA. Nutritional vitamin D deficiency rickets in Sudanese children.
Ann Trop Paediatr 15: 69 76, 1995.
304. Elalouf JM, Roinel N, de Rouffignac C. ADH-like effects of calcitonin on electrolyte
transport by Henles loop of rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 246:
F213F220, 1984.
305. Ellman P, Irwin DB. Osteomalacia following gastrectomy. Postgrad Med J 35: 358
361, 1959.
306. Emmelin N, Kahlson GS. Histamine as a physiological excitant of acid gastric secretion. Acta Physiol Scand 8: 289 304, 1944.
307. Engelhardt W, Grohmann C, Schwille PO, Geus A. Calcium absorption in the rat as
influenced by highly selective vagotomy with special regard to endogenous gastrin.
Res Exp Med 180: 19, 1982.
308. Engelhardt W, Rumenapf G, Schwille PO. Effects of highly selective vagotomy on
small intestinal calcium transport in the rat. Miner Electrolyte Metab 10: 239 243,
1984.

310. Ericsson P, Hakanson R, Rehfeld JF, Norlen P. Gastrin release: antrum microdialysis
reveals a complex neural control. Regul Pept 161: 2232, 2010.
311. Erler I, Hirnet D, Wissenbach U, Flockerzi V, Niemeyer BA. Ca2-selective transient receptor potential V channel architecture and function require a specific
ankyrin repeat. J Biol Chem 279: 34456 34463, 2004.

323. Feldberg W, Toh CC. Distribution of 5-hydroxytryptamine (serotonin, enteramine)


in the wall of the digestive tract. J Physiol 119: 352362, 1953.
324. Fellenius E, Berglindh T, Sachs G, Olbe L, Elander B, Sjostrand SE, Wallmark B.
Substituted benzimidazoles inhibit gastric acid secretion by blocking (H
K)ATPase. Nature 290: 159 161, 1981.
325. Feng J, Petersen CD, Coy DH, Jiang JK, Thomas CJ, Pollak MR, Wank SA. Calciumsensing receptor is a physiologic multimodal chemosensor regulating gastric G-cell
growth and gastrin secretion. Proc Natl Acad Sci USA 107: 1779117796, 2010.
326. Ferrari SL, Behar V, Chorev M, Rosenblatt M, Bisello A. Endocytosis of ligand-human
parathyroid hormone receptor 1 complexes is protein kinase C-dependent and
involves beta-arrestin2. Real-time monitoring by fluorescence microscopy. J Biol
Chem 274: 29968 29975, 1999.
327. Ferrari SL, Pierroz DD, Glatt V, Goddard DS, Bianchi EN, Lin FT, Manen D, Bouxsein ML. Bone response to intermittent parathyroid hormone is altered in mice null
for -Arrestin2. Endocrinology 146: 1854 1862, 2005.
328. Findlay DM, Michelangeli VP, Orlowski RC, Martin TJ. Biological activities and receptor interactions of des-Leu16 salmon and des-Phe16 human calcitonin. Endocrinology 112: 1288 1291, 1983.
329. Firsov D, Bellanger AC, Marsy S, Elalouf JM. Quantitative RT-PCR analysis of calcitonin receptor mRNAs in the rat nephron. Am J Physiol Renal Fluid Electrolyte Physiol
269: F702F709, 1995.
330. Flannery PJ, Spurney RF. Domains of the parathyroid hormone (PTH) receptor
required for regulation by G protein-coupled receptor kinases (GRKs). Biochem
Pharmacol 62: 10471058, 2001.
331. Fleming JV, Wang TC. Amino- and carboxy-terminal PEST domains mediate gastrin
stabilization of rat L-histidine decarboxylase isoforms. Mol Cell Biol 20: 4932 4947, 2000.

312. Esplugues JV, Barrachina MD, Calatayud S, Pique JM, Whittle BJ. Nitric oxide mediates the inhibition by interleukin-1 beta of pentagastrin-stimulated rat gastric acid
secretion. Br J Pharmacol 108: 9 10, 1993.

332. Force T, Bonventre JV, Flannery MR, Gorn AH, Yamin M, Goldring SR. A cloned
porcine renal calcitonin receptor couples to adenylyl cyclase and phospholipase C.
Am J Physiol Renal Fluid Electrolyte Physiol 262: F1110 F1115, 1992.

313. Fahrmann M, Kaufhold M, Pfeiffer AF, Seidler U. Protein kinase C-alpha attenuates
cholinergically stimulated gastric acid secretion of rabbit parietal cells. Br J Pharmacol
139: 545554, 2003.

333. Foresta C, Strapazzon G, De Toni L, Perilli L, Di Mambro A, Muciaccia B, Sartori L,


Selice R. Bone mineral density and testicular failure: evidence for a role of vitamin D
25-hydroxylase in human testis. J Clin Endocrinol Metab 96: E646 E652, 2011.

314. Fahrmann M, Kaufhold M, Rieg T, Seidler U. Different actions of protein kinase C


isoforms alpha and epsilon on gastric acid secretion. Br J Pharmacol 136: 938 946,
2002.

334. Forster IC, Hernando N, Biber J, Murer H. Proximal tubular handling of phosphate:
a molecular perspective. Kidney Int 70: 1548 1559, 2006.

315. Fahrmann M, Mohlig M, Schatz H, Pfeiffer A. Purification and characterization of a


Ca2/calmodulin-dependent protein kinase II from hog gastric mucosa using a protein-protein affinity chromatographic technique. Eur J Biochem 255: 516 525, 1998.
316. Fahrmann M, Pfeiffer A. Copurification of two holoenzyme-forming calcium/calmodulin-dependent protein kinase II isoforms as holoenzyme from porcine stomach. Arch Biochem Biophys 380: 151158, 2000.
317. Fasciotto BH, Gorr SU, DeFranco DJ, Levine MA, Cohn DV. Pancreastatin, a presumed product of chromogranin-A (secretory protein-I) processing, inhibits secretion from porcine parathyroid cells in culture. Endocrinology 125: 16171622, 1989.
318. Favus MJ, Angeid-Backman E. Effects of 1,25(OH)2D3 and calcium channel blockers
on cecal calcium transport in the rat. Am J Physiol Gastrointest Liver Physiol 248:
G676 G681, 1985.

335. Forte JG, Forte GM, Saltman P. K-stimulated phosphatase of microsomes from
gastric mucosa. J Cell Physiol 69: 293304, 1967.
336. Forte JG, Zhu L. Apical recycling of the gastric parietal cell H-K-ATPase. Annu Rev
Physiol 72: 273296, 2010.
337. Fox J, Green DT. Direct effects of calcium channel blockers on duodenal calcium
transport in vivo. Eur J Pharmacol 129: 159 164, 1986.
338. Fraser DR, Kodicek E. Regulation of 25-hydroxycholecalciferol-1-hydroxylase activity in kidney by parathyroid hormone. Nat New Biol 241: 163166, 1973.
339. Fraser DR, Kodicek E. Unique biosynthesis by kidney of a biological active vitamin D
metabolite. Nature 228: 764 766, 1970.
340. Freeman MW, Wiren KM, Rapoport A, Lazar M, Potts JT Jr, Kronenberg HM.
Consequences of amino-terminal deletions of preproparathyroid hormone signal
sequence. Mol Endocrinol 1: 628 638, 1987.

319. FDA. FDA Drug Safety Communication: possible increased risk of fractures of the
hip, wrist, and spine with the use of proton pump inhibitors Food and Drug Administration. http://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformation
forpatientsandproviders/ucm213206.htm [retrieved July 2011].

341. Freeman TC, Howard A, Bentsen BS, Legon S, Walters JR. Cellular and regional
expression of transcripts of the plasma membrane calcium pump PMCA1 in rabbit
intestine. Am J Physiol Gastrointest Liver Physiol 269: G126 G131, 1995.

320. Federico A, Dotti MT, Lore F, Nuti R. Cerebrotendinous xanthomatosis: pathophysiological study on bone metabolism. J Neurol Sci 115: 6770, 1993.

342. Friedman J, Au WY, Raisz LG. Responses of fetal rat bone to thyrocalcitonin in tissue
culture. Endocrinology 82: 149 156, 1968.

321. Feher JJ. Facilitated calcium diffusion by intestinal calcium-binding protein. Am J


Physiol Cell Physiol 244: C303C307, 1983.

343. Friedman J, Raisz LG. Thyrocalcitonin: inhibitor of bone resorption in tissue culture.
Science 150: 14651467, 1965.

246

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

309. Engqvist-Goldstein AE, Warren RA, Kessels MM, Keen JH, Heuser J, Drubin DG.
The actin-binding protein Hip1R associates with clathrin during early stages of
endocytosis and promotes clathrin assembly in vitro. J Cell Biol 154: 1209 1223,
2001.

322. Feher JJ, Fullmer CS, Wasserman RH. Role of facilitated diffusion of calcium by
calbindin in intestinal calcium absorption. Am J Physiol Cell Physiol 262: C517C526,
1992.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


344. Friedman PA. Basal and hormone-activated calcium absorption in mouse renal thick
ascending limbs. Am J Physiol Renal Fluid Electrolyte Physiol 254: F62F70, 1988.
345. Fries W, Rumenapf G, Schwille PO. Disturbances of mineral and bone metabolism
following gastric antrectomy in the rat. Bone Miner 19: 245256, 1992.
346. Friis-Hansen L, Schjerling CK, de la Cour CD, Hakanson R, Rehfeld JF. Characteristics of
gastrin controlled ECL cell specific gene expression. Regul Pept 140: 153161, 2007.
347. Friis-Hansen L, Sundler F, Li Y, Gillespie PJ, Saunders TL, Greenson JK, Owyang C,
Rehfeld JF, Samuelson LC. Impaired gastric acid secretion in gastrin-deficient mice.
Am J Physiol Gastrointest Liver Physiol 274: G561G568, 1998.
348. Frolik CA, Black EC, Cain RL, Satterwhite JH, Brown-Augsburger PL, Sato M, Hock
JM. Anabolic and catabolic bone effects of human parathyroid hormone (134) are
predicted by duration of hormone exposure. Bone 33: 372379, 2003.
349. Fu GK, Lin D, Zhang MY, Bikle DD, Shackleton CH, Miller WL, Portale AA. Cloning
of human 25-hydroxyvitamin D-1 alpha-hydroxylase and mutations causing vitamin
D-dependent rickets type 1. Mol Endocrinol 11: 19611970, 1997.

364. Garland FC, Garland CF, Gorham ED, Young JF. Geographic variation in breast
cancer mortality in the United States: a hypothesis involving exposure to solar radiation. Prev Med 19: 614 622, 1990.
365. Garner SC, Pi M, Tu Q, Quarles LD. Rickets in cation-sensing receptor-deficient
mice: an unexpected skeletal phenotype. Endocrinology 142: 3996 4005, 2001.
366. Garrett JE, Capuano IV, Hammerland LG, Hung BC, Brown EM, Hebert SC, Nemeth EF, Fuller F. Molecular cloning and functional expression of human parathyroid
calcium receptor cDNAs. J Biol Chem 270: 12919 12925, 1995.
367. Garrett JE, Tamir H, Kifor O, Simin RT, Rogers KV, Mithal A, Gagel RF, Brown EM.
Calcitonin-secreting cells of the thyroid express an extracellular calcium receptor
gene. Endocrinology 136: 52025211, 1995.
368. Garrick R, Ireland AW, Posen S. Bone abnormalities after gastric surgery. A prospective histological study. Ann Intern Med 75: 221225, 1971.
369. Garuti R, Croce MA, Piccinini L, Tiozzo R, Bertolini S, Calandra S. Functional analysis
of the promoter of human sterol 27-hydroxylase gene in HepG2 cells. Gene 283:
133143, 2002.
370. Gascon-Barre M, Demers C, Ghrab O, Theodoropoulos C, Lapointe R, Jones G,
Valiquette L, Menard D. Expression of CYP27A, a gene encoding a vitamin D-25
hydroxylase in human liver and kidney. Clin Endocrinol 54: 107115, 2001.

351. Fudge NJ, Kovacs CS. Physiological studies in heterozygous calcium sensing receptor
(CaSR) gene-ablated mice confirm that the CaSR regulates calcitonin release in vivo.
BMC Physiol 4: 5, 2004.

371. Geibel J, Sritharan K, Geibel R, Geibel P, Persing JS, Seeger A, Roepke TK, Deichstetter M, Prinz C, Cheng SX, Martin D, Hebert SC. Calcium-sensing receptor
abrogates secretagogue-induced increases in intestinal net fluid secretion by enhancing cyclic nucleotide destruction. Proc Natl Acad Sci USA 103: 9390 9397, 2006.

352. Fujii T, Takahashi Y, Ikari A, Morii M, Tabuchi Y, Tsukada K, Takeguchi N, Sakai H.


Functional association between K-Cl cotransporter-4 and H,K-ATPase in the
apical canalicular membrane of gastric parietal cells. J Biol Chem 284: 619 629, 2009.

372. Geibel JP, Hebert SC. The functions and roles of the extracellular Ca2-sensing
receptor along the gastrointestinal tract. Annu Rev Physiol 71: 205217, 2009.

353. Fujita A, Horio Y, Higashi K, Mouri T, Hata F, Takeguchi N, Kurachi Y. Specific


localization of an inwardly rectifying K() channel, Kir4.1, at the apical membrane of
rat gastric parietal cells; its possible involvement in K() recycling for the H()K()-pump. J Physiol 540: 8592, 2002.
354. Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, Uchiyama Y, Yamamoto Y,
Wada T, Kojima T, Yokozaki H, Yamashita T, Kato S, Sawada N, Chiba H. Tight
junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2 absorption between enterocytes. Mol Biol Cell 19: 19121921, 2008.
355. Fukumoto K, Nakahara K, Katayama T, Miyazatao M, Kangawa K, Murakami N.
Synergistic action of gastrin and ghrelin on gastric acid secretion in rats. Biochem
Biophys Res Commun 374: 60 63, 2008.
356. Fukushima M, Suzuki Y, Tohira Y, Nishii Y, Suzuki M. 25-Hydroxylation of 1alphahydroxyvitamin D3 in vivo and in perfused rat liver. FEBS Lett 65: 211214, 1976.
357. Fukushima N, Hanada R, Teranishi H, Fukue Y, Tachibana T, Ishikawa H, Takeda S,
Takeuchi Y, Fukumoto S, Kangawa K, Nagata K, Kojima M. Ghrelin directly regulates
bone formation. J Bone Miner Res 20: 790 798, 2005.
358. Fullmer CS, Wasserman RH. Isolation and partial characterization of intestinal calcium-binding proteins from the cow, pig, horse, guinea pig, and chick. Biochim Biophys
Acta 393: 134 142, 1975.
359. Gadelle D, Raibaud P, Sacquet E. beta-Glucuronidase activities of intestinal bacteria
determined both in vitro and in vivo in gnotobiotic rats. Appl Environ Microbiol 49:
682 685, 1985.

373. Geibel JP, Wagner CA, Caroppo R, Qureshi I, Gloeckner J, Manuelidis L, Kirchhoff P,
Radebold K. The stomach divalent ion-sensing receptor scar is a modulator of gastric
acid secretion. J Biol Chem 276: 39549 39552, 2001.
374. Gerber JG, Payne NA. Secretin inhibits canine gastric acid secretion in response to
pentagastrin by modulating gastric histamine release. J Pharmacol Exp Ther 279:
718 723, 1996.
375. Gerbino A, Ruder WC, Curci S, Pozzan T, Zaccolo M, Hofer AM. Termination of
cAMP signals by Ca2 and Gi via extracellular Ca2 sensors: a link to intracellular
Ca2 oscillations. J Cell Biol 171: 303312, 2005.
376. Gerhard M, Neumayer N, Presecan-Siedel E, Zanner R, Lengyel E, Cramer T,
Hocker M, Prinz C. Gastrin induces expression and promoter activity of the vesicular
monoamine transporter subtype 2. Endocrinology 142: 36633672, 2001.
377. Germain N, Galusca B, Le Roux CW, Bossu C, Ghatei MA, Lang F, Bloom SR, Estour
B. Constitutional thinness and lean anorexia nervosa display opposite concentrations
of peptide YY, glucagon-like peptide 1, ghrelin, and leptin. Am J Clin Nutr 85: 967
971, 2007.
378. Gertner JM, Lilburn M, Domenech M. 25-Hydroxycholecalciferol absorption in steatorrhoea and postgastrectomy osteomalacia. Br Med J 1: 1310 1312, 1977.
379. Gesek FA, Friedman PA. On the mechanism of parathyroid hormone stimulation of
calcium uptake by mouse distal convoluted tubule cells. J Clin Invest 90: 749 758,
1992.

360. Gama L, Baxendale-Cox LM, Breitwieser GE. Ca2-sensing receptors in intestinal


epithelium. Am J Physiol Cell Physiol 273: C1168 C1175, 1997.

380. Gesty-Palmer D, Chen M, Reiter E, Ahn S, Nelson CD, Wang S, Eckhardt AE, Cowan
CL, Spurney RF, Luttrell LM, Lefkowitz RJ. Distinct beta-arrestin- and G proteindependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation. J Biol Chem 281: 10856 10864, 2006.

361. Gao XH, Dwivedi PP, Omdahl JL, Morris HA, May BK. Calcitonin stimulates expression of the rat 25-hydroxyvitamin D3-24-hydroxylase (CYP24) promoter in HEK293 cells expressing calcitonin receptor: identification of signaling pathways. J Mol
Endocrinol 32: 8798, 2004.

381. Gesty-Palmer D, Flannery P, Yuan L, Corsino L, Spurney R, Lefkowitz RJ, Luttrell


LM. A beta-arrestin-biased agonist of the parathyroid hormone receptor (PTH1R)
promotes bone formation independent of G protein activation. Sci Transl Med 1:
1ra1, 2009.

362. Garfia B, Canadillas S, Canalejo A, Luque F, Siendones E, Quesada M, Almaden Y,


Aguilera-Tejero E, Rodriguez M. Regulation of parathyroid vitamin D receptor expression by extracellular calcium. J Am Soc Nephrol 13: 29452952, 2002.

382. Ghijsen WE, De Jong MD, Van Os CH. Kinetic properties of Na/Ca2 exchange in
basolateral plasma membranes of rat small intestine. Biochim Biophys Acta 730: 85
94, 1983.

363. Garland C, Shekelle RB, Barrett-Connor E, Criqui MH, Rossof AH, Paul O. Dietary
vitamin D and calcium and risk of colorectal cancer: a 19-year prospective study in
men. Lancet 1: 307309, 1985.

383. Gilardi F, Viviani B, Galmozzi A, Boraso M, Bartesaghi S, Torri A, Caruso D, Crestani


M, Marinovich M, de Fabiani E. Expression of sterol 27-hydroxylase in glial cells and
its regulation by liver X receptor signaling. Neuroscience 164: 530 540, 2009.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

247

Downloaded from on December 8, 2014

350. Fu Q, Jilka RL, Manolagas SC, OBrien CA. Parathyroid hormone stimulates receptor
activator of NFkappa B ligand and inhibits osteoprotegerin expression via protein
kinase A activation of cAMP-response element-binding protein. J Biol Chem 277:
48868 48875, 2002.

SASCHA KOPIC AND JOHN P. GEIBEL


384. Gisbert JP, Gonzalez L, Calvet X, Roque M, Gabriel R, Pajares JM. Proton pump
inhibitors versus H2-antagonists: a meta-analysis of their efficacy in treating bleeding
peptic ulcer. Aliment Pharmacol Ther 15: 917926, 2001.
385. Goldblatt H, Soames KM. Studies on the fat-soluble growth-promoting factor.
Biochem J 17: 446, 1923.
386. Goldenring JR, Shen KR, Vaughan HD, Modlin IM. Identification of a small GTPbinding protein, Rab25, expressed in the gastrointestinal mucosa, kidney, and lung. J
Biol Chem 268: 18419 18422, 1993.
387. Goldenring JR, Soroka CJ, Shen KR, Tang LH, Rodriguez W, Vaughan HD, Stoch SA,
Modlin IM. Enrichment of rab11, a small GTP-binding protein, in gastric parietal cells.
Am J Physiol Gastrointest Liver Physiol 267: G187G194, 1994.
388. Gonnelli S, Caffarelli C, Del Santo K, Cadirni A, Guerriero C, Lucani B, Franci B, Nuti
R. The relationship of ghrelin and adiponectin with bone mineral density and bone
turnover markers in elderly men. Calcif Tissue Int 83: 55 60, 2008.
389. Gorn AH, Rudolph SM, Flannery MR, Morton CC, Weremowicz S, Wang TZ, Krane
SM, Goldring SR. Expression of two human skeletal calcitonin receptor isoforms
cloned from a giant cell tumor of bone. The first intracellular domain modulates
ligand binding and signal transduction. J Clin Invest 95: 2680 2691, 1995.
390. Goss SL, Lemons KA, Kerstetter JE, Bogner RH. Determination of calcium salt
solubility with changes in pH and PCO2, simulating varying gastrointestinal environments. J Pharm Pharmacol 59: 14851492, 2007.
391. Grahammer F, Herling AW, Lang HJ, Schmitt-Graff A, Wittekindt OH, Nitschke R,
Bleich M, Barhanin J, Warth R. The cardiac K channel KCNQ1 is essential for gastric
acid secretion. Gastroenterology 120: 13631371, 2001.

393. Gray RW, Omdahl JL, Ghazarian JG, DeLuca HF Hydroxycholecalciferol-1-hydroxylase. Subcellular location and properties. J Biol Chem 247: 7528 7532, 1972.
394. Gray SL, Lacroix AZ, Larson J, Robbins J, Cauley JA, Manson JE, Chen Z. Proton
pump inhibitor use, hip fracture, and change in bone mineral density in postmenopausal women: results from the womens health initiative. Arch Internal Med 170:
765771, 2010.
395. Graziani G, Badalamenti S, Como G, Gallieni M, Finazzi S, Angelini C, Brancaccio D,
Ponticelli C. Calcium and phosphate plasma levels in dialysis patients after dietary
Ca-P overload: Role of gastric acid secretion. Nephron 91: 474 479, 2002.

406. Haddad JG, Hillman L, Rojanasathit S. Human serum binding capacity and affinity for
25-hydroxyergocalciferol and 25-hydroxycholecalciferol. J Clin Endocrinol Metab 43:
86 91, 1976.
407. Haddad JG, Jennings AS, Aw TC. Vitamin D uptake and metabolism by perfused rat
liver: influences of carrier proteins. Endocrinology 123: 498 504, 1988.
408. Haddad JG Jr, Walgate J. 25-Hydroxyvitamin D transport in human plasma. Isolation
and partial characterization of calcifidiol-binding protein. J Biol Chem 251: 4803
4809, 1976.
409. Haddad JG Jr, Walgate J. Radioimmunoassay of the binding protein for vitamin D and
its metabolites in human serum: concentrations in normal subjects and patients with
disorders of mineral homeostasis. J Clin Invest 58: 12171222, 1976.
410. Hakanson R, Blom H, Carlsson E, Larsson H, Ryberg B, Sundler F. Hypergastrinaemia produces trophic effects in stomach but not in pancreas and intestines. Regul
Pept 13: 225233, 1986.
411. Hakanson R, Ding XQ, Norlen P, Chen D. Circulating pancreastatin is a marker for
the enterochromaffin-like cells of the rat stomach. Gastroenterology 108: 1445
1452, 1995.
412. Hakanson R, Liedberg G. The role of endogenous gastrin in the activation of gastric
histidine decarboxylase in the rat. Effect of antrectomy and vagal denervation. Eur J
Pharmacol 12: 94 103, 1970.
413. Hakanson R, Owman C. Concomitant histochemical demonstration of histamine
and catecholamines in enterochromaffin-like cells of gastric mucosa. Life Sci 6: 759
766, 1967.
414. Hammer RA, Fernandez C, Ertan A, Arimura A. Anesthetic dependence of the
inhibitory effect of neurotensin on pentagastrin-stimulated acid secretion in rats. A
possible role for somatostatin. Life Sci 48: 333339, 1991.
415. Handlogten ME, Huang C, Shiraishi N, Awata H, Miller RT. The Ca2-sensing receptor activates cytosolic phospholipase A2 via a Gqalpha-dependent ERK-independent pathway. J Biol Chem 276: 1394113948, 2001.
416. Handlogten ME, Shiraishi N, Awata H, Huang C, Miller RT. Extracellular Ca2sensing receptor is a promiscuous divalent cation sensor that responds to lead. Am J
Physiol Renal Physiol 279: F1083F1091, 2000.
417. Hanley DA, Ayer LM. Calcium-dependent release of carboxyl-terminal fragments of
parathyroid hormone by hyperplastic human parathyroid tissue in vitro. J Clin Endocrinol Metab 63: 10751079, 1986.

396. Graziani G, Como G, Badalamenti S, Finazzi S, Malesci A, Gallieni M, Brancaccio D,


Ponticelli C. Effect of gastric acid secretion on intestinal phosphate and calcium
absorption in normal subjects. Nephrology Dialysis Transplantation 10: 1376 1380,
1995.

418. Hanley DA, Takatsuki K, Sultan JM, Schneider AB, Sherwood LM. Direct release of
parathyroid hormone fragments from functioning bovine parathyroid glands in vitro.
J Clin Invest 62: 12471254, 1978.

397. Green T, Dockray GJ. Characterization of the peptidergic afferent innervation of the
stomach in the rat, mouse and guinea-pig. Neuroscience 25: 181193, 1988.

419. Hansen CP. The pharmacokinetics and pharmacodynamics of progastrin-derived


peptides. Dan Med Bull 50: 310 319, 2003.

398. Gregory DH, Van Uelft R. Calcium absorption following gastric resection. Am J
Gastroenterol 57: 34 40, 1972.

420. Hansen CP, Goetze JP, Stadil F, Rehfeld JF. Excretion of progastrin products in
human urine. Am J Physiol Gastrointest Liver Physiol 276: G985G992, 1999.

399. Gregory RA, Tracy HJ. The constitution and properties of two gastrins extracted
from hog antral mucosa. Gut 5: 103114, 1964.

421. Hansen KE, Jones AN, Lindstrom MJ, Davis LA, Ziegler TE, Penniston KL, Alvig AL,
Shafer MM. Do proton pump inhibitors decrease calcium absorption? J Bone Miner
Res 25: 2510 2519, 2010.

400. Grisso JA, Kelsey JL, OBrien LA, Miles CG, Sidney S, Maislin G, LaPann K, Moritz D,
Peters B. Risk factors for hip fracture in men. Am J Epidemiol 145: 786 793, 1997.
401. Gunness-Hey M, Hock JM. Increased trabecular bone mass in rats treated with
human synthetic parathyroid hormone. Metab Bone Dis Relat Res 5: 177181, 1984.
402. Guo YD, Strugnell S, Back DW, Jones G. Transfected human liver cytochrome P-450
hydroxylates vitamin D analogs at different side-chain positions. Proc Natl Acad Sci
USA 90: 8668 8672, 1993.
403. Gupta RP, Patrick K, Bell NH. Mutational analysis of CYP27A1: assessment of 27hydroxylation of cholesterol and 25-hydroxylation of vitamin D. Metabolism 56:
1248 1255, 2007.
404. Gyomorey K, Yeger H, Ackerley C, Garami E, Bear CE. Expression of the chloride
channel ClC-2 in the murine small intestine epithelium. Am J Physiol Cell Physiol (could
not determine journal name) 279: 17871794, 2000.

248

422. Hanzlik RP, Fowler SC, Fisher DH. Relative bioavailability of calcium from calcium
formate, calcium citrate, and calcium carbonate. J Pharmacol Exp Ther 313: 1217
1222, 2005.
423. Hardy P, Sechet A, Hottelart C, Oprisiu R, Abighanem O, Said S, Rasombololona M,
Brazier M, Moriniere P, Achard JM, Pruna A, Fournier A. Inhibition of gastric secretion by omeprazole and efficiency of calcium carbonate on the control of hyperphosphatemia in patients on chronic hemodialysis. Artificial Organs 22: 569 573, 1998.
424. Harmeyer J, Deluca HF. Calcium-binding protein and calcium absorption after vitamin D administration. Arch Biochem Biophys 133: 247254, 1969.
425. Hartmann M, Theiss U, Huber R, Luhmann R, Bliesath H, Wurst W, Lucker PW.
Twenty-four-hour intragastric pH profiles and pharmacokinetics following single and
repeated oral administration of the proton pump inhibitor pantoprazole in comparison to omeprazole. Aliment Pharmacol Ther 10: 359 366, 1996.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

392. Grauschopf U, Lilie H, Honold K, Wozny M, Reusch D, Esswein A, Schafer W,


Rucknagel KP, Rudolph R. The N-terminal fragment of human parathyroid hormone
receptor 1 constitutes a hormone binding domain and reveals a distinct disulfide
pattern. Biochemistry 39: 8878 8887, 2000.

405. Habener JF, Singer FR, Deftos LJ, Neer RM, Potts JT Jr. Explanation for unusual
potency of salmon calcitonin. Nat New Biol 232: 9192, 1971.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


426. Hasebe K, Horie S, Komasaka M, Yano S, Watanabe K. Stimulatory effects of nitric
oxide donors on gastric acid secretion in isolated mouse stomach. Eur J Pharmacol
420: 159 164, 2001.
427. Hashizume Y, Waguri S, Watanabe T, Kominami E, Uchiyama Y. Cysteine proteinases in rat parathyroid cells with special reference to their correlation with parathyroid hormone (PTH) in storage granules. J Histochem Cytochem 41: 273282, 1993.

448. Hess AF, Weinstock M. Antirachitic properties imparted to inert fluids by ultraviolet
irradiation. Proc Soc Exp Biol Med 22: 6 7, 1924.
449. Hess AF, Weinstock M. Antirachitic properties imparted to lettuce and to growing
wheat by ultraviolet irradiation. Proc Soc Exp Biol Med 22: 5, 1924.
450. Hess AF, Weinstock M. A further report on imparting antirachitic properties to inert
substances by ultra-violet irradiation. J Biol Chem 63: 297307, 1925.

428. Hattersley G and Chambers TJ. Calcitonin receptors as markers for osteoclastic
differentiation: correlation between generation of bone-resorptive cells and cells
that express calcitonin receptors in mouse bone marrow cultures. Endocrinology
125: 1606 1612, 1989.

451. Hewison M, Burke F, Evans KN, Lammas DA, Sansom DM, Liu P, Modlin RL, Adams
JS. Extra-renal 25-hydroxyvitamin D3-1alpha-hydroxylase in human health and disease. J Steroid Biochem Mol Biol 103: 316 321, 2007.

429. Haussler MR, Myrtle JF, Norman AW. The association of a metabolite of vitamin D3
with intestinal mucosa chromatin in vivo. J Biol Chem 243: 4055 4064, 1968.

452. Hewison M, Freeman L, Hughes SV, Evans KN, Bland R, Eliopoulos AG, Kilby MD,
Moss PA, Chakraverty R. Differential regulation of vitamin D receptor and its ligand
in human monocyte-derived dendritic cells. J Immunol 170: 53825390, 2003.

430. Haussler MR, Norman AW. Chromosomal receptor for a vitamin D metabolite. Proc
Natl Acad Sci USA 62: 155162, 1969.
431. He W, Liu W, Chew CS, Baker SS, Baker RD, Forte JG, Zhu L. Acid secretionassociated translocation of KCNJ15 in gastric parietal cells. Am J Physiol Gastrointest
Liver Physiol 301: G591G600, 2011.
432. Heaney RP, Dowell MS, Bierman J, Hale CA, Bendich A. Absorbability and cost
effectiveness in calcium supplementation. J Am Coll Nutr 20: 239 246, 2001.
433. Heaney RP, Recker RR, Weaver CM. Absorbability of calcium sources: the limited
role of solubility. Calcified Tissue Int 46: 300 304, 1990.
434. Hebert SC, Brown EM, Harris HW. Role of the Ca2-sensing receptor in divalent
mineral ion homeostasis. J Exp Biol 200: 295302, 1997.

436. Heitzmann D, Koren V, Wagner M, Sterner C, Reichold M, Tegtmeier I, Volk T,


Warth R. KCNE beta subunits determine pH sensitivity of KCNQ1 potassium channels. Cell Physiol Biochem 19: 2132, 2007.
437. Helander KG, Bamberg K, Sachs G, Melle D, Helander HF. Localization of mRNA for
the muscarinic M1 receptor in rat stomach. Biochim Biophys Acta 1312: 158 162,
1996.
438. Heller HJ, Greer LG, Haynes SD, Poindexter JR, Pak CY. Pharmacokinetic and
pharmacodynamic comparison of two calcium supplements in postmenopausal
women. J Clin Pharmacol 40: 12371244, 2000.
439. Heller HJ, Stewart A, Haynes S, Pak CY. Pharmacokinetics of calcium absorption
from two commercial calcium supplements. J Clin Pharmacol 39: 11511154, 1999.
440. Hendy GN, Kronenberg HM, Potts JT Jr, Rich A. Nucleotide sequence of cloned
cDNAs encoding human preproparathyroid hormone. Proc Natl Acad Sci USA 78:
73657369, 1981.
441. Hennessy JF, Wells SA Jr, Ontjes DA, Cooper CW. A comparison of pentagastrin
injection and calcium infusion as provocative agents for the detection of medullary
carcinoma of the thyroid. J Clin Endocrinol Metab 39: 487 495, 1974.
442. Henry HL. Parathyroid hormone modulation of 25-hydroxyvitamin D3 metabolism
by cultured chick kidney cells is mimicked and enhanced by forskolin. Endocrinology
116: 503510, 1985.
443. Henry HL. Regulation of the hydroxylation of 25-hydroxyvitamin D3 in vivo and in
primary cultures of chick kidney cells. J Biol Chem 254: 27222729, 1979.
444. Herberth J, Fahrleitner-Pammer A, Obermayer-Pietsch B, Krisper P, Holzer H,
Malluche HH, Dobnig H. Changes in total parathyroid hormone (PTH), PTH-(1 84)
and large C-PTH fragments in different stages of chronic kidney disease. Clin Nephrol
65: 328 334, 2006.
445. Herlong HF, Recker RR, Maddrey WC. Bone disease in primary biliary cirrhosis:
histologic features and response to 25-hydroxyvitamin D. Gastroenterology 83: 103
108, 1982.
446. Hess AF. The prevention and cure of rickets by sunlight. Am J Public Health 12:
104 107, 1922.
447. Hess AF, Unger LJ. The cure of infantile rickets by sunlight. J Am Med Assoc 77: 39,
1921.

454. Heynen G, Brassine A, Daubresse JC, Ligny G, Kanis JA, Gaspar S, Franchimont P.
Lack of clinical and physiological relationship between gastrin and calcitonin in man.
Eur J Clin Invest 11: 331335, 1981.
455. Higham A, Noble P, Thompson DG, Dockray GJ. Increased sensitivity of gastrin cells
to gastric distension following antral denervation in the rat. J Physiol 503: 169 175,
1997.
456. Hildebrand P, Lehmann FS, Ketterer S, Christ AD, Stingelin T, Beltinger J, Gibbons
AH, Coy DH, Calam J, Larsen F, Beglinger C. Regulation of gastric function by
endogenous gastrin releasing peptide in humans: studies with a specific gastrin releasing peptide receptor antagonist. Gut 49: 2328, 2001.
457. Hildmann B, Schmidt A, Murer H. Ca2-transport across basal-lateral plasma membranes from rat small intestinal epithelial cells. J Membr Biol 65: 55 62, 1982.
458. Hill LF, Van den Berg CJ, Mawer EB. Vitamin D metabolism in experimental uraemia:
effects on intestinal transport 45Ca and on formation of 1,25-dihydroxycholecalciferol in rat. Nat New Biol 232: 189 191, 1971.
459. Hills DM, Gerskowitch VP, Roberts SP, Welsh NJ, Shankley NP, Black JW. Pharmacological analysis of the CCKB/gastrin receptors mediating pentagastrin-stimulated
gastric acid secretion in the isolated stomach of the immature rat. Br J Pharmacol 119:
14011410, 1996.
460. Hinata M, Yamamura H, Li L, Watanabe Y, Watano T, Imaizumi Y, Kimura J. Stoichiometry of Na-Ca2 exchange is 3:1 in guinea-pig ventricular myocytes. J Physiol
545: 453 461, 2002.
461. Hirnet D, Olausson J, Fecher-Trost C, Bodding M, Nastainczyk W, Wissenbach U,
Flockerzi V, Freichel M. The TRPV6 gene, cDNA and protein. Cell Calcium 33:
509 518, 2003.
462. Hirsch PF, Baruch H. Is calcitonin an important physiological substance? Endocrine
21: 201208, 2003.
463. Hirsch PF, Gauthier GF, Munson PL. Thyroid hypocalcemic principle and recurrent
laryngeal nerve injury as factors affecting the response to parathyroidectomy in rats.
Endocrinology 73: 244 252, 1963.
464. Hirschfeld J. Immune-electrophoretic demonstration of qualitative differences in
human sera and their relation to the haptoglobins. Acta Pathol Microbiol Scand 47:
160 168, 1959.
465. Hirschfeld J. Individual precipitation patterns of normal rabbit sera. A preliminary
report. Acta Pathol Microbiol Scand 46: 229 238, 1959.
466. Hirschowitz BI, Fong J, Molina E. Effects of pirenzepine and atropine on vagal and
cholinergic gastric secretion and gastrin release and on heart rate in the dog. J
Pharmacol Exp Ther 225: 263268, 1983.
467. Hjalm G, MacLeod RJ, Kifor O, Chattopadhyay N, Brown EM. Filamin-A binds to the
carboxyl-terminal tail of the calcium-sensing receptor, an interaction that participates in CaR-mediated activation of mitogen-activated protein kinase. J Biol Chem
276: 34880 34887, 2001.
468. Hoag HM, Martel J, Gauthier C, Tenenhouse HS. Effects of Npt2 gene ablation and
low-phosphate diet on renal Na/phosphate cotransport and cotransporter gene
expression. J Clin Invest 104: 679 686, 1999.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

249

Downloaded from on December 8, 2014

435. Heersche JN, Marcus R, Aurbach GD. Calcitonin and the formation of 3=,5=-AMP in
bone and kidney. Endocrinology 94: 241247, 1974.

453. Hewison M, Zehnder D, Chakraverty R, Adams JS. Vitamin D and barrier function: a
novel role for extra-renal 1 alpha-hydroxylase. Mol Cell Endocrinol 215: 3138, 2004.

SASCHA KOPIC AND JOHN P. GEIBEL


469. Hock JM, Gera I. Effects of continuous and intermittent administration and inhibition
of resorption on the anabolic response of bone to parathyroid hormone. J Bone Miner
Res 7: 6572, 1992.

droxyvitamin D(3) [1,24-(OH)(2)D(3)], 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] on selected vitamin D-regulated events in the rat. Biochem Pharmacol
60: 701708, 2000.

470. Hocker M, Henihan RJ, Rosewicz S, Riecken EO, Zhang Z, Koh TJ, Wang TC. Gastrin
and phorbol 12-myristate 13-acetate regulate the human histidine decarboxylase
promoter through Raf-dependent activation of extracellular signal-regulated kinaserelated signaling pathways in gastric cancer cells. J Biol Chem 272: 2701527024,
1997.

490. Horsting M, DeLuca HF. In vitro production of 25-hydroxycholecalciferol. Biochem


Biophys Res Commun 36: 251256, 1969.
491. Hosoda H, Kojima M, Matsuo H, Kangawa K. Ghrelin and des-acyl ghrelin: two major
forms of rat ghrelin peptide in gastrointestinal tissue. Biochem Biophys Res Commun
279: 909 913, 2000.

471. Hocker M, John M, Anagnostopoulos J, Buhr HJ, Solimena M, Gasnier B, Henry JP,
Wang TC, Wiedenmann B. Molecular dissection of regulated secretory pathways in
human gastric enterochromaffin-like cells: an immunohistochemical analysis. Histochem Cell Biol 112: 205214, 1999.

492. Howard A, Legon S, Walters JR. Human and rat intestinal plasma membrane calcium
pump isoforms. Am J Physiol Gastrointest Liver Physiol 265: G917G925, 1993.

472. Hocker M, Zhang Z, Fenstermacher DA, Tagerud S, Chulak M, Joseph D, Wang TC.
Rat histidine decarboxylase promoter is regulated by gastrin through a protein kinase
C pathway. Am J Physiol Gastrointest Liver Physiol 270: G619 G633, 1996.

493. Huan J, Olgaard K, Nielsen LB, Lewin E. Parathyroid hormone 7 84 induces hypocalcemia and inhibits the parathyroid hormone 1 84 secretory response to hypocalcemia in rats with intact parathyroid glands. J Am Soc Nephrol 17: 19231930,
2006.

473. Hoenderop JG, Nilius B, Bindels RJ. Calcium absorption across epithelia. Physiol Rev
85: 373 422, 2005.
474. Hoenderop JG, van der Kemp AW, Hartog A, van de Graaf SF, van Os CH, Willems
PH, Bindels RJ. Molecular identification of the apical Ca2 channel in 1,25-dihydroxyvitamin D3-responsive epithelia. J Biol Chem 274: 8375 8378, 1999.
475. Hoenderop JG, Vennekens R, Muller D, Prenen J, Droogmans G, Bindels RJ, Nilius B.
Function and expression of the epithelial Ca2 channel family: comparison of mammalian ECaC1 and 2. J Physiol 537: 747761, 2001.

477. Hohne-Zell B, Galler A, Schepp W, Gratzl M, Prinz C. Functional importance of


synaptobrevin and SNAP-25 during exocytosis of histamine by rat gastric enterochromaffin-like cells. Endocrinology 138: 5518 5526, 1997.
478. Holick MF, Chen TC, Lu Z, Sauter E. Vitamin D and skin physiology: a D-lightful
story. J Bone Miner Res 22 Suppl 2: V28 33, 2007.
479. Holick MF, MacLaughlin JA, Clark MB, Holick SA, Potts JT Jr, Anderson RR, Blank IH,
Parrish JA, Elias P. Photosynthesis of previtamin D3 in human skin and the physiologic
consequences. Science 210: 203205, 1980.
480. Holick MF, Schnoes HK, DeLuca HF, Suda T, Cousins RJ. Isolation and identification
of 1,25-dihydroxycholecalciferol. A metabolite of vitamin D active in intestine. Biochemistry 10: 2799 2804, 1971.
481. Hollande F, Gusdinar T, Bali JP, Magous R. Neurohormonal regulation of histamine
release from isolated rabbit fundic mucosal cells. Agents Actions 38: 149 157, 1993.
482. Hollande F, Imdahl A, Mantamadiotis T, Ciccotosto GD, Shulkes A, Baldwin GS.
Glycine-extended gastrin acts as an autocrine growth factor in a nontransformed
colon cell line. Gastroenterology 113: 1576 1588, 1997.
483. Hollander D, Muralidhara KS, Zimmerman A. Vitamin D-3 intestinal absorption in
vivo: influence of fatty acids, bile salts, and perfusate pH on absorption. Gut 19:
267272, 1978.
484. Hollander D, Truscott TC. Mechanism and site of small intestinal uptake of vitamin
D3 in pharmacological concentrations. Am J Clin Nutr 29: 970 975, 1976.

495. Huang C, Hujer KM, Wu Z, Miller RT. The Ca2-sensing receptor couples to
G12/13 to activate phospholipase D in Madin-Darby canine kidney cells. Am J Physiol
Cell Physiol 286: C22C30, 2004.
496. Huang C, Wu Z, Hujer KM, Miller RT. Silencing of filamin A gene expression inhibits
Ca2-sensing receptor signaling. FEBS Lett 580: 17951800, 2006.
497. Huang JC, Sakata T, Pfleger LL, Bencsik M, Halloran BP, Bikle DD, Nissenson RA.
PTH differentially regulates expression of RANKL and OPG. J Bone Miner Res 19:
235244, 2004.
498. Huang Y, Niwa J, Sobue G, Breitwieser GE. Calcium-sensing receptor ubiquitination
and degradation mediated by the E3 ubiquitin ligase dorfin. J Biol Chem 281: 11610
11617, 2006.
499. Huang Y, Zhou Y, Castiblanco A, Yang W, Brown EM, Yang JJ. Multiple Ca2-binding
sites in the extracellular domain of the Ca2-sensing receptor corresponding to
cooperative Ca2 response. Biochemistry 48: 388 398, 2009.
500. Huang Y, Zhou Y, Yang W, Butters R, Lee HW, Li S, Castiblanco A, Brown EM, Yang
JJ. Identification and dissection of Ca2-binding sites in the extracellular domain of
Ca2-sensing receptor. J Biol Chem 282: 19000 19010, 2007.
501. Huda MS, Durham BH, Wong SP, Dovey TM, McCulloch P, Kerrigan D, Pinkney JH,
Fraser WD, Wilding JP. Lack of an acute effect of ghrelin on markers of bone
turnover in healthy controls and post-gastrectomy subjects. Bone 41: 406 413,
2007.
502. Hughes DA, Tang K, Strotmann R, Schoneberg T, Prenen J, Nilius B, Stoneking M.
Parallel selection on TRPV6 in human populations. PLoS ONE 3: e1686, 2008.
503. Huhtakangas JA, Olivera CJ, Bishop JE, Zanello LP, Norman AW. The vitamin D
receptor is present in caveolae-enriched plasma membranes and binds 1
alpha,25(OH)2-vitamin D3 in vivo and in vitro. Mol Endocrinol 18: 2660 2671, 2004.
504. Huldschinsky K. Heilung von Rachitis durch knstliche Hhensonne. Deutsche Med
Wochenschrift 45: 712, 1919.

485. Holst MC, Kelly JB, Powley TL. Vagal preganglionic projections to the enteric nervous system characterized with Phaseolus vulgaris-leucoagglutinin. J Comp Neurol
381: 81100, 1997.

505. Hume EM, Smith HH. The effect of irradiation of the environment with ultra-violet
light upon the growth and calcification of rats, fed on a diet deficient in fat-soluble
vitamins: the part played by irradiated sawdust. Biochem J 18: 1334 1345, 1924.

486. Holst Pedersen J, Skov Olsen P, Kirkegaard P. Effect of neurotensin and neurotensin
fragments on gastric acid secretion in man. Regul Pept 15: 77 86, 1986.

506. Humphries TJ, Merritt GJ. Review article: drug interactions with agents used to treat
acid-related diseases. Aliment Pharmacol Ther 13 Suppl 3: 18 26, 1999.

487. Holtrop ME, Raisz LG, Simmons HA. The effects of parathyroid hormone, colchicine, and calcitonin on the ultrastructure and the activity of osteoclasts in organ
culture. J Cell Biol 60: 346 355, 1974.

507. Hunyady B, Mezey E, Pacak K, Harta G, Palkovits M. Distribution of muscarinic


receptor mRNAs in the stomachs of normal or immobilized rats. Inflammopharmacology 4: 399 413, 1996.

488. Hori K, Takahashi Y, Horikawa N, Furukawa T, Tsukada K, Takeguchi N, Sakai H. Is


the ClC-2 chloride channel involved in the Cl secretory mechanism of gastric
parietal cells? FEBS Lett 575: 105108, 2004.

508. Ichikawa F, Sato K, Nanjo M, Nishii Y, Shinki T, Takahashi N, Suda T. Mouse primary
osteoblasts express vitamin D3 25-hydroxylase mRNA and convert 1 alpha-hydroxyvitamin D3 into 1 alpha,25-dihydroxyvitamin D3. Bone 16: 129 135, 1995.

489. Horst R, Prapong S, Reinhardt T, Koszewski N, Knutson J, Bishop C. Comparison of


the relative effects of 1,24-dihydroxyvitamin D(2) [1, 24-(OH)(2)D(2)], 1,24-dihy-

509. Imai M. Calcium transport across the rabbit thick ascending limb of Henles loop
perfused in vitro. Pflgers Arch 374: 255263, 1978.

250

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

476. Hoenderop JG, Voets T, Hoefs S, Weidema F, Prenen J, Nilius B, Bindels RJ. Homoand heterotetrameric architecture of the epithelial Ca2 channels TRPV5 and
TRPV6. EMBO J 22: 776 785, 2003.

494. Huang C, Handlogten ME, Miller RT. Parallel activation of phosphatidylinositol 4-kinase and phospholipase C by the extracellular calcium-sensing receptor. J Biol Chem
277: 2029320300, 2002.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


510. Imanishi Y, Kawata T, Kenko T, Wada M, Nagano N, Miki T, Arnold A, Inaba M.
Cinacalcet HCl suppresses Cyclin D1 oncogene-derived parathyroid cell proliferation in a murine model for primary hyperparathyroidism. Calcif Tissue Int 89: 29 35,
2011.
511. Imawari M, Kozawa K, Akanuma Y, Koizumi S, Itakura H, Kosaka K. Serum 25hydroxyvitamin D and vitamin D-binding protein levels and mineral metabolism after
partial and total gastrectomy. Gastroenterology 79: 255258, 1980.
512. IMSHealth. Top Products by U.S. Spending 2010 http://www.imshealth.com/
deployedfiles/ims/Global/Content/Corporate/Press%20Room/Top-line%20
Market%20Data/2010%20Top-line%20Market%20Data/2010_Top_Products_by_
Sales.pdf [retrieved November 2011].
513. Inoue Y, Segawa H, Kaneko I, Yamanaka S, Kusano K, Kawakami E, Furutani J, Ito M,
Kuwahata M, Saito H, Fukushima N, Kato S, Kanayama HO, Miyamoto K. Role of the
vitamin D receptor in FGF23 action on phosphate metabolism. Biochem J 390: 325
331, 2005.
514. Irving AD, Smith G, Catto GR. Does metabolic bone disease follow truncal vagotomy
and gastrojejunostomy? Br Med J 287: 1516, 1983.
515. Isenberg JI, Walsh JH, Grossman MI. Zollinger-Ellison syndrome. Gastroenterology
65: 140 165, 1973.
516. Ishizaki T, Horai Y. Review article: cytochrome P450 and the metabolism of proton
pump inhibitors emphasis on rabeprazole. Aliment Pharmacol Ther 13 Suppl 3: 27
36, 1999.

518. Ishizuya T, Yokose S, Hori M, Noda T, Suda T, Yoshiki S, Yamaguchi A. Parathyroid


hormone exerts disparate effects on osteoblast differentiation depending on exposure time in rat osteoblastic cells. J Clin Invest 99: 29612970, 1997.
519. Ito R, Sato K, Helmer T, Jay G, Agarwal K. Structural analysis of the gene encoding
human gastrin: the large intron contains an Alu sequence. Proc Natl Acad Sci USA 81:
4662 4666, 1984.
520. Ivanovich P, Fellows H, Rich C. The absorption of calcium carbonate. Ann Intern Med
66: 917923, 1967.
521. Jaffe BM, Kopen DF, Lazan DW. Endogenous serotonin in the control of gastric acid
secretion. Surgery 82: 156 163, 1977.
522. Jain RN, Al-Menhali AA, Keeley TM, Ren J, El-Zaatari M, Chen X, Merchant JL, Ross
TS, Chew CS, Samuelson LC. Hip1r is expressed in gastric parietal cells and is
required for tubulovesicle formation and cell survival in mice. J Clin Invest 118:
2459 2470, 2008.

531. Jilka RL, Weinstein RS, Bellido T, Roberson P, Parfitt AM, Manolagas SC. Increased
bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J
Clin Invest 104: 439 446, 1999.
532. Jin HO, Lee KY, Chang TM, Chey WY, Dubois A. Secretin: a physiological regulator
of gastric emptying and acid output in dogs. Am J Physiol Gastrointest Liver Physiol 267:
G702G708, 1994.
533. Johnson JA, Grande JP, Roche PC, Kumar R. Ontogeny of the 1,25-dihydroxyvitamin
D3 receptor in fetal rat bone. J Bone Miner Res 11: 56 61, 1996.
534. Johnson LR, Lichtenberger LM, Copeland EM, Dudrick SJ, Castro GA. Action of
gastrin on gastrointestinal structure and function. Gastroenterology 68: 1184 1192,
1975.
535. Jones B, Jones EL, Bonney SA, Patel HN, Mensenkamp AR, Eichenbaum-Voline S,
Rudling M, Myrdal U, Annesi G, Naik S, Meadows N, Quattrone A, Islam SA, Naoumova RP, Angelin B, Infante R, Levy E, Roy CC, Freemont PS, Scott J, Shoulders CC.
Mutations in a Sar1 GTPase of COPII vesicles are associated with lipid absorption
disorders. Nat Genet 34: 29 31, 2003.
536. Juppner H, Abou-Samra AB, Freeman M, Kong XF, Schipani E, Richards J, Kolakowski LF Jr, Hock J, Potts JT Jr, Kronenberg HM. A G protein-linked receptor for
parathyroid hormone and parathyroid hormone-related peptide. Science 254:
1024 1026, 1991.
537. Jurutka PW, Thompson PD, Whitfield GK, Eichhorst KR, Hall N, Dominguez CE,
Hsieh JC, Haussler CA, Haussler MR. Molecular and functional comparison of 1,25dihydroxyvitamin D3 and the novel vitamin D receptor ligand, lithocholic acid, in
activating transcription of cytochrome P450 3A4. J Cell Biochem 94: 917943, 2005.
538. Juttmann JR, Visser TJ, Buurman C, de Kam E, Birkenhager JC. Seasonal fluctuations
in serum concentrations of vitamin D metabolites in normal subjects. Br Med J 282:
1349 1352, 1981.
539. Kahrilas PJ, Dent J, Lauritsen K, Malfertheiner P, Denison H, Franzen S, Hasselgren
G. A randomized, comparative study of three doses of AZD0865 and esomeprazole
for healing of reflux esophagitis. Clin Gastroenterol Hepatol 5: 13851391, 2007.
540. Kajikawa M, Ishida H, Fujimoto S, Mukai E, Nishimura M, Fujita J, Tsuura Y, Okamoto
Y, Norman AW, Seino Y. An insulinotropic effect of vitamin D analog with increasing
intracellular Ca2 concentration in pancreatic beta-cells through nongenomic signal
transduction. Endocrinology 140: 4706 4712, 1999.
541. Kajimura M, Reuben MA, Sachs G. The muscarinic receptor gene expressed in rabbit
parietal cells is the m3 subtype. Gastroenterology 103: 870 875, 1992.

523. Jain RN, Samuelson LC. Transcriptional profiling of gastrin-regulated genes in mouse
stomach. Physiol Genomics 29: 112, 2007.

542. Kameda T, Mano H, Yamada Y, Takai H, Amizuka N, Kobori M, Izumi N, Kawashima


H, Ozawa H, Ikeda K, Kameda A, Hakeda Y, Kumegawa M. Calcium-sensing receptor in mature osteoclasts, which are bone resorbing cells. Biochem Biophys Res
Commun 245: 419 422, 1998.

524. Jarrousse C, Audousset-Puech MP, Dubrasquet M, Niel H, Martinez J, Bataille D.


Oxyntomodulin (glucagon-37) and its C-terminal octapeptide inhibit gastric acid
secretion. FEBS Lett 188: 81 84, 1985.

543. Kanai S, Hosoya H, Akimoto S, Ohta M, Matsui T, Takiguchi S, Funakoshi A, Miyasaka


K. Gastric acid secretion in cholecystokinin-1 receptor, -2 receptor, and -1, -2
receptor gene knockout mice. J Physiol Sci 59: 2329, 2009.

525. Jarrousse C, Niel H, Audousset-Puech MP, Martinez J, Bataille D. Oxyntomodulin


and its C-terminal octapeptide inhibit liquid meal-stimulated acid secretion. Peptides
7 Suppl 1: 253256, 1986.

544. Kanatani M, Sugimoto T, Kanzawa M, Yano S, Chihara K. High extracellular calcium


inhibits osteoclast-like cell formation by directly acting on the calcium-sensing receptor existing in osteoclast precursor cells. Biochem Biophys Res Commun 261:
144 148, 1999.

526. Jeffery DA, Roufogalis BD, Katz S. The effect of calmodulin on the phosphoprotein
intermediate of Mg2-dependent Ca2-stimulated adenosine triphosphatase in human erythrocyte membranes. Biochem J 194: 481 486, 1981.
527. Jeffery PL, McGuckin MA, Linden SK. Endocrine impact of Helicobacter pylori: focus
on ghrelin and ghrelin o-acyltransferase. World J Gastroenterol 17: 1249 1260, 2011.
528. Jeon TY, Lee S, Kim HH, Kim YJ, Son HC, Kim DH, Sim MS. Changes in plasma
ghrelin concentration immediately after gastrectomy in patients with early gastric
cancer. J Clin Endocrinol Metab 89: 53925396, 2004.
529. Jiang Y, Minet E, Zhang Z, Silver PA, Bai M. Modulation of interprotomer relationships is important for activation of dimeric calcium-sensing receptor. J Biol Chem 279:
1414714156, 2004.

545. Karam SM, Straiton T, Hassan WM, Leblond CP. Defining epithelial cell progenitors
in the human oxyntic mucosa. Stem Cells 21: 322336, 2003.
546. Karaplis AC, Lim SK, Baba H, Arnold A, Kronenberg HM. Inefficient membrane
targeting, translocation, and proteolytic processing by signal peptidase of a mutant
preproparathyroid hormone protein. J Biol Chem 270: 1629 1635, 1995.
547. Karp HJ, Ketola ME, Lamberg-Allardt CJ. Acute effects of calcium carbonate, calcium
citrate and potassium citrate on markers of calcium and bone metabolism in young
women. Br J Nutr 102: 13411347, 2009.
548. Karvar S, Yao X, Crothers JM Jr, Liu Y, Forte JG. Localization and function of soluble
N-ethylmaleimide-sensitive factor attachment protein-25 and vesicle-associated

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

251

Downloaded from on December 8, 2014

517. Ishizawa M, Matsunawa M, Adachi R, Uno S, Ikeda K, Masuno H, Shimizu M, Iwasaki


K, Yamada S, Makishima M. Lithocholic acid derivatives act as selective vitamin D
receptor modulators without inducing hypercalcemia. J Lipid Res 49: 763772, 2008.

530. Jilka RL, OBrien CA, Ali AA, Roberson PK, Weinstein RS, Manolagas SC. Intermittent PTH stimulates periosteal bone formation by actions on post-mitotic preosteoblasts. Bone 44: 275286, 2009.

SASCHA KOPIC AND JOHN P. GEIBEL


membrane protein-2 in functioning gastric parietal cells. J Biol Chem 277: 50030
50035, 2002.
549. Karvar S, Yao X, Duman JG, Hybiske K, Liu Y, Forte JG. Intracellular distribution and
functional importance of vesicle-associated membrane protein 2 in gastric parietal
cells. Gastroenterology 123: 281290, 2002.
550. Karvar S, Zhu L, Crothers J Jr, Wong W, Turkoz M, Forte JG. Cellular localization and
stimulation-associated distribution dynamics of syntaxin-1 and syntaxin-3 in gastric
parietal cells. Traffic 6: 654 666, 2005.
551. Kato K, Hayashizaki Y, Takahashi Y, Himeno S, Matsubara K. Molecular cloning of
the human gastrin gene. Nucleic Acids Res 11: 8197 8203, 1983.
552. Kato K, Himeno S, Takahashi Y, Wakabayashi T, Tarui S, Matsubara K. Molecular
cloning of human gastrin precursor cDNA. Gene 26: 5357, 1983.
553. Kato S, Abe Y, Konishi M, Kuroda N, Takeuchi K. Mechanism of gastric hyperemic
response during acid secretion in rats: relation to nitric oxide, prostaglandins, sensory neurons. J Clin Gastroenterol 25 Suppl 1: S48 55, 1997.
554. Kato S, Aihara E, Yoshii K, Takeuchi K. Dual action of prostaglandin E2 on gastric acid
secretion through different EP-receptor subtypes in the rat. Am J Physiol Gastrointest
Liver Physiol 289: G64 G69, 2005.
555. Kato S, Kitamura M, Korolkiewicz RP, Takeuchi K. Role of nitric oxide in regulation
of gastric acid secretion in rats: effects of NO donors and NO synthase inhibitor. Br
J Pharmacol 123: 839 846, 1998.

557. Kawakami M, Blum CB, Ramakrishnan R, Dell RB, Goodman DS. Turnover of the
plasma binding protein for vitamin D and its metabolites in normal human subjects.
J Clin Endocrinol Metab 53: 1110 1116, 1981.
558. Kawashima K, Ishihara S, Karim Rumi MA, Moriyama N, Kazumori H, Suetsugu H,
Sato H, Fukuda R, Adachi K, Shibata M, Onodera S, Chiba T, Kinoshita Y. Localization
of calcitonin gene-related peptide receptors in rat gastric mucosa. Peptides 23: 955
966, 2002.
559. Kaye JA, Jick H. Proton pump inhibitor use and risk of hip fractures in patients
without major risk factors. Pharmacotherapy 28: 951959, 2008.
560. Kazumori H, Ishihara S, Kawashima K, Fukuda R, Chiba T, Kinoshita Y. Analysis of
gastrin receptor gene expression in proliferating cells in the neck zone of gastric
fundic glands using laser capture microdissection. FEBS Lett 489: 208 214, 2001.
561. Keeley TM, Samuelson LC. Cytodifferentiation of the postnatal mouse stomach in
normal and Huntingtin-interacting protein 1-related-deficient mice. Am J Physiol Gastrointest Liver Physiol 299: G1241G1251, 2010.
562. Keeton TP, Burk SE, Shull GE. Alternative splicing of exons encoding the calmodulinbinding domains and C termini of plasma membrane Ca2-ATPase isoforms 1, 2, 3,
and 4. J Biol Chem 268: 2740 2748, 1993.
563. Kehayoglou AK, Holdsworth CD, Agnew JE, Whelton MJ, Sherlock S. Bone disease
and calcium absorption in primary biliary cirrhosis with special reference to vitamin-D therapy. Lancet 1: 715718, 1968.
564. Keinke O, Ehrlein HJ, Wulschke S. Mechanical factors regulating gastric emptying
examined by the effects of exogenous cholecystokinin and secretin on canine gastroduodenal motility. Can J Physiol Pharmacol 65: 287292, 1987.
565. Kemper B, Habener JF, Mulligan RC, Potts JT Jr, Rich A. Pre-proparathyroid hormone: a direct translation product of parathyroid messenger RNA. Proc Natl Acad Sci
USA 71: 37313735, 1974.

569. Kidd M, Hauso O, Drozdov I, Gustafsson BI, Modlin IM. Delineation of the chemomechanosensory regulation of gastrin secretion using pure rodent G cells. Gastroenterology 137: 231241, 2009.
570. Kidd M, Modlin IM, Black JW, Boyce M, Culler M. A comparison of the effects of
gastrin, somatostatin and dopamine receptor ligands on rat gastric enterochromaffin-like cell secretion and proliferation. Regul Pept 143: 109 117, 2007.
571. Kifor O, Diaz R, Butters R, Kifor I, Brown EM. The calcium-sensing receptor is
localized in caveolin-rich plasma membrane domains of bovine parathyroid cells. J
Biol Chem 273: 21708 21713, 1998.
572. Kifor O, MacLeod RJ, Diaz R, Bai M, Yamaguchi T, Yao T, Kifor I, Brown EM.
Regulation of MAP kinase by calcium-sensing receptor in bovine parathyroid and
CaR-transfected HEK293 cells. Am J Physiol Renal Physiol 280: F291F302, 2001.
573. Kikuchi K, Kikuchi T, Ghishan FK. Characterization of calcium transport by basolateral membrane vesicles of human small intestine. Am J Physiol Gastrointest Liver
Physiol 255: G482G489, 1988.
574. Kim HJ, Yang DK, So I. PDZ domain-containing protein as a physiological modulator
of TRPV6. Biochem Biophys Res Commun 361: 433 438, 2007.
575. Kim MS, Fujiki R, Murayama A, Kitagawa H, Yamaoka K, Yamamoto Y, Mihara M,
Takeyama K, Kato S. 1Alpha,25(OH)2D3-induced transrepression by vitamin D receptor through E-box-type elements in the human parathyroid hormone gene promoter. Mol Endocrinol 21: 334 342, 2007.
576. Kim SW, Her SJ, Park SJ, Kim D, Park KS, Lee HK, Han BH, Kim MS, Shin CS, Kim SY.
Ghrelin stimulates proliferation and differentiation and inhibits apoptosis in osteoblastic MC3T3-E1 cells. Bone 37: 359 369, 2005.
577. Kirchhoff P, Andersson K, Socrates T, Sidani S, Kosiek O, Geibel JP. Characteristics
of the K-competitive H,K-ATPase inhibitor AZD0865 in isolated rat gastric
glands. Am J Physiol Gastrointest Liver Physiol 291: G838 G843, 2006.
578. Kirton CM, Wang T, Dockray GJ. Regulation of parietal cell migration by gastrin in
the mouse. Am J Physiol Gastrointest Liver Physiol 283: G787G793, 2002.
579. Klenk C, Schulz S, Calebiro D, Lohse MJ. Agonist-regulated cleavage of the extracellular domain of parathyroid hormone receptor type 1. J Biol Chem 285: 8665
8674, 2010.
580. Klenk C, Vetter T, Zurn A, Vilardaga JP, Friedman PA, Wang B, Lohse MJ. Formation
of a ternary complex among NHERF1, beta-arrestin, parathyroid hormone receptor. J Biol Chem 285: 3035530362, 2010.
581. Klotz U. Pharmacokinetic considerations in the eradication of Helicobacter pylori.
Clin Pharmacokinet 38: 243270, 2000.
582. Klotz U, Schwab M, Treiber G. CYP2C19 polymorphism and proton pump inhibitors. Basic Clin Pharmacol Toxicol 95: 2 8, 2004.
583. Ko SH, Choi KC, Oh GT, Jeung EB. Effect of dietary calcium and 1,25-(OH)2D3 on
the expression of calcium transport genes in calbindin-D9k and -D28k double knockout mice. Biochem Biophys Res Commun 379: 227232, 2009.
584. Kobayashi T, Tonai S, Ishihara Y, Koga R, Okabe S, Watanabe T. Abnormal functional
and morphological regulation of the gastric mucosa in histamine H2 receptor-deficient mice. J Clin Invest 105: 17411749, 2000.
585. Kocian J, Brodan V. New observations on the absorption of 47Ca in patients with
partial gastrectomy. Digestion 12: 193200, 1975.
586. Kodicek E, Lawson DE, Wilson PW. Biological activity of a polar metabolite of
vitamin D. Nature 228: 763764, 1970.

566. Keusch I, Traebert M, Lotscher M, Kaissling B, Murer H, Biber J. Parathyroid hormone and dietary phosphate provoke a lysosomal routing of the proximal tubular
Na/Pi-cotransporter type II. Kidney Int 54: 1224 1232, 1998.

587. Koh TJ, Field JK, Varro A, Liloglou T, Fielding P, Cui G, Houghton J, Dockray GJ,
Wang TC. Glycine-extended gastrin promotes the growth of lung cancer. Cancer Res
64: 196 201, 2004.

567. Khan M, Santana J, Donnellan C, Preston C, Moayyedi P. Medical treatments in the


short term management of reflux oesophagitis. Cochrane Database Syst Rev
CD003244, 2007.

588. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a


growth-hormone-releasing acylated peptide from stomach. Nature 402: 656 660,
1999.

252

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

556. Kaufhold MA, Krabbenhoft A, Song P, Engelhardt R, Riederer B, Fahrmann M,


Klocker N, Beil W, Manns M, Hagen SJ, Seidler U. Localization, trafficking, and
significance for acid secretion of parietal cell Kir4.1 and KCNQ1 K channels. Gastroenterology 134: 1058 1069, 2008.

568. Khanal RC, Peters TM, Smith NM, Nemere I. Membrane receptor-initiated signaling
in 1,25(OH)2D3-stimulated calcium uptake in intestinal epithelial cells. J Cell Biochem
105: 1109 1116, 2008.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


589. Komaba H, Nakanishi S, Fujimori A, Tanaka M, Shin J, Shibuya K, Nishioka M,
Hasegawa H, Kurosawa T, Fukagawa M. Cinacalcet effectively reduces parathyroid
hormone secretion and gland volume regardless of pretreatment gland size in patients with secondary hyperparathyroidism. Clin J Am Soc Nephrol 5: 23052314,
2010.
590. Komasaka M, Horie S, Watanabe K, Murayama T. Antisecretory effect of somatostatin on gastric acid via inhibition of histamine release in isolated mouse stomach. Eur
J Pharmacol 452: 235243, 2002.
591. Kong XF, Zhu XH, Pei YL, Jackson DM, Holick MF. Molecular cloning, characterization, and promoter analysis of the human 25-hydroxyvitamin D3-1alpha-hydroxylase gene. Proc Natl Acad Sci USA 96: 6988 6993, 1999.
592. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Morony S, Oliveirados-Santos AJ, Van G, Itie A, Khoo W, Wakeham A, Dunstan CR, Lacey DL, Mak TW,
Boyle WJ, Penninger JM. OPGL is a key regulator of osteoclastogenesis, lymphocyte
development and lymph-node organogenesis. Nature 397: 315323, 1999.
593. Konturek JW, Stoll R, Konturek SJ, Domschke W. Cholecystokinin in the control of
gastric acid secretion in man. Gut 34: 321328, 1993.
594. Konturek SJ, Bilski J, Cieszkowski M. Role of cholecystokinin in the intestinal fat- and
acid-induced inhibition of gastric secretion. Regul Pept 42: 97109, 1992.
595. Konturek SJ, Kwiecien N, Obtulowicz W, Mikos E, Sito E, Oleksy J, Popiela T.
Cephalic phase of gastric secretion in healthy subjects and duodenal ulcer patients:
role of vagal innervation. Gut 20: 875 881, 1979.

597. Kopp R, Pfeiffer A. Effects of phorbol ester treatment on dibutyryl cyclic adenosine-5= monophosphate- and carbachol-stimulated aminopyrine accumulation in isolated rat parietal cells. Scand J Gastroenterol 35: 686 693, 2000.
598. Kos CH, Karaplis AC, Peng JB, Hediger MA, Goltzman D, Mohammad KS, Guise TA,
Pollak MR. The calcium-sensing receptor is required for normal calcium homeostasis
independent of parathyroid hormone. J Clin Invest 111: 10211028, 2003.
599. Koshihara Y, Hoshi K, Ishibashi H, Shiraki M. Vitamin K2 promotes 1alpha,25(OH)2
vitamin D3-induced mineralization in human periosteal osteoblasts. Calcif Tissue Int
59: 466 473, 1996.
600. Koushik SV, Wang J, Rogers R, Moskophidis D, Lambert NA, Creazzo TL, Conway
SJ. Targeted inactivation of the sodium-calcium exchanger (Ncx1) results in the lack
of a heartbeat and abnormal myofibrillar organization. FASEB J 15: 1209 1211, 2001.
601. Kozoni V, Tsioulias G, Shiff S, Rigas B. The effect of lithocholic acid on proliferation
and apoptosis during the early stages of colon carcinogenesis: differential effect on
apoptosis in the presence of a colon carcinogen. Carcinogenesis 21: 999 1005, 2000.
602. Kretsinger RH, Mann JE, Simmonds JG. Model of facilitated diffusion of calcium by
the intestinal calcium-binding protein. In: Proceedings of the Fifth Workshop on Vitamin
D, edited by Norman AW, Schaefer K, Herrath DV, Grigoleit HG. Berlin: Debruyter,
1982, p. 232248.
603. Kreutz M, Andreesen R, Krause SW, Szabo A, Ritz E, Reichel H. 1,25-Dihydroxyvitamin D3 production and vitamin D3 receptor expression are developmentally regulated during differentiation of human monocytes into macrophages. Blood 82:
1300 1307, 1993.
604. Krisinger J, Strom M, Darwish HD, Perlman K, Smith C, DeLuca HF. Induction of
calbindin-D 9k mRNA but not calcium transport in rat intestine by 1,25-dihydroxyvitamin D3 24-homologs. J Biol Chem 266: 1910 1913, 1991.
605. Kubler N, Krause U, Wagner PK, Beyer J, Rothmund M. The secretion of parathyroid
hormone and its fragments from dispersed cells of adenomatous parathyroid tissue
at different calcium concentrations. Exp Clin Endocrinol 88: 101108, 1986.
606. Kuestner RE, Elrod RD, Grant FJ, Hagen FS, Kuijper JL, Matthewes SL, OHara PJ,
Sheppard PO, Stroop SD, Thompson DL. Cloning and characterization of an abundant subtype of the human calcitonin receptor. Mol Pharmacol 46: 246 255, 1994.
607. Kuhn B, Schmid A, Harteneck C, Gudermann T, Schultz G. G proteins of the Gq
family couple the H2 histamine receptor to phospholipase C. Mol Endocrinol 10:
16971707, 1996.

609. Kumar R, Cohen WR, Silva P, Epstein FH. Elevated 1,25-dihydroxyvitamin D plasma
levels in normal human pregnancy and lactation. J Clin Invest 63: 342344, 1979.
610. Kumar R, Schaefer J, Grande JP, Roche PC. Immunolocalization of calcitriol receptor,
24-hydroxylase cytochrome P-450, and calbindin D28k in human kidney. Am J Physiol
Renal Fluid Electrolyte Physiol 266: F477F485, 1994.
611. Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi S,
Jingami H, Morikawa K. Structural basis of glutamate recognition by a dimeric
metabotropic glutamate receptor. Nature 407: 971977, 2000.
612. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y,
Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T, Nishikawa S,
Nagai R, Nabeshima YI. Mutation of the mouse klotho gene leads to a syndrome
resembling ageing. Nature 390: 4551, 1997.
613. Kutuzova GD, Akhter S, Christakos S, Vanhooke J, Kimmel-Jehan C, DeLuca HF.
Calbindin D9k knockout mice are indistinguishable from wild-type mice in phenotype and serum calcium level. Proc Natl Acad Sci USA 103: 1237712381, 2006.
614. Kutuzova GD, Deluca HF. Gene expression profiles in rat intestine identify pathways
for 1,25-dihydroxyvitamin D3 stimulated calcium absorption and clarify its immunomodulatory properties. Arch Biochem Biophys 432: 152166, 2004.
615. Kutuzova GD, Sundersingh F, Vaughan J, Tadi BP, Ansay SE, Christakos S, Deluca
HF. TRPV6 is not required for 1alpha,25-dihydroxyvitamin D3-induced intestinal
calcium absorption in vivo. Proc Natl Acad Sci USA 105: 1965519659, 2008.
616. Kwok CS, Yeong JK, Loke YK. Meta-analysis: risk of fractures with acid-suppressing
medication. Bone 48: 768 776, 2011.
617. Labuda M, Labuda D, Korab-Laskowska M, Cole DE, Zietkiewicz E, Weissenbach J,
Popowska E, Pronicka E, Root AW, Glorieux FH. Linkage disequilibrium analysis in
young populations: pseudo-vitamin D-deficiency rickets and the founder effect in
French Canadians. Am J Hum Genet 59: 633 643, 1996.
618. Labuda M, Morgan K, Glorieux FH. Mapping autosomal recessive vitamin D dependency type I to chromosome 12q14 by linkage analysis. Am J Hum Genet 47: 28 36,
1990.
619. Lambers TT, Weidema AF, Nilius B, Hoenderop JG, Bindels RJ. Regulation of the
mouse epithelial Ca2 channel TRPV6 by the Ca2-sensor calmodulin. J Biol Chem
279: 2885528861, 2004.
620. Lamberts R, Stumps D, Plumpe L, Creutzfeldt W. Somatostatin cells in rat antral
mucosa: qualitative and quantitative ultrastructural analyses in different states of
gastric acid secretion. Histochemistry 95: 373382, 1991.
621. Lane NE, Sanchez S, Modin GW, Genant HK, Pierini E, Arnaud CD. Parathyroid
hormone treatment can reverse corticosteroid-induced osteoporosis. Results of a
randomized controlled clinical trial. J Clin Invest 102: 16271633, 1998.
622. Langhans N, Rindi G, Chiu M, Rehfeld JF, Ardman B, Beinborn M, Kopin AS. Abnormal gastric histology and decreased acid production in cholecystokinin-B/gastrin
receptor-deficient mice. Gastroenterology 112: 280 286, 1997.
623. Langub MC, Reinhardt TA, Horst RL, Malluche HH, Koszewski NJ. Characterization
of vitamin D receptor immunoreactivity in human bone cells. Bone 27: 383387,
2000.
624. Lapierre LA, Avant KM, Caldwell CM, Ham AJ, Hill S, Williams JA, Smolka AJ,
Goldenring JR. Characterization of immunoisolated human gastric parietal cells tubulovesicles: identification of regulators of apical recycling. Am J Physiol Gastrointest
Liver Physiol 292: G1249 G1262, 2007.
625. Lark RK, Lester GE, Ontjes DA, Blackwood AD, Hollis BW, Hensler MM, Aris RM.
Diminished and erratic absorption of ergocalciferol in adult cystic fibrosis patients.
Am J Clin Nutr 73: 602 606, 2001.
626. Larsen EH, Nedergaard S, Ussing HH. Role of lateral intercellular space and sodium
recirculation for isotonic transport in leaky epithelia. Rev Physiol Biochem Pharmacol
141: 153212, 2000.
627. Larsen FL, Katz S, Roufogalis BD. Calmodulin regulation of Ca2 transport in human
erythrocytes. Biochem J 200: 185191, 1981.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

253

Downloaded from on December 8, 2014

596. Kopin AS, Lee YM, McBride EW, Miller LJ, Lu M, Lin HY, Kolakowski LF Jr, Beinborn
M. Expression cloning and characterization of the canine parietal cell gastrin receptor. Proc Natl Acad Sci USA 89: 36053609, 1992.

608. Kulaksiz H, Arnold R, Goke B, Maronde E, Meyer M, Fahrenholz F, Forssmann WG,


Eissele R. Expression and cell-specific localization of the cholecystokinin B/gastrin
receptor in the human stomach. Cell Tissue Res 299: 289 298, 2000.

SASCHA KOPIC AND JOHN P. GEIBEL


628. Larsen FL, Vincenzi FF. Calcium transport across the plasma membrane: stimulation
by calmodulin. Science 204: 306 309, 1979.

649. Lepard KJ, Chi J, Mohammed JR, Gidener S, Stephens RL Jr. Gastric antisecretory
effect of serotonin: quantitation of release and site of action. Am J Physiol Endocrinol
Metab 271: E669 E677, 1996.

629. Larsson B, Gritli-Linde A, Norlen P, Lindstrom E, Hakanson R, Linde A. Extracts of


ECL-cell granules/vesicles and of isolated ECL cells from rat oxyntic mucosa evoke a
Ca2 second messenger response in osteoblastic cells. Regul Pept 97: 153161,
2001.

650. LePard KJ, Stephens RL Jr. Serotonin inhibits gastric acid secretion through a 5-hydroxytryptamine1-like receptor in the rat. J Pharmacol Exp Ther 270: 1139 1144,
1994.

630. Larsson B, Norlen P, Lindstrom E, Zhao D, Hakanson R, Linde A. Effects of ECL cell
extracts and granule/vesicle-enriched fractions from rat oxyntic mucosa on cAMP
and IP3 in rat osteoblast-like cells. Regul Pept 106: 1318, 2002.

651. Leth RD, Abrahamsson H, Kilander A, Lundell LR. Malabsorption of fat after partial
gastric resection. A study of pathophysiologic mechanisms. Eur J Surg 157: 205208,
1991.

631. Larsson H, Carlsson E, Ryberg B, Fryklund J, Wallmark B. Rat parietal cell function
after prolonged inhibition of gastric acid secretion. Am J Physiol Gastrointest Liver
Physiol 254: G33G39, 1988.

652. Levant JA, Walsh JH, Isenberg JI. Stimulation of gastric secretion and gastrin release
by single oral doses of calcium carbonate in man. N Engl J Med 289: 555558, 1973.

632. Larsson LI, Goltermann N, de Magistris L, Rehfeld JF, Schwartz TW. Somatostatin
cell processes as pathways for paracrine secretion. Science 205: 13931395, 1979.
633. Lawson DE, Fraser DR, Kodicek E, Morris HR, Williams DH. Identification of 1,25dihydroxycholecalciferol, a new kidney hormone controlling calcium metabolism.
Nature 230: 228 230, 1971.

653. Levi R, Ben-Dov IZ, Lavi-Moshayoff V, Dinur M, Martin D, Naveh-Many T, Silver J.


Increased parathyroid hormone gene expression in secondary hyperparathyroidism
of experimental uremia is reversed by calcimimetics: correlation with posttranslational modification of the trans acting factor AUF1. J Am Soc Nephrol 17: 107112,
2006.
654. Levin F, Edholm T, Ehrstrom M, Wallin B, Schmidt PT, Kirchgessner AM, Hilsted LM,
Hellstrom PM, Naslund E. Effect of peripherally administered ghrelin on gastric
emptying and acid secretion in the rat. Regul Pept 131: 59 65, 2005.

635. Lawton DE, Simcock DC, Candy EJ, Simpson HV. Gastrin secretion by ovine antral
mucosa in vitro. Comp Biochem Physiol A Mol Integr Physiol 126: 233243, 2000.

655. Lewis JJ, Zdon MJ, Adrian TE, Modlin IM. Pancreastatin: a novel peptide inhibitor of
parietal cell secretion. Surgery 104: 10311036, 1988.

636. Lawton GP, Tang LH, Miu K, Gilligan CJ, Absood A, Modlin IM. Adrenergic and
cromolyn sodium modulation of ECL cell histamine secretion. J Surg Res 58: 96 104,
1995.

656. Li P, Chang TM, Chey WY. Secretin inhibits gastric acid secretion via a vagal afferent
pathway in rats. Am J Physiol Gastrointest Liver Physiol 275: G22G28, 1998.

637. Le Moullec JM, Jullienne A, Chenais J, Lasmoles F, Guliana JM, Milhaud G, Moukhtar
MS. The complete sequence of human preprocalcitonin. FEBS Lett 167: 9397, 1984.

657. Li P, Chang TM, Coy D, Chey WY. Inhibition of gastric acid secretion in rat stomach
by PACAP is mediated by secretin, somatostatin, and PGE2. Am J Physiol Gastrointest
Liver Physiol 278: G121G127, 2000.

638. Leclerc R, Pelletier G, Puviani R, Arimura A, Schally AV. Immunohistochemical localization of somatostatin in endocrine cells of the rat stomach. Mol Cell Endocrinol 4:
257261, 1976.
639. Lee GS, Lee KY, Choi KC, Ryu YH, Paik SG, Oh GT, Jeung EB. Phenotype of a
calbindin-D9k gene knockout is compensated for by the induction of other calcium
transporter genes in a mouse model. J Bone Miner Res 22: 1968 1978, 2007.
640. Lee JW, Fuda H, Javitt NB, Strott CA, Rodriguez IR. Expression and localization of
sterol 27-hydroxylase (CYP27A1) in monkey retina. Exp Eye Res 83: 465 469, 2006.
641. Lee MP, Ravenel JD, Hu RJ, Lustig LR, Tomaselli G, Berger RD, Brandenburg SA, Litzi
TJ, Bunton TE, Limb C, Francis H, Gorelikow M, Gu H, Washington K, Argani P,
Goldenring JR, Coffey RJ, Feinberg AP. Targeted disruption of the Kvlqt1 gene
causes deafness and gastric hyperplasia in mice. J Clin Invest 106: 14471455, 2000.
642. Lefkowitz ES, Garland CF. Sunlight, vitamin D, and ovarian cancer mortality rates in
US women. Int J Epidemiol 23: 11331136, 1994.
643. Leheste JR, Melsen F, Wellner M, Jansen P, Schlichting U, Renner-Muller I, Andreassen TT, Wolf E, Bachmann S, Nykjaer A, Willnow TE. Hypocalcemia and osteopathy in mice with kidney-specific megalin gene defect. FASEB J 17: 247249, 2003.
644. Lehto-Axtelius D, Chen D, Surve VV, Hakanson R. Post-gastrectomy osteopenia in
the rat: bone structure is preserved by retaining 10%-30% of the oxyntic gland area.
Scand J Gastroenterol 37: 437 443, 2002.
645. Leichtmann GA, Bengoa JM, Bolt MJ, Sitrin MD. Intestinal absorption of cholecalciferol and 25-hydroxycholecalciferol in patients with both Crohns disease and intestinal resection. Am J Clin Nutr 54: 548 552, 1991.
646. Leitersdorf E, Reshef A, Meiner V, Levitzki R, Schwartz SP, Dann EJ, Berkman N, Cali
JJ, Klapholz L, Berginer VM. Frameshift and splice-junction mutations in the sterol
27-hydroxylase gene cause cerebrotendinous xanthomatosis in Jews or Moroccan
origin. J Clin Invest 91: 2488 2496, 1993.
647. Lemay J, Demers C, Hendy GN, Gascon-Barr M. Oral calcium transiently increases
calbindin(9k) gene expression in adult rat duodena. Calcified Tissue Int 60: 43 47,
1997.
648. Lepage R, Roy L, Brossard JH, Rousseau L, Dorais C, Lazure C, DAmour P. A
non-(1 84) circulating parathyroid hormone (PTH) fragment interferes significantly
with intact PTH commercial assay measurements in uremic samples. Clin Chem 44:
805 809, 1998.

254

658. Li XF, Kraev AS, Lytton J. Molecular cloning of a fourth member of the potassiumdependent sodium-calcium exchanger gene family, NCKX4. J Biol Chem 277:
48410 48417, 2002.
659. Li Y, Wu X, Yao H, Owyang C. Secretin activates vagal primary afferent neurons in
the rat: evidence from electrophysiological and immunohistochemical studies. Am J
Physiol Gastrointest Liver Physiol 289: G745G752, 2005.
660. Li YC. Vitamin D regulation of the renin-angiotensin system. J Cell Biochem 88:
327331, 2003.
661. Li YC, Amling M, Pirro AE, Priemel M, Meuse J, Baron R, Delling G, Demay MB.
Normalization of mineral ion homeostasis by dietary means prevents hyperparathyroidism, rickets, osteomalacia, but not alopecia in vitamin D receptor-ablated mice.
Endocrinology 139: 4391 4396, 1998.
662. Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP. 1,25-Dihydroxyvitamin D3 is a
negative endocrine regulator of the renin-angiotensin system. J Clin Invest 110: 229
238, 2002.
663. Li YC, Pirro AE, Demay MB. Analysis of vitamin D-dependent calcium-binding protein messenger ribonucleic acid expression in mice lacking the vitamin D receptor.
Endocrinology 139: 847 851, 1998.
664. Li YY. Mechanisms for regulation of gastrin and somatostatin release from isolated
rat stomach during gastric distention. World J Gastroenterol 9: 129 133, 2003.
665. Lian J, Stewart C, Puchacz E, Mackowiak S, Shalhoub V, Collart D, Zambetti G, Stein
G. Structure of the rat osteocalcin gene and regulation of vitamin D-dependent
expression. Proc Natl Acad Sci USA 86: 11431147, 1989.
666. Lieben L, Benn BS, Ajibade D, Stockmans I, Moermans K, Hediger MA, Peng JB,
Christakos S, Bouillon R, Carmeliet G. Trpv6 mediates intestinal calcium absorption
during calcium restriction and contributes to bone homeostasis. Bone 47: 301308,
2010.
667. Liedman B, Bosaeus I, Mellstrom D, Lundell L. Osteoporosis after total gastrectomy.
Results of a prospective, clinical study. Scand J Gastroenterol 32: 1090 1095, 1997.
668. Liedman B, Henningsson A, Mellstrom D, Lundell L. Changes in bone metabolism
and body composition after total gastrectomy: results of a longitudinal study. Dig Dis
Sci 45: 819 824, 2000.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

634. Lawson DE, Wilson PW, Kodicek E. New vitamin D metabolite localized in intestinal
cell nuclei. Nature 222: 171172, 1969.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


669. Lin HY, Harris TL, Flannery MS, Aruffo A, Kaji EH, Gorn A, Kolakowski LF Jr, Lodish
HF, Goldring SR. Expression cloning of an adenylate cyclase-coupled calcitonin receptor. Science 254: 10221024, 1991.
670. Lindberg P, Nordberg P, Alminger T, Brandstrom A, Wallmark B. The mechanism of
action of the gastric acid secretion inhibitor omeprazole. J Med Chem 29: 13271329,
1986.
671. Lindsay R, Nieves J, Formica C, Henneman E, Woelfert L, Shen V, Dempster D,
Cosman F. Randomised controlled study of effect of parathyroid hormone on vertebral-bone mass and fracture incidence among postmenopausal women on oestrogen with osteoporosis. Lancet 350: 550 555, 1997.
672. Lindstrom E, Bjorkqvist M, Boketoft A, Chen D, Zhao CM, Kimura K, Hakanson R.
Neurohormonal regulation of histamine and pancreastatin secretion from isolated
rat stomach ECL cells. Regul Pept 71: 73 86, 1997.
673. Lindstrom E, Eliasson L, Bjorkqvist M, Hakanson R. Gastrin and the neuropeptide
PACAP evoke secretion from rat stomach histamine-containing (ECL) cells by stimulating influx of Ca2 through different Ca2 channels. J Physiol 535: 663 677, 2001.

689. Ma YL, Cain RL, Halladay DL, Yang X, Zeng Q, Miles RR, Chandrasekhar S, Martin
TJ, Onyia JE. Catabolic effects of continuous human PTH (138) in vivo is associated
with sustained stimulation of RANKL and inhibition of osteoprotegerin and geneassociated bone formation. Endocrinology 142: 4047 4054, 2001.
690. MacCallum WG, Voegtlin C. On the regulation of the parathyroid to calcium metabolism and the nature of tetany. Bull Johns Hopkins Hosp 19: 91, 1908.
691. Maccarinelli G, Sibilia V, Torsello A, Raimondo F, Pitto M, Giustina A, Netti C, Cocchi
D. Ghrelin regulates proliferation and differentiation of osteoblastic cells. J Endocrinol
184: 249 256, 2005.
692. Mace OJ, Morgan EL, Affleck JA, Lister N, Kellett GL. Calcium absorption by Cav1.3
induces terminal web myosin II phosphorylation and apical GLUT2 insertion in rat
intestine. J Physiol 580: 605 616, 2007.
693. MacGregor RR, Cohn DV, Hamilton JW. The content of carboxyl-terminal fragments of parathormone in extracts of fresh bovine parathyroids. Endocrinology 112:
1019 1025, 1983.
694. Madaus S, Bender H, Schusdziarra V, Kehe K, Munzert G, Weber G, Classen M.
Vagally induced release of gastrin, somatostatin and bombesin-like immunoreactivity
from perfused rat stomach. Effect of stimulation frequency and cholinergic mechanisms. Regul Pept 30: 179 192, 1990.

675. Lipecka J, Bali M, Thomas A, Fanen P, Edelman A, Fritsch J. Distribution of ClC-2


chloride channel in rat and human epithelial tissues. Am J Physiol Cell Physiol 82:
C805C816, 2002.

695. Madaus S, Schusdziarra V, Seufferlein T, Classen M. Effect of galanin on gastrin and


somatostatin release from the rat stomach. Life Sci 42: 23812387, 1988.

676. Liu C, Chen J, Guo Y, Yang L, Zhao C, Bai L. The expression of PTHLH in human
gastric mucosa enterochromaffin-like cells. Dig Dis Sci 56: 993998, 2011.

696. Madhok TC, DeLuca HF. Characteristics of the rat liver microsomal enzyme system
converting cholecalciferol into 25-hydroxycholecalciferol. Evidence for the participation of cytochrome p-450. Biochem J 184: 491 499, 1979.

677. Lloyd KC, Amirmoazzami S, Friedik F, Heynio A, Solomon TE, Walsh JH. Candidate
canine enterogastrones: acid inhibition before and after vagotomy. Am J Physiol
Gastrointest Liver Physiol 272: G1236 G1242, 1997.
678. Lo CW, Paris PW, Clemens TL, Nolan J, Holick MF. Vitamin D absorption in healthy
subjects and in patients with intestinal malabsorption syndromes. Am J Clin Nutr 42:
644 649, 1985.
679. Locklin RM, Khosla S, Turner RT, Riggs BL. Mediators of the biphasic responses of
bone to intermittent and continuously administered parathyroid hormone. J Cell
Biochem 89: 180 190, 2003.
680. Loffing J, Loffing-Cueni D, Valderrabano V, Klausli L, Hebert SC, Rossier BC, Hoenderop JG, Bindels RJ, Kaissling B. Distribution of transcellular calcium and sodium
transport pathways along mouse distal nephron. Am J Physiol Renal Physiol 281:
F1021F1027, 2001.
681. Lorenz S, Frenzel R, Paschke R, Breitwieser GE, Miedlich SU. Functional desensitization of the extracellular calcium-sensing receptor is regulated via distinct mechanisms: role of G protein-coupled receptor kinases, protein kinase C and beta-arrestins. Endocrinology 148: 2398 2404, 2007.
682. Lotscher M, Scarpetta Y, Levi M, Halaihel N, Wang H, Zajicek HK, Biber J, Murer H,
Kaissling B. Rapid downregulation of rat renal Na/Pi cotransporter in response to
parathyroid hormone involves microtubule rearrangement. J Clin Invest 104: 483
494, 1999.
683. Lu P, Boros S, Chang Q, Bindels RJ, Hoenderop JG. The -glucuronidase klotho
exclusively activates the epithelial Ca2 channels TRPV5 and TRPV6. Nephrol Dialysis Transplantation 23: 33973402, 2008.
684. Luiz de Freitas PH, Li M, Ninomiya T, Nakamura M, Ubaidus S, Oda K, Udagawa N,
Maeda T, Takagi R, Amizuka N. Intermittent PTH administration stimulates preosteoblastic proliferation without leading to enhanced bone formation in osteoclastless c-fos(/) mice. J Bone Miner Res 24: 1586 1597, 2009.
685. Lund B, Selnes A. Plasma 1,25-dihydroxyvitamin D levels in pregnancy and lactation.
Acta Endocrinol 92: 330 335, 1979.
686. Lund J, DeLuca HF. Biologically active metabolite of vitamin D3 from bone, liver, and
blood serum. J Lipid Res 7: 739 744, 1966.
687. Lytton J. Na/Ca2 exchangers: three mammalian gene families control Ca2 transport. Biochem J 406: 365382, 2007.
688. Lytton J, Li XF, Dong H, Kraev A. K-dependent Na/Ca2 exchangers in the brain.
Ann NY Acad Sci 976: 382393, 2002.

697. Maeda M, Oshiman K, Tamura S, Futai M. Human gastric-ATPase gene. Similarity to


(Na K)-ATPase genes in exon/intron organization but difference in control
region. J Biol Chem 265: 90279032, 1990.
698. Mahon MJ, Donowitz M, Yun CC, Segre GV. Na/H exchanger regulatory factor 2
directs parathyroid hormone 1 receptor signalling. Nature 417: 858 861, 2002.
699. Mahoney AW, Holbrook RS, Hendricks DG. Effects of calcium solubility on adsorption by rats with induced achlorhydria. Nutr Metab 18: 310 317, 1975.
700. Maier GW, Kreis ME, Zittel TT, Becker HD. Calcium regulation and bone mass loss
after total gastrectomy in pigs. Ann Surg 225: 181192, 1997.
701. Maislos M, Silver J, Fainaru M. Intestinal absorption of vitamin D sterols: differential
absorption into lymph and portal blood in the rat. Gastroenterology 80: 1528 1534,
1981.
702. Maiti A, Beckman MJ. Extracellular calcium is a direct effecter of VDR levels in
proximal tubule epithelial cells that counter-balances effects of PTH on renal Vitamin
D metabolism. J Steroid Biochem Mol Biol 103: 504 508, 2007.
703. Makin G, Lohnes D, Byford V, Ray R, Jones G. Target cell metabolism of 1,25dihydroxyvitamin D3 to calcitroic acid. Evidence for a pathway in kidney and bone
involving 24-oxidation. Biochem J 262: 173180, 1989.
704. Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, Evans RM, Haussler MR,
Mangelsdorf DJ. Vitamin D receptor as an intestinal bile acid sensor. Science 296:
13131316, 2002.
705. Malinowska DH. Cl channel blockers inhibit acid secretion in rabbit parietal cells.
Am J Physiol Cell Physiol 259: C536 C543, 1990.
706. Malinowska DH, Kupert EY, Bahinski A, Sherry AM, Cuppoletti J. Cloning, functional
expression, and characterization of a PKA-activated gastric Cl channel. Am J Physiol
Cell Physiol 268: C191C200, 1995.
707. Malinowska DH, Sherry AM, Tewari KP, Cuppoletti J. Gastric parietal cell secretory
membrane contains PKA- and acid-activated Kir2.1 K channels. Am J Physiol Cell
Physiol Cell Physiol 286: C495C506, 2004.
708. Manela FD, Ren J, Gao J, McGuigan JE, Harty RF. Calcitonin gene-related peptide
modulates acid-mediated regulation of somatostatin and gastrin release from rat
antrum. Gastroenterology 109: 701706, 1995.
709. Manolagas SC, Burton DW, Deftos LJ. 1,25-Dihydroxyvitamin D3 stimulates the
alkaline phosphatase activity of osteoblast-like cells. J Biol Chem 256: 71157117,
1981.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

255

Downloaded from on December 8, 2014

674. Lindstrom E, Hakanson R. Neurohormonal regulation of secretion from isolated rat


stomach ECL cells: a critical reappraisal. Regul Pept 97: 169 180, 2001.

SASCHA KOPIC AND JOHN P. GEIBEL


710. Marcus CS, Lengemann FW. Absorption of Ca45 and Sr85 from solid and liquid food
at various levels of the alimentary tract of the rat. J Nutr 77: 155160, 1962.
711. Marie PJ. The calcium-sensing receptor in bone cells: a potential therapeutic target in
osteoporosis. Bone 46: 571576, 2010.

731. Melander T, Hokfelt T, Rokaeus A, Fahrenkrug J, Tatemoto K, Mutt V. Distribution


of galanin-like immunoreactivity in the gastro-intestinal tract of several mammalian
species. Cell Tissue Res 239: 253270, 1985.
732. Melick RA, Benson JA Jr. Osteomalacia following partial gastrectomy. N Engl J Med
260: 976 978, 1959.

712. Marks HD, Fleet JC, Peleg S. Transgenic expression of the human Vitamin D receptor (hVDR) in the duodenum of VDR-null mice attenuates the age-dependent decline
in calcium absorption. J Steroid Biochem Mol Biol 103: 513516, 2007.

733. Mellanby E. An experimental investigation on rickets. Lancet 193: 407 412, 1919.

713. Martin DL, DeLuca HF. Influence of sodium on calcium transport by the rat small
intestine. Am J Physiol 216: 13511359, 1969.

734. Mentaverri R, Yano S, Chattopadhyay N, Petit L, Kifor O, Kamel S, Terwilliger EF,


Brazier M, Brown EM. The calcium sensing receptor is directly involved in both
osteoclast differentiation and apoptosis. FASEB J 20: 25622564, 2006.

714. Martin SR, Linse S, Johansson C, Bayley PM, Forsen S. Protein surface charges and
Ca2 binding to individual sites in calbindin D9k: stopped-flow studies. Biochemistry
29: 4188 4193, 1990.

735. Meredith SC, Bolt MJ, Rosenberg IH. The supramolecular structure of vitamin D3 in
water. J Colloid Interface Sci 99: 244 255, 1984.

715. Martinez V, Curi AP, Torkian B, Schaeffer JM, Wilkinson HA, Walsh JH, Tache Y.
High basal gastric acid secretion in somatostatin receptor subtype 2 knockout mice.
Gastroenterology 114: 11251132, 1998.

736. Meyer MB, Watanuki M, Kim S, Shevde NK, Pike JW. The human transient receptor
potential vanilloid type 6 distal promoter contains multiple vitamin D receptor binding sites that mediate activation by 1,25-dihydroxyvitamin D3 in intestinal cells. Mol
Endocrinol 20: 14471461, 2006.

716. Martz A, Forte LR, Langeluttig SG. Renal cAMP and 1,25(OH)2D3 synthesis in estrogen-treated chick embryos and hens. Am J Physiol Endocrinol Metab 249: E626
E633, 1985.
717. Massheimer V, Boland R, de Boland AR. Rapid 1,25(OH)2-vitamin D3 stimulation of
calcium uptake by rat intestinal cells involves a dihydropyridine-sensitive cAMPdependent pathway. Cell Signal 6: 299 304, 1994.

719. Masuda Y, Tanaka T, Inomata N, Ohnuma N, Tanaka S, Itoh Z, Hosoda H, Kojima M,


Kangawa K. Ghrelin stimulates gastric acid secretion and motility in rats. Biochem
Biophys Res Commun 276: 905908, 2000.
720. Mate L, Sakamoto T, Greeley GH Jr, Thompson JC. Regulation of gastric acid secretion by secretin and serotonin. Am J Surg 149: 40 45, 1985.
721. Matloff DS, Kaplan MM, Neer RM, Goldberg MJ, Bitman W, Wolfe HJ. Osteoporosis
in primary biliary cirrhosis: effects of 25-hydroxyvitamin D3 treatment. Gastroenterology 83: 97102, 1982.
722. Matsumoto M, Park J, Sugano K, Yamada T. Biological activity of progastrin posttranslational processing intermediates. Am J Physiol Gastrointest Liver Physiol 252:
G315G319, 1987.
723. Matsuoka LY, Ide L, Wortsman J, MacLaughlin JA, Holick MF. Sunscreens suppress
cutaneous vitamin D3 synthesis. J Clin Endocrinol Metab 64: 11651168, 1987.
724. Mawer EB, Taylor CM, Backhouse J, Lumb GA, Stanbury SW. Failure of formation of
1,25-dihydroxycholecalciferol in chronic renal insufficiency. Lancet 1: 626 628,
1973.
725. Mayer G, Arnold R, Feurle G, Fuchs K, Ketterer H, Track NS, Creutzfeldt W.
Influence of feeding and sham feeding upon serum gastrin and gastric acid secretion
in control subjects and duodenal ulcer patients. Scand J Gastroenterol 9: 703710,
1974.
726. Mayer GP, Keaton JA, Hurst JG, Habener JF. Effects of plasma calcium concentration
on the relative proportion of hormone and carboxyl fragments in parathyroid venous
blood. Endocrinology 104: 1778 1784, 1979.
727. McCollum EV, Simmonds N, Becker JE, Shipley PG. Studies on experimental rickets.
XXI. An experimental demonstration of the existence of a vitamin which promotes
calcium deposition. J Biol Chem 53: 293312, 1922.
728. McGehee DS, Aldersberg M, Liu KP, Hsuing S, Heath MJ, Tamir H. Mechanism of
extracellular Ca2 receptor-stimulated hormone release from sheep thyroid parafollicular cells. J Physiol 502: 31 44, 1997.
729. McLaughlin JT, Ai W, Sinclair NF, Colucci R, Raychowdhury R, Koh TJ, Wang TC.
PACAP and gastrin regulate the histidine decarboxylase promoter via distinct mechanisms. Am J Physiol Gastrointest Liver Physiol 286: G51G59, 2004.
730. Mee AP, Hoyland JA, Braidman IP, Freemont AJ, Davies M, Mawer EB. Demonstration of vitamin D receptor transcripts in actively resorbing osteoclasts in bone sections. Bone 18: 295299, 1996.

256

738. Michelangeli F, Ruiz MC, Fernandez E, Ciarrocchi A. Role of Ca2 in H transport by


rabbit gastric glands studied with A23187 and BAPTA, an incorporated Ca2 chelator. Biochim Biophys Acta 983: 8290, 1989.
739. Millen AE, Dodd KW, Subar AF. Use of vitamin, mineral, nonvitamin, and nonmineral
supplements in the United States: The 1987, 1992, and 2000 National Health Interview Survey results. J Am Diet Assoc 104: 942950, 2004.
740. Miller A 3rd, Bronner F. Calcium uptake in isolated brush-border vesicles from rat
small intestine. Biochem J 196: 391 401, 1981.
741. Milne ML, Baran DT. End product inhibition of hepatic 25-hydroxyvitamin D production in the rat: specificity and kinetics. Arch Biochem Biophys 242: 488 492, 1985.
742. Mircheff AK, Wright EM. Analytical isolation of plasma membranes of intestinal
epithelial cells: identification of Na, K-ATPase rich membranes and the distribution
of enzyme activities. J Membr Biol 28: 309 333, 1976.
743. Mitchell HH, Hamiltom TS, Steggerda FR, Bean HW. The chemical composition of
the adult human body and its bearing on the biochemistry of growth. J Biol Chem 158:
625 637, 1945.
744. Mitsuma T, Rhue N, Kayama M, Mori Y, Adachi K, Yokoi Y, Ping J, Nogimori T,
Hirooka Y. Distribution of calcium sensing receptor in rats: an immunohistochemical
study. Endocr Regul 33: 5559, 1999.
745. Miyake N, Hoshi K, Sano Y, Kikuchi K, Tadano K, Koshihara Y. 1,25-Dihydroxyvitamin D3 promotes vitamin K2 metabolism in human osteoblasts. Osteoporos Int 12:
680 687, 2001.
746. Mizobuchi M, Ogata H, Hatamura I, Saji F, Koiwa F, Kinugasa E, Koshikawa S,
Akizawa T. Activation of calcium-sensing receptor accelerates apoptosis in hyperplastic parathyroid cells. Biochem Biophys Res Commun 362: 1116, 2007.
747. Mogard MH, Maxwell V, Sytnik B, Walsh JH. Regulation of gastric acid secretion by
neurotensin in man. Evidence against a hormonal role. J Clin Invest 80: 1064 1067,
1987.
748. Monkawa T, Yoshida T, Wakino S, Shinki T, Anazawa H, Deluca HF, Suda T, Hayashi
M, Saruta T. Molecular cloning of cDNA and genomic DNA for human 25-hydroxyvitamin D3 1 alpha-hydroxylase. Biochem Biophys Res Commun 239: 527533, 1997.
749. Monstein HJ, Nylander AG, Salehi A, Chen D, Lundquist I, Hakanson R. Cholecystokinin-A and cholecystokinin-B/gastrin receptor mRNA expression in the gastrointestinal tract and pancreas of the rat and man. A polymerase chain reaction study. Scand
J Gastroenterol 31: 383390, 1996.
750. Morgan EL, Mace OJ, Affleck J, Kellett GL. Apical GLUT2 and Cav1.3: regulation of
rat intestinal glucose and calcium absorption. J Physiol 580: 593 604, 2007.
751. Morgan EL, Mace OJ, Helliwell PA, Affleck J, Kellett GL. A role for Ca(v)1.3 in rat
intestinal calcium absorption. Biochem Biophys Res Commun 312: 487 493, 2003.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

718. Massry SG, Coburn JW, Chapman LW, Kleeman CR. Role of serum Ca, parathyroid
hormone, NaCl infusion on renal Ca and Na clearances. Am J Physiol 214: 1403
1409, 1968.

737. Miampamba M, Germano PM, Arli S, Wong HH, Scott D, Tache Y, Pisegna JR.
Expression of pituitary adenylate cyclase-activating polypeptide and PACAP type 1
receptor in the rat gastric and colonic myenteric neurons. Regul Pept 105: 145154,
2002.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


752. Morii H, Lund J, Neville PF, DeLuca HF. Biological activity of a vitamin D metabolite.
Arch Biochem Biophys 120: 508 512, 1967.

from ECL carcinoid tumor of Mastomys natalensis. Biochem Biophys Res Commun
187: 11511157, 1992.

753. Mosekilde L, Sogaard CH, Danielsen CC, Torring O. The anabolic effects of human
parathyroid hormone (hPTH) on rat vertebral body mass are also reflected in the
quality of bone, assessed by biomechanical testing: a comparison study between
hPTH-(134) and hPTH-(1 84). Endocrinology 129: 421 428, 1991.

773. Nandi J, Bosche MC, Levine RA. Effects of a phorbol ester and isoquinoline sulfonamides on rabbit parietal cell function. J Pharmacol Exp Ther 279: 97105, 1996.
774. Nandi J, Loo A, Kim SW, Levine RA. Expression and characterization of protein
kinase C in isolated rabbit parietal cells. Int J Mol Med 3: 521526, 1999.

754. Motoyama HI, Friedman PA. Calcium-sensing receptor regulation of PTH-dependent calcium absorption by mouse cortical ascending limbs. Am J Physiol Renal Physiol
283: F399 F406, 2002.

775. Napoli N, Pedone C, Pozzilli P, Lauretani F, Bandinelli S, Ferrucci L, Incalzi RA. Effect
of ghrelin on bone mass density: the InChianti study. Bone 49: 257263, 2011.

755. Muallem S, Fimmel CJ, Pandol SJ, Sachs G. Regulation of free cytosolic Ca2 in the
peptic and parietal cells of the rabbit gastric gland. J Biol Chem 261: 2660 2667,
1986.

776. Naro F, Perez M, Migliaccio S, Galson DL, Orcel P, Teti A, Goldring SR. Phospholipase D- and protein kinase C isoenzyme-dependent signal transduction pathways
activated by the calcitonin receptor. Endocrinology 139: 32413248, 1998.

756. Muallem S, Karlish SJ. Studies on the mechanism of regulation of the red-cell Ca2
pump by calmodulin and ATP. Biochim Biophys Acta 647: 73 86, 1981.

777. Natalucci G, Riedl S, Gleiss A, Zidek T, Frisch H. Spontaneous 24-h ghrelin secretion
pattern in fasting subjects: maintenance of a meal-related pattern. Eur J Endocrinol
152: 845 850, 2005.

757. Muhe L, Lulseged S, Mason KE, Simoes EA. Case-control study of the role of nutritional rickets in the risk of developing pneumonia in Ethiopian children. Lancet 349:
18011804, 1997.
758. Mulderry PK, Ghatei MA, Spokes RA, Jones PM, Pierson AM, Hamid QA, Kanse S,
Amara SG, Burrin JM, Legon S. Differential expression of alpha-CGRP and betaCGRP by primary sensory neurons and enteric autonomic neurons of the rat. Neuroscience 25: 195205, 1988.

760. Mungan Z, Hammer RA, Akarca US, Komaki G, Ertan A, Arimura A. Effect of PACAP
on gastric acid secretion in rats. Peptides 16: 10511056, 1995.
761. Munson K, Garcia R, Sachs G. Inhibitor and ion binding sites on the gastric H-KATPase. Biochemistry 44: 52675284, 2005.
762. Munson K, Law RJ, Sachs G. Analysis of the gastric H,K ATPase for ion pathways and
inhibitor binding sites. Biochemistry 46: 5398 5417, 2007.
763. Murayama A, Takeyama K, Kitanaka S, Kodera Y, Hosoya T, Kato S. The promoter
of the human 25-hydroxyvitamin D3 1 alpha-hydroxylase gene confers positive and
negative responsiveness to PTH, calcitonin, and 1 alpha,25(OH)2D3. Biochem Biophys Res Commun 249: 1116, 1998.
764. Murayama A, Takeyama K, Kitanaka S, Kodera Y, Kawaguchi Y, Hosoya T, Kato S.
Positive and negative regulations of the renal 25-hydroxyvitamin D3 1alpha-hydroxylase gene by parathyroid hormone, calcitonin, and 1alpha,25(OH)2D3 in intact
animals. Endocrinology 140: 2224 2231, 1999.
765. Murer H, Hernando N, Forster I, Biber J. Proximal tubular phosphate reabsorption:
molecular mechanisms. Physiol Rev 80: 13731409, 2000.
766. Murray CB, Morales MM, Flotte TR, McGrath-Morrow SA, Guggino WB, Zeitlin PL.
CIC-2: a developmentally dependent chloride channel expressed in the fetal lung and
downregulated after birth. Am J Respir Cell Mol Biol 12: 597 604, 1995.

779. Nemere I. The 1,25D3-MARRS protein: contribution to steroid stimulated calcium


uptake in chicks and rats. Steroids 70: 455 457, 2005.
780. Nemere I, Norman AW. 1,25-Dihydroxyvitamin D3-mediated vesicular transport of
calcium in intestine: time-course studies. Endocrinology 122: 29622969, 1988.
781. Nemere I, Norman AW. Parathyroid hormone stimulates calcium transport in perfused duodena from normal chicks: comparison with the rapid (transcaltachic) effect
of 1,25-dihydroxyvitamin D3. Endocrinology 119: 1406 1408, 1986.
782. Nemere I, Safford SE, Rohe B, DeSouza MM, Farach-Carson MC. Identification and
characterization of 1,25D3-membrane-associated rapid response, steroid (1,25D3MARRS) binding protein. J Steroid Biochem Mol Biol 89 90: 281285, 2004.
783. Nemere I, Szego CM. Early actions of parathyroid hormone and 1,25-dihydroxycholecalciferol on isolated epithelial cells from rat intestine. I. Limited lysosomal
enzyme release and calcium uptake. Endocrinology 108: 1450 1462, 1981.
784. Nemere I, Szego CM. Early actions of parathyroid hormone and 1,25-dihydroxycholecalciferol on isolated epithelial cells from rat intestine. II. Analyses of additivity,
contribution of calcium, and modulatory influence of indomethacin. Endocrinology
109: 2180 2187, 1981.
785. Nemere I, Yoshimoto Y, Norman AW. Calcium transport in perfused duodena from
normal chicks: enhancement within fourteen minutes of exposure to 1,25-dihydroxyvitamin D3. Endocrinology 115: 1476 1483, 1984.
786. Nguyen-Yamamoto L, Rousseau L, Brossard JH, Lepage R, DAmour P. Synthetic
carboxyl-terminal fragments of parathyroid hormone (PTH) decrease ionized calcium concentration in rats by acting on a receptor different from the PTH/PTHrelated peptide receptor. Endocrinology 142: 1386 1392, 2001.

767. Mutt V, Jorpes JE. Structure of porcine cholecystokinin-pancreozymin. 1. Cleavage


with thrombin and with trypsin. Eur J Biochem 6: 156 162, 1968.

787. Nguyen TM, Lieberherr M, Fritsch J, Guillozo H, Alvarez ML, Fitouri Z, Jehan F,
Garabedian M. The rapid effects of 1,25-dihydroxyvitamin D3 require the vitamin D
receptor and influence 24-hydroxylase activity: studies in human skin fibroblasts
bearing vitamin D receptor mutations. J Biol Chem 279: 75917597, 2004.

768. Nakajima K, Nohtomi K, Sato M, Takano K, Sato K. PTH(7 84) inhibits PTH(134)induced 1,25-(OH)2D3 production in murine renal tubules. Biochem Biophys Res
Commun 381: 283287, 2009.

788. Niall HD, Keutmann H, Sauer R, Hogan M, Dawson B, Aurbach G, Potts J Jr. The
amino acid sequence of bovine parathyroid hormone I. Hoppe-Seylers Z Physiol Chem
351: 1586 1588, 1970.

769. Nakajima T, Konda Y, Izumi Y, Kanai M, Hayashi N, Chiba T, Takeuchi T. Gastrin


stimulates the growth of gastric pit cell precursors by inducing its own receptors. Am
J Physiol Gastrointest Liver Physiol 282: G359 G366, 2002.

789. Nicar MJ, Pak CY. Calcium bioavailability from calcium carbonate and calcium citrate. J Clin Endocrinol Metab 61: 391393, 1985.

770. Nakamura E, Hasumura M, Gabriel AS, Uneyama H, Torii K. Functional role of


calcium-sensing receptor on somatostatin release from rat gastric mucosa (Abstract). Gastroenterology 138: S404, 2010.
771. Nakamura K, Zhou CJ, Parente J, Chew CS. Parietal cell MAP kinases: multiple
activation pathways. Am J Physiol Gastrointest Liver Physiol 271: G640 G649,
1996.
772. Nakata H, Matsui T, Ito M, Taniguchi T, Naribayashi Y, Arima N, Nakamura A,
Kinoshita Y, Chihara K, Hosoda S. Cloning and characterization of gastrin receptor

790. Nicholson GC, Moseley JM, Sexton PM, Mendelsohn FA, Martin TJ. Abundant calcitonin receptors in isolated rat osteoclasts. Biochemical and autoradiographic characterization. J Clin Invest 78: 355360, 1986.
791. Niemeyer BA, Bergs C, Wissenbach U, Flockerzi V, Trost C. Competitive regulation
of CaT-like-mediated Ca2 entry by protein kinase C and calmodulin. Proc Natl Acad
Sci USA 98: 3600 3605, 2001.
792. Nijenhuis T, Hoenderop JG, van der Kemp AW, Bindels RJ. Localization and regulation of the epithelial Ca2 channel TRPV6 in the kidney. J Am Soc Nephrol 14:
27312740, 2003.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

257

Downloaded from on December 8, 2014

759. Mun HC, Franks AH, Culverston EL, Krapcho K, Nemeth EF, Conigrave AD. The
Venus Fly Trap domain of the extracellular Ca2-sensing receptor is required for
L-amino acid sensing. J Biol Chem 279: 51739 51744, 2004.

778. Naveh-Many T, Rahamimov R, Livni N, Silver J. Parathyroid cell proliferation in


normal and chronic renal failure rats. The effects of calcium, phosphate, and vitamin
D. J Clin Invest 96: 1786 1793, 1995.

SASCHA KOPIC AND JOHN P. GEIBEL


793. Nilas L, Christiansen C. Vitamin D deficiency after highly selective vagotomy. Acta
Med Scand 221: 303306, 1987.
794. Nilas L, Christiansen C, Christiansen J. Regulation of vitamin D and calcium metabolism after gastrectomy. Gut 26: 252257, 1985.
795. Nilsson BE, Westlin NE. The fracture incidence after gastrectomy. Acta Chir Scand
137: 533534, 1971.
796. Nishimura M, Yaguti H, Yoshitsugu H, Naito S, Satoh T. Tissue distribution of mRNA
expression of human cytochrome P450 isoforms assessed by high-sensitivity realtime reverse transcription PCR. Yakugaku Zasshi 123: 369 375, 2003.
797. Nordin BE. Measurement and meaning of calcium absorption. Gastroenterology 54:
294 301, 1968.
798. Norlen P, Bernsand M, Konagaya T, Hakanson R. ECL-cell histamine mobilization in conscious rats: effects of locally applied regulatory peptides, candidate
neurotransmitters and inflammatory mediators. Br J Pharmacol 134: 17671777,
2001.
799. Norman AW, Lund J, Deluca HF. Biologically active forms of vitamin D3 in kidney and
intestine. Arch Biochem Biophys 108: 1221, 1964.
800. Norman AW, Myrtle JF, Midgett RJ, Nowicki HG, Williams V, Popjak G. 1,25Dihydroxycholecalciferol: identification of the proposed active form of vitamin D3 in
the intestine. Science 173: 5154, 1971.

802. Norman AW, Olivera CJ, Barreto Silva FR, Bishop JE. A specific binding protein/
receptor for 1alpha,25-dihydroxyvitamin D(3) is present in an intestinal caveolae
membrane fraction. Biochem Biophys Res Commun 298: 414 419, 2002.

814. Okunade GW, Miller ML, Pyne GJ, Sutliff RL, OConnor KT, Neumann JC, Andringa
A, Miller DA, Prasad V, Doetschman T, Paul RJ, Shull GE. Targeted ablation of plasma
membrane Ca2-ATPase (PMCA) 1 and 4 indicates a major housekeeping function
for PMCA1 and a critical role in hyperactivated sperm motility and male fertility for
PMCA4. J Biol Chem 279: 3374233750, 2004.
815. Olesen C, Picard M, Winther AML, Gyrup C, Morth JP, Oxvig C, Moller JV, Nissen
P. The structural basis of calcium transport by the calcium pump. Nature 450:
1036 1042, 2007.
816. Oscarson J, Hakanson R, Liedberg G, Lundqvist G, Sundler F, Thorell J. Variated
serum gastrin concentration: trophic effects on the gastrointestinal tract of the rat.
Acta Physiol Scand Suppl 475: 227, 1979.
817. Owen TA, Aronow MS, Barone LM, Bettencourt B, Stein GS, Lian JB. Pleiotropic
effects of vitamin D on osteoblast gene expression are related to the proliferative
and differentiated state of the bone cell phenotype: dependency upon basal levels of
gene expression, duration of exposure, and bone matrix competency in normal rat
osteoblast cultures. Endocrinology 128: 1496 1504, 1991.
818. Owyang C, Miller LJ, DiMagno EP, Brennan LA Jr, Go VL. Gastrointestinal hormone
profile in renal insufficiency. Mayo Clin Proc 54: 769 773, 1979.
819. Paillard F, Ardaillou R, Malendin H, Fillastre JP, Prier S. Renal effects of salmon
calcitonin in man. J Lab Clin Med 80: 200 216, 1972.
820. Pan Q, Ma J, Zhou Q, Li J, Tang Y, Liu Y, Yang Y, Xiao J, Peng L, Li P, Liang D, Zhang
H, Chen YH. KCNQ1 loss-of-function mutation impairs gastric acid secretion in
mice. Mol Biol Rep 37: 1329 1333, 2010.
821. Panda DK, Miao D, Bolivar I, Li J, Huo R, Hendy GN, Goltzman D. Inactivation of the
25-hydroxyvitamin D 1alpha-hydroxylase and vitamin D receptor demonstrates independent and interdependent effects of calcium and vitamin D on skeletal and
mineral homeostasis. J Biol Chem 279: 16754 16766, 2004.

803. Noto T, Nagasaki M, Endo T. Role of vagus nerves and gastrin in the gastric phase of
acid secretion in male anesthetized rats. Am J Physiol Gastrointest Liver Physiol 272:
G335G339, 1997.

822. Panda DK, Miao D, Tremblay ML, Sirois J, Farookhi R, Hendy GN, Goltzman D.
Targeted ablation of the 25-hydroxyvitamin D 1alpha-hydroxylase enzyme: evidence for skeletal, reproductive, and immune dysfunction. Proc Natl Acad Sci USA 98:
7498 7503, 2001.

804. Nuttall ME, Patton AJ, Olivera DL, Nadeau DP, Gowen M. Human trabecular bone
cells are able to express both osteoblastic and adipocytic phenotype: implications for
osteopenic disorders. J Bone Miner Res 13: 371382, 1998.

823. Pannabecker TL, Chandler JS, Wasserman RH. Vitamin-D-dependent transcriptional


regulation of the intestinal plasma membrane calcium pump. Biochem Biophys Res
Commun 213: 499 505, 1995.

805. Nykjaer A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz J, Melsen F, Christensen EI, Willnow TE. An endocytic pathway essential for renal uptake and activation of the steroid 25-(OH) vitamin D3. Cell 96: 507515, 1999.

824. Pansu D, Bellaton C, Bronner F. Effect of Ca intake on saturable and nonsaturable


components of duodenal Ca transport. Am J Physiol Gastrointest Liver Physiol 240:
G32G37, 1981.

806. Nykjaer A, Fyfe JC, Kozyraki R, Leheste JR, Jacobsen C, Nielsen MS, Verroust PJ,
Aminoff M, de la Chapelle A, Moestrup SK, Ray R, Gliemann J, Willnow TE, Christensen EI. Cubilin dysfunction causes abnormal metabolism of the steroid hormone
25(OH) vitamin D(3). Proc Natl Acad Sci USA 98: 1389513900, 2001.

825. Pansu D, Bellaton C, Roche C, Bronner F. Duodenal and ileal calcium absorption in
the rat and effects of vitamin D. Am J Physiol Gastrointest Liver Physiol 244: G695
G700, 1983.

807. OConnell MB, Madden DM, Murray AM, Heaney RP, Kerzner LJ. Effects of proton
pump inhibitors on calcium carbonate absorption in women: a randomized crossover trial. Am J Med 118: 778 781, 2005.
808. Offermanns S, Iida-Klein A, Segre GV, Simon MI. G alpha q family members couple
parathyroid hormone (PTH)/PTH-related peptide and calcitonin receptors to phospholipase C in COS-7 cells. Mol Endocrinol 10: 566 574, 1996.
809. Oginni LM, Worsfold M, Oyelami OA, Sharp CA, Powell DE, Davie MW. Etiology of
rickets in Nigerian children. J Pediatr 128: 692 694, 1996.
810. Oh DS, Lieu SN, Yamaguchi DJ, Tachiki K, Lambrecht N, Ohning GV, Sachs G,
Germano PM, Pisegna JR. PACAP regulation of secretion and proliferation of pure
populations of gastric ECL cells. J Mol Neurosci 26: 8597, 2005.
811. Oh KW, Lee WY, Rhee EJ, Baek KH, Yoon KH, Kang MI, Yun EJ, Park CY, Ihm SH,
Choi MG, Yoo HJ, Park SW. The relationship between serum resistin, leptin, adiponectin, ghrelin levels and bone mineral density in middle-aged men. Clin Endocrinol
63: 131138, 2005.
812. Ohyama Y, Ozono K, Uchida M, Yoshimura M, Shinki T, Suda T, Yamamoto O.
Functional assessment of two vitamin D-responsive elements in the rat 25-hydroxyvitamin D3 24-hydroxylase gene. J Biol Chem 271: 3038130385, 1996.

258

826. Parhon C, Ureche CS. Untersuchungen uber den Einfluss den die Calcium und
Sodiumsalze auf den Verlauf der experimentellen Tetanie ausuben. Neurol Centr 26:
1099, 1907.
827. Park J, Chiba T, Yamada T. Mechanisms for direct inhibition of canine gastric parietal
cells by somatostatin. J Biol Chem 262: 14190 14196, 1987.
828. Parkinson DB, Thakker RV. A donor splice site mutation in the parathyroid hormone
gene is associated with autosomal recessive hypoparathyroidism. Nat Genet 1: 149
152, 1992.
829. Parthemore JG, Deftos LJ. Calcitonin secretion in normal human subjects. J Clin
Endocrinol Metab 47: 184 188, 1978.
830. Patterson EK, Hodsman AB, Hendy GN, Canaff L, Bringhurst FR, Fraher LJ. Functional analysis of a type 1 parathyroid hormone receptor intracellular tail mutant
[KRK(484 6)AAA]: effects on second messenger generation and cellular targeting.
Bone 46: 1180 1187, 2010.
831. Patton MB. Further experiments on the utilization of calcium from salts by college
women. J Nutr 55: 519 526, 1955.
832. Patton MB, Sutton TS. The utilization of calcium from lactate, gluconate, sulfate and
carbonate salts by young college women. J Nutr 48: 443 452, 1952.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

801. Norman AW, Okamura WH, Hammond MW, Bishop JE, Dormanen MC, Bouillon R, van Baelen H, Ridall AL, Daane E, Khoury R, Farach-Carson MC. Comparison of 6-s-cis- and 6-s-trans-locked analogs of 1alpha,25-dihydroxyvitamin
D3 indicates that the 6-s-cis conformation is preferred for rapid nongenomic
biological responses and that neither 6-s-cis- nor 6-s-trans-locked analogs are
preferred for genomic biological responses. Mol Endocrinol 11: 1518 1531,
1997.

813. Okamoto CT, Karam SM, Jeng YY, Forte JG, Goldenring JR. Identification of clathrin
and clathrin adaptors on tubulovesicles of gastric acid secretory (oxyntic) cells. Am J
Physiol Cell Physiol 274: C1017C1029, 1998.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


833. Pawlow JP, Schumowa-Simanowskaja EO. Beitrage zur Physiologie der Absonderungen. Die Innervation der Magendrusen beim Hunde. Arch Anat Physiol 54 69,
1895.

854. Pi M, Zhang L, Lei SF, Huang MZ, Zhu W, Zhang J, Shen H, Deng HW, Quarles LD.
Impaired osteoblast function in GPRC6A null mice. J Bone Miner Res 25: 10921102,
2010.

834. Pearce SH, Bai M, Quinn SJ, Kifor O, Brown EM, Thakker RV. Functional characterization of calcium-sensing receptor mutations expressed in human embryonic kidney
cells. J Clin Invest 98: 1860 1866, 1996.

855. Picard N, Capuano P, Stange G, Mihailova M, Kaissling B, Murer H, Biber J, Wagner


CA. Acute parathyroid hormone differentially regulates renal brush border membrane phosphate cotransporters. Pflgers Arch 460: 677 687, 2010.

835. Pedersen JI, Holmberg I, Bjorkhem I. Reconstitution of vitamin D3 25-hydroxylase


activity with a cytochrome P-450 preparation from rat liver mitochondria. FEBS Lett
98: 394 398, 1979.

856. Pickard BW, Hodsman AB, Fraher LJ, Watson PH. Type 1 parathyroid hormone
receptor (PTH1R) nuclear trafficking: association of PTH1R with importin alpha1
and beta. Endocrinology 147: 3326 3332, 2006.

836. Peng JB, Brown EM, Hediger MA. Structural conservation of the genes encoding
CaT1, CaT2, and related cation channels. Genomics 76: 99 109, 2001.
837. Peng JB, Chen XZ, Berger UV, Vassilev PM, Brown EM, Hediger MA. A rat kidneyspecific calcium transporter in the distal nephron. J Biol Chem 275: 28186 28194,
2000.
838. Peng JB, Chen XZ, Berger UV, Vassilev PM, Tsukaguchi H, Brown EM, Hediger MA.
Molecular cloning and characterization of a channel-like transporter mediating intestinal calcium absorption. J Biol Chem 274: 22739 22746, 1999.
839. Peng JB, Zhuang L, Berger UV, Adam RM, Williams BJ, Brown EM, Hediger MA,
Freeman MR. CaT1 expression correlates with tumor grade in prostate cancer.
Biochem Biophys Res Commun 282: 729 734, 2001.
840. Perez JF, Ruiz MC, Michelangeli F. Simultaneous measurement and imaging of intracellular Ca2 and H transport in isolated rabbit gastric glands. J Physiol 537: 735
745, 2001.

842. Persson P, Gagnemo-Persson R, Orberg J, Chen D, Hakanson R. Effects of gastrin on


calcium homeostasis in chickens. Endocrinology 129: 11621166, 1991.
843. Persson P, Grunditz T, Axelson J, Sundler F, Hakanson R. Cholecystokinins but not
gastrin-17 release calcitonin from thyroid C-cells in the rat. Regul Pept 21: 4556,
1988.
844. Persson P, Hakanson R, Axelson J, Sundler F. Gastrin releases a blood calciumlowering peptide from the acid-producing part of the rat stomach. Proc Natl Acad Sci
USA 86: 2834 2838, 1989.
845. Petrovic S, Wang Z, Ma L, Seidler U, Forte JG, Shull GE, Soleimani M. Colocalization
of the apical Cl/HCO3 exchanger PAT1 and gastric H-K-ATPase in stomach
parietal cells. Am J Physiol Gastrointest Liver Physiol 283: G1207G1216, 2002.
846. Pfeiffer A, Rochlitz H, Noelke B, Tacke R, Moser U, Mutschler E, Lambrecht G.
Muscarinic receptors mediating acid secretion in isolated rat gastric parietal cells are
of M3 type. Gastroenterology 98: 218 222, 1990.
847. Pfister MF, Lederer E, Forgo J, Ziegler U, Lotscher M, Quabius ES, Biber J, Murer H.
Parathyroid hormone-dependent degradation of type II Na/Pi cotransporters. J Biol
Chem 272: 2012520130, 1997.
848. Phelps CB, Huang RJ, Lishko PV, Wang RR, Gaudet R. Structural analyses of the
ankyrin repeat domain of TRPV6 and related TRPV ion channels. Biochemistry 47:
2476 2484, 2008.
849. Pi M, Chen L, Huang M, Luo Q, Quarles LD. Parathyroid-specific interaction of the
calcium-sensing receptor and G alpha q. Kidney Int 74: 1548 1556, 2008.
850. Pi M, Chen L, Huang MZ, Zhu W, Ringhofer B, Luo J, Christenson L, Li B, Zhang J,
Jackson PD, Faber P, Brunden KR, Harrington JJ, Quarles LD. GPRC6A null mice
exhibit osteopenia, feminization and metabolic syndrome. PLoS One 3: e3858, 2008.
851. Pi M, Faber P, Ekema G, Jackson PD, Ting A, Wang N, Fontilla-Poole M, Mays RW,
Brunden KR, Harrington JJ, Quarles LD. Identification of a novel extracellular cationsensing G-protein-coupled receptor. J Biol Chem 280: 40201 40209, 2005.
852. Pi M, Garner SC, Flannery P, Spurney RF, Quarles LD. Sensing of extracellular
cations in CasR-deficient osteoblasts. Evidence for a novel cation-sensing mechanism. J Biol Chem 275: 3256 3263, 2000.
853. Pi M, Oakley RH, Gesty-Palmer D, Cruickshank RD, Spurney RF, Luttrell LM, Quarles LD. Beta-arrestin- and G protein receptor kinase-mediated calcium-sensing receptor desensitization. Mol Endocrinol 19: 1078 1087, 2005.

858. Pidasheva S, Grant M, Canaff L, Ercan O, Kumar U, Hendy GN. Calcium-sensing


receptor dimerizes in the endoplasmic reticulum: biochemical and biophysical characterization of CASR mutants retained intracellularly. Hum Mol Genet 15: 2200
2209, 2006.
859. Pines M, Fukayama S, Costas K, Meurer E, Goldsmith PK, Xu X, Muallem S, Behar V,
Chorev M, Rosenblatt M, Tashjian AH Jr, Suva LJ. Inositol 1-,4-,5-trisphosphatedependent Ca2 signaling by the recombinant human PTH/PTHrP receptor stably
expressed in a human kidney cell line. Bone 18: 381389, 1996.
860. Pioszak AA, Harikumar KG, Parker NR, Miller LJ, Xu HE. Dimeric arrangement of
the parathyroid hormone receptor and a structural mechanism for ligand-induced
dissociation. J Biol Chem 285: 1243512444, 2010.
861. Pioszak AA, Xu HE. Molecular recognition of parathyroid hormone by its G proteincoupled receptor. Proc Natl Acad Sci USA 105: 5034 5039, 2008.
862. Piqueras L, Tache Y, Martinez V. Peripheral PACAP inhibits gastric acid secretion
through somatostatin release in mice. Br J Pharmacol 142: 6778, 2004.
863. Polak JM, Sullivan SN, Bloom SR, Buchan AM, Facer P, Brown MR, Pearse AG.
Specific localisation of neurotensin to the N cell in human intestine by radioimmunoassay and immunocytochemistry. Nature 270: 183184, 1977.
864. Ponchon G, DeLuca HF. The role of the liver in the metabolism of vitamin D. J Clin
Invest 48: 12731279, 1969.
865. Ponchon G, Kennan AL, DeLuca HF. Activation of vitamin D by the liver. J Clin
Invest 48: 20322037, 1969.
866. Popielski L. Beta-imidazolylathylamin und die Organextrakte. Erster Teil: beta-imidazolylathylamin als machtiger Erreger der Magendrusen. Pflgers Arch 178: 214
236, 1920.
867. Potts JT Jr, Tregear GW, Keutmann HT, Niall HD, Sauer R, Deftos LJ, Dawson BF,
Hogan ML, Aurbach GD. Synthesis of a biologically active N-terminal tetratriacontapeptide of parathyroid hormone. Proc Natl Acad Sci USA 68: 63 67, 1971.
868. Pouwels S, Lalmohamed A, Souverein P, Cooper C, Veldt BJ, Leufkens HG, De
Boer A, Van Staa T, De Vries F. Use of proton pump inhibitors and risk of
hip/femur fracture: A population-based case-control study. Osteoporosis Int 22:
903910, 2011.
869. Prader A, Illig R, Heierli E. Eine besondere Form der primren Vitamin D resistenten
Tachitis mit Hypocalcemie und autosomal dominantem Erbgang: die hereditre
Pseudomangelrachitis. Helv Paediatr Acta 16: 452 468, 1961.
870. Prince CW, Butler WT. 1,25-Dihydroxyvitamin D3 regulates the biosynthesis of
osteopontin, a bone-derived cell attachment protein, in clonal osteoblast-like osteosarcoma cells. Coll Relat Res 7: 305313, 1987.
871. Prinz C, Kajimura M, Scott DR, Mercier F, Helander HF, Sachs G. Histamine secretion from rat enterochromaffinlike cells. Gastroenterology 105: 449 461, 1993.
872. Prinz C, Neumayer N, Mahr S, Classen M, Schepp W. Functional impairment of rat
enterochromaffin-like cells by interleukin 1 beta. Gastroenterology 112: 364 375,
1997.
873. Prinz C, Sachs G, Walsh JH, Coy DH, Wu SV. The somatostatin receptor subtype on
rat enterochromaffinlike cells. Gastroenterology 107: 10671074, 1994.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

259

Downloaded from on December 8, 2014

841. Persson P, Gagnemo-Persson R, Chen D, Axelson J, Nylander AG, Johnell O, Hakanson R. Gastrectomy causes bone loss in the rat: is lack of gastric acid responsible?
Scand J Gastroenterol 28: 301306, 1993.

857. Pickard BW, Hodsman AB, Fraher LJ, Watson PH. Type 1 parathyroid hormone
receptor (PTH1R) nuclear trafficking: regulation of PTH1R nuclear-cytoplasmic
shuttling by importin-alpha/beta and chromosomal region maintenance 1/exportin 1.
Endocrinology 148: 22822289, 2007.

SASCHA KOPIC AND JOHN P. GEIBEL


874. Prosser DE, Guo Y, Jia Z, Jones G. Structural motif-based homology modeling of
CYP27A1 and site-directed mutational analyses affecting vitamin D hydroxylation.
Biophys J 90: 3389 3409, 2006.
875. Prunier JH, Bearn AG, Cleve H. Site of formation of the group-specific component
and certain other serum proteins. Proc Soc Exp Biol Med 115: 10051007, 1964.
876. Pyrah LN, Smith IB. Osteomalacia following gastrectomy. Lancet 270: 935937,
1956.

897. Reeve J, Meunier PJ, Parsons JA, Bernat M, Bijvoet OL, Courpron P, Edouard C,
Klenerman L, Neer RM, Renier JC, Slovik D, Vismans FJ, Potts JT Jr. Anabolic effect
of human parathyroid hormone fragment on trabecular bone in involutional osteoporosis: a multicentre trial. Br Med J 280: 1340 1344, 1980.
898. Reiss AB, Martin KO, Rojer DE, Iyer S, Grossi EA, Galloway AC, Javitt NB. Sterol
27-hydroxylase: expression in human arterial endothelium. J Lipid Res 38: 1254
1260, 1997.

877. Quamme GA. Effect of calcitonin on calcium and magnesium transport in rat
nephron. Am J Physiol Endocrinol Metab 238: E573E578, 1980.

899. Remy C, Kirchhoff P, Hafner P, Busque SM, Mueller MK, Geibel JP, Wagner CA.
Stimulatory pathways of the Calcium-sensing receptor on acid secretion in freshly
isolated human gastric glands. Cell Physiol Biochem 19: 33 42, 2007.

878. Quinn JM, Morfis M, Lam MH, Elliott J, Kartsogiannis V, Williams ED, Gillespie MT,
Martin TJ, Sexton PM. Calcitonin receptor antibodies in the identification of osteoclasts. Bone 25: 1 8, 1999.

900. Rengifo-Cam W, Umar S, Sarkar S, Singh P. Antiapoptotic effects of progastrin on


pancreatic cancer cells are mediated by sustained activation of nuclear factor-B.
Cancer Res 67: 7266 7274, 2007.

879. Quinn SJ, Bai M, Brown EM. pH Sensing by the calcium-sensing receptor. J Biol Chem
279: 3724137249, 2004.

901. Renkema KY, Velic A, Dijkman HB, Verkaart S, van der Kemp AW, Nowik M,
Timmermans K, Doucet A, Wagner CA, Bindels RJ, Hoenderop JG. The calciumsensing receptor promotes urinary acidification to prevent nephrolithiasis. J Am Soc
Nephrol 20: 17051713, 2009.

880. Quinn SJ, Ye CP, Diaz R, Kifor O, Bai M, Vassilev P, Brown E. The Ca2-sensing
receptor: a target for polyamines. Am J Physiol Cell Physiol 273: C1315C1323, 1997.
881. Raczynski J. Recherches exprimentales sur le manque daction du soleil comme
cause du rachitisme. In: C R Assoc Internat Pediatrie. Paris: 1912, p. 308 309.
882. Rakopoulos M, Ikegame M, Findlay DM, Martin TJ, Moseley JM. Short treatment of
osteoclasts in bone marrow culture with calcitonin causes prolonged suppression of
calcitonin receptor mRNA. Bone 17: 447 453, 1995.

884. Rautureau M, Rambaud JC. Aqueous solubilisation of vitamin D3 in normal man. Gut
22: 393397, 1981.
885. Ravazzola M, Benoit R, Ling N, Orci L. Prosomatostatin-derived antrin is present in
gastric D cells and in portal blood. J Clin Invest 83: 362366, 1989.
886. Ray JM, Squires PE, Curtis SB, Meloche MR, Buchan AM. Expression of the calciumsensing receptor on human antral gastrin cells in culture. J Clin Invest 99: 2328 2333,
1997.
887. Ray K, Clapp P, Goldsmith PK, Spiegel AM. Identification of the sites of N-linked
glycosylation on the human calcium receptor and assessment of their role in cell
surface expression and signal transduction. J Biol Chem 273: 34558 34567, 1998.
888. Ray K, Hauschild BC, Steinbach PJ, Goldsmith PK, Hauache O, Spiegel AM. Identification of the cysteine residues in the amino-terminal extracellular domain of the
human Ca2 receptor critical for dimerization. Implications for function of monomeric Ca2 receptor. J Biol Chem 274: 2764227650, 1999.
889. Raychowdhury R, Fleming JV, McLaughlin JT, Bulitta CJ, Wang TC. Identification and
characterization of a third gastrin response element (GAS-RE3) in the human histidine decarboxylase gene promoter. Biochem Biophys Res Commun 297: 1089 1095,
2002.
890. Raychowdhury R, Zhang Z, Hocker M, Wang TC. Activation of human histidine
decarboxylase gene promoter activity by gastrin is mediated by two distinct nuclear
factors. J Biol Chem 274: 2096120969, 1999.

903. Resnick RH, Adelardi CF, Gray SJ. Stimulation of gastric secretion in man by a
serotonin antagonist. Gastroenterology 42: 2225, 1962.
904. Reubi JC, Waser B, Horisberger U, Halter F, Soroka CJ, Kumar RR, Goldenring JR,
Modlin IM. Identification of somatostatin and gastrin receptors on enterochromaffinlike cells from Mastomys gastric tumors. Endocrinology 131: 166 172, 1992.
905. Reubi JC, Waser B, Laderach U, Stettler C, Friess H, Halter F, Schmassmann A.
Localization of cholecystokinin A and cholecystokinin B-gastrin receptors in the
human stomach. Gastroenterology 112: 11971205, 1997.
906. Riccardi D, Brown EM. Physiology and pathophysiology of the calcium-sensing receptor in the kidney. Am J Physiol Renal Physiol 298: F485F499, 2010.
907. Riccardi D, Hall AE, Chattopadhyay N, Xu JZ, Brown EM, Hebert SC. Localization of
the extracellular Ca2/polyvalent cation-sensing protein in rat kidney. Am J Physiol
Renal Physiol 274: F611F622, 1998.
908. Riccardi D, Lee WS, Lee K, Segre GV, Brown EM, Hebert SC. Localization of the
extracellular Ca2-sensing receptor and PTH/PTHrP receptor in rat kidney. Am J
Physiol Renal Fluid Electrolyte Physiol 271: F951F956, 1996.
909. Riccardi D, Traebert M, Ward DT, Kaissling B, Biber J, Hebert SC, Murer H. Dietary
phosphate and parathyroid hormone alter the expression of the calcium-sensing
receptor (CaR) and the Na-dependent Pi transporter (NaPi-2) in the rat proximal
tubule. Pflgers Arch 441: 379 387, 2000.
910. Richardson CT, Walsh JH, Cooper KA, Feldman M, Fordtran JS. Studies on the role
of cephalic-vagal stimulation in the acid secretory response to eating in normal
human subjects. J Clin Invest 60: 435 441, 1977.

891. Recker RR. Calcium absorption and achlorhydria. N Engl J Med 313: 70 73, 1985.

911. Ridefelt P, Hellman P, Stridsberg M, Akerstrom G, Rastad J. Different secretory


actions of pancreastatin in bovine and human parathyroid cells. Biosci Rep 14: 221
229, 1994.

892. Reddy GS, Norman AW, Willis DM, Goltzman D, Guyda H, Solomon S, Philips DR,
Bishop JE, Mayer E. Regulation of vitamin D metabolism in normal human pregnancy.
J Clin Endocrinol Metab 56: 363370, 1983.

912. Robert A, Olafsson AS, Lancaster C, Zhang WR. Interleukin-1 is cytoprotective,


antisecretory, stimulates PGE2 synthesis by the stomach, retards gastric emptying.
Life Sci 48: 123134, 1991.

893. Reddy GS, Tserng KY. Calcitroic acid, end product of renal metabolism of 1,25dihydroxyvitamin D3 through C-24 oxidation pathway. Biochemistry 28: 17631769,
1989.

913. Robinson CJ, Spanos E, James MF, Pike JW, Haussler MR, Makeen AM, Hillyard CJ,
MacIntyre I. Role of prolactin in vitamin D metabolism and calcium absorption during
lactation in the rat. J Endocrinol 94: 443 453, 1982.

894. Reed JS, Meredith SC, Nemchausky BA, Rosenberg IH, Boyer JL. Bone disease in
primary biliary cirrhosis: reversal of osteomalacia with oral 25-hydroxyvitamin D.
Gastroenterology 78: 512517, 1980.

914. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D. The crystal structure of the
nuclear receptor for vitamin D bound to its natural ligand. Mol Cell 5: 173179, 2000.

895. Reenstra WW, Forte JG. Characterization of K and Cl conductances in apical


membrane vesicles from stimulated rabbit oxyntic cells. Am J Physiol Gastrointest
Liver Physiol 259: G850 G858, 1990.
896. Reerink EH, Van Wijk W. The vitamin D problem. Biochem J 25: 10011009, 1931.

260

915. Rodriguez L, Tu C, Cheng Z, Chen TH, Bikle D, Shoback D, Chang W. Expression


and functional assessment of an alternatively spliced extracellular Ca2-sensing receptor in growth plate chondrocytes. Endocrinology 146: 5294 5303, 2005.
916. Rodriguez ME, Almaden Y, Canadillas S, Canalejo A, Siendones E, Lopez I, AguileraTejero E, Martin D, Rodriguez M. The calcimimetic R-568 increases vitamin D

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

883. Rasmussen H, Anast C, Arnaud C. Thyrocalcitonin, EGTA, and urinary electrolyte


excretion. J Clin Invest 46: 746 752, 1967.

902. Repa JJ, Lund EG, Horton JD, Leitersdorf E, Russell DW, Dietschy JM, Turley SD.
Disruption of the sterol 27-hydroxylase gene in mice results in hepatomegaly and
hypertriglyceridemia. Reversal by cholic acid feeding. J Biol Chem 275: 3968539692,
2000.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


receptor expression in rat parathyroid glands. Am J Physiol Renal Physiol 292: F1390
F1395, 2007.

936. Sachs G, Shin JM, Briving C, Wallmark B, Hersey S. The pharmacology of the gastric
acid pump: the H,K ATPase. Annu Rev Pharmacol Toxicol 35: 277305, 1995.

917. Roepke TK, Anantharam A, Kirchhoff P, Busque SM, Young JB, Geibel JP, Lerner DJ,
Abbott GW. The KCNE2 potassium channel ancillary subunit is essential for gastric
acid secretion. J Biol Chem 281: 23740 23747, 2006.

937. Safadi FF, Thornton P, Magiera H, Hollis BW, Gentile M, Haddad JG, Liebhaber SA,
Cooke NE. Osteopathy and resistance to vitamin D toxicity in mice null for vitamin
D binding protein. J Clin Invest 103: 239 251, 1999.

918. Rosen H, Reshef A, Maeda N, Lippoldt A, Shpizen S, Triger L, Eggertsen G, Bjorkhem I, Leitersdorf E. Markedly reduced bile acid synthesis but maintained levels of
cholesterol and vitamin D metabolites in mice with disrupted sterol 27-hydroxylase
gene. J Biol Chem 273: 1480514812, 1998.

938. Sakurada T, Ro S, Onouchi T, Ohno S, Aoyama T, Chinen K, Takabayashi H, Kato S,


Takayama K, Yakabi K. Comparison of the actions of acylated and desacylated
ghrelin on acid secretion in the rat stomach. J Gastroenterol 45: 11111120, 2010.
939. Salvesen HA. The function of the parathyroids. J Biol Chem 56: 443 456, 1923.

919. Rosenblatt M, Segre GV, Tyler GA, Shepard GL, Nussbaum SR, Potts JT Jr. Identification of a receptor-binding region in parathyroid hormone. Endocrinology 107:
545550, 1980.
920. Ross TK, Darwish HM, Moss VE, DeLuca HF. Vitamin D-influenced gene expression
via a ligand-independent, receptor-DNA complex intermediate. Proc Natl Acad Sci
USA 90: 92579260, 1993.
921. Rost CR, Bikle DD, Kaplan RA. In vitro stimulation of 25-hydroxycholecalciferol 1
alpha-hydroxylation by parathyroid hormone in chick kidney slices: evidence for a
role for adenosine 3=,5=-monophosphate. Endocrinology 108: 10021006, 1981.
922. Roth J, Thorens B, Hunziker W, Norman AW, Orci L. Vitamin D-dependent calcium
binding protein: immunocytochemical localization in chick kidney. Science 214: 197
200, 1981.
923. Roux C, Briot K, Gossec L, Kolta S, Blenk T, Felsenberg D, Reid DM, Eastell R, Gler
CC. Increase in vertebral fracture risk in postmenopausal women using omeprazole.
Calcified Tissue Int 84: 1319, 2009.

925. Rowe DW, Kream BE. Regulation of collagen synthesis in fetal rat calvaria by 1,25dihydroxyvitamin D3. J Biol Chem 257: 8009 8015, 1982.
926. Rowling MJ, Kemmis CM, Taffany DA, Welsh J. Megalin-mediated endocytosis of
vitamin D binding protein correlates with 25-hydroxycholecalciferol actions in human mammary cells. J Nutr 136: 2754 2759, 2006.
927. Rumenapf G, Schwille PO, Erben RG, Schreiber M, Berge B, Fries W, Schmiedl A,
Koroma S, Hohenberger W. Gastric fundectomy in the rat: effects on mineral and
bone metabolism, with emphasis on the gastrin-calcitonin-parathyroid hormonevitamin D axis. Calcif Tissue Int 63: 433 441, 1998.
928. Rumenapf G, Schwille PO, Wagner W, Tiecks FP, Fries W, Galewski D. Highly
selective vagotomy in the rat: effects on bone and mineral metabolism. Scand J
Gastroenterol 29: 232237, 1994.
929. Rune SJ. Acid-base parameters of duodenal contents in man. Gastroenterology 62:
533539, 1972.
930. Russell J, Bar A, Sherwood LM, Hurwitz S. Interaction between calcium and 1,25dihydroxyvitamin D3 in the regulation of preproparathyroid hormone and vitamin D
receptor messenger ribonucleic acid in avian parathyroids. Endocrinology 132: 2639
2644, 1993.
931. Russell J, Lettieri D, Sherwood LM. Suppression by 1,25(OH)2D3 of transcription of
the pre-proparathyroid hormone gene. Endocrinology 119: 2864 2866, 1986.
932. Rutten MJ, Bacon KD, Marlink KL, Stoney M, Meichsner CL, Lee FP, Hobson SA,
Rodland KD, Sheppard BC, Trunkey DD, Deveney KE, Deveney CW. Identification
of a functional Ca2-sensing receptor in normal human gastric mucous epithelial
cells. Am J Physiol Gastrointest Liver Physiol 277: G662G670, 1999.
933. Saarem K, Pedersen JI. Sex differences in the hydroxylation of cholecalciferol and of
5 beta-cholestane-3 alpha,7 alpha,12 alpha-triol in rat liver. Biochem J 247: 7378,
1987.

941. Sandor A, Kidd M, Lawton GP, Miu K, Tang LH, Modlin IM. Neurohormonal modulation of rat enterochromaffin-like cell histamine secretion. Gastroenterology 110:
1084 1092, 1996.
942. Sands JM, Flores FX, Kato A, Baum MA, Brown EM, Ward DT, Hebert SC, Harris
HW. Vasopressin-elicited water and urea permeabilities are altered in IMCD in
hypercalcemic rats. Am J Physiol Renal Physiol 274: F978 F985, 1998.
943. Sands JM, Naruse M, Baum M, Jo I, Hebert SC, Brown EM, Harris HW. Apical
extracellular calcium/polyvalent cation-sensing receptor regulates vasopressin-elicited water permeability in rat kidney inner medullary collecting duct. J Clin Invest 99:
1399 1405, 1997.
944. Sandvik AK, Cui G, Bakke I, Munkvold B, Waldum HL. PACAP stimulates gastric acid
secretion in the rat by inducing histamine release. Am J Physiol Gastrointest Liver
Physiol 281: G997G1003, 2001.
945. Sandvik AK, Dimaline R, Forster ER, Evans D, Dockray GJ. Differential control of
somatostatin messenger RNA in rat gastric corpus and antrum. Role of acid, food,
and capsaicin-sensitive afferent neurons. J Clin Invest 91: 244 250, 1993.
946. Sandvik AK, Kleveland PM, Waldum HL. Muscarinic M2 stimulation releases histamine in the totally isolated, vascularly perfused rat stomach. Scand J Gastroenterol 23:
1049 1056, 1988.
947. Saperas E, Cominelli F, Tache Y. Potent inhibition of gastric acid secretion by intravenous interleukin-1 beta and -1 alpha in rats. Peptides 13: 221226, 1992.
948. Saperas E, Yang H, Tache Y. Interleukin-1 beta acts at hypothalamic sites to inhibit
gastric acid secretion in rats. Am J Physiol Gastrointest Liver Physiol 263: G414 G418,
1992.
949. Saperas ES, Yang H, Rivier C, Tache Y. Central action of recombinant interleukin-1
to inhibit acid secretion in rats. Gastroenterology 99: 1599 1606, 1990.
950. Sawada N, Sakaki T, Ohta M, Inouye K. Metabolism of vitamin D3 by human
CYP27A1. Biochem Biophys Res Commun 273: 977984, 2000.
951. Schachter D. Toward a molecular description of active transport. In: Biological Membranes, edited by Dowben RM. Boston: Little, Brown, 1969, p. 157176.
952. Schachter D, Dowdle EB, Schenker H. Active transport of calcium by the small
intestine of the rat. Am J Physiol 198: 263268, 1960.
953. Schachter D, Finkelstein JD, Kowarski S. Metabolism of vitamin D. I. Preparation of
radioactive vitamin D and its intestinal absorption in the rat. J Clin Invest 43: 787796,
1964.
954. Schachter D, Rosen SM. Active transport of Ca45 by the small intestine and its
dependence on vitamin D. Am J Physiol 196: 357362, 1959.
955. Schaffalitzky de Muckadell OB, Fahrenkrug J. Secretion pattern of secretin in man:
regulation by gastric acid. Gut 19: 812 818, 1978.
956. Schatzmann HJ. ATP-dependent Ca2-extrusion from human red cells. Experientia
22: 364 365, 1966.

934. Sabbieti MG, Agas D, Xiao L, Marchetti L, Coffin JD, Doetschman T, Hurley MM.
Endogenous FGF-2 is critically important in PTH anabolic effects on bone. J Cell
Physiol 219: 143151, 2009.

957. Schayer RW. Formation and binding of histamine by rat tissues in vitro. Am J Physiol
187: 63 65, 1956.

935. Saccomani G, Psarras CG, Smith PR, Kirk KL, Shoemaker RL. Histamine-induced
chloride channels in apical membrane of isolated rabbit parietal cells. Am J Physiol Cell
Physiol 260: C1000 C1011, 1991.

958. Schepp W, Dehne K, Herrmuth H, Pfeffer K, Prinz C. Identification and functional


importance of IL-1 receptors on rat parietal cells. Am J Physiol Gastrointest Liver
Physiol 275: G1094 G1105, 1998.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

261

Downloaded from on December 8, 2014

924. Rovner AJ, Stallings VA, Schall JI, Leonard MB, Zemel BS. Vitamin D insufficiency in
children, adolescents, and young adults with cystic fibrosis despite routine oral supplementation. Am J Clin Nutr 86: 1694 1699, 2007.

940. Sandle GI, Fraser G, Long S, Warhurst G. A cAMP-activated chloride channel in the
plasma membrane of cultured human gastric cells (HGT-1). Pflgers Arch 417: 259
263, 1990.

SASCHA KOPIC AND JOHN P. GEIBEL


959. Schepp W, Dehne K, Riedel T, Schmidtler J, Schaffer K, Classen M. Oxyntomodulin:
a cAMP-dependent stimulus of rat parietal cell function via the receptor for glucagon-like peptide-1 (736)NH2. Digestion 57: 398 405, 1996.
960. Schepp W, Prinz C, Hakanson R, Schusdziarra V, Classen M. Bombesin-like peptides
stimulate gastrin release from isolated rat G-cells. Regul Pept 28: 241253, 1990.
961. Schepp W, Prinz C, Tatge C, Hakanson R, Schusdziarra V, Classen M. Galanin inhibits
gastrin release from isolated rat gastric G-cells. Am J Physiol Gastrointest Liver Physiol
258: G596 G602, 1990.
962. Schiller LR, Walsh JH, Feldman M. Distention-induced gastrin release: effects of
luminal acidification and intravenous atropine. Gastroenterology 78: 912917, 1980.
963. Schinke T, Schilling AF, Baranowsky A, Seitz S, Marshall RP, Linn T, Blaeker M,
Huebner AK, Schulz A, Simon R, Gebauer M, Priemel M, Kornak U, Perkovic S,
Barvencik F, Beil FT, Del Fattore A, Frattini A, Streichert T, Pueschel K, Villa A,
Debatin KM, Rueger JM, Teti A, Zustin J, Sauter G, Amling M. Impaired gastric
acidification negatively affects calcium homeostasis and bone mass. Nature Med 15:
674 681, 2009.
964. Schipani E, Karga H, Karaplis AC, Potts JT Jr, Kronenberg HM, Segre GV, AbouSamra AB, Juppner H. Identical complementary deoxyribonucleic acids encode a
human renal and bone parathyroid hormone (PTH)/PTH-related peptide receptor.
Endocrinology 132: 21572165, 1993.
965. Schjoldager BT, Baldissera FG, Mortensen PE, Holst JJ, Christiansen J. Oxyntomodulin: a potential hormone from the distal gut. Pharmacokinetics and effects on gastric
acid and insulin secretion in man. Eur J Clin Invest 18: 499 503, 1988.

967. Schmitz F, Goke MN, Otte JM, Schrader H, Reimann B, Kruse ML, Siegel EG, Peters
J, Herzig KH, Folsch UR, Schmidt WE. Cellular expression of CCK-A and CCK-B/
gastrin receptors in human gastric mucosa. Regul Pept 102: 101110, 2001.
968. Schmorl G. (Editor). Verhandlungen der Deutschen Pathologischen Gesellschaft. Jena:
Gustav Fischer, 1909.
969. Schnoke M, Midura SB, Midura RJ. Parathyroid hormone suppresses osteoblast
apoptosis by augmenting DNA repair. Bone 45: 590 602, 2009.
970. Schoeber JP, Topala CN, Wang X, Diepens RJ, Lambers TT, Hoenderop JG, Bindels
RJ. RGS2 inhibits the epithelial Ca2 channel TRPV6. J Biol Chem 281: 29669 29674,
2006.
971. Schoeber JPH, Topala CN, Lee KP, Lambers TT, Ricard G, Van Der Kemp AWCM,
Huynen MA, Hoenderop JGJ, Bindels RJM. Identification of Nipsnap1 as a novel
auxiliary protein inhibiting TRPV6 activity. Pflgers Arch 457: 91101, 2008.
972. Schoen MS, Lindenbaum J, Roginsky MS, Holt PR. Significance of serum level of
25-hydroxycholecalciferol in gastrointestinal disease. Am J Dig Dis 23: 137142,
1978.
973. Schofield GC, Ito S, Bolender RP. Changes in membrane surface areas in mouse
parietal cells in relation to high levels of acid secretion. J Anat 128: 669 692, 1979.
974. Scholz D, Schwille PO, Schley HW, Hanisch E, Bieger D, Zeuner E, Engelhardt W.
Mineral metabolism and vitamin D status before and up to five years following highly
selective vagotomy in duodenal ulcer patients. Hepatogastroenterology 30: 102106,
1983.
975. Schubert ML, Edwards NF, Makhlouf GM. Regulation of gastric somatostatin secretion in the mouse by luminal acidity: a local feedback mechanism. Gastroenterology
94: 317322, 1988.
976. Schubert ML, Jong MJ, Makhlouf GM. Bombesin/GRP-stimulated somatostatin secretion is mediated by gastrin in the antrum and intrinsic neurons in the fundus. Am
J Physiol Gastrointest Liver Physiol 261: G885G889, 1991.
977. Schubert ML, Makhlouf GM. Gastrin secretion induced by distention is mediated by
gastric cholinergic and vasoactive intestinal peptide neurons in rats. Gastroenterology
104: 834 839, 1993.
978. Schubert ML, Saffouri B, Walsh JH, Makhlouf GM. Inhibition of neurally mediated
gastrin secretion by bombesin antiserum. Am J Physiol Gastrointest Liver Physiol 248:
G456 G462, 1985.

262

980. Schulak JA, Kaplan EL. Gastrin-induced hypocalcemia in thyroparathyroidectomized


rats. Metabolism 23: 11031106, 1974.
981. Schulak JA, Kaplan EL. The importance of the stomach in gastrin-induced hypocalcemia in the rat. Endocrinology 96: 12171220, 1975.
982. Schusdziarra V, Harris V, Conlon JM, Arimura A, Unger R. Pancreatic and gastric
somatostatin release in response to intragastric and intraduodenal nutrients and HCl
in the dog. J Clin Invest 62: 509 518, 1978.
983. Schwab M, Klotz U, Hofmann U, Schaeffeler E, Leodolter A, Malfertheiner P,
Treiber G. Esomeprazole-induced healing of gastroesophageal reflux disease is unrelated to the genotype of CYP2C19: evidence from clinical and pharmacokinetic
data. Clin Pharmacol Ther 78: 627 634, 2005.
984. Schwartz GG, Wang MH, Zang M, Singh RK, Siegal GP. 1,25-Dihydroxyvitamin D
(calcitriol) inhibits the invasiveness of human prostate cancer cells. Cancer Epidemiol
Biomarkers Prev 6: 727732, 1997.
985. Seal A, Liu E, Buchan A, Brown J. Immunoneutralization of somatostatin and neurotensin: effect on gastric acid secretion. Am J Physiol Gastrointest Liver Physiol 255:
G40 G45, 1988.
986. Segawa H, Kaneko I, Takahashi A, Kuwahata M, Ito M, Ohkido I, Tatsumi S, Miyamoto K. Growth-related renal type II Na/Pi cotransporter. J Biol Chem 277: 19665
19672, 2002.
987. Segawa H, Yamanaka S, Onitsuka A, Tomoe Y, Kuwahata M, Ito M, Taketani Y,
Miyamoto K. Parathyroid hormone-dependent endocytosis of renal type IIc Na-Pi
cotransporter. Am J Physiol Renal Physiol 292: F395F403, 2007.
988. Segev H, Honigman A, Rosen H, Leitersdorf E. Transcriptional regulation of the
human sterol 27-hydroxylase gene (CYP27) and promoter mapping. Atherosclerosis
156: 339 347, 2001.
989. Segre GV, DAmour P, Hultman A, Potts JT Jr. Effects of hepatectomy, nephrectomy, nephrectomy/uremia on the metabolism of parathyroid hormone in the rat. J
Clin Invest 67: 439 448, 1981.
990. Selye H. Anesthetic effect of steroid hormones. Proc Soc Exp Biol Med 46: 116, 1941.
991. Serfaty-Lacrosniere C, Wood RJ, Voytko D, Saltzman JR, Pedrosa M, Sepe TE,
Russell RR. Hypochlorhydria from short-term omeprazole treatment does not inhibit intestinal absorption of calcium, phosphorus, magnesium or zinc from food in
humans. J Am Coll Nutr 14: 364 368, 1995.
992. Seroussi E, Pan HQ, Kedra D, Roe BA, Dumanski JP. Characterization of the human
NIPSNAP1 gene from 22q12: a member of a novel gene family. Gene 212: 1320,
1998.
993. Setoguchi T, Salen G, Tint GS, Mosbach EH. A biochemical abnormality in cerebrotendinous xanthomatosis. Impairment of bile acid biosynthesis associated with incomplete degradation of the cholesterol side chain. J Clin Invest 53: 13931401,
1974.
994. Seva C, Dickinson CJ, Yamada T. Growth-promoting effects of glycine-extended
progastrin. Science 265: 410 412, 1994.
995. Shareghi GR, Stoner LC. Calcium transport across segments of the rabbit distal
nephron in vitro. Am J Physiol Renal Fluid Electrolyte Physiol 235: F367F375, 1978.
996. Sheikh MS, Ramirez A, Emmett M, Santa Ana C, Schiller LR, Fordtran JS. Role of
vitamin D-dependent and vitamin D-independent mechanisms in absorption of food
calcium. J Clin Invest 81: 126 132, 1988.
997. Sheikh MS, Santa Ana CA, Nicar MJ, Schiller LR, Fordtran JS. Gastrointestinal absorption of calcium from milk and calcium salts. N Engl J Med 317: 532536, 1987.
998. Sheinin Y, Kallay E, Wrba F, Kriwanek S, Peterlik M, Cross HS. Immunocytochemical
localization of the extracellular calcium-sensing receptor in normal and malignant
human large intestinal mucosa. J Histochem Cytochem 48: 595 602, 2000.
999. Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junction pore and leak
pathways: a dynamic duo. Annu Rev Physiol 73: 283309, 2011.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

966. Schmidt WE, Schenk S, Nustede R, Holst JJ, Folsch UR, Creutzfeldt W. Cholecystokinin is a negative regulator of gastric acid secretion and postprandial release of
gastrin in humans. Gastroenterology 107: 1610 1620, 1994.

979. Schuessler M, Astecker N, Herzig G, Vorisek G, Schuster I. Skin is an autonomous


organ in synthesis, two-step activation and degradation of vitamin D(3): CYP27 in
epidermis completes the set of essential vitamin D(3)-hydroxylases. Steroids 66:
399 408, 2001.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


1000. Shen LP, Pictet RL, Rutter WJ. Human somatostatin I: sequence of the cDNA. Proc
Natl Acad Sci USA 79: 4575 4579, 1982.
1001. Sherry AM, Malinowska DH, Morris RE, Ciraolo GM, Cuppoletti J. Localization of
ClC-2 Cl channels in rabbit gastric mucosa. Am J Physiol Cell Physiol 280: C1599
C1606, 2001.
1002. Shibata N, Matsui H, Yokota T, Matsuura B, Maeyama K, Onji M. Direct effects of
nitric oxide on histamine release from rat enterochromaffin-like cells. Eur J Pharmacol
535: 2533, 2006.
1003. Shimizu T, Yoshitomi K, Nakamura M, Imai M. Effects of PTH, calcitonin, cAMP on
calcium transport in rabbit distal nephron segments. Am J Physiol Renal Fluid Electrolyte Physiol 259: F408 F414, 1990.
1004. Shimoi K, Saka N, Nozawa R, Sato M, Amano I, Nakayama T, Kinae N. Deglucuronidation of a flavonoid, luteolin monoglucuronide, during inflammation. Drug Metab
Dispos 29: 15211524, 2001.
1005. Shin JM, Besancon M, Prinz C, Simon A, Sachs G. Continuing development of acid
pump inhibitors: site of action of pantoprazole. Aliment Pharmacol Ther 8 Suppl 1:
1123, 1994.
1006. Shin JM, Besancon M, Simon A, Sachs G. The site of action of pantoprazole in the
gastric H/K-ATPase. Biochim Biophys Acta 1148: 223233, 1993.

1007. Shin JM, Cho YM, Sachs G. Chemistry of covalent inhibition of the gastric (H ,
K)-ATPase by proton pump inhibitors. J Am Chem Soc 126: 7800 7811, 2004.

1022. Sneddon WB, Syme CA, Bisello A, Magyar CE, Rochdi MD, Parent JL, Weinman EJ,
Abou-Samra AB, Friedman PA. Activation-independent parathyroid hormone receptor internalization is regulated by NHERF1 (EBP50). J Biol Chem 278: 43787
43796, 2003.
1023. Soejima M, Tachida H, Koda Y. Sequence analysis of human TRPV6 suggests positive
selection outside Africa. Biochem Genet 47: 147153, 2009.
1024. Soll AH. The interaction of histamine with gastrin and carbamylcholine on oxygen
uptake by isolated mammalian parietal cells. J Clin Invest 61: 381389, 1978.
1025. Soll AH. Potentiating interactions of gastric stimulants on [14C]aminopyrine accumulation by isolated canine parietal cells. Gastroenterology 83: 216 223, 1982.
1026. Soll AH, Wollin A. Histamine and cyclic AMP in isolated canine parietal cells. Am J
Physiol Endocrinol Metab Gastrointest Physiol 237: E444 E450, 1979.
1027. Sone T, Kerner S, Pike JW. Vitamin D receptor interaction with specific DNA.
Association as a 1,25-dihydroxyvitamin D3-modulated heterodimer. J Biol Chem 266:
23296 23305, 1991.
1028. Song P, Groos S, Riederer B, Feng Z, Krabbenhoft A, Manns MP, Smolka A, Hagen SJ,
Neusch C, Seidler U. Kir4.1 channel expression is essential for parietal cell control of
acid secretion. J Biol Chem 286: 14120 14128, 2011.
1029. Song P, Groos S, Riederer B, Feng Z, Krabbenhoft A, Smolka A, Seidler U. KCNQ1
is the luminal K recycling channel during stimulation of gastric acid secretion. J
Physiol 587: 39553965, 2009.

1009. Shinki T, Ueno Y, DeLuca HF, Suda T. Calcitonin is a major regulator for the
expression of renal 25-hydroxyvitamin D3-1alpha-hydroxylase gene in normocalcemic rats. Proc Natl Acad Sci USA 96: 8253 8258, 1999.

1030. Song Y, Peng X, Porta A, Takanaga H, Peng JB, Hediger MA, Fleet JC, Christakos S.
Calcium transporter 1 and epithelial calcium channel messenger ribonucleic acid are
differentially regulated by 1,25 dihydroxyvitamin D3 in the intestine and kidney of
mice. Endocrinology 144: 38853894, 2003.

1010. Shoback DM, Membreno LA, McGhee JG. High calcium and other divalent cations
increase inositol trisphosphate in bovine parathyroid cells. Endocrinology 123: 382
389, 1988.

1031. Sopjani M, Kunert A, Czarkowski K, Klaus F, Laufer J, Fller M, Lang F. Regulation of


the Ca2 Channel TRPV6 by the Kinases SGK1, PKB/Akt, and PIKfyve. J Membr Biol
233: 35 41, 2010.

1011. Shulkes A, Read M. Regulation of somatostatin secretion by gastrin- and acid-dependent mechanisms. Endocrinology 129: 2329 2334, 1991.

1032. Sowa H, Kaji H, Iu MF, Tsukamoto T, Sugimoto T, Chihara K. Parathyroid hormoneSmaD3 axis exerts anti-apoptotic action and augments anabolic action of transforming growth factor beta in osteoblasts. J Biol Chem 278: 52240 52252, 2003.

1012. Shyjan AW, Canfield VA, Levenson R. Evolution of the Na,K- and H-K-ATPase
beta subunit gene family: structure of the murine Na-K-ATPase beta 2 subunit
gene. Genomics 11: 435 442, 1991.

1033. St-Arnaud R. The direct role of vitamin D on bone homeostasis. Arch Biochem Biophys
473: 225230, 2008.

1013. Sidani SM, Kirchhoff P, Socrates T, Stelter L, Ferreira E, Caputo C, Roberts KE, Bell
RL, Egan ME, Geibel JP. DeltaF508 mutation results in impaired gastric acid secretion. J Biol Chem 282: 6068 6074, 2007.

1034. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glorieux FH. The 25-hydroxyvitamin D 1-alpha-hydroxylase gene maps to the pseudovitamin D-deficiency rickets
(PDDR) disease locus. J Bone Miner Res 12: 15521559, 1997.

1014. Silve C, Petrel C, Leroy C, Bruel H, Mallet E, Rognan D, Ruat M. Delineating a Ca2
binding pocket within the venus flytrap module of the human calcium-sensing receptor. J Biol Chem 280: 3791737923, 2005.

1035. Stamp TC, Round JM. Seasonal changes in human plasma levels of 25-hydroxyvitamin
D. Nature 247: 563565, 1974.

1015. Silver J, Fainaru M. Transport of vitamin D sterols in human plasma: effect of excess
vitamin D, 25 hydroxyvitamin D and 1,25 dihydroxyvitamin D. Eur J Clin Invest 9:
433 438, 1979.
1016. Singer FR, Foster GV, Jpolin GF, Nadarajah A, Parkinson DK, Thalassinos N, Woodhouse NJ, Clark MB, Fraser TR, MacIntyre I. Acute effects of administered calcitonin
in man. Calcif Tissue Res Suppl: 20, 1968.
1017. Singh P, Velasco M, Given R, Varro A, Wang TC. Progastrin expression predisposes
mice to colon carcinomas and adenomas in response to a chemical carcinogen.
Gastroenterology 119: 162171, 2000.
1018. Sitrin MD, Bengoa JM. Intestinal absorption of cholecalciferol and 25-hydroxycholecalciferol in chronic cholestatic liver disease. Am J Clin Nutr 46: 10111015, 1987.
1019. Sitrin MD, Pollack KL, Bolt MJ, Rosenberg IH. Comparison of vitamin D and 25hydroxyvitamin D absorption in the rat. Am J Physiol Gastrointest Liver Physiol 242:
G326 G332, 1982.
1020. Sizemore GW, Go VL, Kaplan EL, Sanzenbacher LJ, Holtermuller KH, Arnaud CD.
Relations of calcitonin and gastrin in the Zollinger-Ellison syndrome and medullary
carcinoma of the thyroid. N Engl J Med 288: 641 644, 1973.

1036. Stedman CA, Barclay ML. Review article: comparison of the pharmacokinetics, acid
suppression and efficacy of proton pump inhibitors. Aliment Pharmacol Ther 14:
963978, 2000.
1037. Steenbock H, Black A. Fat-soluble vitamins. XVII. The induction of growth-promoting and calcifying properties in a ration by exposure to ultra-violet light. J Biol Chem
61: 405 422, 1924.
1038. Steggerda FR, Mitchell HH. The effect of the citrate ion on the calcium metabolism
of adult human subjects. J Nutr 31: 423 438, 1946.
1039. Stepan V, Pausawasdi N, Ramamoorthy S, Todisco A. The Akt and MAPK signaltransduction pathways regulate growth factor actions in isolated gastric parietal cells.
Gastroenterology 127: 1150 1161, 2004.
1040. Stepan V, Sugano K, Yamada T, Park J, Dickinson CJ. Gastrin biosynthesis in canine G
cells. Am J Physiol Gastrointest Liver Physiol 282: G766 G775, 2002.
1041. Sternfeld L, Anderie I, Schmid A, Al-Shaldi H, Krause E, Magg T, Schreiner D, Hofer
HW, Schulz I. Identification of tyrosines in the putative regulatory site of the Ca2
channel TRPV6. Cell Calcium 42: 91102, 2007.
1042. Sternfeld L, Krause E, Schmid A, Anderie I, Latas A, Al-Shaldi H, Kohl A, Evers K,
Hofer HW, Schulz I. Tyrosine phosphatase PTP1B interacts with TRPV6 in vivo and

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

263

Downloaded from on December 8, 2014

1008. Shin JM, Homerin M, Domagala F, Ficheux H, Sachs G. Characterization of the


inhibitory activity of tenatoprazole on the gastric H,K-ATPase in vitro and in vivo.
Biochem Pharmacol 71: 837 849, 2006.

1021. Slepchenko BM, Bronner F. Modeling of transcellular Ca transport in rat duodenum


points to coexistence of two mechanisms of apical entry. Am J Physiol Cell Physiol 281:
C270 C281, 2001.

SASCHA KOPIC AND JOHN P. GEIBEL


plays a role in TRPV6-mediated calcium influx in HEK293 cells. Cell Signal 17: 951
960, 2005.
1043. Strehler EE, Zacharias DA. Role of alternative splicing in generating isoform diversity
among plasma membrane calcium pumps. Physiol Rev 81: 2150, 2001.
1044. Strong TV, Boehm K, Collins FS. Localization of cystic fibrosis transmembrane conductance regulator mRNA in the human gastrointestinal tract by in situ hybridization.
J Clin Invest 93: 347354, 1994.
1045. Strushkevich N, Usanov SA, Plotnikov AN, Jones G, Park HW. Structural analysis of
CYP2R1 in complex with vitamin D3. J Mol Biol 380: 95106, 2008.
1046. Stumpf T, Zhang Q, Hirnet D, Lewandrowski U, Sickmann A, Wissenbach U, Dorr
J, Lohr C, Deitmer JW, Fecher-Trost C. The human TRPV6 channel protein is
associated with cyclophilin B in human placenta. J Biol Chem 283: 18086 18098,
2008.
1047. Su HC, Bishop AE, Power RF, Hamada Y, Polak JM. Dual intrinsic and extrinsic
origins of CGRP- and NPY-immunoreactive nerves of rat gut and pancreas. J Neurosci 7: 2674 2687, 1987.
1048. Su P, Rennert H, Shayiq RM, Yamamoto R, Zheng YM, Addya S, Strauss JF 3rd,
Avadhani NG. A cDNA encoding a rat mitochondrial cytochrome P450 catalyzing
both the 26-hydroxylation of cholesterol and 25-hydroxylation of vitamin D3: gonadotropic regulation of the cognate mRNA in ovaries. DNA Cell Biol 9: 657 667,
1990.

1050. Sudo Y, Matsuo K, Tetsuo T, Tsutsumi S, Ohkura M, Nakai J, Uezono Y. Derived


(mutated)-types of TRPV6 channels elicit greater Ca2 influx into the cells than
ancestral-types of TRPV6: evidence from Xenopus oocytes and mammalian cell expression system. J Pharmacol Sci 114: 281291, 2010.
1051. Sugano K, Park J, Dobbins WO, Yamada T. Glycine-extended progastrin processing
intermediates: accumulation and cosecretion with gastrin. Am J Physiol Gastrointest
Liver Physiol 253: G502G507, 1987.
1052. Suki WN, Eknoyan G, Rector FC Jr, Seldin DW. The renal diluting and concentrating
mechanism in hypercalcemia. Nephron 6: 50 61, 1969.
1053. Sulimovici S, Roginsky MS, Duffy JL, Pfeifer RF. Calciferol 25-hydroxylase activity in
smooth and rough endoplasmic reticulum of rat liver. Arch Biochem Biophys 195:
4552, 1979.
1054. Sundler F, Ekblad E, Absood A, Hakanson R, Koves K, Arimura A. Pituitary adenylate
cyclase activating peptide: a novel vasoactive intestinal peptide-like neuropeptide in
the gut. Neuroscience 46: 439 454, 1992.
1055. Sunthornthepvarakul T, Churesigaew S, Ngowngarmratana S. A novel mutation of
the signal peptide of the preproparathyroid hormone gene associated with autosomal recessive familial isolated hypoparathyroidism. J Clin Endocrinol Metab 84:
37923796, 1999.
1056. Suva LJ, Winslow GA, Wettenhall RE, Hammonds RG, Moseley JM, DiefenbachJagger H, Rodda CP, Kemp BE, Rodriguez H, Chen EY. A parathyroid hormonerelated protein implicated in malignant hypercalcemia: cloning and expression. Science 237: 893 896, 1987.
1057. Suzuki H, Nakamura I, Takahashi N, Ikuhara T, Matsuzaki K, Isogai Y, Hori M, Suda
T. Calcitonin-induced changes in the cytoskeleton are mediated by a signal pathway
associated with protein kinase A in osteoclasts. Endocrinology 137: 4685 4690, 1996.
1058. Suzuki M, Ishibashi K, Ooki G, Tsuruoka S, Imai M. Electrophysiologic characteristics
of the Ca-permeable channels, ECaC and CaT, in the kidney. Biochem Biophys Res
Commun 274: 344 349, 2000.
1059. Tache Y, Saperas E. Potent inhibition of gastric acid secretion and ulcer formation by
centrally and peripherally administered interleukin-1. Ann NY Acad Sci 664: 353368,
1992.
1060. Takahashi N, Yamana H, Yoshiki S, Roodman GD, Mundy GR, Jones SJ, Boyde A,
Suda T. Osteoclast-like cell formation and its regulation by osteotropic hormones in
mouse bone marrow cultures. Endocrinology 122: 13731382, 1988.

264

1062. Takeuchi Y, Pausawasdi N, Todisco A. Carbachol activates ERK2 in isolated gastric


parietal cells via multiple signaling pathways. Am J Physiol Gastrointest Liver Physiol
276: G1484 G1492, 1999.
1063. Takeuchi Y, Yamada J, Yamada T, Todisco A. Functional role of extracellular signalregulated protein kinases in gastric acid secretion. Am J Physiol Gastrointest Liver
Physiol 273: G1263G1272, 1997.
1064. Talmage RV, Neuenschwander J, Kraintz L. Evidence for the existence of thyrocalcitonin in the rat. Endocrinology 76: 103107, 1965.
1065. Tam CS, Heersche JN, Murray TM, Parsons JA. Parathyroid hormone stimulates the
bone apposition rate independently of its resorptive action: differential effects of
intermittent and continuous administration. Endocrinology 110: 506 512, 1982.
1066. Tanaka S, Hamada K, Yamada N, Sugita Y, Tonai S, Hunyady B, Palkovits M, Falus A,
Watanabe T, Okabe S, Ohtsu H, Ichikawa A, Nagy A. Gastric acid secretion in
L-histidine decarboxylase-deficient mice. Gastroenterology 122: 145155, 2002.
1067. Tanaka Y, Castillo L, Wineland MJ, DeLuca HF. Synergistic effect of progesterone,
testosterone, estradiol in the stimulation of chick renal 25-hydroxyvitamin D3-1alpha-hydroxylase. Endocrinology 103: 20352039, 1978.
1068. Tanaka Y, Deluca HF. The control of 25-hydroxyvitamin D metabolism by inorganic
phosphorus. Arch Biochem Biophys 154: 566 574, 1973.
1069. Tanaka Y, DeLuca HF. Measurement of mammalian 25-hydroxyvitamin D3 24R-and
1 alpha-hydroxylase. Proc Natl Acad Sci USA 78: 196 199, 1981.
1070. Tang LH, Stoch SA, Modlin IM, Goldenring JR. Identification of rab2 as a tubulovesicle-membrane-associated protein in rabbit gastric parietal cells. Biochem J 285:
715719, 1992.
1071. Tanrattana C, Charoenphandhu N, Limlomwongse L, Krishnamra N. Prolactin directly stimulated the solvent drag-induced calcium transport in the duodenum of
female rats. Biochim Biophys Acta 1665: 8191, 2004.
1072. Tatemoto K, Efendic S, Mutt V, Makk G, Feistner GJ, Barchas JD. Pancreastatin,
a novel pancreatic peptide that inhibits insulin secretion. Nature 324: 476 478,
1986.
1073. Taylor AN, Wasserman RH. Vitamin D3-induced calcium-binding protein: partial
purification, electrophoretic visualization, and tissue distribution. Arch Biochem Biophys 119: 536 540, 1967.
1074. Taylor IL, Byrne WJ, Christie DL, Ament ME, Walsh JH. Effect of individual L-amino
acids on gastric acid secretion and serum gastrin and pancreatic polypeptide release
in humans. Gastroenterology 83: 273278, 1982.
1075. Taylor IL, Sells RA, McConnell RB, Dockray GJ. Serum gastrin in patients with
chronic renal failure. Gut 21: 10621067, 1980.
1076. Teranishi H, Kasuya M, Aoshima K, Kato T, Migita S. Demonstration of vitamin
D-binding protein (Gc-globulin) in the urine of Itai-itai disease patients. Toxicol Lett
15: 712, 1983.
1077. Thamsborg G, Jensen JE, Kollerup G, Hauge EM, Melsen F, Sorensen OH. Effect of
nasal salmon calcitonin on bone remodeling and bone mass in postmenopausal osteoporosis. Bone 18: 207212, 1996.
1078. Theodoropoulos C, Demers C, Petit JL, Gascon-Barre M. High sensitivity of rat
hepatic vitamin D3-25 hydroxylase CYP27A to 1,25-dihydroxyvitamin D3 administration. Am J Physiol Endocrinol Metab 284: E138 E147, 2003.
1079. Thomas WC Jr, Morgan HG, Connor TB, Haddock L, Bills CE, Howard JE. Studies of
antiricketic activity in sera from patients with disorders of calcium metabolism and
preliminary observations on the mode of transport of vitamin D in human serum. J
Clin Invest 38: 1078 1085, 1959.
1080. Thompson GR, Lewis B, Booth CC. Absorption of vitamin D3-3H in control subjects
and patients with intestinal malabsorption. J Clin Invest 45: 94 102, 1966.
1081. Thompson GR, Lewis B, Booth CC. Vitamin-D absorption after partial gastrectomy.
Lancet 1: 457 458, 1966.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

1049. Suda J, Zhu L, Okamoto CT, Karvar S. Rab27b localizes to the tubulovesicle membranes of gastric parietal cells and regulates acid secretion. Gastroenterology 140:
868 878, 2011.

1061. Takahashi S, Goldring S, Katz M, Hilsenbeck S, Williams R, Roodman GD. Downregulation of calcitonin receptor mRNA expression by calcitonin during human osteoclast-like cell differentiation. J Clin Invest 95: 167171, 1995.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


1082. Thompson GR, Ockner RK, Isselbacher KJ. Effect of mixed micellar lipid on the
absorption of cholesterol and vitamin D3 into lymph. J Clin Invest 48: 8795, 1969.

tive hormone and hypocalcemia-induced 1,25(OH)2D synthesis. Kidney Int 72:


1330 1335, 2007.

1083. Thompson PD, Jurutka PW, Haussler CA, Whitfield GK, Haussler MR. Heterodimeric DNA binding by the vitamin D receptor and retinoid X receptors is enhanced
by 1,25-dihydroxyvitamin D3 and inhibited by 9-cis-retinoic acid. Evidence for allosteric receptor interactions. J Biol Chem 273: 8483 8491, 1998.

1103. Usui E, Noshiro M, Okuda K. Molecular cloning of cDNA for vitamin D3 25-hydroxylase from rat liver mitochondria. FEBS Lett 262: 135138, 1990.

1084. Thunberg R. Localization of cells containing and forming histamine in the gastric
mucosa of the rat. Exp Cell Res 47: 108 115, 1967.
1085. Tian XQ, Chen TC, Matsuoka LY, Wortsman J, Holick MF. Kinetic and thermodynamic studies of the conversion of previtamin D3 to vitamin D3 in human skin. J Biol
Chem 268: 14888 14892, 1993.
1086. Tian XQ, Holick MF. A liposomal model that mimics the cutaneous production of
vitamin D3. Studies of the mechanism of the membrane-enhanced thermal isomerization of previtamin D3 to vitamin D3. J Biol Chem 274: 4174 4179, 1999.
1087. Tinel N, Diochot S, Borsotto M, Lazdunski M, Barhanin J. KCNE2 confers background current characteristics to the cardiac KCNQ1 potassium channel. EMBO J
19: 6326 6330, 2000.
1088. Tishkoff DX, Nibbelink KA, Holmberg KH, Dandu L, Simpson RU. Functional vitamin D receptor (VDR) in the t-tubules of cardiac myocytes: VDR knockout cardiomyocyte contractility. Endocrinology 149: 558 564, 2008.
1089. Tondapu P, Provost D, Adams-Huet B, Sims T, Chang C, Sakhaee K. Comparison of
the absorption of calcium carbonate and calcium citrate after Roux-en-Y gastric
bypass. Obes Surg 19: 1256 1261, 2009.

1091. Tovey FI, Karamanolis DG, Godfrey J, Clark CG. Post-gastrectomy nutrition: methods of outpatient screening for early osteomalacia. Hum Nutr Clin Nutr 39: 439 446,
1985.
1092. Toyoshima C, Nomura H. Structural changes in the calcium pump accompanying the
dissociation of calcium. Nature 418: 605 611, 2002.
1093. Toyoshima C, Nomura H, Sugita Y. Crystal structures of Ca2-ATPase in various
physiological states. Ann NY Acad Sci 986: 1 8, 2003.
1094. Tregear GW, Van Rietschoten J, Greene E, Keutmann HT, Niall HD, Reit B, Parsons
JA, Potts JT Jr. Bovine parathyroid hormone: minimum chain length of synthetic
peptide required for biological activity. Endocrinology 93: 1349 1353, 1973.
1095. Tsunoda Y, Funasaka M, Modlin IM, Hidaka H, Fox LM, Goldenring JR. An inhibitor
of Ca2/calmodulin-dependent protein kinase II, KN-62, inhibits cholinergic-stimulated parietal cell secretion. Am J Physiol Gastrointest Liver Physiol 262: G118 G122,
1992.

1105. Vagin O, Denevich S, Sachs G. Plasma membrane delivery of the gastric H-KATPase: the role of beta-subunit glycosylation. Am J Physiol Cell Physiol 285: C968
C976, 2003.
1106. Vagin O, Munson K, Lambrecht N, Karlish SJ, Sachs G. Mutational analysis of the
K-competitive inhibitor site of gastric H-K-ATPase. Biochemistry 40: 7480
7490, 2001.
1107. Vagne M, Andre C. The effect of secretin on gastric emptying in man. Gastroenterology 60: 421 424, 1971.
1108. Van Abel M, Hoenderop JG, van der Kemp AW, Friedlaender MM, van Leeuwen JP,
Bindels RJ. Coordinated control of renal Ca2 transport proteins by parathyroid
hormone. Kidney Int 68: 1708 1721, 2005.
1109. Van Abel M, Hoenderop JG, van der Kemp AW, van Leeuwen JP, Bindels RJ. Regulation of the epithelial Ca2 channels in small intestine as studied by quantitative
mRNA detection. Am J Physiol Gastrointest Liver Physiol 285: G78 G85, 2003.
1110. Van Cromphaut SJ, Dewerchin M, Hoenderop JG, Stockmans I, Van Herck E, Kato S,
Bindels RJ, Collen D, Carmeliet P, Bouillon R, Carmeliet G. Duodenal calcium absorption in vitamin D receptor-knockout mice: functional and molecular aspects.
Proc Natl Acad Sci USA 98: 13324 13329, 2001.
1111. Van de Graaf SF, Chang Q, Mensenkamp AR, Hoenderop JG, Bindels RJ. Direct
interaction with Rab11a targets the epithelial Ca2 channels TRPV5 and TRPV6 to
the plasma membrane. Mol Cell Biol 26: 303312, 2006.
1112. Van de Graaf SF, Hoenderop JG, Gkika D, Lamers D, Prenen J, Rescher U, Gerke V,
Staub O, Nilius B, Bindels RJ. Functional expression of the epithelial Ca2 channels
(TRPV5 and TRPV6) requires association of the S100A10-annexin 2 complex. EMBO
J 22: 1478 1487, 2003.
1113. Van de Graaf SF, Hoenderop JG, van der Kemp AW, Gisler SM, Bindels RJ. Interaction of the epithelial Ca2 channels TRPV5 and TRPV6 with the intestine- and
kidney-enriched PDZ protein NHERF4. Pflgers Arch 452: 407 417, 2006.
1114. Van Leeuwen JP, van Driel M, van den Bemd GJ, Pols HA. Vitamin D control of
osteoblast function and bone extracellular matrix mineralization. Crit Rev Eukaryot
Gene Expr 11: 199 226, 2001.
1115. Varrault A, Pena MS, Goldsmith PK, Mithal A, Brown EM, Spiegel AM. Expression of
G protein alpha-subunits in bovine parathyroid. Endocrinology 136: 4390 4396,
1995.

1096. Tu Q, Pi M, Karsenty G, Simpson L, Liu S, Quarles LD. Rescue of the skeletal


phenotype in CasR-deficient mice by transfer onto the Gcm2 null background. J Clin
Invest 111: 1029 1037, 2003.

1116. Varro A, Green T, Holmes S, Dockray GJ. Calcitonin gene-related peptide in visceral
afferent nerve fibres: quantification by radioimmunoassay and determination of axonal transport rates. Neuroscience 26: 927932, 1988.

1097. Tucker G 3rd, Gagnon RE, Haussler MR. Vitamin D3-25-hydroxylase: tissue occurrence and apparent lack of regulation. Arch Biochem Biophys 155: 4757, 1973.

1117. Velluz L, Amirad G. Chimie organique-equilibre de reaction entre precalciferol et


calciferol. Compt Rend Assoc Anat 288: 853 855, 1949.

1098. Tudpor K, Teerapornpuntakit J, Jantarajit W, Krishnamra N, Charoenphandhu N.


1,25-Dihydroxyvitamin D(3) rapidly stimulates the solvent drag-induced paracellular
calcium transport in the duodenum of female rats. J Physiol Sci 58: 297307, 2008.

1118. Velluz L, Amirad G. Chimie organique-le precalciferol. Compt Rend Assoc Anat 228:
692 694, 1949.

1099. Tzaneva MA. Ultrastructural immunohistochemical localization of gastrin, somatostatin and serotonin in endocrine cells of human antral gastric mucosa. Acta Histochem 105: 191201, 2003.
1100. Uchida M, Teranishi H, Aoshima K, Katoh T, Kasuya M, Inadera H. Elevated urinary
levels of vitamin D-binding protein in the inhabitants of a cadmium polluted area,
Jinzu River basin, Japan. Tohoku J Exp Med 211: 269 274, 2007.
1101. Uehara A, Okumura T, Sekiya C, Okamura K, Takasugi Y, Namiki M. Interleukin-1
inhibits the secretion of gastric acid in rats: possible involvement of prostaglandin.
Biochem Biophys Res Commun 162: 1578 1584, 1989.
1102. Usatii M, Rousseau L, Demers C, Petit JL, Brossard JH, Gascon-Barre M, Lavigne JR,
Zahradnik RJ, Nemeth EF, DAmour P. Parathyroid hormone fragments inhibit ac-

1119. Velluz L, Amirad G. Chimie organique-nouveau precursor de la vitamine D3. Compt


Rend Assoc Anat 228: 10371038, 1949.
1120. Velluz L, Amirad G, Petit A. Le precalciferol: ses relations dequilibre avec le calciferol. Bull Soc Chim France 16: 501507, 1949.
1121. Verboven C, Rabijns A, De Maeyer M, Van Baelen H, Bouillon R, De Ranter C. A
structural basis for the unique binding features of the human vitamin D-binding
protein. Nat Struct Biol 9: 131136, 2002.
1122. Vertino AM, Bula CM, Chen JR, Almeida M, Han L, Bellido T, Kousteni S, Norman
AW, Manolagas SC. Nongenotropic, anti-apoptotic signaling of 1alpha,25(OH)2vitamin D3 and analogs through the ligand binding domain of the vitamin D receptor
in osteoblasts and osteocytes. Mediation by Src, phosphatidylinositol 3-, and JNK
kinases. J Biol Chem 280: 14130 14137, 2005.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

265

Downloaded from on December 8, 2014

1090. Topala CN, Schoeber JP, Searchfield LE, Riccardi D, Hoenderop JG, Bindels RJ.
Activation of the Ca2-sensing receptor stimulates the activity of the epithelial Ca2
channel TRPV5. Cell Calcium 45: 331339, 2009.

1104. Vagin O, Denevich S, Munson K, Sachs G. SCH28080, a K-competitive inhibitor of


the gastric H-K-ATPase, binds near the M5 6 luminal loop, preventing K access
to the ion binding domain. Biochemistry 41: 1275512762, 2002.

SASCHA KOPIC AND JOHN P. GEIBEL


1123. Vilardaga JP, Krasel C, Chauvin S, Bambino T, Lohse MJ, Nissenson RA. Internalization determinants of the parathyroid hormone receptor differentially regulate betaarrestin/receptor association. J Biol Chem 277: 8121 8129, 2002.

1143. Wang LD, Hoeltzel M, Gantz I, Hunter R, Del Valle J. Characterization of the histamine H2 receptor structural components involved in dual signaling. J Pharmacol Exp
Ther 285: 573578, 1998.

1124. Villa-Bellosta R, Ravera S, Sorribas V, Stange G, Levi M, Murer H, Biber J, Forster IC.
The Na-Pi cotransporter PiT-2 (SLC20A2) is expressed in the apical membrane of
rat renal proximal tubules and regulated by dietary Pi. Am J Physiol Renal Physiol 296:
F691F699, 2009.

1144. Wang Q, Curran ME, Splawski I, Burn TC, Millholland JM, VanRaay TJ, Shen J,
Timothy KW, Vincent GM, de Jager T, Schwartz PJ, Toubin JA, Moss AJ, Atkinson
DL, Landes GM, Connors TD, Keating MT. Positional cloning of a novel potassium
channel gene: KVLQT1 mutations cause cardiac arrhythmias. Nat Genet 12: 1723,
1996.

1125. Vinik AI, Gaginella TS, ODorisio TM, Shapiro B, Wagner L. The distribution and
characterization of somatostatin-like immunoreactivity in epithelial cells, submucosa, muscle of the rat stomach and intestine. Endocrinology 109: 19211926, 1981.
1126. Voets T, Janssens A, Prenen J, Droogmans G, Nilius B. Mg2-dependent gating and
strong inward rectification of the cation channel TRPV6. J Gen Physiol 121: 245260,
2003.
1127. Vogelsang H, Schofl R, Tillinger W, Ferenci P, Gangl A. 25-Hydroxyvitamin D absorption in patients with Crohns disease and with pancreatic insufficiency. Wien Klin
Wochenschr 109: 678 682, 1997.
1128. Wada M, Furuya Y, Sakiyama J, Kobayashi N, Miyata S, Ishii H, Nagano N. The
calcimimetic compound NPS R-568 suppresses parathyroid cell proliferation in rats
with renal insufficiency. Control of parathyroid cell growth via a calcium receptor. J
Clin Invest 100: 29772983, 1997.
1129. Wada S, Martin TJ, Findlay DM. Homologous regulation of the calcitonin receptor in
mouse osteoclast-like cells and human breast cancer T47D cells. Endocrinology 136:
26112621, 1995.

1131. Waldum HL, Sandvik AK, Brenna E, Petersen H. Gastrin-histamine sequence in the
regulation of gastric acid secretion. Gut 32: 698 701, 1991.
1132. Wallace JL, Cucala M, Mugridge K, Parente L. Secretagogue-specific effects of interleukin-1 on gastric acid secretion. Am J Physiol Gastrointest Liver Physiol 261: G559
G564, 1991.

1146. Wang TJ, Zhang F, Richards JB, Kestenbaum B, van Meurs JB, Berry D, Kiel DP, Streeten
EA, Ohlsson C, Koller DL, Peltonen L, Cooper JD, OReilly PF, Houston DK, Glazer NL,
Vandenput L, Peacock M, Shi J, Rivadeneira F, McCarthy MI, Anneli P, de Boer IH,
Mangino M, Kato B, Smyth DJ, Booth SL, Jacques PF, Burke GL, Goodarzi M, Cheung
CL, Wolf M, Rice K, Goltzman D, Hidiroglou N, Ladouceur M, Wareham NJ, Hocking LJ,
Hart D, Arden NK, Cooper C, Malik S, Fraser WD, Hartikainen AL, Zhai G, Macdonald
HM, Forouhi NG, Loos RJ, Reid DM, Hakim A, Dennison E, Liu Y, Power C, Stevens HE,
Jaana L, Vasan RS, Soranzo N, Bojunga J, Psaty BM, Lorentzon M, Foroud T, Harris TB,
Hofman A, Jansson JO, Cauley JA, Uitterlinden AG, Gibson Q, Jarvelin MR, Karasik D,
Siscovick DS, Econs MJ, Kritchevsky SB, Florez JC, Todd JA, Dupuis J, Hypponen E,
Spector TD. Common genetic determinants of vitamin D insufficiency: a genomewide association study. Lancet 376: 180 188, 2010.
1147. Wang W, Lu M, Balazy M, Hebert SC. Phospholipase A2 is involved in mediating the
effect of extracellular Ca2 on apical K channels in rat TAL. Am J Physiol Renal
Physiol 273: F421F429, 1997.
1148. Wang WH, Lu M, Hebert SC. Cytochrome P-450 metabolites mediate extracellular
Ca2-induced inhibition of apical K channels in the TAL. Am J Physiol Cell Physiol
271: C103C111, 1996.
1149. Wasserman RH, Corradino RA, Taylor AN. Vitamin D-dependent calcium-binding
protein. Purification and some properties. J Biol Chem 243: 3978 3986, 1968.
1150. Wasserman RH, Kallfelz FA, Comar CL. Active transport of calcium by rat duodenum in vivo. Science 133: 883 884, 1961.

1133. Walling MW, Rothman SS. Apparent increase in carrier affinity for intestinal calcium
transport following dietary calcium restriction. J Biol Chem 245: 50075011, 1970.

1151. Wasserman RH, Taylor AN. Vitamin D3-induced calcium-binding protein in chick
intestinal mucosa. Science 152: 791793, 1966.

1134. Walling MW, Rothman SS. Phosphate-independent, carrier-mediated active transport of calcium by rat intestine. Am J Physiol 217: 1144 1148, 1969.

1152. Wasserman RH, Taylor AN. Vitamin D-dependent calcium-binding protein. Response to some physiological and nutritional variables. J Biol Chem 243: 39873993,
1968.

1135. Wallmark B, Brandstrom A, Larsson H. Evidence for acid-induced transformation of


omeprazole into an active inhibitor of (H K)-ATPase within the parietal cell.
Biochim Biophys Acta 778: 549 558, 1984.
1136. Walters JR. Calbindin-D9k stimulates the calcium pump in rat enterocyte basolateral
membranes. Am J Physiol Gastrointest Liver Physiol 256: G124 G128, 1989.
1137. Walters JR, Howard A, Charpin MV, Gniecko KC, Brodin P, Thulin E, Forsen S.
Stimulation of intestinal basolateral membrane calcium-pump activity by recombinant synthetic calbindin-D9k and specific mutants. Biochem Biophys Res Commun
170: 603 608, 1990.
1138. Walters JR, Howard A, Lowery LJ, Mawer EB, Legon S. Expression of genes involved
in calcium absorption in human duodenum. Eur J Clin Invest 29: 214 219, 1999.
1139. Wang B, Bisello A, Yang Y, Romero GG, Friedman PA. NHERF1 regulates parathyroid hormone receptor membrane retention without affecting recycling. J Biol Chem
282: 36214 36222, 2007.
1140. Wang B, Yang Y, Abou-Samra AB, Friedman PA. NHERF1 regulates parathyroid
hormone receptor desensitization: interference with beta-arrestin binding. Mol
Pharmacol 75: 1189 1197, 2009.
1141. Wang DH, Hu YS, Du JJ, Hu YY, Zhong WD, Qin WJ. Ghrelin stimulates proliferation
of human osteoblastic TE85 cells via NO/cGMP signaling pathway. Endocrine 35:
112117, 2009.
1142. Wang L, Gantz I, DelValle J. Histamine H2 receptor activates adenylate cyclase and
PLC via separate GTP-dependent pathways. Am J Physiol Gastrointest Liver Physiol
271: G613G620, 1996.

266

1153. Watanabe S, Chey WY, Lee KY, Chang TM. Secretin is released by digestive products of fat in dogs. Gastroenterology 90: 1008 1017, 1986.
1154. Watson F, Kiernan RS, Deavall DG, Varro A, Dimaline R. Transcriptional activation of
the rat vesicular monoamine transporter 2 promoter in gastric epithelial cells: regulation by gastrin. J Biol Chem 276: 76617671, 2001.
1155. Watson PH, Fraher LJ, Hendy GN, Chung UI, Kisiel M, Natale BV, Hodsman AB.
Nuclear localization of the type 1 PTH/PTHrP receptor in rat tissues. J Bone Miner
Res 15: 10331044, 2000.
1156. Webb AR, Kline L, Holick MF. Influence of season and latitude on the cutaneous
synthesis of vitamin D3: exposure to winter sunlight in Boston and Edmonton will not
promote vitamin D3 synthesis in human skin. J Clin Endocrinol Metab 67: 373378,
1988.
1157. Weiss LA, Langenberg C, Barrett-Connor E. Ghrelin and bone: is there an association in older adults?: the Rancho Bernardo study. J Bone Miner Res 21: 752757, 2006.
1158. Weitkamp LR, Rucknagel DL, Gershowitz H. Genetic linkage between structural loci
for albumin and group specific component (Gc). Am J Hum Genet 18: 559 571, 1966.
1159. Wettschureck N, Lee E, Libutti SK, Offermanns S, Robey PG, Spiegel AM. Parathyroid-specific double knockout of Gq and G11 alpha-subunits leads to a phenotype
resembling germline knockout of the extracellular Ca2-sensing receptor. Mol Endocrinol 21: 274 280, 2007.
1160. Whalen EJ, Rajagopal S, Lefkowitz RJ. Therapeutic potential of beta-arrestin- and G
protein-biased agonists. Trends Mol Med 17: 126 139, 2011.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

1130. Wada S, Udagawa N, Nagata N, Martin TJ, Findlay DM. Calcitonin receptor downregulation relates to calcitonin resistance in mature mouse osteoclasts. Endocrinology
137: 10421048, 1996.

1145. Wang TC, Koh TJ, Varro A, Cahill RJ, Dangler CA, Fox JG, Dockray GJ. Processing
and proliferative effects of human progastrin in transgenic mice. J Clin Invest 98:
1918 1929, 1996.

GASTRIC ACID, CALCIUM ABSORPTION, AND BONE HEALTH


1161. Wiborg O, Berglund L, Boel E, Norris F, Norris K, Rehfeld JF, Marcker KA, Vuust J.
Structure of a human gastrin gene. Proc Natl Acad Sci USA 81: 10671069, 1984.
1162. Wikvall K. Hydroxylations in biosynthesis of bile acids. Isolation of a cytochrome
P-450 from rabbit liver mitochondria catalyzing 26-hydroxylation of C27-steroids. J
Biol Chem 259: 3800 3804, 1984.
1163. Wilkes JM, Kajimura M, Scott DR, Hersey SJ, Sachs G. Muscarinic responses of gastric
parietal cells. J Membr Biol 122: 97110, 1991.
1164. Windaus A. The chemistry of irradiated ergosterol. Proc R Soc Lond 108: 568 575,
1931.
1165. Windaus A, Bock F. ber das Provitamin aus dem Sterin der Schweineschwarte.
Hoppe-Seylers Zeitschr physiol Chem 245: 168 170, 1936.
1166. Windaus A, Hess A. Sterine und antirachitisches Vitamin. In: Nachrichten von der
Gesellschaft der Wissenschaften zu Gttingen aus dem Jahre 1926. Berlin: Weidmannsche Buchhandlung, 1927, p. 175184.
1167. Windaus A, Lttringhaus A, Deppe M. ber das krystallisierte Vitamin D1. Just Lieb
Ann Chem 489: 252269, 1931.
1168. Wissenbach U, Niemeyer BA, Fixemer T, Schneidewind A, Trost C, Cavalie A, Reus
K, Meese E, Bonkhoff H, Flockerzi V. Expression of CaT-like, a novel calciumselective channel, correlates with the malignancy of prostate cancer. J Biol Chem 276:
1946119468, 2001.

1169. Wolosin JM, Forte JG. Stimulation of oxyntic cell triggers K and Cl conductances
in apical H-K-ATPase membrane. Am J Physiol Cell Physiol 246: C537C545, 1984.

1182. Yamada T, Soll AH, Park J, Elashoff J. Autonomic regulation of somatostatin release:
studies with primary cultures of canine fundic mucosal cells. Am J Physiol Gastrointest
Liver Physiol 247: G567G573, 1984.
1183. Yamaguchi T, Chattopadhyay N, Kifor O, Brown EM. Extracellular calcium
[Ca2(o)]-sensing receptor in a murine bone marrow-derived stromal cell line
(ST2): potential mediator of the actions of Ca2(o) on the function of ST2 cells.
Endocrinology 139: 35613568, 1998.
1184. Yamaguchi T, Chattopadhyay N, Kifor O, Butters RR Jr, Sugimoto T, Brown EM.
Mouse osteoblastic cell line (MC3T3E1) expresses extracellular calcium (Ca2o)sensing receptor and its agonists stimulate chemotaxis and proliferation of
MC3T3-E1 cells. J Bone Miner Res 13: 1530 1538, 1998.
1185. Yamaguchi T, Chattopadhyay N, Kifor O, Ye C, Vassilev PM, Sanders JL, Brown EM.
Expression of extracellular calcium-sensing receptor in human osteoblastic MG-63
cell line. Am J Physiol Cell Physiol 280: C382C393, 2001.
1186. Yamauchi M, Yamaguchi T, Kaji H, Sugimoto T, Chihara K. Involvement of calciumsensing receptor in osteoblastic differentiation of mouse MC3T3E1 cells. Am J
Physiol Endocrinol Metab 288: E608 E616, 2005.
1187. Yang F, Brune JL, Naylor SL, Cupples RL, Naberhaus KH, Bowman BH. Human
group-specific component (Gc) is a member of the albumin family. Proc Natl Acad Sci
USA 82: 7994 7998, 1985.
1188. Yang F, Luna VJ, McAnelly RD, Naberhaus KH, Cupples RL, Bowman BH. Evolutionary and structural relationships among the group-specific component, albumin and
alpha-fetoprotein. Nucleic Acids Res 13: 8007 8017, 1985.
1189. Yang W, Friedman PA, Kumar R, Omdahl JL, May BK, Siu-Caldera ML, Reddy GS,
Christakos S. Expression of 25(OH)D3 24-hydroxylase in distal nephron: coordinate
regulation by 1,25(OH)2D3 and cAMP or PTH. Am J Physiol Endocrinol Metab 276:
E793E805, 1999.

1171. Wortsman J, Pak CY, Bartter FC, Deftos L, Delea CS. Pathogenesis of osteomalacia
in secondary hyperparathyroidism after gastrectomy. Am J Med 52: 556 564, 1972.

1190. Yang YX, Lewis JD, Epstein S, Metz DC. Long-term proton pump inhibitor therapy
and risk of hip fracture. JAMA 296: 29472953, 2006.

1172. Wrenn RW, Biddulph DM. Parathyroid hormone-induced calcium efflux from isolated renal cortical tubules: evidence for cyclic AMP mediation. Mol Cell Endocrinol
15: 29 40, 1979.

1191. Yao X, Karam SM, Ramilo M, Rong Q, Thibodeau A, Forte JG. Stimulation of gastric
acid secretion by cAMP in a novel alpha-toxin-permeabilized gland model. Am J
Physiol Cell Physiol 271: C61C73, 1996.

1173. Wright MJ, Sullivan RR, Gaffney-Stomberg E, Caseria DM, OBrien KO, Proctor DD,
Simpson CA, Kerstetter JE, Insogna KL. Inhibiting gastric acid production does not
affect intestinal calcium absorption in young, healthy individuals: a randomized,
crossover, controlled clinical trial. J Bone Miner Res 25: 22052211, 2010.

1192. Ye CP, Yamaguchi T, Chattopadhyay N, Sanders JL, Vassilev PM, Brown EM. Extracellular calcium-sensing-receptor (CaR)-mediated opening of an outward K channel in murine MC3T3-E1 osteoblastic cells: evidence for expression of a functional
CaR. Bone 27: 2127, 2000.

1174. Wu G, Burzon DT, di SantAgnese PA, Schoen S, Deftos LJ, Gershagen S, Cockett
AT. Calcitonin receptor mRNA expression in the human prostate. Urology 47: 376
381, 1996.

1193. Yergey AL, Abrams SA, Vieira NE, Aldroubi A, Marini J, Sidbury JB. Determination of
fractional absorption of dietary calcium in humans. J Nutr 124: 674 682, 1994.

1175. Wu S, Grieff M, Brown AJ. Regulation of renal vitamin D-24-hydroxylase by phosphate: effects of hypophysectomy, growth hormone and insulin-like growth factor I.
Biochem Biophys Res Commun 233: 813 817, 1997.
1176. Wuster C, Raue F, Meyer C, Bergmann M, Ziegler R. Long-term excess of endogenous calcitonin in patients with medullary thyroid carcinoma does not affect bone
mineral density. J Endocrinol 134: 141147, 1992.
1177. Wyatt MA, Jarvie E, Feniuk W, Humphrey PP. Somatostatin sst2 receptor-mediated
inhibition of parietal cell function in rat isolated gastric mucosa. Br J Pharmacol 119:
905910, 1996.
1178. Xu H, Zhao H, Tian W, Yoshida K, Roullet JB, Cohen DM. Regulation of a transient
receptor potential (TRP) channel by tyrosine phosphorylation. SRC family kinasedependent tyrosine phosphorylation of TRPV4 on TYR-253 mediates its response to
hypotonic stress. J Biol Chem 278: 11520 11527, 2003.

1194. Yoshizawa T, Handa Y, Uematsu Y, Takeda S, Sekine K, Yoshihara Y, Kawakami T,


Arioka K, Sato H, Uchiyama Y, Masushige S, Fukamizu A, Matsumoto T, Kato S. Mice
lacking the vitamin D receptor exhibit impaired bone formation, uterine hypoplasia
and growth retardation after weaning. Nat Genet 16: 391396, 1997.
1195. Yu EW, Bauer SR, Bain PA, Bauer DC. Proton pump inhibitors and risk of fractures:
a meta-analysis of 11 international studies. Am J Med 124: 519 526, 2011.
1196. Yu PL, Fujimura M, Hayashi N, Nakamura T, Fujimiya M. Mechanisms in regulating
the release of serotonin from the perfused rat stomach. Am J Physiol Gastrointest Liver
Physiol 280: G1099 G1105, 2001.
1197. Yue L, Peng JB, Hediger MA, Clapham DE. CaT1 manifests the pore properties of
the calcium-release-activated calcium channel. Nature 410: 705709, 2001.
1198. Zacharias DA, Kappen C. Developmental expression of the four plasma membrane
calcium ATPase (Pmca) genes in the mouse. Biochim Biophys Acta 1428: 397 405,
1999.

1179. Xu J, Henriksnas J, Barone S, Witte D, Shull GE, Forte JG, Holm L, Soleimani M.
SLC26A9 is expressed in gastric surface epithelial cells, mediates Cl/HCO3 exchange, is inhibited by NH4. Am J Physiol Cell Physiol 289: C493C505, 2005.

1199. Zaidi M, Chambers TJ, Gaines Das RE, Morris HR, MacIntyre I. A direct action of
human calcitonin gene-related peptide on isolated osteoclasts. J Endocrinol 115:
511518, 1987.

1180. Xu J, Song P, Miller ML, Borgese F, Barone S, Riederer B, Wang Z, Alper SL, Forte JG,
Shull GE, Ehrenfeld J, Seidler U, Soleimani M. Deletion of the chloride transporter
Slc26a9 causes loss of tubulovesicles in parietal cells and impairs acid secretion in the
stomach. Proc Natl Acad Sci USA 105: 1795517960, 2008.

1200. Zanello LP, Norman AW. Rapid modulation of osteoblast ion channel responses by
1alpha,25(OH)2-vitamin D3 requires the presence of a functional vitamin D nuclear
receptor. Proc Natl Acad Sci USA 101: 1589 1594, 2004.

1181. Xue Y, Fleet JC. Intestinal vitamin D receptor is required for normal calcium and
bone metabolism in mice. Gastroenterology 136: 13171327, e13111312, 2009.

1201. Zanner R, Hapfelmeier G, Gratzl M, Prinz C. Intracellular signal transduction during


gastrin-induced histamine secretion in rat gastric ECL cells. Am J Physiol Cell Physiol
282: C374 C382, 2002.

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

267

Downloaded from on December 8, 2014

1170. Woodrow JP, Sharpe CJ, Fudge NJ, Hoff AO, Gagel RF, Kovacs CS. Calcitonin plays
a critical role in regulating skeletal mineral metabolism during lactation. Endocrinology
147: 4010 4021, 2006.

SASCHA KOPIC AND JOHN P. GEIBEL


1202. Zavros Y, Fleming WR, Hardy KJ, Shulkes A. Regulation of fundic and antral somatostatin secretion by CCK and gastrin. Am J Physiol Gastrointest Liver Physiol 274:
G742G750, 1998.

1214. Zhao CM, Chen D, Yamada H, Dornonville de la Cour C, Lindstrom E, Persson L,


Hakanson R. Rat stomach ECL cells: mode of activation of histidine decarboxylase.
Regul Pept 114: 2127, 2003.

1203. Zavros Y, Shulkes A. Cholecystokinin (CCK) regulates somatostatin secretion through both
the CCK-A and CCK-B/gastrin receptors in sheep. J Physiol 505: 811821, 1997.

1215. Zhao CM, Jacobsson G, Chen D, Hakanson R, Meister B. Exocytotic proteins in


enterochromaffin-like (ECL) cells of the rat stomach. Cell Tissue Res 290: 539 551,
1997.

1204. Zehnder D, Bland R, Chana RS, Wheeler DC, Howie AJ, Williams MC, Stewart PM,
Hewison M. Synthesis of 1,25-dihydroxyvitamin D3 by human endothelial cells is
regulated by inflammatory cytokines: a novel autocrine determinant of vascular cell
adhesion. J Am Soc Nephrol 13: 621 629, 2002.
1205. Zehnder D, Bland R, Williams MC, McNinch RW, Howie AJ, Stewart PM, Hewison
M. Extrarenal expression of 25-hydroxyvitamin d(3)-1 alpha-hydroxylase. J Clin Endocrinol Metab 86: 888 894, 2001.
1206. Zehnder D, Evans KN, Kilby MD, Bulmer JN, Innes BA, Stewart PM, Hewison M.
The ontogeny of 25-hydroxyvitamin D(3) 1alpha-hydroxylase expression in human
placenta and decidua. Am J Pathol 161: 105114, 2002.
1207. Zeng N, Athmann C, Kang T, Lyu RM, Walsh JH, Ohning GV, Sachs G, Pisegna JR.
PACAP type I receptor activation regulates ECL cells and gastric acid secretion. J Clin
Invest 104: 13831391, 1999.
1208. Zeng N, Kang T, Lyu RM, Wong H, Wen Y, Walsh JH, Sachs G, Pisegna JR. The
pituitary adenylate cyclase activating polypeptide type 1 receptor (PAC1-R) is expressed on gastric ECL cells: evidence by immunocytochemistry and RT-PCR. Ann
NY Acad Sci 865: 147156, 1998.
1209. Zeng N, Kang T, Wen Y, Wong H, Walsh J, Sachs G. Galanin inhibition of enterochromaffin-like cell function. Gastroenterology 115: 330 339, 1998.

1216. Zhao G, Simpson RU. Membrane localization, Caveolin-3 association and rapid
actions of vitamin D receptor in cardiac myocytes. Steroids 75: 555559,
2010.
1217. Zhong Y, Armbrecht HJ, Christakos S. Calcitonin, a regulator of the 25-hydroxyvitamin D3 1alpha-hydroxylase gene. J Biol Chem 284: 11059 11069, 2009.
1218. Zhou AT, Assil I, Abou-Samra AB. Role of asparagine-linked oligosaccharides in the
function of the rat PTH/PTHrP receptor. Biochemistry 39: 6514 6520, 2000.
1219. Zhou C, Lu F, Cao K, Xu D, Goltzman D, Miao D. Calcium-independent and
1,25(OH)2D3-dependent regulation of the renin-angiotensin system in 1alpha-hydroxylase knockout mice. Kidney Int 74: 170 179, 2008.
1220. Zhuang L, Peng JB, Tou L, Takanaga H, Adam RM, Hediger MA, Freeman MR.
Calcium-selective ion channel, CaT1, is apically localized in gastrointestinal tract
epithelia and is aberrantly expressed in human malignancies. Lab Invest 82: 1755
1764, 2002.
1221. Zhuang X, Adipietro KA, Datta S, Northup JK, Ray K. Rab1 small GTP-binding
protein regulates cell surface trafficking of the human calcium-sensing receptor.
Endocrinology 151: 5114 5123, 2010.
1222. Zierold C, Mings JA, DeLuca HF. Parathyroid hormone regulates 25-hydroxyvitamin
D(3)-24-hydroxylase mRNA by altering its stability. Proc Natl Acad Sci USA 98:
1357213576, 2001.

1211. Zhang JX, Fasciotto BH, Darling DS, Cohn DV. Pancreastatin, a chromogranin Aderived peptide, inhibits transcription of the parathyroid hormone and chromogranin A genes and decreases the stability of the respective messenger ribonucleic
acids in parathyroid cells in culture. Endocrinology 134: 1310 1316, 1994.

1223. Zietkiewicz E, Labuda M, Sinnett D, Glorieux FH, Labuda D. Linkage mapping by


simultaneous screening of multiple polymorphic loci using Alu oligonucleotide-directed PCR. Proc Natl Acad Sci USA 89: 8448 8451, 1992.

1212. Zhang W, Na T, Wu G, Jing H, Peng JB. Down-regulation of intestinal apical calcium entry
channel TRPV6 by ubiquitin E3 ligase Nedd42. J Biol Chem 285: 3658636596, 2010.

1224. Zornitzer AE, Bronner F. In situ studies of calcium absorption in rats. Am J Physiol
220: 12611266, 1971.

1213. Zhang Z, Sun S, Quinn SJ, Brown EM, Bai M. The extracellular calcium-sensing
receptor dimerizes through multiple types of intermolecular interactions. J Biol Chem
276: 5316 5322, 2001.

1225. Zuo Q, Claveau D, Hilal G, Leclerc M, Brunette MG. Effect of calcitonin on calcium
transport by the luminal and basolateral membranes of the rabbit nephron. Kidney Int
51: 19911999, 1997.

268

Physiol Rev VOL 93 JANUARY 2013 www.prv.org

Downloaded from on December 8, 2014

1210. Zeng N, Walsh JH, Kang T, Wu SV, Sachs G. Peptide YY inhibition of rat gastric
enterochromaffin-like cell function. Gastroenterology 112: 127135, 1997.

Gastric Acid, Calcium Absorption, and Their Impact


on Bone Health
Sascha Kopic and John P. Geibel

Physiol Rev 93:189-268, 2013. doi:10.1152/physrev.00015.2012


You might find this additional info useful...
This article cites 1211 articles, 454 of which can be accessed free at:
/content/93/1/189.full.html#ref-list-1
This article has been cited by 1 other HighWire hosted articles
Hypomagnesaemia associated with long-term use of proton pump inhibitors
James Wei Tatt Toh, Evonne Ong and Robert Wilson
Gastroenterol. Rep., August 19, 2014; .
[Abstract] [Full Text] [PDF]
Updated information and services including high resolution figures, can be found at:
/content/93/1/189.full.html

This information is current as of December 8, 2014.

Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is
published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda
MD 20814-3991. Copyright 2013 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our
website at http://www.the-aps.org/.

Downloaded from on December 8, 2014

Additional material and information about Physiological Reviews can be found at:
http://www.the-aps.org/publications/prv

Das könnte Ihnen auch gefallen