You are on page 1of 27

Intestinal Calcium Absorption

Kannikar Wongdee,1,2 Krittikan Chanpaisaeng,2,3 Jarinthorn Teerapornpuntakit,2,4 and


Narattaphol Charoenphandhu*2,5,6,7

ABSTRACT
In this article, we focus on mammalian calcium absorption across the intestinal epithelium in
normal physiology. Intestinal calcium transport is essential for supplying calcium for metabolism
and bone mineralization. Dietary calcium is transported across the mucosal epithelia via saturable
transcellular and nonsaturable paracellular pathways, both of which are under the regulation of
1,25-dihydroxyvitamin D3 and several other endocrine and paracrine factors, such as parathyroid
hormone, prolactin, 17β-estradiol, calcitonin, and fibroblast growth factor-23. Calcium absorption
occurs in several segments of the small and large intestine with varying rates and capacities.
Segmental heterogeneity also includes differential expression of calcium transporters/carriers
(e.g., transient receptor potential cation channel and calbindin-D9k ) and the presence of favorable
factors (e.g., pH, luminal contents, and gut motility). Other proteins and transporters (e.g., plasma
membrane vitamin D receptor and voltage-dependent calcium channels), as well as vesicular
calcium transport that probably contributes to intestinal calcium absorption, are also discussed.
© 2021 American Physiological Society. Compr Physiol 11:1-27, 2021.

Didactic Synopsis • Neural regulation of intestinal calcium absorption by


enteric nervous system is plausible but direct evidence to
Major Teaching Points support this notion is scant and inconclusive.
• Polarity is an important aspect of epithelia. Epithelial
cells including intestinal enterocytes have two domains,
that is, apical and basolateral domains. Apical domain is Introduction
exposed to lumen or external environment and respon- Calcium absorption from the intestine is the sole supply of
sible for absorption and protection. Basolateral domain calcium for various body functions including bone miner-
is associated with neighboring epithelial cells and the alization, neural transmission, cardiomyocyte contraction,
basement membrane. hormonal secretion, and blood coagulation. Daily dietary
calcium requirements vary according to demographics, for
• Calcium (Ca2+ —as free-ionized calcium) traverses the
example, age, gender, ethnicity, and physiological conditions
intestinal epithelia via transcellular and paracellular
pathways.
*Correspondence to: naratt@narattsys.com
• Transcellular Ca2+ transport is a three-step process 1 Faculty of Allied Health Sciences, Burapha University, Chonburi,
consisting of TRPV6-mediated apical calcium uptake, Thailand
2 Center of Calcium and Bone Research (COCAB), Faculty of Science,
cytoplasmic diffusion of Ca2+ -laden calbindin-D9k ,
and PMCA1b - and NCX1-mediated basolateral calcium Mahidol University, Bangkok, Thailand
3 Functional Ingredients and Food Innovation Research Group,
extrusion.
National Center for Genetic Engineering and Biotechnology (BIOTEC),
• Paracellular Ca2+ transport is determined by tight junc- National Science and Technology Development Agency (NSTDA),
Pathum Thani, Thailand
tion permselectivity and the expression of tight junction
4 Department of Physiology, Faculty of Medical Science, Naresuan
proteins, for example, claudin-2, -12, or -15 as well as per-
University, Phitsanulok, Thailand
ijunctional actomyosin ring remodeling. 5 Department of Physiology, Faculty of Science, Mahidol University,

• The calciotropic hormone 1,25-dihydroxyvitamin D3 (an Bangkok, Thailand


6 Institute of Molecular Biosciences, Mahidol University, Nakhon
active form of vitamin D) is a potent positive regulator of
Pathom, Thailand
transcellular and paracellular calcium absorption. 7 The Academy of Science, The Royal Society of Thailand, Bangkok,

• A number of humoral factors (e.g., fibroblast growth Thailand


factor-23 and calcitonin) and certain luminal factors Published online, May 2021 (comprehensivephysiology.com)
(e.g., iron and alkaline pH in the bulk phase of intestinal DOI:10.1002/cphy.c200014
lumen) can reduce intestinal calcium absorption. Copyright © American Physiological Society.

Volume 11, May 2021 1


Intestinal Calcium Absorption Comprehensive Physiology

such as pregnancy and lactation, and exposure to sunlight absorption, intestinal sojourn time, and solubility of dietary
or ultraviolet B radiation (for reviews, please see Refs. 42, calcium. The following section will address how these factors
201, 230). In adults, it ranges from 1000 to 1300 mg elemen- affect the amount and efficiency of calcium absorption in a
tal calcium per day (42). In rodents, females normally have given segment (Figures 1 and 2).
higher rates of intestinal calcium absorption than males (212).
Higher calcium demand is apparent in certain conditions, for
example, bone calcium accretion during adolescence, placen- Stomach
tal calcium transfer during pregnancy, and milk production Calcium absorption is generally absent in the stomach (155).
in lactating women (36, 157, 235). On the other hand, the However, the stomach plays a role in providing an acidic
rate of intestinal calcium absorption declines with age due in environment for calcium solubilization (for review, please
part to downregulation of nuclear vitamin D receptor (VDR) see Ref. 127). Previous preclinical and clinical observations
expression and the decrease in responsiveness of target tissues consistently showed that gastrectomy resulted in impaired
to 1,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ] (82). calcium absorption and low bone mass (23, 41, 153). Stomach
Regarding calcium absorption, the gastrointestinal mucosal acid produced by gastric parietal cells increases dissolution
epithelia exhibit varying degrees of heterogeneity. Most mam- of calcium salts and allows calcium ions to form complexes
mals including humans, rats, mice, horses, and ruminants with other food constituents. As a result, soluble calcium can
absorb dietary calcium predominantly in the small intestine be absorbed when it passes into the intestine. A variety of
in a vitamin D-dependent manner (69, 173, 238). However, transporters and gastrin receptors in the gastric mucosa are
each intestinal segment utilizes different cellular mechanisms involved in maintaining normal gastric acidification. Animals
to absorb calcium. For example, the proximal small intestine with genetic disruption of genes expressing proteins involved
of the rat—duodenum and proximal jejunum—is capable of in the stomach acid production, for example, Cckbr, Tcirg1,
transporting calcium via both transcellular and paracellular Snx10, had defect in gastric acid secretion, hypocalcemia,
pathways, whereas the paracellular route is more important and osteopathy (83, 205, 252). These findings implicate the
in the distal small intestine (121). In addition, the intestine importance of normal stomach acid secretion in calcium
also shows heterogeneity along the crypt-villus axis, that homeostasis. In support of this, extensive investigations in
is, calcium secretion predominantly takes place in the crypt individuals with decreased gastric acidity (e.g., elderly and
region, whereas calcium absorption mainly occurs in the atrophic gastritis patients) revealed that insoluble calcium
upper two-thirds of the villus which corresponds to the abun- salts had reduced calcium solubility in these subjects (243).
dant expression of calcium-binding protein calbindin-D9k Taken together, these data strongly suggest that gastric acidi-
(also known as S100 calcium-binding protein G or S100G) at fication is indispensable for intestinal calcium absorption and
the villous tip (100). helps maintain normocalcemia.
In this article, we aim to elaborate on the cellular and
molecular mechanisms of calcium transport across the mam-
malian intestinal epithelium as well as its local and systemic Duodenum
regulation by paracrine and endocrine factors. Dietary nutri- When acidic chyme enters the duodenum, it will be neu-
ents and luminal factors affecting calcium absorption are also tralized by bicarbonate and alkaline mucus secreted from
discussed. Brunner’s glands—also known as the submucosal duodenal
glands. Because of a high amount of free-ionized calcium,
the immediate change of pH in the intestinal environment
Segmental Heterogeneity of Calcium does not impact the amount of soluble calcium, and calcium
Absorption solubility is highest in the duodenum as compared to other
distal intestinal segments (60, 73). In addition, food in a liquid
Calcium absorption occurs throughout the gastrointestinal form yields a higher amount of available calcium in a shorter
tract yet there is heterogeneity in the efficiency of intestinal time compared to the solid food (155). However, with only
calcium absorption and transport pathways used in each 2 to 3 min sojourn time of food in the duodenum (60), the
segment. The efficiency of calcium absorption partly depends amount of calcium absorbed in the duodenum accounts for
on luminal calcium concentration that is mostly attributed to approximately 8% of the total intestinal calcium absorption
the dietary intake. Calcium is transported across the epithe- (44, 60, 159).
lium through two primary mechanisms, that is, transcellular Both active transcellular and paracellular transport mech-
[saturable, 1,25(OH)2 D3 -, and energy-dependent] and para- anisms take place in the duodenum (Figure 3B). When
cellular (non-saturable, concentration-dependent) transport. individuals receive inadequate calcium intake, transcellular
The cellular machinery for these two transport pathways transport predominates in the duodenum to increase the
is discussed in the section titled “Cellular Mechanisms of efficiency of calcium absorption. It is well established that
Gastrointestinal Calcium Absorption”. When calcium intake 1,25(OH)2 D3 increases transcellular absorption against a
is adequate or high, the efficiency of calcium absorption in chemical gradient (26, 71, 115, 116, 183) by upregulat-
any given segment is determined by the mechanism used for ing calcium-selective transporters [e.g., transient receptor

2 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Stomach

• Producing acidic environment


for calcium solubilization
Duodenum • JCa, detectable but unclear
physiological significance
Jnet +++ Percent absorbed 8% (probably negligible)

Jejunum

Jnet + Percent absorbed 17%


Colon

Jnet ++ Percent absorbed 15%

Ileum
Percent absorbed
Jnet +
65%–88%
Cecum

Jnet ++++ Percent absorbed 10%


Rectum
Producing acidic environment Unclear physiological significance
from microfloral fermentation (probably negligible)

Figure 1 Segmental heterogeneity of calcium absorption. Key features including magnitude of net absorptive calcium flux
rate (Jnet = Jmucosa-to-serosa − Jserosa-to-mucosa ) and percentage of total absorbed calcium under normal (calcium- and vitamin D-
replete) conditions are presented for each intestinal segment. The number of “+” signs represents the amount of net calcium
flux compared to cecum which has the highest net flux marked with “++++”.

potential cation channel, subfamily V, member 6 (TRPV6) the expression of the tight junction proteins claudin-2 and
and a lesser extent TRPV5] and calcium-binding proteins -12 in the jejunum. Specifically, the mRNA and protein
(e.g., calbindin-D9k in mammals and calbindin-D28k in levels of these claudins decreased significantly (3–4 fold) in
birds). Because this transport mechanism can be saturated, the jejunum of VDR knockout mice compared to wild-type
consequent absorption across the enterocytes decreases with mice (79). The reduced expression of these proteins was also
increased calcium intake (183). On the other hand, the para- observed in the duodenum, ileum, and colon of the VDR
cellular transport of calcium is nonsaturable and is driven by knockout animals, but to a lesser extent than in the jejunum
the electrochemical gradient of calcium and solvent drags (79). Claudin-2 is a tight junction protein in intestinal epithe-
(3, 116, 238). Accordingly, our investigation of rat duodenum lial cells that possesses paracellular cation-selectivity (162,
showed that when luminal concentration of free-ionized 218). A recent study using claudin-2 knockout animals
calcium is greater than 5 mmol/L, the paracellular transport showed that the absence of this protein led to a decrease in
serves as the major pathway (up to 10:1) as compared to the paracellular calcium absorption (46), thus emphasizing the
transcellular transport (31). importance of claudin-2 in this transport pathway.
While paracellular calcium flux takes place throughout the
jejunum, small saturable component of calcium absorption
Jejunum (∼15%) exists mostly in the proximal jejunum (183). Steady
Calcium ions that enter the jejunum are progressively exposed state perfusion studies in healthy adults with exogenous
to higher pH as they approach the distal part of jejunum (pH 1,25(OH)2 D3 administration showed an increase in jejunal
∼6.6–8), resulting in an increase in insoluble calcium in calcium absorption (131). However, recent reports showed
the lumen (60). In rats fed high-calcium diets, the sojourn the expression of TRPV6 was much lower in the jejunum
time in the entire jejunum is 40 to 45 min (20, 60), which is of adult mice compared to newborns (13, 195). Therefore,
20 times longer than the time spent in the duodenum. Thus, transcellular calcium absorption occurs in the jejunum is
the absorbable amount of dietary calcium is higher than postulated to play a role in ensuring a positive calcium
in the duodenum (17% vs. 7%) (44). The jejunal calcium balance in postnatal development when the transcellular
transport occurs transcellularly and paracellularly. In the mechanism in the duodenum is not fully developed. Using
rat jejunum, paracellular transport is responsible for more the Ussing chamber technique, Beggs et al. (13) measured
than 80% of calcium absorption (183). Interestingly, there is transcellular calcium flux across jejunum of TRPV6 mutant
evidence suggesting that 1,25(OH)2 D3 treatment upregulated and Cav 1.3 (L-type calcium channel) knockout mice in

Volume 11, May 2021 3


Intestinal Calcium Absorption Comprehensive Physiology

)
in
(m

9k
e

-D
tim

in
y
lit

nd

5
Intestinal segments

1b
rn

-2

-1

-1
bi

A
PV
u

bi

n
lu

R
jo

ld

ld

ld
al

PM

VD
TR
So
So

C
Stomach 240–300 ++++ – N/A N/A N/A N/A N/A +

++++ ++++ ++++ + ++ +++ ++++


Duodenum 3 ++ ++ +++ +++ ++ ++ +++

++, – – + + + + ++++
Jejunum 40–45 ++
++ ++ ++ ++ +++ +++

– – + + ++ (S) ++ ++++
Ileum ~130 +
++ + ++ ++ ++ ++

++++, ++ + +++ (S) + + + ++


Proxmimal

Cecum 92 +
colon

++++ ++++ +++ ++ ++ ++


+ + +++ +++ + + ++
Proximal
50 + ++ +++ +++ ++++ ++ ++
colon
Cecum

+ + ++ ++ ++ + ++
Distal colon 42 +
+++ +++ ++ +++ ++ ++

(20, 60) (60) (185, 195) (195) (195) (195) (195) (195) (53)
References
(219) (219) (219) (219) (219) (219)

Figure 2 Intestinal segment-specific sojourn time, calcium solubility, and expression profile of genes/proteins related to
calcium transport. The number of “+” signifies the relative abundance of a feature for a given intestinal segment. A “–”
indicates that an mRNA expression is absent in a segment. The sojourn time in the table represents time of chyme. The
lower lines of each segment (red text) represent distribution of mRNA expression of genes related to calcium transport in
female rats refer to our previous study (219). N/A, not available; S, sexual disparity in the expression profile; Cldn, claudin;
PMCA1b , plasma membrane Ca2+ ATPase-1b; TRPV6, transient receptor potential cation channel, subfamily V, member 6;
VDR, vitamin D receptor.

suckling period. They found a significant increase in TRPV6- it has a more basic environment, that is, pH > 8, that reduces
and Cav 1.3-mediated transcellular calcium absorption in the calcium solubility and limits the amount of calcium available
jejunum of 2-week-old suckling mice, whereas these effects for absorption, the total amount of calcium absorbed is
disappeared in the 2-month-old mice. Furthermore, in vivo not reduced (60). This is because when a small amount of
perfusion studies (1.25 mmol/L luminal calcium) showed the ionized calcium in the lumen is absorbed, more insoluble
activity of Cav 1.3 in the proximal jejunum to mid-ileum. This calcium will be ionized and become available for absorption.
suggests that when luminal calcium is high, Cav 1.3-mediated Because the food bolus spends the longest transit time in
pathway may enhance calcium absorption in addition to the the ileum (121.5 min) (60), the slow, yet steady process
paracellular pathway (159); however, its exact contribution of paracellular absorption yields a relatively large amount
requires further studies. of absorbable calcium (65%–88%) (44, 155). Duflos et al.
Despite a relatively high mucosa-to-serosa calcium flux, (60) have shown that, in young adult rats fed 1.5% dietary
the serosa-to-mucosa calcium flux in the jejunum is twice as calcium, the sojourn time of the duodenum, mid-jejunum
much leading to net ionized calcium secretion into the intesti- and distal ileum were 2.5, 12, and 58 min, respectively.
nal lumen (Figure 3B) (116, 195). It is believed that this secre- Therefore, a larger amount of calcium is absorbed in the
tory process may account for the endogenous fecal calcium
ileum even though the “rate” of calcium transport in the
observed in calcium balance studies (232). Taken together, it
ileum is much slower than in the cecum and duodenum
is likely that the calcium absorbed from the jejunum does not
(117). Similarly, in healthy premenopausal women, the
contribute much to the plasma calcium pool.
mouth-to-cecum transit time appeared to be positively cor-
related with the efficiency of calcium absorption (12). The
Ileum ileal mucosa-to-serosa calcium flux mostly occurs through
Among different segments of the small intestine, the ileum is the paracellular pathway and exhibits a limited capacity
where most of ionized calcium is absorbed (155). Although for active transcellular absorption (183). Claudin-2 and -12

4 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

(A) (B)

Lumen Villous tip


c
Calbindin-D9k expression
Villi

a –4 mV

Lacteal
0 mV

e Blood
vessels
f

b Basement
Crypt membrane
Goblet cell
d

a = net Ca2+ absorption c = Transcellular transport


b = net Ca2+ secretion d = Paracellular transport
e = Voltage-dependent Ca2+ secretion
(almost negligible)
f = Unstirred water layer/acid microclimate

Figure 3 Diagrams of intestinal villi. (A) Longitudinal section of villous showing sites of net calcium absorption at
villous (a) and net calcium secretion at crypts of intestine (b). The heterogeneity of calbindin-D9k (S100 calcium-binding
protein G, S100G) expression and the primary direction of calcium flux are presented along the crypt-villous axis.
(B) Transverse section of villous tip featuring transcellular (c) and paracellular calcium absorption (d), secretion (e),
and the existence of unstirred water layer and acid microclimate (f).

which are tight junction proteins involved in the paracellular Similar to the jejunum, calcium secretion in the ileum
transport are highly expressed in the ileum compared to other was also demonstrated in Ussing chamber experiments over
segments (78). On the other hand, the expression of TRPV6 a range of 0.125 to 11.2 mmol/L calcium concentration in
and calbindin-D9k is virtually absent and the expression of the bathing solution (119, 165, 232). The serosa-to-mucosa
plasma membrane Ca2+ ATPase-1b (PMCA1b ) was very low flux was proposed to be generated by hydrostatic driving
in the ileum of mice and rats (21, 99, 183, 185, 195). In force resulting in paracellular calcium movement (14, 119,
addition, studies in animals given 1,25(OH)2 D3 12 h prior 165). By using in situ loop study in female rats, Krishnamra
to calcium absorption test using everted gut sac or in situ et al. (132) demonstrated calcium secretion along the small
intestinal loop techniques showed that ileal calcium absorp- and large intestine and found that the calcium secretion was
tion was not changed (14, 15, 116, 183). Despite evidence directly proportional to the plasma calcium concentration.
illustrating the absence of transcellular calcium transport in For example, calcium secretion in hypercalcemic rats was
the ileum, another line of evidence has suggested that the increased while secretion was decreased in hypocalcemic rats.
ileum probably has the capacity to actively transport calcium
(234). When stimulated with 1,25(OH)2 D3 , the mRNA and
protein expression of calbindin-D9k and PMCA1b in the ileum Cecum
of aged rats were upregulated (8, 9). The explanation for this It has been suggested that the transcellular mechanism of
discrepancy could be the temporal responsive nature of the calcium transport is prominent in the cecum. Findings sup-
ileum to 1,25(OH)2 D3 treatment and very low concentration porting this notion showed that rat and mouse cecum exhibited
of calbindin-D9k (protein level in ileum vs. duodenum at high mRNA expression levels (second to duodenum) of many
16 h post-treatment: ∼2.7 vs. 45 μg/mg) and basolateral transporters, for example, TRPV6 and PMCA1b , while other
calcium pump protein in the ileum (8). Though transcellular intestinal segments had extremely low expression levels
calcium transport proteins are present in the ileum, more of these proteins (195, 219). Furthermore, Dhawan et al.
studies are needed to determine whether this pathway in the (53) showed that the transgenic expression of human VDR
ileum significantly contributes to the total intestinal calcium exclusively in ileum, cecum, and colon (distal intestine) could
absorption. rescue global VDR knockout mice from hypocalcemia and

Volume 11, May 2021 5


Intestinal Calcium Absorption Comprehensive Physiology

abnormal bone mineralization. With 1,25(OH)2 D3 injection, Paracellular calcium absorption is proposed to be the major
the mRNA expression level of TRPV6 and Cyp24A1—the calcium transport pathway in the colon. By using in situ loop
genes known to be regulated by 1,25(OH)2 D3 —was highest technique, Krishnamra et al. (132) showed that calcium
in the cecum (53). Other studies showed significant response absorption in the colon of normocalcemic and hypercalcemic
in cecal calcium flux and expression of transcellular calcium rats was similar as opposed to the reduction in absorption in
transport proteins in response to vitamin D treatment (117, other intestinal segments of hypercalcemic rats. Interestingly,
187). These findings highlight the importance of vitamin only thyroparathyroidectomized (TPTX)-induced hypocal-
D-dependent transcellular calcium transport in the cecum and cemic rats showed a significantly lower rate of calcium
its significance in whole-body calcium homeostasis. absorption in the colon (132). Such a reduction of calcium
When bidirectional calcium fluxes were measured using absorption in the colon of TPTX-induced hypocalcemia was
Ussing chamber technique, cecum showed a significantly hypothesized to be due to a decrease in circulating level of
high net calcium absorption. This supports the importance parathyroid hormone (PTH) or a decrease in 1,25(OH)2 D3
of this segment in intestinal calcium absorption in mice after TPTX (132).
(195) and rats (34, 117, 164). However, some studies showed In addition, Jongwattanapisan et al. (111) demonstrated
different or opposite findings. For example, Brommage et al. the compensatory colonic calcium hyperabsorption in
(19) measured intestinal calcium absorption in cecectomized cecectomized rats. Therein, a calcium balance study was
rats receiving intraperitoneal 1,25(OH)2 D3 injection. They demonstrated in 8-week-old cecectomized female rats.
found that fractional calcium absorption of 47 Ca/47 Sc in the While calcium loss was presented in the first week after the
control and cecectomized rats was not significantly differ- operation, the colonic calcium absorption was significantly
ent. Thus, physiological significance of the cecal calcium elevated. These data suggest that there is a tight regulation
absorption remains unclear and requires further investiga- in the intestinal tract to ensure overall preservation of body
tion. Indeed, the overall calcium absorption might exhibit calcium. Furthermore, colonic perfusion studies in healthy
redundancy or compensatory response—that is, other intesti- human showed that exogenous 1,25(OH)2 D3 administration
nal segments can compensate for the loss of cecal calcium increased net colonic calcium movement from zero toward a
uptake. positive net absorption (92). These studies illustrate that what
we observed in preclinical studies can likely be extrapolated
to humans (111, 187). However, it could be said that in normal
Colon conditions, the colon probably contributes very little to the
The large intestine has been shown to account for less than overall calcium economy but appears to have the machinery
10% of total intestinal calcium absorption (20, 44), with to compensate should the need arise.
calcium transport mostly occurs in the ascending colon and
not at all in the transverse colon (20). By the time, the food
bolus arrives at the colon, only a small amount of calcium is
still available for absorption. Duflos et al. (60) studied cal- Cellular Mechanisms of Gastrointestinal
cium absorption along the entire intestine of 8-week-old male Calcium Absorption
Sprague Dawley rats and reported that approximately 15%
of the total soluble calcium was found in the large intestine. Transcellular and paracellular pathways are two primary
Since a quarter of intestinal sojourn time is spent in the colon cellular mechanisms of calcium transport. In the past, these
(92 min vs. 371 min total time) (20), an appreciable amount two systems were generally viewed as separate entities,
of calcium is absorbed at the colon via both transcellular and with the former mainly responsible for calcium transport in
paracellular pathways. During high calcium intake, the colon times of calcium deficiency. Until recently, new evidence
participates in calcium absorption via paracellular pathway suggests that transcellular and paracellular transport systems
(118, 129, 187, 195). may cooperate to maintain calcium homeostasis. Whereas
Transport proteins involved in the transcellular (TRPV6, the paracellular calcium transport is predominant during
calbindin-D9k , and PMCA1b ) and paracellular (claudin-2, high calcium intake when luminal calcium concentration is
-12, and -15) pathways are expressed in both proximal and much greater than plasma ionized calcium, the transcellular
distal colon (20, 195). In response to vitamin D2 treatment calcium transport becomes important during high calcium
(2 doses of 20,000 IU), the colon of vitamin D-deficient demand (e.g., pregnancy and lactation) or low calcium intake.
mice showed high mucosa-to-serosa flux and low serosa- The cellular machinery for these two transport pathways is
to-mucosa transport resulting in net calcium absorption discussed below.
(187). Transcellular calcium transport is suggested to occur
specifically in the descending colon (64). However, due to a
multiple-fold decrease in expression of transcellular transport Transcellular calcium transport
proteins as compared to the duodenum, the active pathway Free-ionized calcium cannot freely traverse the cell mem-
in the colon is postulated to contribute much less to overall brane of the intestinal enterocytes. Therefore, they require
calcium absorption. specific transporters or calcium channels on both apical

6 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Apical Ca2+ K+ Basolateral


NCKX 4Na+
Trifluoperazine
PMCA1b Vanadate

Ca2+ Ca2+
Ruthenium red TRPV6
Calbindin-D9k
Ca2+
Ca2+ KB-R7943
CAM 3Na+ NCX1
TRPV5
S100G
3Na+

NKA
2K+

Ouabain

Figure 4 Mechanism of transcellular calcium transport across the enterocyte. Calcium is trans-
ported across apical membrane through transient receptor potential cation channel, subfamily
V, member 6 (TRPV6), and, to a lesser extent, TRPV5, facilitated diffusion through the cytosol,
and active basolateral extrusion by the plasma membrane Ca2+ ATPase-1b (PMCA1b ) and other
transporters [e.g., Na+ /Ca2+ exchanger 1 (NCX1) and K+ -dependent Na+ /Ca2+ exchanger
(NCKX)]. Some inhibitors of each calcium transporter are also indicated. Calcium-binding pro-
teins including calbindin-D9k (CaBP-9k) and calmodulin (CAM) may play a role in a buffering
system to prevent intracellular calcium overload as well as translocate calcium ions from the
mucosa to the serosa. The stoichiometry for NCX1 is 4 Na+ to 1-2 Ca2+ moved per transport
cycle; however, an average stoichiometry is 3 Na+ :1 Ca2+ . The stoichiometry of NKA is 3 Na+ :2
K+ . TRPV6, NKA, and NCX1 activities are inhibited by ruthenium red, ouabain, and KB-R7943,
respectively. PMCA1b activity is inhibited by trifluoperazine and vanadate.

and basolateral membranes to facilitate calcium movement goblet cells (251). Because of a much higher expression of
from the intestinal mucosa to the serosal side. This calcium TRPV6 than TRPV5 in the duodenum, TRPV5 is not critical
transport is a vitamin D-regulated process (26, 81, 98) that to the intestinal calcium absorption (169).
consists of three major steps: (i) apical calcium entry that Under normal conditions, TRPV6 plays a critical role in
is mainly facilitated by TRPV6; (ii) intracellular calcium apical calcium entry. Since TRPV6 fully functions under a
translocation mediated by calcium-binding proteins; and (iii) hyperpolarized potential (120), the electrochemical gradient
basolateral calcium extrusion (Figure 4). in the duodenum favors mucosa-to-serosa calcium ion flux.
Consistently, the inducible transgenic TRPV6 expression
in the duodenum of the VDR knockout mice increased
Apical calcium entry calcium absorption and bone density (45). Moreover, when
TRPV channels are the main regulators for apical calcium human intestinal-like Caco-2 cells were examined using
entry. This subfamily of ion channels was first discov- chromatin immunoprecipitation, five putative vitamin D
ered in an extensive screening of cloned DNA from apical response elements (VDRE) were identified upstream to
calcium transporters in rat duodenum using the model of TRPV6 coding region, and mutagenesis of these VDRE
Xenopus laevis oocytes (124). Though all six members of abolished the response to 1,25(OH)2 D3 (156). By binding
TRPV subfamily are capable of aiding calcium flux, only to VDRE and subsequently upregulating VDR-mediated
TRPV5 (also known as CaT2 or ECAC1) and TRPV6 (also transcription, both exogenous and endogenously produced
known as CaT1 or ECAC2) are highly selective to calcium 1,25(OH)2 D3 lead to upregulation of genes involved in active
(PCa / PNa > 100) (99). Both TRPV5 and TRPV6 are epithelial calcium absorption, for example, TRPV6, and calbindin-D9k
calcium channels that mainly facilitate calcium reabsorption (8, 213, 214). Multiple studies exemplified the importance
in the renal tubules and calcium absorption in the intestine, of intestinal VDR in calcium homeostasis. For example,
respectively (94, 158). In the duodenum where mostly active a transgenic intestinal-specific expression of VDR in the
calcium transport takes place, both proteins were expressed in VDR knockout mice could restore their calcium absorp-
humans, whereas only TRPV6 was predominantly expressed tion to normal (248). Not only does 1,25(OH)2 D3 directly
in the mouse and rat (50). In laying hens, the TRPV6 pro- upregulates intestinal calcium transporters, but it could also
tein expression was also abundant in the proximal small indirectly modulates other factors that promote calcium
intestine—duodenum and jejunum—particularly in the apical absorption such as mucosal morphology and increased sur-
membrane of villous cells, but not in the intestinal crypts and face area (49, 204). Specifically, in VDR knockout mice, the

Volume 11, May 2021 7


Intestinal Calcium Absorption Comprehensive Physiology

apical membrane of duodenal enterocytes exhibited elonga- flux and imply that other unidentified mechanisms for active
tion of microvilli—perhaps as a compensatory mechanism transcellular transport may exist.
to improve calcium absorption rate—with no change in Another transport protein Cav 1.3 encoded by CACNA1D
duodenal villous height or crypt depth (133). These data gene has been proposed to be involved in active transcel-
strongly support the importance of VDR and subsequently lular calcium absorption. In contrast to TRPV6 which
the critical role of 1,25(OH)2 D3 in the regulation of calcium normally opens at hyperpolarized potential of less than
absorption. −50 mV, Cav 1.3—as a voltage-dependent L-type calcium
Despite the strong evidence supporting TRPV6 as a pri- channel—has relatively low threshold and opens at sub-
mary transporter in transcellular calcium transport, another threshold potential or slightly depolarized potential of
line of research suggests that the transcellular calcium trans- approximately −40 mV (the resting potential of the apical
port mechanism exhibits redundancy since the absence of membrane of enterocytes is approximately −47 mV) (120).
certain transporters—for example, in TRPV6 knockout mice The nifedipine/verapamil-sensitive Cav 1.3-mediated calcium
and TRPV5/TRPV6 double knockdown Caco-2 cells—did absorption was shown to be coupled with transcellular glu-
cose absorption (151), thus being consistent with the previous
not completely abolish calcium transport (134, 135, 163).
finding that luminal glucose was apparently a potent enhancer
Furthermore, Benn et al. (15) explored whether TRPV6 was
of transcellular calcium absorption (24). During the epithelial
indispensable to active transcellular calcium transport by
glucose uptake in the duodenum and proximal jejunum
using wild-type and TRPV6 knockout mice fed either a low
via Na+ -dependent glucose transporter (SGLT)-1, sodium
(0.02%) or high (1%) calcium diet and the everted gut sac
entry induces slight depolarization of the apical membrane,
assay. Under high calcium conditions where active calcium
thereby triggering Cav 1.3 opening (Figure 5) (120, 238).
transport was limited, wild-type and TRPV6 knockout mice Interestingly, the Cav 1.3-mediated calcium transport led to
showed similarly low transcellular calcium flux (15). In terminal web myosin II phosphorylation, which, in turn,
contrast, TRPV6 knockout mice under low dietary calcium initiated insertion of glucose transporter isoform GLUT2
conditions had a significant decrease in intestinal calcium into the apical membrane for additional glucose uptake in
transport when compared to wild-type mice. Despite this rats (151).
decrease, TRPV6 knockout mice fed the low calcium diet While this line of research supports the role of Cav 1.3 in
still demonstrated a significant 2.9-fold increase in transcellu- calcium absorption and its potential coupling with glucose,
lar calcium transport as compared to TRPV6 knockout mice Reyes-Fernandez and Fleet (193) did not observe the effect of
fed the high calcium diet (15). Moreover, active intestinal glucose (25 mmol/L) in enhancing calcium absorption or any
calcium absorption of TRPV6 knockout mice could still be changes in the mRNA expression levels of Cav 1.3 in duode-
increased by 1,25(OH)2 D3 injection (15, 135). These findings num, jejunum, or ileum of 9-week-old male C57BL/6J mice
have challenged the dogma that TRPV6 is the only trans- fed either a high or low calcium diet for 1 week. Furthermore,
porter necessary for 1,25(OH)2 D3 -mediated apical calcium Li et al. (143) reported that the mRNA expression of Cav 1.3

Apical Depolarization Basolateral


Na+
ΔVt
NKA
PMCA1b
Na+
SGLT1 K+
Opening at subthreshold Glu Ca2+
potential (~ –40 mV)
Dihydropyridines Cav1.3 Calbindin-D9k
Ca2+
Ca2+
TRPV6 Na+ NCX1
CAM
Ca2+
TRPV5 Na+

NKA
Resting potential K+
–47 mV

Figure 5 Effect of apical membrane potential changes on transcellular intestinal calcium


absorption. During glucose uptake via Na+ -dependent glucose transporter (SGLT)-1, sodium
entry induces slight depolarization of the apical membrane, thereby triggering Cav 1.3 open-
ing at slight depolarized potential of approximately −40 mV as compared with −47 mV resting
potential of the apical membrane. Cav 1.3 activity is inhibited by dihydropyridine. PMCA1b ,
plasma membrane Ca2+ ATPase-1b; NCX1, Na+ /Ca2+ exchanger 1; NKA, Na+ /K+ -ATPase;
calbindin-D9 k (S100 calcium-binding protein G, S100G); CAM, calmodulin.

8 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

in male wild-type mice was greater than those in female. By transduction while preventing cell apoptosis. Once a bulk
using Cav 1.3 null mutant mice, they found that only bone of ionized calcium enters the enterocytes, cells must have
parameters of male mice, but not female mice, were nega- mechanisms for buffering the extra calcium as well as translo-
tively affected by loss of Cav 1.3 when compared to wild-type cating these calcium ions from the mucosa to the serosal side.
mice (143). Taken together, Cav 1.3 may contribute to intesti- The intracellular calcium translocation is mediated primarily
nal calcium absorption in a sex-specific manner, which could by calcium-binding proteins. Other mechanisms including
also be governed by species specificity. Therefore, future intraorganellar tunneling, vesicular calcium transport, and
research examining the effect of Cav 1.3 in both genders of free-ionized calcium diffusion may also be used and are
rats, mice, or humans is required to resolve this question. discussed in the following section.
Although the physiological significance of Cav -mediated Calbindin-D9k is the primary calcium-binding protein in
calcium absorption under normal conditions—particularly in the enterocytes with small high-affinity EF-hand domains
vitamin D-replete conditions—remains unclear, Cav appears that can bind 2 moles of calcium per mole of protein (207).
to be the salient calcium transporter during pregnancy and An increase in the concentration of this cytosolic calcium-
lactation when prolactin becomes a major calcium-regulating binding protein was detected at weaning in mice or rats along
hormone (32, 163). A study in Caco-2 monolayer further with increase in the nonsaturable transcellular pathway of
showed that prolactin directly enhanced the Cav -mediated absorption (224). Calbindin-D9k is prominently expressed in
calcium transport via PI3K and PKC𝜁 pathways (221). the small intestine and is accumulated in the villi of duodenal
Furthermore, pre-weaning suckling and post-weaning mice enterocytes, especially at the villous tip (Figure 3A) (100).
differentially exhibited the TRPV6- and Cav 1.3-mediated cal- It has been very well established that calbindin-D9k is
cium transport mechanisms in different segments of the small regulated at the transcriptional level by 1,25(OH)2 D3 medi-
intestine. Specifically, in 2-week-old, pre-weaning suck- ated through vitamin D signaling (48, 71, 198). VDRE were
ling mice, Cav 1.3-mediated calcium uptake in the jejunum identified in the 5′ -flanking regions of calbindin-D9k gene
and ileum was most prominent, whereas in 2-month-old, in humans and rats (48, 109). Fleet and colleagues (72)
post-weaning mice, the TRPV6-mediated calcium uptake in tested whether the intestinal-specific overexpression of VDR
the duodenum was the main pathway for intestinal calcium (HV2 mice) would improve transcellular calcium transport in
absorption (13). The impaired bone mineralization observed mice fed low calcium diets. The investigators observed that
in CACNA1D knockout pups supported that Cav 1.3 was 1,25(OH)2 D3 injection significantly increased calbindin-D9k
essential for maintaining positive calcium balance and bone in duodenum of wild-type mice with no significant increase
calcium accretion in the pre-weaning period (13). in HV2 mice (72). This confirms that the transcription of
Some evidence suggests that Cav may play a role in calbindin-D9k is under the regulation of vitamin D.
calcium absorption in patients with diabetes mellitus, liver Another line of research indicated that intestinal expression
cirrhosis, and inflammatory bowel diseases (IBD) who of calbindin-D9k could be modulated by other factors besides
have high plasma levels of advanced oxidation protein 1,25(OH)2 D3 . Wang and co-workers, by using the Caco-2
products. By using cultured Caco-2 cells in the presence clone TC7 cells, reported that calbindin-D9k gene was upreg-
of advanced oxidation protein products, Wu et al. showed ulated more strongly by differentiation of the enterocytes
that the mRNA and protein expression levels of all calcium than 1,25(OH)2 D3 (>100-fold vs. 2-fold) (233). Deletion-
transport proteins [i.e., TRPV6, calbindin-D9k , PMCA1b , mutation studies showed that the promoter sequences for
and Na+ /Ca2+ exchanger 1 (NCX1)], except Cav 1.3, were Cdx-2 (a homeobox protein) and hepatocyte nuclear factor
downregulated (245). These findings suggest that the Cav 1.3- (HNF)-1 had a strong impact on calbindin-D9k gene expres-
mediated calcium absorption was spared and could be used sion during differentiation (233). In addition, an increase in
along with other alternative intracellular calcium translo- calcium influx via TRPV6 could also upregulate the mRNA
cation pathways (e.g., calcium shunting or tunneling) to and protein levels of calbindin-D9k even in mice lacking VDR
facilitate intestinal calcium flux in these conditions. Although (45). These findings suggested that while 1,25(OH)2 D3 could
one study reported downregulation of Cav 1.3 in the colon of promote the transcription of calbindin-D9k , other factors
patients with severe IBD and in mice with trinitrobenzene (e.g., increased calcium flux, Cdx-2, and HNF-1) are also
sulfonic acid-induced colitis (192), these observations do important for basal calbindin-D9k expression.
not completely rule out the plausible involvement of Cav in Even though calbindin-D9k is believed to be an important
intestinal calcium absorption. Thus, more investigations on player in transcellular pathway, the level of calbindin-D9k
Cav 1.3 in the intestine of rodents and humans of different age mRNA does not always correspond with the degree of active
groups and under physiological and pathological conditions calcium absorption (139). When compared with wild-type
are still warranted. mice, calbindin-D9k knockout mice showed similar serum
calcium and no change in calcium absorption either with or
without 1,25(OH)2 D3 treatment (2, 15, 134). Lee et al. (139)
Intracellular calcium translocation
demonstrated mRNA expression of TRPV6 and PMCA1b in
The intracellular calcium concentration must be maintained the duodenum of calbindin-D9k knockout mice were upreg-
below 10−7 mol/L in order to facilitate intracellular signal ulated compared to wild-type mice, suggesting alternative

Volume 11, May 2021 9


Intestinal Calcium Absorption Comprehensive Physiology

Apical Basolateral
Vesicular transport Ca2+
2+
Ca
Cai Free diffusion PMCA1b
2+
Ca
TRPV6 IP3R
STIM1 Ca2+
ER
Ca2+

Unidentified Tunneling RyR


Ca2+ channel SERCA

Figure 6 Model of vesicular and reticular transport of calcium. Diagram shows


vesicular calcium transport and calcium tunneling through the meshwork of endoplas-
mic reticulum (ER). Calcium enters the cell through TRPV6 or other calcium channels.
Intracellular calcium pool (Cai ) in the apical compartment is moved into the vesi-
cles or pumped into ER lumen by sarco-endoplasmic reticulum Ca2+ -ATPase (SERCA).
Calcium diffuses along the ER tunnel to the basolateral side and released into the
cytoplasm by 1,4,5-trisphosphate receptor (IP3 R) and ryanodine receptor (RyR) and
extrudes by the plasma membrane Ca2+ ATPase-1b (PMCA1b ). In cellular calcium
depleted condition, a calcium sensor stromal interacting molecule 1 (STIM1) is translo-
cated to the apical membrane to induce electrogenic calcium influx. The pathways
with red texts (i.e., STIM1, SERCA, ER, IP3R, and RyR) are hypothetical from indirect
evidence and need more investigations to confirm their physiological significance.
TRPV6, transient receptor potential cation channel, subfamily V, member 6.

compensatory mechanisms to maintain normal transcellular in an increased expression of the tight junction proteins in the
calcium transport. Lastly, genetic inactivation of calbindin- kidney suggesting a compensatory increase in renal paracel-
D9k did not change the serum calcium level (2, 139). These lular calcium reabsorption (104). Further research is required
data suggest that calbindin-D9k may not be the only calcium- to gain insight into a coordination between transcellular and
binding protein responsible for the intracellular calcium paracellular calcium transport pathways.
translocation. Besides the passive buffering of cytoplasmic calcium by
In addition to calbindin-D9k , several other calcium-binding calbindin-D9k and other calcium-binding proteins (Figure 6),
proteins, such as parvalbumin, calmodulin, and soluble other mechanisms of intracellular calcium mobilization
resistance-related calcium-binding protein (i.e., sorcin) might have been reported. Endoplasmic reticulum (ER) has been
have a role in translocating calcium in the cytoplasm (244). hypothesized to play an important role in the active buffering,
A DNA microarray analysis in Caco-2 cells showed that presumably as a part of the store-operated calcium entry via
sorcin expression was upregulated by 1,25(OH)2 D3 (244). TRPV6 (186). The ER lumen stretches between the apical and
In addition, parvalbumin, calmodulin, and sorcin may also
basolateral membrane and the sarco-endoplasmic reticulum
bind to and directly regulate the activities of the basolateral
Ca2+ -ATPase (SERCA) is localized to the apical compart-
PMCA1b and NCX1 (17, 253). Therefore, these proteins may
ment where it pumps cytoplasmic calcium into the ER lumen.
modulate the amount of calcium being translocated across
Thus, calcium can diffuse along the ER tunnel network to the
the cell in correspondence to the rate of basolateral calcium
extrusion. basolateral region where inositol 1,4,5-trisphosphate (IP3)
Recently, new evidence has shown that transcellular and and ryanodine receptors serve to release calcium into the
paracellular transport systems might work cooperatively. cytoplasm. Onodera et al. (179) reported in a study of rat
In a study of calbindin-D9k knockout mice, Hwang et al. colonic epithelium and human colonic HT29/B6 cell line
(105) showed that, under normal calcium diet, the expression that stromal interacting molecule 1 (STIM1) and Orai1—a
of tight junction genes occludin, claudin-2, and claudin-15 calcium sensor and a calcium release-activated calcium
in the duodenum was significantly increased compared to channel, respectively—were crucial for the apical calcium
wild-type mice. On the contrary, the calbindin-D9k knock- uptake into the colonic cells. When calcium store is depleted,
out mice under low calcium and low vitamin D diet had STIM1 is translocated to the apical membrane (179) where
reduced expression of occludin, zonula occludens (ZO)-1, it forms complexes with Orai1 and induces electrogenic
and claudin-15 as compared to wild-type animals (105). The calcium influx. TRPV6 and reverse-mode NCX1 might also
adaptation to the absence of calbindin-D9k was also reflected contribute to calcium entry during calcium store depletion

10 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

(179, 186). Nevertheless, it remains unclear whether intraor- activated by serine/threonine phosphorylation, cytosolic cal-
ganellar tunneling of calcium significantly contributes to the cium, calmodulin, and calbindin-D9k while it can be inhibited
overall intestinal calcium absorption or is merely a part of by vanadate (209). Trifluoperazine—an antipsychotic drug
intracellular calcium signaling for enhancing transport of that acts as a calmodulin inhibitor—is capable of blocking
other ions such as chloride and potassium. PMCA1b -mediated calcium efflux (114) and preventing
Various intracellular membrane-bound vesicles, such as PMCA1b activation in rat duodenum and descending colon
lysosome and endocytic vesicles, have also been postulated to (31, 63). Unlike wild-type mice, mice with intestine-specific
contribute to a bulk transport of calcium and some other tran- deletion of Pmca1 did not show an increase in active calcium
sition elements (e.g., iron and copper) across the cytoplasm absorption in response to intraperitoneal administration of
(160, 167). Lysosome-like vesicles near the apical mem- 1,25(OH)2 D3 . Moreover, these mice also showed marked
brane also accumulate some other ions, including aluminum, reduction in growth and bone mineralization (202). Recently,
gadolinium, and terbium (1). Calcium probably enters the 4.1R protein is suggested to be crucial for the PMCA1b
absorptive cells during endocytosis, which is partially con- activity. Liu et al. (148) demonstrated that the 4.1R protein
trolled by 1,25(OH)2 D3 through its membrane receptor co-localized with PMCA1b and the 4.1R knockout mice had
known as 1,25(OH)2 D3 -MARRS (membrane-associated, complex phenotypes that include hypocalcemia, elevated
rapid response, steroid-binding) protein (166, 167). Alter- serum 1,25(OH)2 D3, and PTH, impaired calcium absorption
natively, calcium may first traverse the apical membrane in the small intestine, downregulation of PMCA1b expression
via TRPV6 or other calcium channels before being moved and osteopenia. Thereby, this suggests that 4.1R protein may
into the vesicles by unidentified transporter(s). The amount be an interacting protein required for action of PMCA1b .
of accumulated calcium ions is dependent on vesicular Collectively, these findings indicate that pharmacological
acidic pH; therefore, a disruption of vesicular acidification inhibition and genetic deficiency of PMCA1b , as well as
by chloroquine halts the process (167). Calbindin in the 4.1R protein, can have a consequence in calcium and bone
vesicles probably helps buffer ionized calcium and facilitate homeostasis.
accumulation of calcium (137). Although passive diffusion The NCX1-mediated calcium transport is responsible for
of ionized calcium in the apical-to-basolateral direction is approximately 20% of basolateral calcium flux (86, 98).
possible especially during 1,25(OH)2 D3 -stimulated con- Nevertheless, both PMCA1b and NCX1 work together in
ditions with steep calcium gradients, its diffusion rate is an interdependent manner. A complete inhibition of NCX1
much slower (∼1/70) than facilitated diffusion mediated by by small molecules such as KB-R7943 led to cessation
Ca2+ -laden proteins (e.g., calbindin-D9k and parvalbumin) of PMCA1b activity and vice versa (57). It is plausible
(65, 66). Indeed, an increase in cytosolic ionized calcium that blockade of one basolateral transporter culminates in
during the enhanced calcium absorption probably signals increased cytoplasmic calcium, which halts the entire process
most calcium transporters particularly PMCA1b to accelerate of transcellular calcium transport. This hypothesis is in line
calcium efflux into the extracellular space (223). with the observations that elevated cytoplasmic calcium
Since most acidified lysosome-like vesicles have a limited levels in the vicinity of TRPV6 and Cav calcium channels
volume, other metals particularly iron, have been hypothe- led to inactivation of these channels, thereby decreasing
sized to compete with calcium for uptake into the vesicles apical calcium influx (51, 120, 138, 170). However, whether
(130). In other words, iron-bound transferrin may occupy compensatory upregulation of PMCA1b in nonfunctional
space in the lysosome, leaving less space for calcium accu- NCX1 condition exists requires further investigations. Under
mulation. This phenomenon partially explains how high iron normal conditions, the electrogenic NCX1 in the duodenal
intake inhibits intestinal calcium absorption (141). enterocytes extrudes calcium with a stoichiometry of 3 Na+ :1
Ca2+ (forward mode) and by coupling with Na+ /K+ -ATPase,
sodium concentration gradient across the basolateral mem-
Calcium transport across the basolateral membrane brane could be maintained (98). A change in sodium gradient
may trigger NCX1-induced calcium entry (reverse mode).
The basolateral calcium efflux is a cellular energy-dependent Dong et al. (55) have shown that a reverse-mode NCX1 was
process. Several transporters, particularly P-type primary essential for the duodenal HCO3 − secretion, which required
active transporter ATP2B1 (i.e., PMCA1b ) and secondary an increase in intracellular calcium.
active transporter SLC8A1 (i.e., NCX1), predominantly move NCKX is a plasma membrane transporter that appears to
cytoplasmic calcium across the basolateral membrane of the mediate Ca2+ extrusion along with K+ and the Na+ comes
intestinal absorptive cells (69, 98). Many other transporters into the cell. Among five isoforms of NCKX (i.e., NCKX1–5)
such as ATP2B4 and K+ -dependent Na+ /Ca2+ exchanger (4, 5), the NCKX1 is the first isoform that was initially discov-
(SLC24 or NCKX) may also contribute to calcium extrusion ered in the outer segment of retinal rod (25, 206). Recently,
(5, 101). Yang et al. (250) reported that male NCKX3 knockout mice
PMCA1b is the major calcium transporter at the baso- had a significantly decreased mRNA expression of TRPV6
lateral membrane as its activity contributes up to 80% of and calbindin-D9k in the duodenal mucosal cells despite hav-
the basolateral calcium efflux (54, 86). PMCA1b is potently ing normal serum calcium levels (250). However, the NCKX3

Volume 11, May 2021 11


Intestinal Calcium Absorption Comprehensive Physiology

knockout mice had a significant increase of plasma PTH and (96, 225), thereby strengthening solvent drag-induced cal-
a decrease in bone mineral content (250), suggesting that cal- cium transport. The apical glucose uptake together with
cium might have been drawn from the bone to compensate Na+ influx is likely to modulate the transcellular calcium
the decreased extracellular calcium pool. These data implicate transport, presumably by inducing the apical membrane
NCKX3 as another basolateral transporter involved in intesti- depolarization that opens Cav 1.3 and providing substrates
nal calcium absorption; however, their physiological signifi- for ATP production from oxidation of glucose (120). As far
cance remains to be investigated. as the coordination between NHE3 and CFTR is concerned,
Lastly, calcium-rich vesicles localized at the basolateral the underlying mechanism of how NHE3-CFTR system
compartment of the enterocytes are able to fuse with the modulates calcium absorption remains unclear
basolateral membrane for calcium extrusion (137). However, An appropriate sodium gradient across the basolateral
under stimulated conditions with increased cytoplasmic membrane is tightly controlled by Na+ /K+ ATPase, which
calcium, some vesicular cation-selective channels, such as is expressed predominantly in the lateral membrane and
two-pore channels (TPCs), can induce calcium release into to a lesser extent in the basal membrane (6). This gradient
the cytoplasm close to the basolateral membrane (255). TPCs provides a driving force for NCX1 as well as NHE1. NHE1 is
also help maintain proper vesicular osmolality, volume, and a salient intracellular pH regulator that maintains H+ gradient
endocytic vesicle trafficking (75). Meanwhile, an increase and can modulate the activity of various membrane proteins
in cytoplasmic calcium near basolateral membrane induces including protein 4.1R, NCX1, and perhaps PMCA1b (145,
basolateral calcium extrusion via PMCA1b and NCX1. 174). For example, NHE1 is required for an interaction
between NCX1 and its modulator calmodulin (145). More
investigations are required to gain more understanding in
Other transporters associated with calcium transport
the interplay among these plasma membrane proteins and
Several investigations have suggested that a number of trans- transporters.
porters, for example, Na+ /H+ exchanger 3 (NHE3), Na+ /K+ -
ATPase, and cystic fibrosis transmembrane conductance
regulator (CFTR), contribute to intestinal calcium absorption. Paracellular calcium transport
For example, NHE3 knockout mice exhibited the impairment It was previously believed that the paracellular space—also
in calcium, sodium, and water absorption across the intestine, known as lateral intercellular space—is merely a simple con-
particularly in the cecum (195) where the rate of active duit in which water and ions can diffuse passively along the
transcellular calcium transport is much higher than other electrochemical gradient. Although the paracellular calcium
intestinal segments (111, 117). We previously reported that flux is nonsaturable and independent of the amount of cal-
100 nM tenapanor—a potent NHE3 inhibitor—completely cium intake (182), several lines of evidence have confirmed
abolished hepcidin-induced duodenal calcium absorption in that fluid and ion movement across this space is actively
hemizygous β-globin knockout thalassemic mice (27) as well controlled by the absorptive cells as well as systemic and
as leucine- and leucine tripeptide-induced calcium transport local mediators such as 1,25(OH)2 D3 and cytokines produced
in the rat duodenum (220). by immune cells (22, 95, 116, 221). Paracellular transport
NHE3 has been postulated to profoundly affect pericellu- pathway in human intestinal epithelium-like Caco-2 mono-
lar and intracellular microenvironment required for calcium layer also showed rectification since under the voltage-clamp
absorption. Since intracellular acidic pH can reduce transcel- experiments, the apical-to-basolateral calcium flux was much
lular calcium transport (35), NHE3 inhibition that leads to greater than flux in the opposite direction (221).
H+ accumulation in the cytoplasm is expected to disrupt cal- Generally, paracellular calcium diffusion is dependent
cium transport process. Apical Na+ uptake by NHE3 is also on voltage gradient or transepithelial potential difference
crucial for a number of processes pertaining to paracellular (PD), and its concentration gradient. Owing to electrogenic
calcium transport (222, 240). For instance, high Na+ uptake transport of cations—notably sodium, the lumen compart-
and accumulation in the paracellular space are able to increase ment of all intestinal segments has negative potential as
solvent drag or convective water flux that favors paracellular compared to the plasma compartment (30, 115). Therefore,
calcium movement in an ATP-dependent manner (3). Further- the voltage gradient often favors calcium transport in the
more, because of its activity, the apical H+ efflux is increased. basolateral-to-apical direction, that is, calcium secretion into
NHE3 is probably important for maintaining high calcium the lumen (115, 116). Nevertheless, the voltage-dependent
solubility in the acid microclimate near the mucosal surface calcium transport in the rat duodenum is considered negli-
(172, 181, 195, 222). Failure to maintain this luminal acidic gible because of a relatively small PD of approximately 3 to
environment close to the microvilli might render calcium salt 4 mV (30).
to precipitate into insoluble complexes, thus compromising The calcium concentration gradient is considerable in the
calcium absorption. proximal part of the small intestine, that is, duodenum and
NHE3 also couples or directly interacts with other transport proximal jejunum. Dietary calcium salts, as well as many
systems, for example, SGLT1 and CFTR. Coordination of other minerals, become ionized in the acidic gastric juice
SGLT1-NHE3 system ensures continuous uptake of sodium before they pass into the duodenum (60, 149). Luminal

12 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Apical Tight junction Basolateral can form heteromers to produce an anastomosing meshwork
of tight junction ring (7). The extracellular loops with charged
amino acids protrude into the paracellular space to create
channel-like pores with different permselectivity (39). For
example, claudin-2, -12, and -15 contain negatively charged
amino acids in their extracellular loops, which attract cations
Occludin
including Ca2+ and Na+ (78, 228).
E-cadherin
Claudins The expression of intestinal claudins can be regulated by
Standing osmotic gradient vitamin D signaling and 1,25(OH)2 D3 . Fujita et al. (79) have
Ca2+ Ca2+
demonstrated that VDR knockout mice had a reduced expres-
K+
Na+ NKA sion of claudin-2 and -12 in the jejunum, ileum, and colon,
ZO-1 whereas 1,25(OH)2 D3 markedly upregulated claudin-2 and
-12 protein expression in Caco-2 cells. Consistently, by using
Actin cytoskeleton TRPV6/calbindin-D9k double knockout, Christakos et al.
(38) reported that 1,25(OH)2 D3 increased the expression of
claudin-2 and -12 while downregulated the expression of
cadherin-17. Even though calbindin-D9k knockout mice did
Luminal [Ca2+] Plasma ionized [Ca2+]
>5 mM 1.25 mM
not show reduced intestinal calcium absorption, the mRNA
expression levels of claudin-2 and -15 were upregulated,
Figure 7 Paracellular intestinal calcium transport. Although calcium presumably to allow more calcium entry via paracellular
concentration gradient is a driving force for paracellular calcium trans- pathway (105). Furthermore, siRNA knockdown of claudin-
port, the tight junction (i.e., claudins, occludin, ZO-1) acts as a barrier
to restricts paracellular ion movement in a charge- and size-selective
15 expression in Caco-2 monolayer was found to diminish
manner. Calcium and small water-soluble molecules move along the prolactin-induced paracellular calcium transport (32), while
stream of water in apical to basolateral direction (solvent drag-induced lactating rats with calcium hyperabsorption exhibited upreg-
calcium absorption). Elevation of NKA activity increases the driving
force for the solvent drag-induced calcium transport. ZO-1, zonula
ulation of claudin-15 protein expression in the duodenum
occludens-1. (237). Thus, intestinal claudins play an important role in
promoting paracellular calcium transport and may serve as a
compensatory mechanism when transcellular calcium trans-
calcium concentration may be as high as approximately 3 to port is compromised or when there is an increased calcium
10 mmol/L versus 1.2 to 1.3 mmol/L free-ionized calcium demand.
in the interstitium, and it is certainly elevated after oral cal- Recently, a rare missense mutation in CLDN2 gene
cium supplementation or ingestion of calcium-rich diet (60). encoding claudin-2 was identified in an Iranian family with
Experiments using ex vivo rat duodenum and epithelium-like azoospermia, renal stones, and hypercalciuria, the latter of
Caco-2 monolayer showed a significant increase in paracel- which resulted from a decrease in the claudin-2-mediated
lular calcium flux when the luminal calcium concentrations paracellular calcium reabsorption in the proximal renal
exceeded 5 mmol/L (31, 108). In the distal small intestine tubule (10, 46). Claudin-2 knockout mice also manifested
like ileum, the calcium uptake appeared to be 1,25(OH)2 D3 - a marked reduction in the colonic paracellular permeability
independent (184). Pansu et al. (184) determined calcium to calcium; however, a calcium balance study in claudin-2
absorption in duodenum and ileum of vitamin D-deficient knockout mice revealed greater net calcium absorption than
rats before and after intraperitoneal 1,25(OH)2 D3 injection. in wild-type animals (46). While the loss of claudin-2 led to
Treatment of 1,25(OH)2 D3 induced transcellular calcium reduced colonic permeability to calcium along with decreased
absorption only in the duodenum, but not the ileum. passive calcium secretion in the colon, calcium secretion in
The intestinal absorptive cells regulate the rate of para- duodenum and ileum were similar between the genotypes,
cellular calcium flux by various mechanisms. In mice, the thus leading to greater net calcium absorption in claudin-2
tight junction width between adjacent duodenal epithelial knockout mice (46). In addition, Gloux et al. (90) showed that
cells is 16 to 18 nm, similar to that found in humans (133). the mRNA level of claudin-2 in the duodenum and jejunum of
Perijunctional localization of polymeric F-actin filaments laying hens was significantly increased, presumably to meet
and contraction of actomyosin ring complex near the tight an increased calcium demand for eggshell biomineralization
junction alter its size-selective property and also enhance dif- (∼2 g/day) while minimizing resorption of medullary bone
fusion rate of calcium (128, 226). In addition, the absorptive (91). Collectively, claudin-2 is one of the major tight junction
cells are able to change the expression of claudins and some proteins that form calcium-selective paracellular pores in
other transmembrane tight junction proteins, for example, human, rodent, and possibly in avian.
occludin, to modulate size and charge selectivity of the tight Moreover, the intestinal absorptive cells can promote
junction (Figure 7) (7, 74). paracellular calcium flux by increasing solvent drag. In a
In humans and rodents, the claudin family of transmem- leaky epithelium with transepithelial resistance (TER) of
brane tight junction proteins consists of >20 members, which ≤100 Ω⋅cm2 like the intestinal epithelium, a significant

Volume 11, May 2021 13


Intestinal Calcium Absorption Comprehensive Physiology

volume of water can flow through the paracellular pathway. However, direct evidence supporting the local conversion of
Calcium and small water-soluble molecules dissolved in vitamin D is still needed.
luminal fluid thus move along the stream of water in the Based on the transcaltachia model—a model of rapid
apical-to-basolateral direction—a phenomenon known as stimulation of calcium transport by hormones—in mammal
the solvent drag-induced calcium absorption (28). Since the and avian, treating the perfused duodenum from chicks
paracellular water flow is dependent on sodium accumulation with PTH and 1,25(OH)2 D3 directly enhanced the vectorial
and hyperosmotic milieu in the paracellular space distal to vesicular calcium transport (137, 168). Indeed, 1,25(OH)2 D3
the tight junction, the intestinal absorptive cells may increase is able to induce a rapid response and enhance vesicular
the activity of Na+ /K+ -ATPase lining the lateral membrane calcium transport within a few minutes (68). The underly-
to transport more sodium. Consequently, this increases the ing cellular mechanism of this rapid response is not well
driving force for the solvent drag-induced calcium transport understood but it has been postulated to be mediated by
(28, 98). Under normal conditions, solvent drag-induced 1,25(OH)2 D3 -MARRS protein or VDR, the latter of which
calcium transport contributes up to 75% to 80% of the total is recruited to the caveolin-1-rich plasma membrane or
calcium absorption across the proximal small intestine, where caveolae (102). Moreover, Sterling and Nemere (215) further
a great deal of glucose and sodium is absorbed. demonstrated that both PTH 1-34 (an active peptide fragment
of PTH with 34 amino acids) and 25(OH)D3 were capable
of enhancing vesicular calcium transport in isolated chick
Systemic Controls of Mucosal Calcium duodenal epithelial cells. It is likely that the rapid action
of 1,25(OH)2 D3 is mediated by nongenomic mechanisms
Uptake involving phospholipase C (e.g., PLC-γ), c-Src kinase, and
Endocrine regulation ERK1/2 (184).
Paracellular calcium transport is markedly decreased in
Canonical parathyroid gland-kidney-intestine axis vitamin D deficiency, in part by the downregulated expres-
Mucosal calcium uptake is controlled by several factors such sion of claudin-2 and -12 (79). VDR knockout mice also
as endocrine and luminal factors. PTH and 1,25(OH)2 D3 manifested a decrease in the expression level of claudin-2
are the two most important hormones that maintain cal- in the duodenal mucosa although the mucosal permeability
cium homeostasis by exerting their actions on three organs, to FITC-D 4000 (a large-size paracellular marker) remained
that is, bone, kidney, and intestine (for reviews, please see unaltered (133). Besides the effect on the intestine, Kladnit-
(69, 127). As depicted in Figure 8, the canonical hormonal sky et al. (125) used a luciferase reporter assay in HEK 293
axis, a decrease in plasma ionized calcium is detected by and OK (opossum kidney) cells to show that 1,25(OH)2 D3
calcium-sensing receptor (CaSR)—a member of C fam- suppressed the human CLDN16 promoter activity and this
ily G protein-coupled receptor—in the parathyroid chief is consistent with a decrease in renal CLDN16 expression
cells, which, in turn, release PTH to indirectly promote observed in mice received parenteral 1,25(OH)2 D3 . These
25-hydroxyvitamin D3 1α-hydroxylase (CYP27B1) in the findings reveal the suppressive role of this hormone on
proximal renal tubular cells (127). CYP27B1 catalyzes the CLDN16 at the transcriptional level. Since 1,25(OH)2 D3
hydroxylation of 25(OH)D3 to 1,25(OH)2 D3 . This active form decreases renal claudin 16 transcription and claudin-16
of vitamin D is a potent calciotropic hormone that directly promotes calcium and magnesium reabsorption in the thick
stimulates the intestinal calcium absorption (for review, ascending limb of Henle’s loop (TAL), it is tempting to
please see Ref. 238). In humans and rodents, the small and speculate that an acute 1,25(OH)2 D3 administration and
large intestine abundantly express nuclear VDR and are well the resultant intestinal hyperabsorption of calcium is prob-
responsive to 1,25(OH)2 D3 . The binding of 1,25(OH)2 D3 to ably associated with calciuria. In contrast to the effect of
the VDR response element promotes expression of calcium 1,25(OH)2 D3 in mediating transcellular calcium reabsorption
transport machinery including TRPV5, TRPV6, calbindin- in the distal convoluted tubule, 1,25(OH)2 D3 possibly inhibits
D9k , NCX1, PMCA1b , claudin-2, and -12. Furthermore, the paracellular calcium reabsorption in the TAL—through the
1,25(OH)2 D3 -mediated induction of TRPV6 expression has downregulation of claudin-16—in response to intestinal
been shown to precede an increase in duodenal calcium hyperabsorption of calcium. This further suggests that
absorption in mice (214). Therefore, the VDR-mediated 1,25(OH)2 D3 also helps maintain a balance between the
signaling could enhance both transcellular and paracellular intestinal calcium/magnesium absorption and renal excretion.
calcium transport (79, 238). The expression of TRPV5,
TRPV6, and calbindin-D9k in the duodenal epithelial cells Roles of bone-intestinal axis and other endocrine
of rats injected subcutaneously with 2 μg/kg 1,25(OH)2 D3
factors
were markedly elevated as early as 3 h post-injection (122).
Since the enterocytes also produce CYP27B1 and CYP24A1, The bone-intestinal axis for regulating mineral metabolism
the latter of which is a mitochondrial monooxygenase for has recently been reported. The very first example is the
1,25(OH)2 D3 catabolism (178, 203), they probably respond role of osteoblast- and osteocyte-derived fibroblast growth
to 1,25(OH)2 D3 and, in turn, modulate calcium transporters. factor (FGF)-23 in the regulation of intestinal phosphate

14 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Ca2+ deposition in bone


Calcitonin (inhibition of osteoclastic
bone resorption)

Thyroid gland
(C cell)

Rising Ca2+ levels in blood


Blood Ca2+ levels decrease
Increase Ca2+ excretion

Homeostasis
Normal blood Ca2+ level

mM mg/dL
Total calcium 2.0–2.5 8–10
Ionized calcium 1.2–1.3 4.5–5.5

Rising Ca2+ levels in blood

Bone Ca2+ release Blood Ca2+ levels decrease

Increase Ca2+ absorption

1,25(OH)2D3 PTH

PTH

Cyp27B1
ER
Mitochondria
CaSR Ca2+
Proximal tubular cell
Parathyroid chief cell

Figure 8 Systemic control of mammalian calcium homeostasis by three organs (i.e., bone, kidney, and intestine).
The details are fully explained in the section titled “Systemic Controls Mucosal Calcium Absorption”. Cyp27B1, 25-
hydroxyvitamin D3 1α-hydroxylase; PTH, parathyroid hormone.

absorption. Since FGF-23 is a phosphaturic and hypophos- CYP24A1 activity and decreases CYP27B1 activity in the
phatemic hormone, its actions are mainly to counterbalance kidney and some other extrarenal cells (e.g., monocytes),
the effects of 1,25(OH)2 D3 on intestinal phosphate absorp- thereby reducing 1,25(OH)2 D3 actions (11).
tion and directly reduce phosphate reabsorption in the kidney In addition to FGF-23, several other hormones have been
(191). Recently, intravenous FGF-23 injection has been reported to participate in the systemic regulation of calcium
reported to inhibit 1,25(OH)2 D3 -induced intestinal calcium absorption such as calcitonin and prolactin. Calcitonin is
transport in rodents (122), indicating that FGF-23 is also a major calcium-regulating hormone that reduces plasma
a calcium-regulating hormone. Several circulating factors, calcium concentration. The primary site of calcitonin produc-
for example, plasma ionized calcium, inorganic phosphate, tion is parafollicular cells of the thyroid gland. After being
and 1,25(OH)2 D3 , have been reported to upregulate FGF-23 released into the circulation, calcitonin binds the osteoclasts
production and secretion (190, 191). FGF-23 also increases to inhibit bone resorption (76, 77). Although known for

Volume 11, May 2021 15


Intestinal Calcium Absorption Comprehensive Physiology

this mechanism in bone, the role of calcitonin in intestinal hormones appear to contribute to calcium homeostasis by
calcium absorption is still controversial and most studies synchronizing the intestinal calcium supply with the rate of
were performed in animals (216). Some in vitro evidence bone calcium accretion.
revealed inhibitory effect of calcitonin on intestinal calcium In summary, several hormones either produced from the
absorption, but animal studies showed otherwise. Employing canonical parathyroid gland-kidney-intestine axis or those
isolated small intestine from vitamin D-replete rats, Olson produced from other tissues like FGF-23, prolactin, 17β-
and colleagues (177) reported the immediate reduction of estradiol, and GH are involved in the regulation of intestinal
intestinal calcium absorption after acute infusion of low calcium absorption (Figure 9). The multitude of hormonal
dose of calcitonin (10 mU) while a high dose (500 mU) regulators ensures that the plasma ionized calcium level
enhanced calcium absorption. However, these effects were is strictly maintained within the physiologic range while
absent in the intestine from vitamin D-deficient rats (177). adequately supplying calcium to meet certain needs (e.g.,
On the other hand, physiological and pharmacological doses growth, lactation) at different stages of life.
of calcitonin did not alter intestinal calcium absorption in
thyroparathyroidectomized rat, a calcitonin-deficient model,
(43) and a 12-day infusion of 200-mU calcitonin in these
rats conversely increased plasma calcium by 50%, which
Local Controls of Mucosal Calcium
was likely resulted from an increase in intestinal calcium Uptake
absorption (107). Based on these data, the effect of calcitonin
on intestinal calcium absorption seems to be dose- and vita- Possible roles of FGF-23 and CaSR
min D-dependent. Although current evidence suggests the As mentioned earlier, FGF-23 is well known as a bone-
involvement of this hormone in intestinal calcium transport, derived phosphate-regulating systemic factor that promotes
inconsistency observed in the findings emphasizes the need phosphate excretion in the kidney and increases 1,25(OH)2 D3
to investigate the exact roles and underlying mechanisms of catabolism (93, 113, 229). To prevent hyperphosphatemia
calcitonin in well-controlled mammalian models and humans. that causes ectopic calcification (110), the increased circu-
Prolactin and 17β-estradiol are potent stimulators of lating level of phosphate stimulates secretion of PTH and
intestinal calcium absorption (28, 163, 227). Both hormones FGF-23, which, in turn, enhances phosphate excretion. In
are able to directly upregulate the expression of transcellular regard to direct actions of FGF-23 on physiological renal
calcium transporters, such as TRPV6 (163, 227). Meanwhile, phosphate excretion, circulating FGF-23 binds to a recep-
17β-estradiol increased VDR expression in the rat duodenum, tor complex of fibroblast growth factor receptor (FGFR)
thus augmenting 1,25(OH)2 D3 responsiveness (147). As a and α-Klotho in the renal tubular epithelial cells. There
lactogenic hormone, prolactin has been shown to enhance are four major FGFR isoforms, that is, FGFR1-4. Under
intestinal calcium absorption during pregnancy and lactation, physiological conditions, FGFR1 appears to be the main
which are reproductive periods with high calcium demand FGF-23 receptor that works with membrane-associated
for fetal bone development and lactogenesis (32). Prolactin Klotho and activates a signaling cascade involving ERK1/2
not only stimulates transcellular calcium transport but also and serum/glucocorticoid-regulated kinase-1 (SGK1) (194).
markedly increases paracellular calcium flux, in part by This signaling cascade results in phosphorylation of the
modulating the expression and function of tight junction scaffolding protein Na+ /H+ exchange regulatory cofactor
protein claudin-15 (32, 241). The activity of transcellular (NHERF)-1 and internalization of sodium/phosphate cotrans-
calcium-transporting proteins (e.g., calbindin-D9k , PMCA1b , porter (NaPi)-2a and -2c in proximal renal tubules and reduces
and Cav 1.3) is also dependent on prolactin (163, 221, 242). the tubular phosphate reabsorption (62, 229). FGF-23 also
A transcriptome analysis of the duodenal epithelial cells from indirectly decreases phosphate absorption by suppressing
pituitary-grafted hyperprolactinemic rats further revealed 1,25(OH)2 D3 synthesis. In the same time, FGF-23 promotes
a number of upregulated calcium channel transcripts, for 1,25(OH)2 D3 catabolism by upregulating CYP24A1. This
example, Cacnb1, Cacna1h, and Cacna1g (37) although reduces the circulating active 1,25(OH)2 D3 levels and, in
whether these calcium channels contribute to the duodenal turn, decreasing intestinal phosphate uptake (113, 126, 211,
calcium absorption remains unclear. These data support 229).
the importance of prolactin and 17β-estradiol in regulating Besides being produced by bone cells, FGF-23 transcript
intestinal calcium absorption. has been shown to express in cells of other tissues such
Hormones that promote skeletal growth and development as kidney, brain, lung, liver, and spleen (122, 126). Thus,
[e.g., growth hormone (GH), insulin-like growth factor it may also act as a local factor to control certain local
1, placental lactogen, and thyroxine] have been reported metabolism. Several studies showed that FGF-23 and FGFRs
to increase calcium absorption across the small intestinal were expressed in small and large intestinal enterocytes (161,
mucosa (70, 154, 180, 254). In aged female rats (16 months 217, 239, 242). Specifically, by using immunohistochemistry
old), 12-day GH injection was found to increase the duodenal in the duodenum of female rats, Khuituan et al. reported
calbindin-D9k levels, and a combination of GH and PTH that FGFR1-4, but not α- and β-Klotho, were expressed in
treatment showed an additive effect (70). Interestingly, these the duodenal enterocytes (122). Although the expression

16 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Neurocrine factors

VIP
Substance P
Dopamine
Serotonin

Enteric VIPergic neuron


Luminal factors
Ca2+
Stimulators TTX sensitive

Amino acids
Glucose/Galactose
SCFAs
Paracrine
Prebiotics Cldn-2, -3, -12
Inhibitors Ca2+
NKA Neuroendocrine
2+ 3+
Fe and Fe PMCA1b cell
Zinc Na+
Copper Ca2+ K+ Ca2+ Endocrine factors
TRPV6
Ca2+ Stimulatory Inhibitory
Ca2+
TRPV5 Calbindin-D9k Na + NCX1 1,25(OH)2D3 FGF-23
Prolactin Calcitonin
Ca2+ Na+
Cav1.3 Estrogen Corticosteroids
NKA GH
K+ IGF-1
1,25(OH)2D3-dependent Thyroid hormone

Prolactin-dependent
Apical Basolateral

Figure 9 Extrinsic (humoral and neural) and intrinsic (luminal) regulators of intestinal calcium absorption. The luminal and endocrine
factors that act as stimulator and inhibitor on intestinal calcium absorption showing in the green and orange boxes, respectively. Although
the presented luminal and endocrine factors are known to modulate calcium absorption, the involvement of neurocrine factors (marked as
italic text), or factors released from neuroendocrine cells in the enteric nervous system is in the emerging area of interest and needs more
investigations to confirm the physiological significance. Cldn, claudin; FGF-23, fibroblast growth factor-23; GH, growth hormone; IGF-1,
insulin-like growth factor-1; SCFAs, short-chain fatty acids; TTX, tetrodotoxin; VIP, vasoactive intestinal peptide; NCX1, Na+ /Ca2+ exchanger
1; NKA, Na+ /K+ -ATPase; PMCA1b , plasma membrane Ca2+ ATPase-1b; calbindin-D9k (S100 calcium-binding protein G, S100G); TRPV,
transient receptor potential cation channel, subfamily V.

of membrane-associated Klotho has not been reported in the 1,25(OH)2 D3 -mediated transcellular and paracellular
the intestinal enterocytes, a body of research supports the calcium absorption (122, 123). Although the exact molecular
role of FGF-23 in regulating intestinal calcium absorption mechanism and signaling pathway(s) by which FGF-23
through both systemic and local (paracrine) mechanisms reduces calcium transport remains elusive, several signaling
(for its systemic actions, please see section titled “Roles of proteins, such as MAPK/ERK, p38 MAPK, and protein
Bone-Intestinal Axis and Other Endocrine Factors”). It is kinase C might be involved in the process (122, 123).
noteworthy that soluble Klotho in the circulation probably High calcium and 1,25(OH)2 D3 exposures appear to be the
contributes to FGF-23 signaling in the intestine. driving force for FGF-23 secretion and the resultant suppres-
An investigation in Caco-2 cell culture further sug- sion of calcium transport. The pathway in the enterocytes may
gested that intestinal epithelial cells could secrete FGF-23 involve the CaSR, which plays a role in monitoring calcium
peptide into the apical and basolateral compartments in transport across the enterocytes (239). By using Caco-2 cells,
response to 1,25(OH)2 D3 and apical exposure to high cal- Wongdee et al. (239) reported that CaSR inhibitors were
cium concentration, which were conditions that mimicked capable of suppressing the 1,25(OH)2 D3 -induced upregula-
1,25(OH)2 D3 -induced calcium hyperabsorption and high- tion of FGF-23. Furthermore, similar to apical high-calcium
calcium intake, respectively (199, 239). Functional studies exposure, the apical CaSR activation by cinacalcet and
in the duodenum of female rats confirmed that FGF-23 AC-265347 diminished the 1,25(OH)2 D3 -mediated transcel-
exerted a direct action on the intestine by directly inhibiting lular calcium transport presumably via the upregulation of

Volume 11, May 2021 17


Intestinal Calcium Absorption Comprehensive Physiology

(A) Apical Basolateral

Transcellular
1,25(OH)2D3
FGF-23
FGF-23

Paracellular

CaSR
FGF-23
Activators FGF-23
CaSR
2+
Ca
TRPV6
Transcellular

(B)

SCFAs

PMCA1b
Luminal pH 2
TRPV6
Ca2+ Ca2+
Calcium complexes Calbindin-D9k
[e.g., CaHPO4, Ca(H2PO4)2]
Na+ NCX1

Na+
1 Free Ca2+ NKA
K+
1 SCFAs enhance free ionized calcium
2 SCFAs increase expression of TRPV6

Figure 10 The emerging topic of intestinal calcium absorption. (A) A schematic diagram
shows possible mechanism of fibroblast growth factor (FGF)-23 and calcium-sensing recep-
tor (CaSR) on local negative feedback regulation of calcium absorption. (B) A schematic
diagram showing a possible mechanism of how short-chain fatty acids (SCFAs) enhances
transcellular calcium transport in the intestine. Hypothetical pathways or pathways with
indirect evidence are presented as dashed lines. NCX1, Na+ /Ca2+ exchanger 1; NKA,
Na+ /K+ -ATPase; PMCA1b , plasma membrane Ca2+ ATPase-1b; calbindin-D9k (S100
calcium-binding protein G, S100G); TRPV, transient receptor potential cation channel,
subfamily V.

FGF-23. These findings corroborated the involvement of was unchanged compared with pretreatment state, thereby
FGF-23 and CaSR in an ultrashort negative feedback loop for indicating that the CaSR-mediated reduction of calcium
preventing excessive calcium absorption, particularly during absorption may work through attenuation of TRPV6 expres-
prolonged 1,25(OH)2 D3 treatment and high-calcium intake sion on the apical membrane or TRPV6 function (140).
(Figure 10A) (239). Taken together, these data support that CaSR in the apical
Another mechanism that has been proposed as a safeguard and/or basolateral membrane could play a role in regulating
to prevent serum calcium overload is basolateral CaSR sig- calcium homeostasis but may act through different molecular
naling. CaSR is indeed expressed throughout the intestine pathways.
in both the apical and basolateral membrane of enterocytes.
Lee et al. (140) applied CaSR activator cinacalcet to either
apical or basolateral hemichamber of the Ussing chamber
and demonstrated that only activation of basolateral CaSR
Factors Affecting Calcium Absorption
led to a decrease in transcellular calcium absorption (140). Besides being controlled by hormones, the intestinal calcium
They further tested the effects of basolateral cinacalcet absorption is also affected by the local environment in the gut
treatment in the proximal colon of nonfunctional TRPV6 lumen such as the presence of luminal nutrients, amino acids,
(TRPV6D541A ) mice and found that the net calcium flux sugars, or microbiota-derived metabolites.

18 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

Luminal pH and unstirred water layer across Caco-2 monolayer (146), suggesting that these bio-
logically active peptides may directly enhance calcium
An in vitro digestion study revealed that various forms of
transporter activity or act as slow-release calcium complexes
calcium sources, for example, calcium carbonate, calcium
and/or prevent precipitation of ionized calcium into an
phosphate, and calcium citrate malate, are mostly soluble
insoluble form.
under gastric pH, but become insoluble (∼20%–36% bioac-
In addition to small peptides and amino acids, monosac-
cessibility) under small intestinal pH (149). The apical
charides have ability to modulate calcium absorption. For
domain of enterocytes is seemingly close to luminal fluid,
example, the SGLT-1 substrates such as glucose and galactose
which contains variety of nutrients and minerals. However,
exhibit a stimulatory effect on intestinal calcium absorption.
there is a thin layer of gel-like fluid covering the microvilli
On the other hand, fructose has an inhibitory effect on
that blocks the enterocytes from exposing directly to the lumi-
calcium absorption (59, 200). High fructose intake has been
nal fluid. This layer is called unstirred water layer that helps
shown to indirectly diminish the intestinal calcium absorp-
maintain particular physicochemical profiles of ion and water
tion in growing and lactating rats by increasing 1,25(OH)2 D3
(97, 142). The unstirred water layer can create an anomalous
catabolism and decreasing 1,25(OH)2 D3 synthesis (58, 59).
ion gradient that makes the diffusion rate unpredictable, thus
In vitro and in vivo studies in growing rats revealed that fruc-
becoming a pre-epithelial diffusion barrier. Bulk flow of
tose diminished intestinal calcium absorption, by reducing
luminal fluid over more viscous fluid of the unstirred water
circulating 1,25(OH)2 D3 levels, TRPV6, and calbindin-D9K
layer might create Saffman-Taylor instability—also known
expression, thereby decreasing transcellular calcium transport
as viscous fingering—at the interface. This unstable interface
(59). The underlying mechanism of fructose-induced reduc-
allows luminal nutrients to penetrate deep into the unstirred
tion of 1,25(OH)2 D3 might be associated with FGF-23—a
water layer similar to the way that H+ penetrates through the
regulatory hormone of 1,25(OH)2 D3 production. Douard
mucus gel layer produced by gastric mucin (16). Furthermore,
et al. (59) found that fructose increased circulating FGF-23
the fluid layer near the microvilli is an acid microclimate,
levels, which inhibited CYP27B1 and stimulated CYP24A1
which is maintained by H+ efflux into a mucin-rich milieu
mRNA expression. As a consequence, this fructose-mediated
by NHE3, thereby providing H+ gradient or a source of H+
elevation of FGF-23 reduced 1,25(OH)2 D3 production.
for protonation of certain anions and solubility of minerals
Several minerals, such as nonheme iron and zinc, can
including calcium (222). The unstirred water layer in the
also indirectly hinder the transepithelial calcium transport
small intestine has approximately 500 to 800 μm thickness
(67, 130). In alkalinized luminal fluid, zinc has a tendency
with constant weak acid microenvironment (pH 6–7). This
to precipitate with calcium and phosphate into an insoluble
layer is comprised of water (∼95%) and other solutes, for
complex. The formation of these complexes is experimentally
example, mucin or glycans, secretory proteins, ions, extracel-
prevented by casein phosphopeptides (67). High iron intake
lular part of transporters, and receptors (97, 210). Neurogenic
and certain conditions with duodenal iron hyperabsorption,
chloride secretion, locomotion activity, or gut motility can
for example, β-thalassemia, are associated with a reduction
reduce the thickness of unstirred water layer to as thin as thin
in duodenal calcium transport (130). However, investigations
as <300 μm and this thinner layer is expected to enhance
pertaining to the effects of transition metals (e.g., copper)
absorption of nutrients and minerals (40, 142).
on intestinal calcium absorption are scant. A study in Ross
308 chicks has suggested that copper nanocolloid feeding
Luminal nutrients and copper accumulation in the small intestinal cells led to
Generally, Na+ -coupled nutrient cotransport induces myosin a significant decrease in calcium absorption; however, the
light chain kinase-mediated remodeling of perijunctional underlying cellular mechanism remains elusive (175). Since
actomyosin ring complex (95). This phenomenon can the intestinal oxidative stress caused by reactive oxygen
increase the paracellular permeability to small ions including species (ROS) often results in the downregulation of cal-
Na+ and Ca2+ . Indeed, phosphorylation of myosin II reg- bindins, PMCA1b and/or NCX1 as well as inhibition of
ulatory light chain and ZO-1 reorganization are crucial for calcium absorption (196, 197, 247), it is possible that iron
the overall barrier function of tight junction, thus increasing and copper diminish intestinal calcium absorption, in part, by
paracellular transport of many molecules and ions (95). inducing ROS production.
Luminal nutrients can also modulate intestinal calcium
transport (Figure 9). A number of l-amino acids, especially
leucine, proline, hydroxyproline, isoleucine, alanine, and Microbiota-derived metabolites
lysine, are capable of enhancing intestinal calcium trans- Microbiota produce large quantities of small acidic molecules,
port (220). Dipeptides and tripeptides—as substrates of which increase the solubility of calcium complexes and thus
H+ -dependent peptide transporter (PEPT)-1—also directly directly stimulates calcium absorption (Figure 9) (47). Since
stimulate calcium absorption in NHE3-dependent fashion only ionized calcium is readily absorbed by the intestinal
(220). Furthermore, certain small peptides, such as peptides absorptive cells or easily diffuses across the paracellular
from tilapia bone collagen hydrolysate, were found to exhibit space, the efficient sites of absorption as indicated by the
calcium-chelating capacity and increase calcium uptake relatively high transport rate are segments with acidic pH, that

Volume 11, May 2021 19


Intestinal Calcium Absorption Comprehensive Physiology

is, duodenum and proximal large intestine (60), the latter of was found to inhibit NHE3 activity in Caco-2 cells (88).
which (precisely cecum and proximal colon) is the major site Although there is no direct evidence of neural regulation
of microfloral fermentation. Gut microbiota produces several of intestinal calcium transport, the enteric neurons may
metabolites from digested foods and prebiotics. Among these indirectly modulate calcium absorption through alteration
molecules, short-chain fatty acids (SCFAs) are known to of sodium absorption or NHE3 activity. Changes in the
modulate intestinal calcium absorption (47). It has long been intestinal HCO3 − secretion by enteric neurons might alter
demonstrated that SCFAs, for example, acetate, propionate, the luminal pH, thereby affecting calcium solubility (29,
and butyrate, were capable of increasing calcium absorption 136). Moreover, the submucosal secretomotor neurons also
in the rat colon (150). Consistently, a randomized controlled produce bradykinin and arachidonic acid metabolites, which
trial study has also suggested the use of soluble corn fiber to are known to induce neurogenic chloride secretion across the
increase calcium absorption in female adolescents. Soluble small intestinal epithelium (189). In the kidney, bradykinin
corn fiber is a corn-derived nondigestible carbohydrate that activated TRPV5 for tubular calcium reabsorption (89), but
promotes SCFAs production and expansion of gut micro- whether it is able to activate the intestinal TRPV5 or TRPV6
biota such as Bifidobacterium and Parabacteroides (236), remains to be investigated.
thus suggesting that the gut microbiota may play a role in VIP is another important neurotransmitter that directly
calcium homeostasis and bone health. At the cellular level modulates paracellular calcium transport. A study in Caco-2
(Figure 10B), SCFAs (∼2 mmol/L) were reported to induce and HT29-Cl.19A monolayers suggested that direct exposure
SCFA response element and transcription of TRPV6 gene in to VIP on the basolateral side markedly increased transep-
Caco-2 cells (80). However, future research is needed to iden- ithelial resistance while decreasing paracellular calcium
tify the exact mechanisms of action of SCFAs on intestinal transport (18). In the feline gallbladder mucosa, intravenous
calcium absorption, calcium homeostasis, and bone strength. VIP infusion led to net secretion of calcium into the lumen,
Prebiotics, probiotics, and synbiotics have become impor- whereas sympathetic stimulation enhanced net calcium
tant ingredients for nutraceutical supplements for promotion absorption (171). It is noteworthy that direct innervation
of calcium absorption and bone health, especially in certain of enteric VIPergic neurons to the mouse ileal mucosa has
subgroups of individuals with osteopathy, such as insulin been shown to be essential for production of new goblet cells
resistance, diabetes mellitus, or dyslipidemia. For example, (208). These goblet cells secrete mucin into unstirred water
the probiotics Lactobacillus paracasei HII01, prebiotics layer, thus probably modulating diffusion of luminal ions
xylooligosaccharide, and synbiotics mixture of both have near the brush border.
been shown to augment osteoblastic bone formation in obese, Substance P, a neurotransmitter in the ENS, possibly
insulin-resistant rats (61). In addition, one of the widely used contributes to the regulation of calcium permeability despite
nutraceutical ingredients is fructooligosaccharide, which has having minimal effect on net calcium absorption across
been reported to increase calcium and magnesium absorption the rat ileum (231). Furthermore, dopamine was found to
across the colonic and rectal epithelia in rats (176). Unlike reduce calcium secretion across the rabbit ileum, thereby
fructose, a monosaccharide, that is readily absorbed in the decreasing total ileal calcium content and perhaps mitigat-
small intestine, fructooligosaccharide is indigestible and ing intestinal calcium wasting (56). We also observed that
its absorption is negligible. Thus it travels along the small tetrodotoxin—a potent sodium channel blocker and inhibitor
intestine as an intact molecule to the colon where it serves as of neural transmission—significantly diminished the rat duo-
substrate for bacterial fermentation in the colon and stimulates denal calcium transport (Charoenphandhu and Thammayon,
the growth of bacterial microflora. As a result, SCFAs are unpublished observations). Taken together, it is tempting to
produced from the fermentation process and these SCFAs are speculate that neural control of calcium transporter activity
likely to contribute to enhance bone formation and calcium exists in the intestine.
absorption as discussed in the aforementioned paragraph.

Gut-derived serotonin
Emerging Areas of Interest ENS and some special cell types in the intestine have potential
to secrete local regulatory factors to control calcium transport
Neural control of calcium absorption such as gut-derived serotonin. Approximately 90% of total
Since enteric nervous system (ENS) is formed by submucosal serotonin in our body is produced by cells in the gastrointesti-
and myenteric plexuses and consists of many secretomotor nal tract, for example, the duodenal enterochromaffin cells.
neurons, these neurons can directly innervate the enterocytes The remaining serotonin which is synthesized by serotoner-
and control epithelial transport of major ions, especially Na+ gic neurons in brain stem does not cross blood-brain barrier
and Cl− (40). Several neurotransmitters and neurohormones and thus has no role in regulating intestinal calcium transport.
such as acetylcholine, serotonin, nitric oxide, bradykinin, Normally, the gut-derived serotonin has a role in controlling
and vasoactive intestinal peptide (VIP) have been shown to gut motility and fluid and electrolyte transport (47, 249).
regulate intestinal chloride secretion (Figure 9) (40, 189). Tryptophan hydroxylase (TPH) is the rate-limiting enzyme
In addition, activation of 5-HT4 receptor by serotonin essential for serotonin biosynthesis. There are two isoforms of

20 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

this enzyme. TPH1 is mainly synthesized in enterochromaffin calcium transport has been well established especially in
cells, while TPH2 is produced in neurons (144). Serotonin humans and rodents, the cellular and molecular mechanisms
exerts its effects via serotonin receptors and reuptake into of paracellular calcium transport remain unclear and are
the cells via serotonin reuptake transporters (SERTs) that mostly based on in vitro studies. For example, it is unclear
are expressed apically on the intestinal epithelial cells (85). whether the calcium-selective pores formed by claudin het-
The activity of SERTs is dependent on extracellular sodium erodimers really exist in the intestine. More investigations
and chloride and is inhibited by selective serotonin reuptake are required to demonstrate the underlying mechanisms of
inhibitor (SSRI) such as fluoxetine (52). Enterochromaffin- factors affecting paracellular calcium absorption. These fac-
derived serotonin has reported to stimulate peristaltic reflexes tors include unstirred water layer, extracellular pH, luminal
and modify intestinal motility in normal and pathological and pericellular calcium, other nutrients as well as endocrine
conditions (84). In contrast to enterochromaffin cells, sero- and paracrine factors. Although the PTH-vitamin D system is
tonergic neurons appear to be crucial for controlling gut the most important regulator of calcium absorption, the role
motility. An experiment in TPH2 null mice showed longer of several other hormones including prolactin, GH, estro-
total gastrointestinal transit time than in wild-type controls, gen, and thyroid hormone could predominate under specific
especially in the colon (144). conditions. For instance, prolactin is required to promote
Although the effects of gut-derived serotonin on gut motil- systemic responses to meet a huge demand of calcium for
ity are extensively studied, evidence on the direct effects of lactogenesis (37).
serotonin on intestinal calcium absorption is still scant. Some The information pertaining to negative regulators or
investigations have indirectly suggested responsiveness of counterbalancing factors is scant. Local and systemic FGF-
the intestine to serotonin. Gill et al. (87) reported that, among 23 system, as well as CaSR, expressed in the enterocytes
different intestinal segments, SERT showed the highest probably help prevent calcium hyperabsorption as part of
expression level in the ileum, while duodenum, jejunum, a negative feedback loop, but little is known on how these
and colon had lower mRNA levels, respectively. Moreover, mediators regulate the function of calcium transporters in the
increases in extracellular serotonin induced by fluoxetine absorptive villous epithelial cells. Since the rate of calcium
treatment has been shown to upregulate the expression of absorption is dependent on nutrient contents in meals, such
transcellular calcium transporters (e.g., TRPV6, Cav 1.3, as sodium, amino acids, oligopeptides, prebiotic derivatives,
and NCX1) in male rats subjected to restraint stress. These and small acidic molecules (60, 80, 95, 240), the cellular
data suggest potential regulation of gut-derived serotonin on mechanisms of calcium absorption must be different among
intestinal calcium absorption (33). different human populations and species in different habitats
with a wide variety of diets. However, intestinal calcium
absorption of some species (e.g., Cryptomys damarensis and
Stanniocalcin (STC) Heterocephalus glaber) has been reported to be independent
In bony fish, the intestinal calcium transport is under the of vitamin D (188). Therefore, there are other calciotropic
control of STC1. STC1 is a glycosylated peptide hormone factors yet to be identified.
that first discovered in the corpuscles of Stannius—a small
endocrine gland in the kidney of teleostean and holostean
fish. STC1 was discovered to have similar function to calci- Acknowledgements
tonin which appears to prevent hypercalcemia by preventing
calcium entry into the gill and intestine of fish (106, 112). The authors thank Professor Nateetip Krishnamra for valu-
In mammals, human recombinant STC has been reported to able comments on this article and Thitapha Kiattisirichai for
decrease calcium absorption across the rat duodenum (152). artwork. Our research was supported by grants from Mahi-
This observed phenomenon might be due to a downregulation dol University – Multidisciplinary Research Cluster Grant
of TRPV5 and TRPV6 expression, but not NCX1, PMCA1b , (to NC), the Thailand Research Fund (TRF) through the TRF
or VDR expression (246). Hung et al. (103) demonstrated Senior Research Scholar Grant (RTA6080007 to NC), TRF
that the expression of STC1 in renal proximal tubular cells International Research Network Program (IRN60W0001 to
of rats was positively regulated by 1,25(OH)2 D3 and PTH, KW and NC), TRF – Office of the Higher Education Com-
presumably as part of a negative feedback loop. mission Research Grant for New Scholar (MRG6280198 to
JT), Faculty of Allied Health Sciences, Burapha University
(to KW), National Research Council of Thailand (NRCT, to
NC), and Faculty of Science, Mahidol University (CIF and
Conclusions and Perspectives CNI grant, to NC).
Intestinal calcium absorption is a complex cellular energy-
dependent process that more than 20 protein families, for References
example, TRPV5/6, Cav 1.3, calbindins, PMCAs, NCX1, and
claudins, are involved in the mechanisms of transcellular 1. Ahlem A, Ali el H, Leila T. Lysosomes of the liver and the duodenal
enterocytes, a site of handling of two rare earths. Ultrastructural and
and paracellular calcium transport. While transcellular active microanalytical study. Biol Trace Elem Res 124: 40-51, 2008.

Volume 11, May 2021 21


Intestinal Calcium Absorption Comprehensive Physiology

2. Akhter S, Kutuzova GD, Christakos S, DeLuca HF. Calbindin D9k is 3 inhibitor diminishes hepcidin-enhanced duodenal calcium transport
not required for 1,25-dihydroxyvitamin D3 -mediated Ca2+ absorption in hemizygous β-globin knockout thalassemic mice. Mol Cell Biochem
in small intestine. Arch Biochem Biophys 460: 227-232, 2007. 427: 201-208, 2017.
3. Alexander RT, Rievaj J, Dimke H. Paracellular calcium transport across 28. Charoenphandhu N, Krishnamra N. Prolactin is an important regulator
renal and intestinal epithelia. Biochem Cell Biol 92: 467-480, 2014. of intestinal calcium transport. Can J Physiol Pharmacol 85: 569-581,
4. Altimimi HF, Schnetkamp PP. Examining Ca2+ extrusion of 2007.
Na+ /Ca2+ -K+ exchangers. Ann NY Acad Sci 1099: 29-33, 2007. 29. Charoenphandhu N, Laohapitakworn S, Kraidith K, Nakkrasae LI,
5. Altimimi HF, Schnetkamp PP. Na+ /Ca2+ -K+ exchangers (NCKX): Jongwattanapisan P, Tharabenjasin P, Krishnamra N. Electrogenic
Functional properties and physiological roles. Channels (Austin) 1: Na+ /HCO3 – co-transporter-1 is essential for the parathyroid hormone-
62-69, 2007. stimulated intestinal HCO3 – secretion. Biochem Biophys Res Commun
6. Amerongen HM, Mack JA, Wilson JM, Neutra MR. Membrane 409: 775-779, 2011.
domains of intestinal epithelial cells: Distribution of Na+ ,K+ -ATPase 30. Charoenphandhu N, Limlomwongse L, Krishnamra N. Prolactin
and the membrane skeleton in adult rat intestine during fetal devel- directly stimulates transcellular active calcium transport in the duode-
opment and after epithelial isolation. J Cell Biol 109: 2129-2138, num of female rats. Can J Physiol Pharmacol 79: 430-438, 2001.
1989. 31. Charoenphandhu N, Limlomwongse L, Krishnamra N. Prolactin
7. Anderson JM, Van Itallie CM. Physiology and function of the tight directly enhanced Na+ /K+ - and Ca2+ -ATPase activities in the
junction. Cold Spring Harb Perspect Biol 1: a002584, 2009. duodenum of female rats. Can J Physiol Pharmacol 84: 555-563,
8. Armbrecht HJ, Boltz MA, Christakos S, Bruns ME. Capacity of 1,25- 2006.
dihydroxyvitamin D to stimulate expression of calbindin D changes 32. Charoenphandhu N, Nakkrasae LI, Kraidith K, Teerapornpuntakit J,
with age in the rat. Arch Biochem Biophys 352: 159-164, 1998. Thongchote K, Thongon N, Krishnamra N. Two-step stimulation of
9. Armbrecht HJ, Boltz MA, Kumar VB. Intestinal plasma membrane cal- intestinal Ca2+ absorption during lactation by long-term prolactin expo-
cium pump protein and its induction by 1,25(OH)2 D3 decrease with sure and suckling-induced prolactin surge. Am J Physiol Endocrinol
age. Am J Physiol 277: G41-G47, 1999. Metab 297: E609-E619, 2009.
10. Askari M, Karamzadeh R, Ansari-Pour N, Karimi-Jafari MH, 33. Charoenphandhu N, Suntornsaratoon P, Jongwattanapisan P, Wongdee
Almadani N, Sadighi Gilani MA, Gourabi H, Vosough Taghi Dizaj K, Krishnamra N. Enhanced trabecular bone resorption and microstruc-
A, Mohseni Meybodi A, Sadeghi M, Bashamboo A, McElreavey tural bone changes in rats after removal of the cecum. Am J Physiol
K, Totonchi M. Identification of a missense variant in CLDN2 in Endocrinol Metab 303: E1069-E1075, 2012.
obstructive azoospermia. J Hum Genet 64: 1023-1032, 2019. 34. Charoenphandhu N, Teerapornpuntakit J, Lapmanee S, Krishnamra N,
11. Bacchetta J, Sea JL, Chun RF, Lisse TS, Wesseling-Perry K, Gales B, Charoenphandhu J. Duodenal calcium transporter mRNA expression
Adams JS, Salusky IB, Hewison M. Fibroblast growth factor 23 inhibits in stressed male rats treated with diazepam, fluoxetine, reboxetine, or
extrarenal synthesis of 1,25-dihydroxyvitamin D in human monocytes. venlafaxine. Mol Cell Biochem 369: 87-94, 2012.
J Bone Miner Res 28: 46-55, 2013. 35. Charoenphandhu N, Tudpor K, Pulsook N, Krishnamra N. Chronic
12. Barger-Lux MJ, Heaney RP, Lanspa SJ, Healy JC, DeLuca HF. An metabolic acidosis stimulated transcellular and solvent drag-induced
investigation of sources of variation in calcium absorption efficiency. calcium transport in the duodenum of female rats. Am J Physiol
J Clin Endocrinol Metab 80: 406-411, 1995. Gastrointest Liver Physiol 291: G446-G455, 2006.
13. Beggs MR, Lee JJ, Busch K, Raza A, Dimke H, Weissgerber P, Engel 36. Charoenphandhu N, Wongdee K, Krishnamra N. Is prolactin the
J, Flockerzi V, Alexander RT. TRPV6 and Cav 1.3 mediate distal small cardinal calciotropic maternal hormone? Trends Endocrinol Metab 21:
intestine calcium absorption before weaning. Cell Mol Gastroenterol 395-401, 2010.
Hepatol 8: 625-642, 2019. 37. Charoenphandhu N, Wongdee K, Teerapornpuntakit J, Thongchote K,
14. Behar J, Kerstein MD. Intestinal calcium absorption: Differences in Krishnamra N. Transcriptome responses of duodenal epithelial cells
transport between duodenum and ileum. Am J Physiol 230: 1255-1260, to prolactin in pituitary-grafted rats. Mol Cell Endocrinol 296: 41-52,
1976. 2008.
15. Benn BS, Ajibade D, Porta A, Dhawan P, Hediger M, Peng JB, Jiang 38. Christakos S, Dhawan P, Ajibade D, Benn BS, Feng J, Joshi SS. Mech-
Y, Oh GT, Jeung EB, Lieben L, Bouillon R, Carmeliet G, Christakos S. anisms involved in vitamin D mediated intestinal calcium absorption
Active intestinal calcium transport in the absence of transient receptor and in non-classical actions of vitamin D. J Steroid Biochem Mol Biol
potential vanilloid type 6 and calbindin-D9k . Endocrinology 149: 3196- 121: 183-187, 2010.
3205, 2008. 39. Colegio OR, Van Itallie C, Rahner C, Anderson JM. Claudin extracel-
16. Bhaskar KR, Garik P, Turner BS, Bradley JD, Bansil R, Stanley HE, lular domains determine paracellular charge selectivity and resistance
LaMont JT. Viscous fingering of HCl through gastric mucin. Nature
360: 458-461, 1992. but not tight junction fibril architecture. Am J Physiol Cell Physiol 284:
17. Bindels RJ, Timmermans JA, Hartog A, Coers W, van Os CH. C1346-C1354, 2003.
Calbindin-D9k and parvalbumin are exclusively located along basolat- 40. Cooke HJ. Neurotransmitters in neuronal reflexes regulating intestinal
eral membranes in rat distal nephron. J Am Soc Nephrol 2: 1122-1129, secretion. Ann NY Acad Sci 915: 77-80, 2000.
1991. 41. Corbeels K, Verlinden L, Lannoo M, Simoens C, Matthys C, Verstuyf
18. Blais A, Aymard P, Lacour B. Paracellular calcium transport across A, Meulemans A, Carmeliet G, Van der Schueren B. Thin bones: Vita-
Caco-2 and HT29 cell monolayers. Pflugers Arch 434: 300-305, 1997. min D and calcium handling after bariatric surgery. Bone Rep 8: 57-63,
19. Brommage R, Binacua C, Carrie AL. The cecum does not participate in 2018.
the stimulation of intestinal calcium absorption by calcitriol. J Steroid 42. Cormick G, Belizan JM. Calcium intake and health. Nutrients 11: 1606,
Biochem Mol Biol 54: 71-73, 1995. 2019.
20. Bronner F, Pansu D. Nutritional aspects of calcium absorption. J Nutr 43. Cramer CF. Effect of salmon calcitonin on in vivo calcium absorption
129: 9-12, 1999. in rats. Calcif Tissue Res 13: 169-172, 1973.
21. Bronner F, Pansu D, Stein WD. An analysis of intestinal calcium trans- 44. Cramer CF, Copp DH. Progress and rate of absorption of radiostron-
port across the rat intestine. Am J Physiol 250: G561-G569, 1986. tium through intestinal tracts of rats. Proc Soc Exp Biol Med 102: 514-
22. Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hop- 517, 1959.
kins AM, Nusrat A. Proinflammatory cytokines disrupt epithelial bar- 45. Cui M, Li Q, Johnson R, Fleet JC. Villin promoter-mediated transgenic
rier function by apoptosis-independent mechanisms. J Immunol 171: expression of transient receptor potential cation channel, subfamily V,
6164-6172, 2003. member 6 (TRPV6) increases intestinal calcium absorption in wild-
23. Carrasco F, Basfi-Fer K, Rojas P, Csendes A, Papapietro K, Codoceo type and vitamin D receptor knockout mice. J Bone Miner Res 27:
J, Inostroza J, Krebs NF, Westcott JL, Miller LV, Ruz M. Calcium 2097-2107, 2012.
absorption may be affected after either sleeve gastrectomy or Roux-en- 46. Curry JN, Saurette M, Askari M, Pei L, Filla MB, Beggs MR, Rowe PS,
Y gastric bypass in premenopausal women: A 2-y prospective study. Fields T, Sommer AJ, Tanikawa C, Kamatani Y, Evan AP, Totonchi M,
Am J Clin Nutr 108: 24-32, 2018. Alexander RT, Matsuda K, Yu AS. Claudin-2 deficiency associates with
24. Carroll KM, Wood RJ, Chang EB, Rosenberg IH. Glucose enhance- hypercalciuria in mice and human kidney stone disease. J Clin Invest
ment of transcellular calcium transport in the intestine. Am J Physiol 130: 1948-1960, 2020.
255: G339-G345, 1988. 47. D’Amelio P, Sassi F. Gut microbiota, immune system, and bone. Calcif
25. Cervetto L, Lagnado L, Perry RJ, Robinson DW, McNaughton PA. Tissue Int 102: 415-425, 2018.
Extrusion of calcium from rod outer segments is driven by both sodium 48. Darwish HM, DeLuca HF. Identification of a 1,25-dihydroxyvitamin
and potassium gradients. Nature 337: 740-743, 1989. D3 -response element in the 5’-flanking region of the rat calbindin D-9k
26. Chandra S, Fullmer CS, Smith CA, Wasserman RH, Morrison GH. Ion gene. Proc Natl Acad Sci USA 89: 603-607, 1992.
microscopic imaging of calcium transport in the intestinal tissue of vita- 49. Deevi RK, McClements J, McCloskey KD, Fatehullah A, Tkocz
min D-deficient and vitamin D-replete chickens: A 44 Ca stable isotope D, Javadi A, Higginson R, Marsh Durban V, Jansen M, Clarke A,
study. Proc Natl Acad Sci USA 87: 5715-5719, 1990. Loughrey MB, Campbell FC. Vitamin D3 suppresses morphological
27. Charoenphandhu N, Kraidith K, Lertsuwan K, Sripong C, Suntorn- evolution of the cribriform cancerous phenotype. Oncotarget 7:
saratoon P, Svasti S, Krishnamra N, Wongdee K. Na+ /H+ exchanger 49042-49064, 2016.

22 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

50. den Dekker E, Hoenderop JG, Nilius B, Bindels RJ. The epithelial 75. Freeman SA, Uderhardt S, Saric A, Collins RF, Buckley CM, Mylva-
calcium channels, TRPV5 & TRPV6: From identification towards ganam S, Boroumand P, Plumb J, Germain RN, Ren D, Grinstein S.
regulation. Cell Calcium 33: 497-507, 2003. Lipid-gated monovalent ion fluxes regulate endocytic traffic and sup-
51. Derler I, Hofbauer M, Kahr H, Fritsch R, Muik M, Kepplinger K, Hack port immune surveillance. Science 367: 301-305, 2020.
ME, Moritz S, Schindl R, Groschner K, Romanin C. Dynamic but 76. Friedman J, Au WY, Raisz LG. Responses of fetal rat bone to thyro-
not constitutive association of calmodulin with rat TRPV6 channels calcitonin in tissue culture. Endocrinology 82: 149-156, 1968.
enables fine tuning of Ca2+ -dependent inactivation. J Physiol 577: 77. Friedman J, Raisz LG. Thyrocalcitonin: Inhibitor of bone resorption in
31-44, 2006. tissue culture. Science 150: 1465-1467, 1965.
52. Descarries L, Riad M. Effects of the antidepressant fluoxetine on the 78. Fujita H, Chiba H, Yokozaki H, Sakai N, Sugimoto K, Wada T, Kojima
subcellular localization of 5-HT1A receptors and SERT. Philos Trans T, Yamashita T, Sawada N. Differential expression and subcellular
R Soc B 367: 2416-2425, 2012. localization of claudin-7, -8, -12, -13, and -15 along the mouse
53. Dhawan P, Veldurthy V, Yehia G, Hsaio C, Porta A, Kim KI, Patel intestine. J Histochem Cytochem 54: 933-944, 2006.
N, Lieben L, Verlinden L, Carmeliet G, Christakos S. Transgenic 79. Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, Uchiyama Y,
expression of the vitamin D receptor restricted to the ileum, cecum, Yamamoto Y, Wada T, Kojima T, Yokozaki H, Yamashita T, Kato
and colon of vitamin D receptor knockout mice rescues vitamin D S, Sawada N, Chiba H. Tight junction proteins claudin-2 and -12 are
receptor-dependent rickets. Endocrinology 158: 3792-3804, 2017. critical for vitamin D-dependent Ca2+ absorption between enterocytes.
54. Diaz de Barboza G, Guizzardi S, Tolosa de Talamoni N. Molecular Mol Biol Cell 19: 1912-1921, 2008.
aspects of intestinal calcium absorption. World J Gastroenterol 21: 80. Fukushima A, Aizaki Y, Sakuma K. Short-chain fatty acids induce
7142-7154, 2015. intestinal transient receptor potential vanilloid type 6 expression in rats
55. Dong H, Sellers ZM, Smith A, Chow JY, Barrett KE. Na+ /Ca2+ and Caco-2 cells. J Nutr 139: 20-25, 2009.
exchange regulates Ca2+ -dependent duodenal mucosal ion transport 81. Fullmer CS, Chandra S, Smith CA, Morrison GH, Wasserman RH.
and HCO3 – secretion in mice. Am J Physiol Gastrointest Liver Physiol Ion microscopic imaging of calcium during 1,25-dihydroxyvitamin
288: G457-G465, 2005. D-mediated intestinal absorption. Histochem Cell Biol 106: 215-222,
56. Donowitz M, Cusolito S, Battisti L, Fogel R, Sharp GW. Dopamine 1996.
stimulation of active Na and Cl absorption in rabbit ileum: Interaction 82. Gallagher JC. Vitamin D and aging. Endocrinol Metab Clin North Am
with α2-adrenergic and specific dopamine receptors. J Clin Invest 69: 42: 319-332, 2013.
1008-1016, 1982. 83. Gawenis LR, Ledoussal C, Judd LM, Prasad V, Alper SL, Stuart-Tilley
57. Dorkkam N, Wongdee K, Suntornsaratoon P, Krishnamra N, Charoen- A, Woo AL, Grisham C, Sanford LP, Doetschman T, Miller ML, Shull
phandhu N. Prolactin stimulates the L-type calcium channel-mediated GE. Mice with a targeted disruption of the AE2 Cl– /HCO3 – exchanger
transepithelial calcium transport in the duodenum of male rats. Biochem are achlorhydric. J Biol Chem 279: 30531-30539, 2004.
Biophys Res Commun 430: 711-716, 2013. 84. Gershon MD. 5-Hydroxytryptamine (serotonin) in the gastrointestinal
58. Douard V, Asgerally A, Sabbagh Y, Sugiura S, Shapses SA, Casirola tract. Curr Opin Endocrinol Diabetes Obes 20: 14-21, 2013.
D, Ferraris RP. Dietary fructose inhibits intestinal calcium absorption 85. Ghia JE, Li N, Wang H, Collins M, Deng Y, El-Sharkawy RT, Cote F,
and induces vitamin D insufficiency in CKD. J Am Soc Nephrol 21: Mallet J, Khan WI. Serotonin has a key role in pathogenesis of experi-
261-271, 2010. mental colitis. Gastroenterology 137: 1649-1660, 2009.
59. Douard V, Sabbagh Y, Lee J, Patel C, Kemp FW, Bogden JD, Lin S, 86. Ghijsen WE, De Jong MD, Van Os CH. Kinetic properties of
Ferraris RP. Excessive fructose intake causes 1,25-(OH)2 D3 -dependent Na+ /Ca2+ exchange in basolateral plasma membranes of rat small
inhibition of intestinal and renal calcium transport in growing rats. Am intestine. Biochim Biophys Acta 730: 85-94, 1983.
J Physiol Endocrinol Metab 304: E1303-E1313, 2013. 87. Gill RK, Pant N, Saksena S, Singla A, Nazir TM, Vohwinkel L,
60. Duflos C, Bellaton C, Pansu D, Bronner F. Calcium solubility, intestinal Turner JR, Goldstein J, Alrefai WA, Dudeja PK. Function, expression,
sojourn time and paracellular permeability codetermine passive cal- and characterization of the serotonin transporter in the native human
cium absorption in rats. J Nutr 125: 2348-2355, 1995. intestine. Am J Physiol Gastrointest Liver Physiol 294: G254-G262,
61. Eaimworawuthikul S, Tunapong W, Chunchai T, Suntornsaratoon P, 2008.
Charoenphandhu N, Thiennimitr P, Chattipakorn N, Chattipakorn SC. 88. Gill RK, Saksena S, Tyagi S, Alrefai WA, Malakooti J, Sarwar Z, Turner
Altered gut microbiota ameliorates bone pathology in the mandible of JR, Ramaswamy K, Dudeja PK. Serotonin inhibits Na+ /H+ exchange
obese-insulin-resistant rats. Eur J Nutr 59: 1453-1462, 2020. activity via 5-HT4 receptors and activation of PKCα in human intestinal
62. Erben RG, Andrukhova O. FGF23-Klotho signaling axis in the kidney.
Bone 100: 62-68, 2017. epithelial cells. Gastroenterology 128: 962-974, 2005.
63. Favus MJ, Angeid-Backman E, Breyer MD, Coe FL. Effects of triflu- 89. Gkika D, Topala CN, Chang Q, Picard N, Thebault S, Houillier P,
operazine, ouabain, and ethacrynic acid on intestinal calcium transport. Hoenderop JG, Bindels RJ. Tissue kallikrein stimulates Ca2+ reabsorp-
Am J Physiol 244: G111-G115, 1983. tion via PKC-dependent plasma membrane accumulation of TRPV5.
64. Favus MJ, Kathpalia SC, Coe FL. Kinetic characteristics of calcium EMBO J 25: 4707-4716, 2006.
absorption and secretion by rat colon. Am J Physiol 240: G350-G354, 90. Gloux A, Le Roy N, Brionne A, Bonin E, Juanchich A, Benzoni G,
1981. Piketty ML, Prie D, Nys Y, Gautron J, Narcy A, Duclos MJ. Candidate
65. Feher JJ. Facilitated calcium diffusion by intestinal calcium-binding genes of the transcellular and paracellular calcium absorption pathways
protein. Am J Physiol 244: C303-C307, 1983. in the small intestine of laying hens. Poult Sci 98: 6005-6018, 2019.
66. Feher JJ, Fullmer CS, Wasserman RH. Role of facilitated diffusion of 91. Gloux A, Le Roy N, Ezagal J, Meme N, Hennequet-Antier C, Piketty
calcium by calbindin in intestinal calcium absorption. Am J Physiol ML, Prie D, Benzoni G, Gautron J, Nys Y, Narcy A, Duclos MJ. Possi-
262: C517-C526, 1992. ble roles of parathyroid hormone, 1.25(OH)2 D3 , and fibroblast growth
67. Feng Y, Zhang J, Miao Y, Guo W, Feng G, Yang Y, Guo T, Wu H, factor 23 on genes controlling calcium metabolism across different tis-
Zeng M. Prevention of zinc precipitation with calcium phosphate by sues of the laying hen. Domest Anim Endocrinol 72: 106407, 2019.
casein hydrolysate improves zinc absorption in mouse small intestine 92. Grinstead WC, Pak CY, Krejs GJ. Effect of 1,25-dihydroxyvitamin D3
ex vivo via a nanoparticle-mediated mechanism. J Agric Food Chem on calcium absorption in the colon of healthy humans. Am J Physiol
68: 652-659, 2020. 247: G189-G192, 1984.
68. Fleet JC. Rapid, membrane-initiated actions of 1,25 dihydroxyvitamin 93. Guo YC, Yuan Q. Fibroblast growth factor 23 and bone mineralisation.
D: What are they and what do they mean? J Nutr 134: 3215-3218, 2004. Int J Oral Sci 7: 8-13, 2015.
69. Fleet JC. Regulation of intestinal calcium and phosphate absorption. 94. Hasan R, Zhang X. Ca2+ regulation of TRP ion channels. Int J Mol Sci
In: Feldman D, editor. Vitamin D. London, UK: Academic Press, 2018, 19: 1256, 2018.
p. 329-342. 95. He WQ, Wang J, Sheng JY, Zha JM, Graham WV, Turner JR. Contribu-
70. Fleet JC, Bruns ME, Hock JM, Wood RJ. Growth hormone and parathy- tions of myosin light chain kinase to regulation of epithelial paracellular
roid hormone stimulate intestinal calcium absorption in aged female permeability and mucosal homeostasis. Int J Mol Sci 21: 993, 2020.
rats. Endocrinology 134: 1755-1760, 1994. 96. Herrmann JR, Turner JR. Beyond Ussing’s chambers: Contemporary
71. Fleet JC, Eksir F, Hance KW, Wood RJ. Vitamin D-inducible calcium thoughts on integration of transepithelial transport. Am J Physiol Cell
transport and gene expression in three Caco-2 cell lines. Am J Physiol Physiol 310: C423-C431, 2016.
Gastrointest Liver Physiol 283: G618-G625, 2002. 97. Hibino H, Takai M, Noguchi H, Sawamura S, Takahashi Y, Sakai H,
72. Fleet JC, Reyes-Fernandez P. Intestinal responses to 1,25 dihydroxyvi- Shiku H. An approach to the research on ion and water properties in
tamin D are not improved by higher intestinal VDR levels resulting the interphase between the plasma membrane and bulk extracellular
from intestine-specific transgenic expression of VDR in mice. J Steroid solution. J Physiol Sci 67: 439-445, 2017.
Biochem Mol Biol 200: 105670, 2020. 98. Hoenderop JG, Nilius B, Bindels RJ. Calcium absorption across epithe-
73. Fleet JC, Schoch RD. Molecular mechanisms for regulation of intesti- lia. Physiol Rev 85: 373-422, 2005.
nal calcium absorption by vitamin D and other factors. Crit Rev Clin 99. Hoenderop JG, Vennekens R, Muller D, Prenen J, Droogmans G,
Lab Sci 47: 181-195, 2010. Bindels RJ, Nilius B. Function and expression of the epithelial Ca2+
74. Förster C. Tight junctions and the modulation of barrier function in channel family: Comparison of mammalian ECaC1 and 2. J Physiol
disease. Histochem Cell Biol 130: 55-70, 2008. 537: 747-761, 2001.

Volume 11, May 2021 23


Intestinal Calcium Absorption Comprehensive Physiology

100. Hong EJ, Jeung EB. Biological significance of calbindin-D9k within 124. Kiela PR, Ghishan FK. Molecular mechanisms of intestinal trans-
duodenal epithelium. Int J Mol Sci 14: 23330-23340, 2013. port of calcium, phosphate, and magnesium. In: Said H, editor.
101. Howard A, Legon S, Walters JR. Human and rat intestinal plasma mem- Physiology of the Gastrointestinal Tract. London, UK: Elsevier Inc.,
brane calcium pump isoforms. Am J Physiol 265: G917-G925, 1993. 2018, p. 1405-1449.
102. Huhtakangas JA, Olivera CJ, Bishop JE, Zanello LP, Norman AW. The 125. Kladnitsky O, Rozenfeld J, Azulay-Debby H, Efrati E, Zelikovic
vitamin D receptor is present in caveolae-enriched plasma membranes I. The claudin-16 channel gene is transcriptionally inhibited by
and binds 1α,25(OH)2 -vitamin D3 in vivo and in vitro. Mol Endocrinol 1,25-dihydroxyvitamin D. Exp Physiol 100: 79-94, 2015.
18: 2660-2671, 2004. 126. Kolek OI, Hines ER, Jones MD, LeSueur LK, Lipko MA, Kiela PR,
103. Hung NT, Yamamoto H, Takei Y, Masuda M, Otani A, Kozai M, Ikeda Collins JF, Haussler MR, Ghishan FK. 1α,25-Dihydroxyvitamin D3
S, Nakahashi O, Tanaka S, Taketani Y, Takeda E. Up-regulation of upregulates FGF23 gene expression in bone: The final link in a renal-
stanniocalcin 1 expression by 1,25-dihydroxy vitamin D3 and parathy- gastrointestinal-skeletal axis that controls phosphate transport. Am J
roid hormone in renal proximal tubular cells. J Clin Biochem Nutr 50: Physiol Gastrointest Liver Physiol 289: G1036-G1042, 2005.
227-233, 2012. 127. Kopic S, Geibel JP. Gastric acid, calcium absorption, and their impact
104. Hwang I, Hong EJ, Yang H, Kang HS, Ahn C, An BS, Jeung EB. on bone health. Physiol Rev 93: 189-268, 2013.
Regulation of tight junction gene expression in the kidney of calbindin- 128. Kovbasnjuk ON, Szmulowicz U, Spring KR. Regulation of the MDCK
D9k and/or -D28k knockout mice after consumption of a calcium- or a cell tight junction. J Membr Biol 161: 93-104, 1998.
calcium/vitamin D-deficient diet. BMC Biochem 15: 6, 2014. 129. Kraidith K, Jantarajit W, Teerapornpuntakit J, Nakkrasae LI, Krish-
105. Hwang I, Yang H, Kang HS, Ahn C, Hong EJ, An BS, Jeung EB. namra N, Charoenphandhu N. Direct stimulation of the transcellular
Alteration of tight junction gene expression by calcium- and vitamin and paracellular calcium transport in the rat cecum by prolactin.
D-deficient diet in the duodenum of calbindin-null mice. Int J Mol Sci Pflugers Arch 458: 993-1005, 2009.
14: 22997-23010, 2013. 130. Kraidith K, Svasti S, Teerapornpuntakit J, Vadolas J, Chaimana
106. Ishibashi K, Imai M. Prospect of a stanniocalcin endocrine/paracrine R, Lapmanee S, Suntornsaratoon P, Krishnamra N, Fucharoen S,
system in mammals. Am J Physiol Renal Physiol 282: F367-F375, Charoenphandhu N. Hepcidin and 1,25(OH)2 D3 effectively restore
2002. Ca2+ transport in beta-thalassemic mice: Reciprocal phenomenon
107. Jaeger P, Jones W, Clemens TL, Hayslett JP. Evidence that calcitonin of Fe2+ and Ca2+ absorption. Am J Physiol Endocrinol Metab 311:
stimulates 1,25-dihydroxyvitamin D production and intestinal absorp- E214-E223, 2016.
tion of calcium in vivo. J Clin Invest 78: 456-461, 1986. 131. Krejs GJ, Nicar MJ, Zerwekh JE, Norman DA, Kane MG, Pak CY.
108. Jantarajit W, Thongon N, Pandaranandaka J, Teerapornpuntakit J, Effect of 1,25-dihydroxyvitamin D3 on calcium and magnesium
Krishnamra N, Charoenphandhu N. Prolactin-stimulated transep- absorption in the healthy human jejunum and ileum. Am J Med 75:
ithelial calcium transport in duodenum and Caco-2 monolayer are 973-976, 1983.
mediated by the phosphoinositide 3-kinase pathway. Am J Physiol 132. Krishnamra N, Angkanaporn K, Deenoi T. Comparison of calcium
Endocrinol Metab 293: E372-E384, 2007. absorptive and secretory capacities of segments of intact or function-
109. Jeung EB, Leung PC, Krisinger J. The human calbindin-D9k gene. ally resected intestine during normo-, hypo-, and hyper-calcemia. Can
Complete structure and implications on steroid hormone regulation. J Physiol Pharmacol 72: 764-770, 1994.
J Mol Biol 235: 1231-1238, 1994. 133. Kühne H, Hause G, Grundmann SM, Schutkowski A, Brandsch
110. Jimbo R, Kawakami-Mori F, Mu S, Hirohama D, Majtan B, Shimizu C, Stangl GI. Vitamin D receptor knockout mice exhibit elongated
Y, Yatomi Y, Fukumoto S, Fujita T, Shimosawa T. Fibroblast growth intestinal microvilli and increased ezrin expression. Nutr Res 36:
factor 23 accelerates phosphate-induced vascular calcification in the 184-192, 2016.
absence of Klotho deficiency. Kidney Int 85: 1103-1111, 2014. 134. Kutuzova GD, Akhter S, Christakos S, Vanhooke J, Kimmel-Jehan C,
111. Jongwattanapisan P, Suntornsaratoon P, Wongdee K, Dorkkam Deluca HF. Calbindin D9k knockout mice are indistinguishable from
N, Krishnamra N, Charoenphandhu N. Impaired body calcium wild-type mice in phenotype and serum calcium level. Proc Natl Acad
metabolism with low bone density and compensatory colonic calcium Sci USA 103: 12377-12381, 2006.
absorption in cecectomized rats. Am J Physiol Endocrinol Metab 302: 135. Kutuzova GD, Sundersingh F, Vaughan J, Tadi BP, Ansay SE, Chris-
E852-E863, 2012. takos S, Deluca HF. TRPV6 is not required for 1α,25-dihydroxyvitamin
112. Joshi AD. New insights into physiological and pathophysiological D3 -induced intestinal calcium absorption in vivo. Proc Natl Acad Sci
functions of stanniocalcin 2. Front Endocrinol (Lausanne) 11: 172, USA 105: 19655-19659, 2008.
2020. 136. Laohapitakworn S, Thongbunchoo J, Nakkrasae LI, Krishnamra N,
113. Jüppner H. Phosphate and FGF-23. Kidney Int Suppl 79: S24-S27, Charoenphandhu N. Parathyroid hormone (PTH) rapidly enhances
2011. CFTR-mediated HCO3 – secretion in intestinal epithelium-like Caco-2
114. Kang S, Hong J, Lee JM, Moon HE, Jeon B, Choi J, Yoon NA, Paek monolayer: A novel ion regulatory action of PTH. Am J Physiol Cell
SH, Roh EJ, Lee CJ, Kang SS. Trifluoperazine, a well-known antipsy- Physiol 301: C137-C149, 2011.
chotic, inhibits glioblastoma invasion by binding to calmodulin and 137. Larsson D, Nemere I. Vectorial transcellular calcium transport in intes-
disinhibiting calcium release channel IP3R. Mol Cancer Ther 16: 217- tine: Integration of current models. J Biomed Biotechnol 2: 117-119,
227, 2017. 2002.
115. Karbach U. Segmental heterogeneity of cellular and paracellular cal- 138. Lee D, Obukhov AG, Shen Q, Liu Y, Dhawan P, Nowycky MC, Chris-
cium transport across the rat duodenum and jejunum. Gastroenterology takos S. Calbindin-D28k decreases L-type calcium channel activity
100: 47-58, 1991. and modulates intracellular calcium homeostasis in response to K+
116. Karbach U. Paracellular calcium transport across the small intestine. depolarization in a rat beta cell line RINr1046-38. Cell Calcium 39:
J Nutr 122: 672-677, 1992. 475-485, 2006.
117. Karbach U, Feldmeier H. The cecum is the site with the highest calcium 139. Lee GS, Lee KY, Choi KC, Ryu YH, Paik SG, Oh GT, Jeung EB. Phe-
absorption in rat intestine. Dig Dis Sci 38: 1815-1824, 1993. notype of a calbindin-D9k gene knockout is compensated for by the
118. Karbach U, Rummel W. Calcium transport across the colon ascendens induction of other calcium transporter genes in a mouse model. J Bone
and the influence of 1,25-dihydroxyvitamin D3 and dexamethasone. Miner Res 22: 1968-1978, 2007.
Eur J Clin Invest 17: 368-374, 1987. 140. Lee JJ, Liu X, O’Neill D, Beggs MR, Weissgerber P, Flockerzi V,
119. Karbach U, Rummel W. Cellular and paracellular calcium transport Chen XZ, Dimke H, Alexander RT. Activation of the calcium sensing
in the rat ileum and the influence of 1α,25-dihydroxyvitamin D3 receptor attenuates TRPV6-dependent intestinal calcium absorption.
and dexamethasone. Naunyn Schmiedebergs Arch Pharmacol 336: JCI Insight 5: e128013, 2019.
117-124, 1987. 141. Lertsuwan K, Wongdee K, Teerapornpuntakit J, Charoenphandhu N.
120. Kellett GL. Alternative perspective on intestinal calcium absorption: Intestinal calcium transport and its regulation in thalassemia: Interac-
Proposed complementary actions of Cav 1.3 and TRPV6. Nutr Rev 69: tion between calcium and iron metabolism. J Physiol Sci 68: 221-232,
347-370, 2011. 2018.
121. Khanal RC, Nemere I. Regulation of intestinal calcium transport. Annu 142. Levitt MD, Furne JK, Levitt DG. Shaking of the intact rat and intestinal
Rev Nutr 28: 179-196, 2008. angulation diminish the jejunal unstirred layer. Gastroenterology 103:
122. Khuituan P, Teerapornpuntakit J, Wongdee K, Suntornsaratoon P, Kon- 1460-1466, 1992.
thapakdee N, Sangsaksri J, Sripong C, Krishnamra N, Charoenphandhu 143. Li J, Zhao L, Ferries IK, Jiang L, Desta MZ, Yu X, Yang Z, Duncan
N. Fibroblast growth factor-23 abolishes 1,25-dihydroxyvitamin D3 - RL, Turner CH. Skeletal phenotype of mice with a null mutation in
enhanced duodenal calcium transport in male mice. Am J Physiol Cav 1.3 L-type calcium channel. J Musculoskelet Neuronal Interact 10:
Endocrinol Metab 302: E903-E913, 2012. 180-187, 2010.
123. Khuituan P, Wongdee K, Jantarajit W, Suntornsaratoon P, Krish- 144. Li Z, Chalazonitis A, Huang YY, Mann JJ, Margolis KG, Yang
namra N, Charoenphandhu N. Fibroblast growth factor-23 negates QM, Kim DO, Cote F, Mallet J, Gershon MD. Essential roles of
1,25(OH)2 D3 -induced intestinal calcium transport by reducing the enteric neuronal serotonin in gastrointestinal motility and the devel-
transcellular and paracellular calcium fluxes. Arch Biochem Biophys opment/survival of enteric dopaminergic neurons. J Neurosci 31:
536: 46-52, 2013. 8998-9009, 2011.

24 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

145. Liao QS, Du Q, Lou J, Xu JY, Xie R. Roles of Na+ /Ca2+ exchanger 170. Nilius B, Prenen J, Vennekens R, Hoenderop JG, Bindels RJ, Droog-
1 in digestive system physiology and pathophysiology. World J Gas- mans G. Modulation of the epithelial calcium channel, ECaC, by intra-
troenterol 25: 287-299, 2019. cellular Ca2+ . Cell Calcium 29: 417-428, 2001.
146. Liao W, Chen H, Jin W, Yang Z, Cao Y, Miao J. Three newly iso- 171. Nilsson B, Delbro D, Friman S, Thune A, Svanvik J. Sympathetic and
lated calcium-chelating peptides from tilapia bone collagen hydrolysate VIP-ergic control of calcium and bicarbonate transport in the feline gall
enhance calcium absorption activity in intestinal Caco-2 cells. J Agric bladder mucosa in vivo. J Auton Nerv Syst 60: 49-55, 1996.
Food Chem 68: 2091-2098, 2020. 172. Noonan WT, Woo AL, Nieman ML, Prasad V, Schultheis PJ, Shull GE,
147. Liel Y, Shany S, Smirnoff P, Schwartz B. Estrogen increases 1,25- Lorenz JN. Blood pressure maintenance in NHE3-deficient mice with
dihydroxyvitamin D receptors expression and bioresponse in the rat transgenic expression of NHE3 in small intestine. Am J Physiol Regul
duodenal mucosa. Endocrinology 140: 280-285, 1999. Integr Comp Physiol 288: R685-R691, 2005.
148. Liu C, Weng H, Chen L, Yang S, Wang H, Debnath G, Guo X, Wu L, 173. Norris SA, Pettifor JM, Gray DA, Buffenstein R. Calcium metabolism
Mohandas N, An X. Impaired intestinal calcium absorption in protein and bone mass in female rabbits during skeletal maturation: Effects of
4.1R-deficient mice due to altered expression of plasma membrane cal- dietary calcium intake. Bone 29: 62-69, 2001.
cium ATPase 1b (PMCA1b). J Biol Chem 288: 11407-11415, 2013. 174. Nunomura W, Denker SP, Barber DL, Takakuwa Y, Gascard P. Charac-
149. Lorieau L, Le Roux L, Gaucheron F, Ligneul A, Hazart E, Dupont D, terization of cytoskeletal protein 4.1R interaction with NHE1 (Na+ /H+
Floury J. Bioaccessibility of four calcium sources in different whey- exchanger isoform 1). Biochem J 446: 427-435, 2012.
based dairy matrices assessed by in vitro digestion. Food Chem 245: 175. Ognik K, Stepniowska A, Cholewinska E, Kozlowski K. The effect of
454-462, 2018. administration of copper nanoparticles to chickens in drinking water on
150. Lutz T, Scharrer E. Effect of short-chain fatty acids on calcium absorp- estimated intestinal absorption of iron, zinc, and calcium. Poult Sci 95:
tion by the rat colon. Exp Physiol 76: 615-618, 1991. 2045-2051, 2016.
151. Mace OJ, Morgan EL, Affleck JA, Lister N, Kellett GL. Calcium 176. Ohta A, Ohtsuki M, Baba S, Adachi T, Sakata T, Sakaguchi E. Calcium
absorption by Cav 1.3 induces terminal web myosin II phosphorylation and magnesium absorption from the colon and rectum are increased in
and apical GLUT2 insertion in rat intestine. J Physiol 580: 605-616, rats fed fructooligosaccharides. J Nutr 125: 2417-2424, 1995.
2007. 177. Olson EB Jr, Deluca HF, Potts JT Jr. Calcitonin inhibition of vitamin
152. Madsen KL, Tavernini MM, Yachimec C, Mendrick DL, Alfonso D-induced intestinal calcium absorption. Endocrinology 90: 151-157,
PJ, Buergin M, Olsen HS, Antonaccio MJ, Thomson AB, Fedorak 1972.
RN. Stanniocalcin: A novel protein regulating calcium and phosphate 178. Omdahl JL, Morris HA, May BK. Hydroxylase enzymes of the vitamin
transport across mammalian intestine. Am J Physiol 274: G96-G102, D pathway: Expression, function, and regulation. Annu Rev Nutr 22:
1998. 139-166, 2002.
153. Maier GW, Kreis ME, Zittel TT, Becker HD. Calcium regulation and 179. Onodera K, Pouokam E, Diener M. STIM1-regulated Ca2+ influx
bone mass loss after total gastrectomy in pigs. Ann Surg 225: 181-192, across the apical and the basolateral membrane in colonic epithelium.
1997. J Membr Biol 246: 271-285, 2013.
154. Mainoya JR. Effects of bovine growth hormone, human placental lac- 180. Orihuela D. Inhibitory effect of aluminium on calcium absorption in
togen and ovine prolactin on intestinal fluid and ion transport in the rat. small intestine of rats with different thyroid hormone status. J Inorg
Endocrinology 96: 1165-1170, 1975. Biochem 103: 1542-1547, 2009.
155. Marcus CS, Lengemann FW. Absorption of Ca45 and Sr85 from solid 181. Pan W, Borovac J, Spicer Z, Hoenderop JG, Bindels RJ, Shull GE,
and liquid food at various levels of the alimentary tract of the rat. J Nutr Doschak MR, Cordat E, Alexander RT. The epithelial sodium/proton
77: 155-160, 1962. exchanger, NHE3, is necessary for renal and intestinal calcium
156. Meyer MB, Watanuki M, Kim S, Shevde NK, Pike JW. The human (re)absorption. Am J Physiol Renal Physiol 302: F943-F956, 2012.
transient receptor potential vanilloid type 6 distal promoter contains 182. Pansu D, Bellaton C, Bronner F. Effect of Ca intake on saturable and
multiple vitamin D receptor binding sites that mediate activation by nonsaturable components of duodenal Ca transport. Am J Physiol 240:
1,25-dihydroxyvitamin D3 in intestinal cells. Mol Endocrinol 20: 1447- G32-G37, 1981.
1461, 2006. 183. Pansu D, Bellaton C, Roche C, Bronner F. Duodenal and ileal calcium
157. Misra M. Effects of hypogonadism on bone metabolism in female absorption in the rat and effects of vitamin D. Am J Physiol 244: G695-
adolescents and young adults. Nat Rev Endocrinol 8: 395-404, 2012. G700, 1983.
158. Montell C. Physiology, phylogeny, and functions of the TRP superfam-
ily of cation channels. Sci STKE 2001: re1, 2001. 184. Pardo VG, de Boland AR. Tyrosine phosphorylation signalling depen-
159. Morgan EL, Mace OJ, Helliwell PA, Affleck J, Kellett GL. A role for dent on 1α,25(OH)2 -vitamin D3 in rat intestinal cells: Effect of ageing.
Cav 1.3 in rat intestinal calcium absorption. Biochem Biophys Res Com- Int J Biochem Cell Biol 36: 489-504, 2004.
mun 312: 487-493, 2003. 185. Peng JB, Chen XZ, Berger UV, Vassilev PM, Tsukaguchi H, Brown
160. Moriya M, Linder MC. Vesicular transport and apotransferrin in intesti- EM, Hediger MA. Molecular cloning and characterization of a channel-
nal iron absorption, as shown in the Caco-2 cell model. Am J Physiol like transporter mediating intestinal calcium absorption. J Biol Chem
Gastrointest Liver Physiol 290: G301-G309, 2006. 274: 22739-22746, 1999.
161. Murgue B, Tsunekawa S, Rosenberg I, deBeaumont M, Podolsky DK. 186. Petersen OH, Fedirko NV. Calcium signalling: Store-operated channel
Identification of a novel variant form of fibroblast growth factor recep- found at last. Curr Biol 11: R520-R523, 2001.
tor 3 (FGFR3 IIIb) in human colonic epithelium. Cancer Res 54: 5206- 187. Petith MM, Wilson HD, Schedl HP. Vitamin D dependence of in vivo
5211, 1994. calcium transport and mucosal calcium binding protein in rat large
162. Muto S, Hata M, Taniguchi J, Tsuruoka S, Moriwaki K, Saitou M, intestine. Gastroenterology 76: 99-104, 1979.
Furuse K, Sasaki H, Fujimura A, Imai M, Kusano E, Tsukita S, 188. Pitcher T, Buffenstein R. Intestinal calcium transport in mole-rats
Furuse M. Claudin-2-deficient mice are defective in the leaky and (Cryptomys damarensis and Heterocephalus glaber) is independent of
cation-selective paracellular permeability properties of renal proximal both genomic and non-genomic vitamin D mediation. Exp Physiol 80:
tubules. Proc Natl Acad Sci USA 107: 8011-8016, 2010. 597-608, 1995.
163. Nakkrasae LI, Thongon N, Thongbunchoo J, Krishnamra N, Charoen- 189. Qu MH, Ji WS, Zhao TK, Fang CY, Mao SM, Gao ZQ. Neurophysiol-
phandhu N. Transepithelial calcium transport in prolactin-exposed ogical mechanisms of bradykinin-evoked mucosal chloride secretion in
intestine-like Caco-2 monolayer after combinatorial knockdown of guinea pig small intestine. World J Gastrointest Pathophysiol 7: 150-
TRPV5, TRPV6 and Cav 1.3. J Physiol Sci 60: 9-17, 2010. 159, 2016.
164. Nellans HN, Goldsmith RS. Transepithelial calcium transport by rat 190. Quarles LD. Endocrine functions of bone in mineral metabolism regu-
cecum: High-efficiency absorptive site. Am J Physiol 240: G424-G431, lation. J Clin Invest 118: 3820-3828, 2008.
1981. 191. Quarles LD. Skeletal secretion of FGF-23 regulates phosphate and vita-
165. Nellans HN, Kimberg DV. Anomalous calcium secretion in rat ileum: min D metabolism. Nat Rev Endocrinol 8: 276-286, 2012.
Role of paracellular pathway. Am J Physiol 236: E473-E481, 1979. 192. Radhakrishnan VM, Gilpatrick MM, Parsa NA, Kiela PR, Ghishan FK.
166. Nemere I. The 1,25D3 -MARRS protein: Contribution to steroid stimu- Expression of Cav 1.3 calcium channel in the human and mouse colon:
lated calcium uptake in chicks and rats. Steroids 70: 455-457, 2005. Posttranscriptional inhibition by IFNgamma. Am J Physiol Gastroin-
167. Nemere I, Leathers V, Norman AW. 1,25-Dihydroxyvitamin D3 - test Liver Physiol 312: G77-G84, 2017.
mediated intestinal calcium transport. Biochemical identification of 193. Reyes-Fernandez PC, Fleet JC. Luminal glucose does not enhance
lysosomes containing calcium and calcium-binding protein (calbindin- active intestinal calcium absorption in mice: Evidence against a role
D28K ). J Biol Chem 261: 16106-16114, 1986. for Cav 1.3 as a mediator of calcium uptake during absorption. Nutr
168. Nemere I, Norman AW. Parathyroid hormone stimulates calcium trans- Res 35: 1009-1015, 2015.
port in perfused duodena from normal chicks: Comparison with the 194. Richter B, Faul C. FGF23 Actions on target tissues-with and without
rapid (transcaltachic) effect of 1,25-dihydroxyvitamin D3 . Endocrinol- Klotho. Front Endocrinol (Lausanne) 9: 189, 2018.
ogy 119: 1406-1408, 1986. 195. Rievaj J, Pan W, Cordat E, Alexander RT. The Na+ /H+ exchanger iso-
169. Nijenhuis T, Hoenderop JG, Bindels RJ. TRPV5 and TRPV6 in Ca2+ form 3 is required for active paracellular and transcellular Ca2+ trans-
(re)absorption: Regulating Ca2+ entry at the gate. Pflugers Arch 451: port across murine cecum. Am J Physiol Gastrointest Liver Physiol 305:
181-192, 2005. G303-G313, 2013.

Volume 11, May 2021 25


Intestinal Calcium Absorption Comprehensive Physiology

196. Rivoira M, Rodriguez V, Lopez MP, Tolosa de Talamoni N. Time 218. Tamura A, Hayashi H, Imasato M, Yamazaki Y, Hagiwara A, Wada M,
dependent changes in the intestinal Ca2+ absorption in rats with type I Noda T, Watanabe M, Suzuki Y, Tsukita S. Loss of claudin-15, but not
diabetes mellitus are associated with alterations in the intestinal redox claudin-2, causes Na+ deficiency and glucose malabsorption in mouse
state. Biochim Biophys Acta 1852: 386-394, 2015. small intestine. Gastroenterology 140: 913-923, 2011.
197. Rivoira MA, Marchionatti AM, Centeno VA, Diaz de Barboza GE, 219. Teerapornpuntakit J, Dorkkam N, Wongdee K, Krishnamra N,
Peralta Lopez ME, Tolosa de Talamoni NG. Sodium deoxycholate Charoenphandhu N. Endurance swimming stimulates transepithelial
inhibits chick duodenal calcium absorption through oxidative stress calcium transport and alters the expression of genes related to calcium
and apoptosis. Comp Biochem Physiol A Mol Integr Physiol 162: absorption in the intestine of rats. Am J Physiol Endocrinol Metab
397-405, 2012. 296: E775-E786, 2009.
198. Roche C, Bellaton C, Pansu D, Miller A 3rd, Bronner F. Localization of 220. Thammayon N, Wongdee K, Lertsuwan K, Suntornsaratoon P, Thong-
vitamin D-dependent active Ca2+ transport in rat duodenum and rela- bunchoo J, Krishnamra N, Charoenphandhu N. Na+ /H+ exchanger 3
tion to CaBP. Am J Physiol 251: G314-G320, 1986. inhibitor diminishes the amino-acid-enhanced transepithelial calcium
199. Rodrat M, Wongdee K, Panupinthu N, Thongbunchoo J, Teeraporn- transport across the rat duodenum. Amino Acids 49: 725-734, 2017.
puntakit J, Krishnamra N, Charoenphandhu N. Prolonged exposure to 221. Thongon N, Nakkrasae LI, Thongbunchoo J, Krishnamra N, Charoen-
1,25(OH)2 D3 and high ionized calcium induces FGF-23 production phandhu N. Enhancement of calcium transport in Caco-2 monolayer
in intestinal epithelium-like Caco-2 monolayer: A local negative feed- through PKCzeta-dependent Cav 1.3-mediated transcellular and recti-
back for preventing excessive calcium transport. Arch Biochem Biophys fying paracellular pathways by prolactin. Am J Physiol Cell Physiol
640: 10-16, 2018. 296: C1373-C1382, 2009.
200. Rodríguez V, Rivoira M, Guizzardi S, Tolosa de Talamoni N. Naringin 222. Thwaites DT, Anderson CM. H+ -coupled nutrient, micronutrient and
prevents the inhibition of intestinal Ca2+ absorption induced by a fruc- drug transporters in the mammalian small intestine. Exp Physiol 92:
tose rich diet. Arch Biochem Biophys 636: 1-10, 2017. 603-619, 2007.
201. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton 223. Timmermans JA, Kaune R, Bindels RJ, van Os CH. Quantification of
SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Ca2+ -ATPases in porcine duodenum. Effects of 1,25(OH)2 D3
Mayne ST, Rosen CJ, Shapses SA. The 2011 report on dietary reference deficiency. Biochim Biophys Acta 1065: 177-184, 1991.
intakes for calcium and vitamin D from the Institute of Medicine: What 224. Toverud SU, Dostal LA. Calcium absorption during development:
clinicians need to know. J Clin Endocrinol Metab 96: 53-58, 2011. Experimental studies of the rat small intestine. J Pediatr Gastroenterol
202. Ryan ZC, Craig TA, Filoteo AG, Westendorf JJ, Cartwright EJ, Ney- Nutr 5: 688-695, 1986.
ses L, Strehler EE, Kumar R. Deletion of the intestinal plasma mem- 225. Turner JR, Black ED. NHE3-dependent cytoplasmic alkalinization
brane calcium pump, isoform 1, Atp2b1, in mice is associated with is triggered by Na+ -glucose cotransport in intestinal epithelia. Am J
Physiol Cell Physiol 281: C1533-C1541, 2001.
decreased bone mineral density and impaired responsiveness to 1, 25- 226. Turner JR, Rill BK, Carlson SL, Carnes D, Kerner R, Mrsny RJ, Madara
dihydroxyvitamin D3 . Biochem Biophys Res Commun 467: 152-156, JL. Physiological regulation of epithelial tight junctions is associated
2015. with myosin light-chain phosphorylation. Am J Physiol 273: C1378-
203. Sakaki T, Sawada N, Komai K, Shiozawa S, Yamada S, Yamamoto K, C1385, 1997.
Ohyama Y, Inouye K. Dual metabolic pathway of 25-hydroxyvitamin 227. Van Cromphaut SJ, Rummens K, Stockmans I, Van Herck E, Dijcks
D3 catalyzed by human CYP24. Eur J Biochem 267: 6158-6165, 2000. FA, Ederveen AG, Carmeliet P, Verhaeghe J, Bouillon R, Carmeliet
204. Sampson HW, Krawitt EL. A morphometric investigation of the duode- G. Intestinal calcium transporter genes are upregulated by estrogens
nal mucosa of normal, vitamin D-deficient, and vitamin D-replete rats. and the reproductive cycle through vitamin D receptor-independent
Calcif Tissue Res 21: 213-218, 1976. mechanisms. J Bone Miner Res 18: 1725-1736, 2003.
205. Schinke T, Schilling AF, Baranowsky A, Seitz S, Marshall RP, Linn 228. Van Itallie CM, Fanning AS, Anderson JM. Reversal of charge selectiv-
T, Blaeker M, Huebner AK, Schulz A, Simon R, Gebauer M, Priemel ity in cation or anion-selective epithelial lines by expression of different
M, Kornak U, Perkovic S, Barvencik F, Beil FT, Del Fattore A, Frat- claudins. Am J Physiol Renal Physiol 285: F1078-F1084, 2003.
tini A, Streichert T, Pueschel K, Villa A, Debatin KM, Rueger JM, 229. Vervloet M. Renal and extrarenal effects of fibroblast growth factor 23.
Teti A, Zustin J, Sauter G, Amling M. Impaired gastric acidification Nat Rev Nephrol 15: 109-120, 2019.
negatively affects calcium homeostasis and bone mass. Nat Med 15: 230. Wallace TC, Reider C, Fulgoni VL 3rd. Calcium and vitamin D dispari-
674-681, 2009. ties are related to gender, age, race, household income level, and weight
206. Schnetkamp PP, Basu DK, Szerencsei RT. Na+ -Ca2+ exchange in classification but not vegetarian status in the United States: Analysis
bovine rod outer segments requires and transports K+ . Am J Physiol of the NHANES 2001-2008 data set. J Am Coll Nutr 32: 321-330, 2013.
257: C153-C157, 1989. 231. Walling MW, Brasitus TA, Kimberg DV. Effects of calcitonin and sub-
207. Schwaller B. Cytosolic Ca2+ buffers. Cold Spring Harb Perspect Biol stance P on the transport of Ca, Na and Cl across rat ileum in vitro.
2: a004051, 2010. Gastroenterology 73: 89-94, 1977.
208. Schwerdtfeger LA, Tobet SA. Vasoactive intestinal peptide regulates 232. Walling MW, Kimberg DV. Active secretion of calcium by adult rat
ileal goblet cell production in mice. Physiol Rep 8: e14363, 2020. ileum and jejunum in vitro. Am J Physiol 225: 415-422, 1973.
209. Sharma RV, Butters CA, McEldoon JP, Bhalla RC. Characterization 233. Wang L, Klopot A, Freund JN, Dowling LN, Krasinski SD, Fleet JC.
of Ca2+ uptake in plasma membrane vesicles isolated from guinea pig Control of differentiation-induced calbindin-D9k gene expression in
ileum smooth muscle. Cell Calcium 8: 65-77, 1987. Caco-2 cells by cdx-2 and HNF-1α. Am J Physiol Gastrointest Liver
210. Shekhawat PB, Pokharkar VB. Understanding peroral absorption: Reg- Physiol 287: G943-G953, 2004.
ulatory aspects and contemporary approaches to tackling solubility and 234. Wasserman RH. Vitamin D and the dual processes of intestinal calcium
permeability hurdles. Acta Pharm Sin B 7: 260-280, 2017. absorption. J Nutr 134: 3137-3139, 2004.
211. Shimada T, Yamazaki Y, Takahashi M, Hasegawa H, Urakawa I, 235. Weaver CM, Gordon CM, Janz KF, Kalkwarf HJ, Lappe JM, Lewis
Oshima T, Ono K, Kakitani M, Tomizuka K, Fujita T, Fukumoto R, O’Karma M, Wallace TC, Zemel BS. The National Osteoporosis
S, Yamashita T. Vitamin D receptor-independent FGF23 actions in Foundation’s position statement on peak bone mass development and
regulating phosphate and vitamin D metabolism. Am J Physiol Renal lifestyle factors: A systematic review and implementation recommen-
Physiol 289: F1088-F1095, 2005. dations. Osteoporos Int 27: 1281-1386, 2016.
212. Song Y, Fleet JC. 1,25 dihydroxycholecalciferol-mediated calcium 236. Whisner CM, Martin BR, Nakatsu CH, Story JA, MacDonald-Clarke
absorption and gene expression are higher in female than in male mice. CJ, McCabe LD, McCabe GP, Weaver CM. Soluble corn fiber increases
J Nutr 134: 1857-1861, 2004. calcium absorption associated with shifts in the gut microbiome: A ran-
213. Song Y, Kato S, Fleet JC. Vitamin D receptor (VDR) knockout mice domized dose-response trial in free-living pubertal females. J Nutr 146:
reveal VDR-independent regulation of intestinal calcium absorption 1298-1306, 2016.
and ECaC2 and calbindin D9k mRNA. J Nutr 133: 374-380, 2003. 237. Wongdee K, Charoenphandhu N. Regulation of epithelial calcium
214. Song Y, Peng X, Porta A, Takanaga H, Peng JB, Hediger MA, Fleet transport by prolactin: From fish to mammals. Gen Comp Endocrinol
JC, Christakos S. Calcium transporter 1 and epithelial calcium channel 181: 235-240, 2013.
messenger ribonucleic acid are differentially regulated by 1,25 dihy- 238. Wongdee K, Charoenphandhu N. Vitamin D-enhanced duodenal cal-
droxyvitamin D3 in the intestine and kidney of mice. Endocrinology cium transport. Vitam Horm 98: 407-440, 2015.
144: 3885-3894, 2003. 239. Wongdee K, Rodrat M, Keadsai C, Jantarajit W, Teerapornpuntakit J,
215. Sterling TM, Nemere I. Calcium uptake and membrane trafficking in Thongbunchoo J, Charoenphandhu N. Activation of calcium-sensing
response to PTH or 25(OH)D3 in polarized intestinal epithelial cells. receptor by allosteric agonists cinacalcet and AC-265347 abolishes the
Steroids 72: 151-157, 2007. 1,25(OH)2 D3 -induced Ca2+ transport: Evidence that explains how the
216. Swaminathan R, Ker J, Care D. Calcitonin and intestinal calcium intestine prevents excessive Ca2+ absorption. Arch Biochem Biophys
absorption. J Endocrinol 61: 83-94, 1974. 657: 15-22, 2018.
217. Takaishi S, Sawada M, Morita Y, Seno H, Fukuzawa H, Chiba T. 240. Wongdee K, Rodrat M, Teerapornpuntakit J, Krishnamra N, Charoen-
Identification of a novel alternative splicing of human FGF receptor phandhu N. Factors inhibiting intestinal calcium absorption: Hormones
4: Soluble-form splice variant expressed in human gastrointestinal and luminal factors that prevent excessive calcium uptake. J Physiol Sci
epithelial cells. Biochem Biophys Res Commun 267: 658-662, 2000. 69: 683-696, 2019.

26 Volume 11, May 2021


Comprehensive Physiology Intestinal Calcium Absorption

241. Wongdee K, Teerapornpuntakit J, Siangpro C, Chaipai S, Charoen- 249. Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, Confavreux C, Kle-
phandhu N. Duodenal villous hypertrophy and upregulation of menhagen KC, Tanaka KF, Gingrich JA, Guo XE, Tecott LH, Mann
claudin-15 protein expression in lactating rats. J Mol Histol 44: JJ, Hen R, Horvath TL, Karsenty G. A serotonin-dependent mecha-
103-109, 2013. nism explains the leptin regulation of bone mass, appetite, and energy
242. Wongdee K, Teerapornpuntakit J, Sripong C, Longkunan A, Chankam- expenditure. Cell 138: 976-989, 2009.
ngoen W, Keadsai C, Kraidith K, Krishnamra N, Charoenphandhu N. 250. Yang H, Ahn C, Shin EK, Lee JS, An BS, Jeung EB. NCKX3 was
Intestinal mucosal changes and upregulated calcium transporter and compensated by calcium transporting genes and bone resorption in a
FGF-23 expression during lactation: Contribution of lactogenic hor- NCKX3 KO mouse model. Mol Cell Endocrinol 454: 93-102, 2017.
mone prolactin. Arch Biochem Biophys 590: 109-117, 2016. 251. Yang JH, Hou JF, Farquharson C, Zhou ZL, Deng YF, Wang L, Yu Y.
243. Wood RJ, Serfaty-Lacrosniere C. Gastric acidity, atrophic gastritis, and Localisation and expression of TRPV6 in all intestinal segments and
calcium absorption. Nutr Rev 50: 33-40, 1992. kidney of laying hens. Br Poult Sci 52: 507-516, 2011.
244. Wood RJ, Tchack L, Angelo G, Pratt RE, Sonna LA. DNA microar- 252. Ye L, Morse LR, Zhang L, Sasaki H, Mills JC, Odgren PR, Sibbel G,
ray analysis of vitamin D-induced gene expression in a human colon Stanley JR, Wong G, Zamarioli A, Battaglino RA. Osteopetrorickets
carcinoma cell line. Physiol Genomics 17: 122-129, 2004. due to Snx10 deficiency in mice results from both failed osteoclast
245. Wu P, Xie F, Xue M, Xu X, He S, Lin M, Bai L. Advanced oxidation activity and loss of gastric acid-dependent calcium absorption. PLoS
protein products decrease the expression of calcium transport channels Genet 11: e1005057, 2015.
in small intestinal epithelium via the p44/42 MAPK signaling pathway. 253. Zamparelli C, Macquaide N, Colotti G, Verzili D, Seidler T, Smith GL,
Eur J Cell Biol 94: 190-203, 2015. Chiancone E. Activation of the cardiac Na+ -Ca2+ exchanger by sorcin
246. Xiang J, Guo R, Wan C, Wu L, Yang S, Guo D. Regulation of intestinal via the interaction of the respective Ca2+ -binding domains. J Mol Cell
epithelial calcium transport proteins by stanniocalcin-1 in Caco2 cells. Cardiol 49: 132-141, 2010.
Int J Mol Sci 17: 1095, 2016. 254. Zhang Q, Wastney ME, Rosen CJ, Beamer WG, Weaver CM. Insulin-
247. Xiao Y, Cui J, Shi YH, Sun J, Wang ZP, Le GW. Effects of duode- like growth factor-1 increases bone calcium accumulation only during
nal redox status on calcium absorption and related genes expression in rapid growth in female rats. J Nutr 141: 2010-2016, 2011.
high-fat diet-fed mice. Nutrition 26: 1188-1194, 2010. 255. Zhu MX, Evans AM, Ma J, Parrington J, Galione A. Two-pore channels
248. Xue Y, Fleet JC. Intestinal vitamin D receptor is required for normal for integrative Ca signaling. Commun Integr Biol 3: 12-17, 2010.
calcium and bone metabolism in mice. Gastroenterology 136: 1317-
1327, 2009.

Volume 11, May 2021 27

You might also like