<<

The Journal of Physiological Sciences (2019) 69:683–696 https://doi.org/10.1007/s12576-019-00688-3

REVIEW

Factors inhibiting intestinal absorption: hormones and luminal factors that prevent excessive calcium uptake

Kannikar Wongdee1,2 · Mayuree Rodrat2,3 · Jarinthorn Teerapornpuntakit2,4 · Nateetip Krishnamra2,3 · Narattaphol Charoenphandhu2,3,5,6

Received: 4 March 2019 / Accepted: 9 June 2019 / Published online: 20 June 2019 © The Physiological Society of Japan and Springer Japan KK, part of Springer Nature 2019

Abstract

Besides the two canonical calciotropic hormones, namely and 1,25-dihydroxyvitamin D [1,25(OH)2D3], there are several other endocrine and paracrine factors, such as prolactin, estrogen, and -like growth factor that have been known to directly stimulate intestinal calcium absorption. Generally, to maintain an optimal plasma calcium level, these positive regulators enhance calcium absorption, which is indirectly counterbalanced by a long-loop mechanism, i.e., through calcium-sensing receptor in the parathyroid chief cells. However, several lines of recent evidence have revealed the presence of calcium absorption inhibitors present in the intestinal lumen and extracellular fuid in close vicinity to , which could also directly compromise calcium absorption. For example, luminal iron, circulating fbroblast growth factor (FGF)-23, and stanniocalcin can decrease calcium absorption, thereby preventing excessive calcium uptake under certain conditions. Interestingly, the intestinal epithelial cells themselves could lower their rate of calcium uptake after exposure to high luminal calcium concentration, suggesting a presence of an ultra-short negative feedback loop independent of systemic hormones. The existence of neural regulation is also plausible but this requires more supporting evidence. In the present review, we elaborate on the physiological signifcance of these negative feedback regulators of calcium absorption, and provide evidence to show how our body can efciently restrict a food of calcium infux in order to maintain calcium .

Keywords Calcium absorption · Calcium-sensing receptor (CaSR) · Fibroblast growth factor (FGF)-23 · Iron transport · Parathyroid hormone (PTH) ·

Introduction

Besides being the major inorganic component in , cal- cium is an essential element that has roles in several func- * Narattaphol Charoenphandhu tions, e.g., neurotransmitter release, , [email protected] blood , and intracellular signal transduction. Ninety-nine percent of body calcium is stored in bone in 1 Faculty of Allied Health Sciences, Burapha University, the form of crystal ­[Ca (PO ) (OH) ], while Chonburi, Thailand 10 4 6 2 the remaining 1% is distributed in the plasma, interstitium, 2 Center of Calcium and Bone Research (COCAB), Faculty intracellular fuid, and within the cells in mitochondria and of Science, Mahidol University, Bangkok, Thailand . The intracellular calcium is main- 3 Department of , Faculty of Science, Mahidol tained at concentration as low as 0.1 µM, which is lower than University, Rama VI Road, Bangkok 10400, Thailand the extracellular calcium concentration (free ionized cal- 4 Department of Physiology, Faculty of Medical Science, cium of 1.1–1.3 mM) by ~ 1000-fold. An excess of calcium Naresuan University, Phitsanulok, Thailand in either the intracellular or extracellular fuid is extremely 5 Institute of Molecular Biosciences, Mahidol University, dangerous. Because free ionized calcium is toxic to the cell, Nakhon Pathom, Thailand a prolonged rise in the intracellular calcium can lead to cell 6 The Academy of Science, The Royal Society of Thailand, death by activating various enzymes, such as protein kinase Dusit, Bangkok, Thailand

Vol.:(0123456789)1 3 684 The Journal of Physiological Sciences (2019) 69:683–696

C, caspases, phospholipases, proteases, and endonucleases in the ingested foods, e.g., iron, phytate, oxalate, and as well as apoptotic process [59]. Therefore, the intestine, tannin (Fig. 1), can inhibit calcium absorption, the which is the only site for calcium entry into the body, must physiological significance of these substances is not have mechanisms to regulate calcium uptake. Besides, the fully understood. The present article thus focuses on intestinal epithelial cells themselves also need to tightly the regulatory roles of these intestine- and nutrient- control their intracellular calcium concentration to prevent derived factors and their feedback mechanisms in the superfuous calcium uptake, which may damage themselves suppression of intestinal calcium absorption. It is gen- as well as other cells in the body [57, 59, 106]. erally accepted that both stimulatory and inhibitory Since the intestine is the only route for calcium regulators of calcium absorption are humoral factors uptake, it is subjected to local and systemic regulation, with autocrine, paracrine, and endocrine functions, which helps protect against inadequate as well as exces- as depicted in Fig. 1. Apparently, the possible role of sive absorption of calcium. Both stimuli and inhibitors neural regulation may also exist and may be analogous of calcium absorption have been described, with the to the splanchnic nerve regulation of calcium uptake plasma calcium-PTH-vitamin D feedback loop as the across the gallbladder mucosa [73]. The overview of most prominent feedback regulation. Although local factors controlling intestinal calcium transport is sum- hormones, secretory factors, and some components marized in Table 1.

Fig. 1 Intrinsic (humoral and neural) and extrinsic (luminal) regulators of intestinal calcium absorption (please see text for details)

1 3 The Journal of Physiological Sciences (2019) 69:683–696 685

Table 1 Summary of possible factors afecting calcium absorption across the intestinal Factors Transcellular Paracellular Net ­Ca2+ absorption References

Extrinsic/luminal Ca2+ (long-term low luminal Increase expression of TRPV5/6, N/A ↑ [4, 8, 10] 2+ ­Ca or low calcium diet) CaBP-D9k, NCX1, ­PMCA1b, Increase activity of ­PMCA1b and NCX1 Ca2+ (prolong exposure to high Possibly by increase FGF-23 N/A ↓ [84] luminal Ca2+) expression ­(Ca2+ > 30 mM) and inhibit transcellular transport by unknown mechanism Iron N/A N/A ↓ [58]

Glucose/galactose (SGLT1 Decrease ­Cav1.3 activity Increase solvent drag-induced ↑ [52, 91] substrates) paracellular calcium fow Amino acids N/A Increase NHE activity ↑ [96] Increase nutrient-induced paracel- lular calcium fow Fructose Decrease expression of TRPV6 and N/A ↓ [27, 28] CaBP-D9k Intrinsic Hormone

1,25(OH)2D3 Increase expression of TRPV5/6, Stimulate nongenomic signaling ↑ [32, 40] CaBP-D9k, NCX1, ­PMCA1b pathways involving PI3K, PKC, and MEK to enhance calcium transport Increase expression of claudin-2, 12 PTH Increase calcium uptake N/A ↑ [71] (Unknown mechanism)

FGF-23 Decrease 1,25(OH)2D3-enhanced Decrease 1,25(OH)2D3-enhanced ↓ [53, 54] expressions of TRPV5/6, CaBP- paracellular calcium transport D9k and calcium permeability Paracrine FGF-23 Increase CaSR activity N/A ↓ [84] Stanniocalcin-1 Decrease expression of TRPV5/6, N/A ↓ [107] CaBP-D9k No change in the expression of NCX1, ­PMCA1b Neural (ENS) VIP N/A N/A ↑ [73] (Indirect evidence)

CaBP-D9k -D9k, CaSR calcium-sensing receptor, Cav1.3 L-type voltage-gated calcium channel, FGF-23 fbroblast growth factor 23, ENS enteric nervous system, N/A not available, NCX1 calcium exchanger 1, TJ tight junction, VIP vasoactive intestinal peptide

Mechanisms of intestinal calcium absorption humans and rodents, the vitamin D-dependent transcellu- in mammals lar calcium transport is predominant in the proximal small intestine, particularly the , and is of importance In mammals, calcium is absorbed through the intestinal in low-calcium intake conditions [3, 7, 103]. A regular diet epithelial cells via two major pathways, i.e., transcellular without dairy products is generally considered as a low-nor- and paracellular pathways [24]. The relative contribution of mal calcium diet, which requires an active calcium transport transcellular and paracellular calcium absorption depends mechanism. on several factors including the amount of calcium intake, The uphill transcellular is composed of solubility and chyme alkalinity, bioavailability, and segment three steps, i.e., (1) apical vitamin D-dependent calcium transit time [52]. Although both calcium transport mecha- entry via the transient receptor potential cation channel, nisms are found along the entire length of the intestine in subfamily V, member 6 (TRPV6), and to a lesser extent

1 3 686 The Journal of Physiological Sciences (2019) 69:683–696

TRPV5, (2) cytoplasmic calcium translocation (bound to luminal calcium > 5 mM), calcium prefers to difuse into calbindin-D9k), and (3) basolateral calcium extrusion via the body. On the other hand, an extremely low calcium con- 2+ + plasma membrane ­Ca -ATPase 1b ­(PMCA1b) and ­Na / centration in the lumen may aggravate calcium secretion, Ca2+-exchanger 1 (NCX1) [13, 24]. Regarding calcium which probably results in net calcium loss during claudin-2 entry into the enterocytes, besides TRPV5/6 channels, the overexpression. However, further experiment is required to apical voltage-dependent L-type calcium channel subtype confrm the latter hypothesis. 1.3 ­(Cav1.3) is also involved in calcium uptake, particularly in the presence of depolarizing nutrients in the lumen, such as glucose, galactose, and some amino acids [52, 68, 96]. Possible feedback loops for intestinal Once calcium enters the cells, it is bufered by the cyto- calcium absorption plasmic calcium-binding protein, calbindin-D9k [24, 86]. Other calcium-binding proteins such as may also To maintain calcium homeostasis, the amount of calcium contribute, albeit to a lesser extent, to the intracellular cal- absorbed by the intestine is fne-tuned to match the body cium translocation [24]. Finally, calcium is extruded across calcium requirement by several factors, including hor- the basolateral membrane through several transporters, e.g., mones and luminal nutrients [57]. Since calcium absorp- + + + 2+ PMCA­ 1b, NCX1 coupled to ­Na /K -ATPase, and ­Na /Ca / tion is relatively low under normal conditions (~ 25–35% K+-exchanger (NCKX), the latter of which extrudes one ­K+ of total calcium intake) as compared with other minerals and one ­Ca2+ with infux of four sodium ions to maintain (such as magnesium and ), most regulatory low intracellular calcium concentration [52, 57, 88]. factors are stimulators for enhancing calcium absorption. The second major route is the paracellular pathway, which However, little is known regarding how calcium absorption allows calcium to traverse the lateral intercellular (paracel- is regulated when faced with excessive calcium intake and lular) space. Calcium transport via this route can be both a potential risk of calcium toxicity. Normally, the level of cellular energy-dependent active or gradient-dependent pas- plasma calcium is considered as a component of the nega- sive transport. The paracellular active transport is generally tive feedback regulation to suppress parathyroid hormone mediated by solvent drag, which is induced by sodium efux (PTH) release and production of 1,25-dihydroxycholecal- + + by ­Na /K -ATPase that creates hyperosmotic environment ciferol [1,25(OH)2D3], thereby slowing down the intestinal in the paracellular space. This subsequently induces water calcium absorption as discussed below. difusion in a lumen-to-plasma direction, dragging along with it small permeable molecules, including calcium [6, Roles of hormones 24, 52]. Regarding the paracellular passive calcium transport The classical hormones involved in the positive regu- driven by the concentration gradient of calcium between the lation of intestinal calcium absorption are PTH and lumen and plasma, it occurs more readily in the condition of 1,25(OH)2D3, the latter of which is synthesized in the high-calcium intake—generally considered signifcant when by the renal proximal tubule and which directly luminal calcium > 5 mM—and can occur throughout the enhances the intestinal calcium absorption through active entire length of the intestine [24, 30]. Paracellular transport transcellular and passive paracellular pathways in both is also determined notably by the size- and charge-selective genomic and non-genomic fashion (for review, please properties of the tight junction, which creates specifc bar- see Ref. [57]). For the active transcellular pathway, riers for ions, such as calcium, sodium, and chloride [112]. 1,25(OH)2D3 exerts genomic actions by binding to vita- Claudins is the major family of the tight junction-associated min D receptor (VDR), thus increasing the expression proteins responsible for the paracellular size- and charge- of calcium transport machinery, i.e., TRPV6, calbindin- selective properties. Claudin-2, -12, and -15 in particular D9k, ­PMCA1b, and NCX1 [11, 20, 24, 104]. In addition, have been shown to have signifcant roles in the regulation 1,25(OH)2D3 probably exerts a non-genomic action of paracellular calcium transport across the intestinal epi- to rapidly enhance calcium transport by binding to the thelia [14, 24, 32]. Claudin-2 has negatively charged amino plasma membrane receptor 1,25(OH)2D3-MARRS (mem- acids (e.g., aspartate) in the extracellular domains, which brane-associated, rapid response steroid-binding) pro- protrude into the paracellular space to form a tight junction tein [72]. In brief, the non-genomic action occurs when pore. With these negative charges, claudin-2 is permeable to 1,25(OH)2D3 binds to the membrane-bound 1,25(OH)2D3- cations [21, 110]. In the presence of upregulated claudin-2 MARRS instead of the intracellular/nuclear VDR, which expression, the luminal calcium is expected to easily difuse is a transcription factor [29]. This membrane-bound across the paracellular space in both lumen-to-plasma and receptor can activate certain second messengers, includ- plasma-to-lumen directions. When the luminal concentra- ing phospholipase A2 (PLA2) and protein kinase C tion is greater than the plasma ionized calcium level (e.g., (PKC) [26, 87]. Regarding the paracellular absorption,

1 3 The Journal of Physiological Sciences (2019) 69:683–696 687

1,25(OH)2D3 enhances calcium transport through both Roles of calcium‑sensing receptor (CaSR) passive diffusion and solvent drag mechanisms, in part by modifying the charge- and size-selective properties Parathyroid chief cells, renal tubular cells, and of the tight junction proteins, particularly claudin-2 and are known to express CaSR for sensing pericellular calcium. -12 [32]. Normally, CaSR also senses other positively charged ions PTH is widely known as a hypercalcemic and (e.g., ­Mg2+, ­Cd2+, ­Ba2+, ­La3+ and ­Gd3+), cationic molecules hypophosphatemic hormone upstream to 1,25(OH)2D3. It (e.g., γ-glutathione), and amino acids and polyamines (e.g., elevates the plasma calcium levels by enhancing osteo- tryptophan, l-phenylalanine, spermidine, and spermine) [12, clastic bone resorption, renal calcium reabsorption, and 22]. Thus, an increase in plasma calcium level is detect- increasing intestinal calcium uptake [37]. However, the able by parathyroid chief cells, resulting in an inhibition of role of PTH on intestinal calcium absorption is normally PTH release and decrease in calcium absorption (Fig. 2a). indirect through renal 1,25(OH)2D3 production. Specif- However, up until now, the role of CaSR in the intestinal cally, PTH stimulates the transcription of the CYP27B1 epithelial cells has been elusive, and whether it can directly gene for renal enzyme 1α-hydroxylase, and suppresses modulate intestinal calcium absorption independent of PTH CYP24A1 for renal 24-hydroxylase, an enzyme respon- is largely unknown. Indeed, CaSR may potentially play a sible for 1,25(OH)2D3 degradation [111], thus elevating role in local regulation of intestinal calcium absorption the plasma level of 1,25(OH)2D3. For the direct action of since it is abundantly expressed in both apical and baso- PTH on the intestinal calcium absorption, it was reported lateral membranes of the enterocytes [17]. However, most that N-terminal fragment 1–34 of PTH could stimulate studies on the intestinal CaSR have been performed in the calcium transport in perfused duodenal loops from nor- large intestine rather than the small intestine where calcium mal chicks transcaltachia (rapid, non-genomic)—a rapid absorption is most active [5, 18, 41]. Direct activation of hormonal stimulation of intestinal calcium absorption [71, this receptor in the apical membrane can increase calcium 75]. Moreover, the presence of PTH receptor 1 (PTH1R) in absorption in the large intestine [18, 50], while reducing the basolateral membrane of rat intestinal epithelial cells calcium uptake in the small intestinal-like Caco-2 monolayer suggests possible direct action of PTH in the intestine. (Fig. 2b). Rodrat et al. have reported that CaSR probably However, this study did not demonstrate the regulation of sensed high apical calcium which in turn increased FGF-23 PTH on intestinal calcium absorption [36]. Indeed, direct expression to suppress calcium transport [84]. The roles of exposure to PTH not only increases calcium absorption intestinal CaSR were also studied in Casr intestinal-specifc but also promotes transport of other ions, e.g., potassium, knockout mice. Although Casr-defcient mice manifested an chloride, and bicarbonate, across the intestinal epithelium. impaired intestinal integrity, altered composition of the gut There was evidence that PTH regulation of bicarbonate microbiota and infammation [19, 77], the function related secretion is not an adaptive mechanism but is more like to the regulation of calcium transport was not determined. transcaltachia (rapid, non-genomic). These studies used Similarly, the renal tubular epithelia also use local CaSR Caco-2 cells, which may refect a colonic phenotype unless to modulate calcium reabsorption. Activation of CaSR on the results are confrmed in small intestine [16, 61]. Up till the luminal side of renal proximal tubular cells is capable of now, the physiological signifcance of the PTH-regulated stimulating ­Na+/H+-exchanger 3 (NHE3), thereby enhanc- ion transport in the gut is not clearly known. ing the paracellular calcium transport via a solvent drag- Another hormone that was recently shown to have a mediated mechanism (Fig. 2c) [101]. On the other hand, the calcium-regulating function is the bone-derived fbroblast basolateral CaSR plays a role in the inhibition of the para- growth factor (FGF)-23 [53, 54, 79]. FGF-23 was gener- cellular calcium reabsorption in the thick ascending limb of ally recognized as a phosphaturic hormone produced and Henle’s loop independent of PTH action [98]. secreted from osteocytes and osteoblasts. Its secretion is enhanced by high serum , 1,25(OH)2D3, and perhaps calcium [64, 79, 100]. The systemic actions of Stimulation of intestinal calcium absorption FGF-23 are to increase urinary phosphate excretion and by luminal nutrients suppress 1,25(OH)2D3 production. FGF-23 suppresses CYP27B1 gene expression and stimulates CYP24A1 gene It has also been reported that a number of nutrients, includ- expression, leading to a decreased circulating 1,25(OH)2D3 ing amino acids, oligosaccharides, disaccharides, and mono- [78, 89]. Furthermore, FGF-23 has recently been shown to saccharides (glucose and galactose), can stimulate calcium act directly on the intestine to abolish the 1,25(OH)2D3- absorption [91, 92, 96, 109], however, it remains largely stimulated intestinal calcium absorption [53, 54]. The cel- unknown how this process is counterbalanced. Generally, lular mechanism of FGF-23 as a local inhibitor of calcium there are at least three possible mechanisms to explain how absorption is elaborated in “intrinsic inhibitory factors”. the enterocytes enhance calcium transport in response to

1 3 688 The Journal of Physiological Sciences (2019) 69:683–696

Fig. 2 Responses of calcium- A sensing receptor (CaSR) to extracellular calcium in a para- chief cells, b intestinal epithelial cells, and c renal tubular epithelial cells (please see text for details)

B

C

luminal nutrients, i.e., nutrient-induced paracellular calcium Therefore, this calcium transport is referred to as the sol- fow, ­Cav1.3 activation, and NHE3 activation. vent drag-induced calcium absorption [52, 91]. In addi- tion, the fermented dairy products and incomplete digested Solvent drag and nutrient‑induced paracellular carbohydrate, e.g., short-chain fatty acid, oligosaccharides, calcium fow and polysaccharides, can also enhance calcium absorption in the small and large intestine in rats by increasing tight Glucose and/or galactose enter the intestinal cells via api- junction permeability to calcium [80, 108]. Although the cal sodium-dependent glucose transporter-1 (SGLT1), solvent drag was not directly determined in most studies, after which sodium is pumped out into the paracellular the solvent drag-induced paracellular calcium transport space by ­Na+/K+-ATPase especially those at the lateral should contribute to the process since the uptake of these membrane. As explained earlier, the increase in the para- small organic molecules is often sodium-dependent. How- cellular osmolality together with relative permeability ever, the direct efect of microbiota-derived organic mol- of tight junction to water drives paracellular water fow ecules on the transcellular calcium transporters cannot be from luminal to the blood side, bringing calcium with it. ruled out and requires future investigation.

1 3 The Journal of Physiological Sciences (2019) 69:683–696 689

Cav1.3 activation Counterregulatory and inhibitory factors for intestinal calcium absorption Under normal conditions, the resting membrane potential of the intestinal epithelial cells is approximately − 47 mV. Although the body has a calcium-lowering hormone, calci- Infux of sodium together with glucose can depolarize the tonin, and the kidney to prevent hypercalcemia through sup- apical membrane, thereby activating ­Cav1.3 for transcellular pression of bone resorption and enhancement of urinary cal- calcium uptake particularly when the transmembrane poten- cium excretion, respectively. These mechanisms take place tial reaches the ­Cav1.3 threshold of approximately − 20 mV only after calcium has already been taken up into the body [52]. Lactose—the major disaccharide in milk—also stimu- and raise its plasma concentration. It is tempting to postulate lates intestinal calcium absorption [1], but the mechanism that the intestine as a front line to encounter calcium has a remains unclear. It is possible that the efect of lactose is monitoring mechanism to prevent fooding of calcium into due to the elevated concentrations of glucose and galactose the enterocytes. Several lines of evidence have suggested following lactose digestion by lactase in the intestinal lumen. the presence of counterregulatory or inhibitory factors for Both glucose and galactose are substrates of SGLT1, which limiting calcium absorption, e.g., hormones, luminal ions, can induce sodium entry, thereby depolarizing the apical or byproduct of nutrient digestion [9, 60]. In this review, plasma membrane and activating apical calcium channel, we subdivide these counterregulatory factors into two major ­Cav1.3, for calcium to difuse down its concentration gradi- groups, i.e., factors in the lumen (extrinsic) and those in the ent into the cell [52]. plasma (intrinsic), as shown in Fig. 1.

NHE3 activation Extrinsic inhibitory factors

Although some amino acids are transported by sodium- The extrinsic or luminal factors are defned as luminal con- dependent amino acid transporter (e.g., B­ 0AT1) similar to tent-derived factors that may be either nutrient or non-nutri- monosaccharide absorption by SGLT1 [47], oligopeptides ent substances with inhibitory efect on calcium absorption, are taken up into the enterocytes by H­ +-coupled peptide e.g., high luminal calcium concentration, iron, sugars, and transporter (PepT1) [102]. It is possible that ­H+ is extruded byproducts of nutrient digestion (e.g., phytate, oxalate, and from the cell back into the lumen via NHE3, allowing only tannin), as follows. sodium to enter the cell down its concentration gradient and to exit the cell by the basolateral ­Na+/K+-ATPase into the Prolonged exposure to high luminal calcium paracellular space, thus generating solvent drag along the paracellular space. Previously, calcium has been known to exert indirect nega- The exact mechanism by which NHE3 modulates cal- tive feedback on calcium absorption. Briefy, CaSR has been cium absorption is largely unknown, but the NHE3 inhibi- reported in the apical and basolateral plasma membrane [17, tor, tenapanor, was found to completely abolish intestinal 18]. It is postulated that CaSR exerts its signaling through calcium transport [96]. Interestingly, NHE3 not only acts as activation of phosphodiesterases (PDE) which in turn a carrier for sodium uptake but also plays an important role enhances cyclic nucleotide degradation [35, 94]. Although in the regulation of intestinal pH. Apical NHE3 transports information on specifc PDE in the intestinal epithelial cells ­H+ into the lumen in exchange with Na­ +, thus preventing is scant, PDE might be an important mediator in the nega- intracellular acidifcation, and at the same time making the tive regulatory process because of the existence of PDE1–5 lumen more acid, which is known as acid microclimate [97]. isoforms in human colonic cancer cells [76, 94]. A slight luminal acidifcation promotes the generation of Interestingly, Rodrat et al. [84] recently demonstrated ionized calcium, which might subsequently increase calcium that transepithelial calcium transport across Caco-2 mon- absorption [31]. NHE3 knockout animals were found to have olayer was diminished after direct exposure to high-dose impaired absorption of calcium, sodium, and water, as well 1,25(OH)2D3 (10 nM) or high concentration of apical ion- as acid/base imbalance [74, 81]. This fnding could be par- ized calcium (30 mM CaCl­ 2). Since CaSR inhibitors (calhex tially explained by the fact that NHE3 normally transports 231 and NPS2143) could prevent the high luminal calcium- ­H+ to the lumen while uptaking Na­ + into the cytoplasm. A induced suppression of calcium transport, it is likely that reduction in the transcellular calcium absorption in NHE3 apical CaSR acts as a component of an anticipatory or feed- null mice was mainly due to a decrease in calcium fux in back mechanism to prevent excessive calcium infux into mucosal-to-serosal direction, consistent with the downregu- the cell, while CaSR in the basolateral membrane probably lation of TRPV6 expression [81]. helps detect the amount of absorbed calcium ions that accu- mulate in the immediate vicinity of the basal membrane or

1 3 690 The Journal of Physiological Sciences (2019) 69:683–696 paracellular space. In other words, the intestinal epithelial Nigerian children fed phytase-treated or untreated meal cells are able to closely monitor ionized calcium levels at showed no diference in their fractional calcium absorption both entry and exit points by using CaSR which in turn acti- [95]. Nevertheless, a correlation analysis on calcium and vates secondary negative regulators, e.g., FGF-23, to slow inhibitory factors, including oxalate, tannin, phytate, and down calcium absorption, thus preventing excessive calcium dietary fber revealed that oxalate had the greatest negative uptake into the body [53, 54, 84]. Detail of the inhibitory efect on calcium bioavailability [2]. efect of FGF23 is further described in the following section. Besides nutrient and non-nutrient substances, other lumi- nal compounds can inhibit calcium absorption, for instance, Iron lithocholic acid (LCA), deoxycholic acid or its salt, sodium deoxycholate (NaDOC). LCA and NaDOC are secondary It has long been known that high oral calcium intake inter- bile acids that are formed by enzymes of the intestinal fora. feres with heme and nonheme iron absorption [63, 76], A high concentration of NaDOC not only damages liver but whether high iron intake causes a reduction in calcium tissue and promotes colon cancer [92], but also impairs absorption is still uncertain. A recent investigation in iron intestinal calcium absorption [46, 66, 83]. Determination hyperabsorptive β-thalassemic mice showed that duodenal of calcium absorption in chick duodenum revealed that calcium absorption had an inverse correlation with tran- NaDOC suppressed calcium absorption by downregulating sepithelial iron transport [58]. Interestingly, after injection transcellular calcium transport machinery, i.e., PMCA­ 1b, of —a liver-derived inhibitory factor of intestinal calbindin-D28k, and NCX1. Such negative efects might be iron absorption [38]—the thalassemia-induced impair- a consequence of oxidative stress as suggested by concurrent ment of calcium absorption was alleviated [58]. However, increases reactive oxygen species (ROS) production, glu- the exact cellular and molecular mechanisms of iron-asso- tathione reduction, and mitochondrial swelling. Indeed, ROS ciated impairment of calcium absorption require further has been shown to inhibit calcium absorption [83]. More investigation. studies are required to show if NaDOC-induced impaired intestinal calcium absorption would be of clinical concern Sugars on bone health.

Although some monosaccharides, particularly SGLT1 sub- Intrinsic inhibitory factors strates (e.g., glucose and galactose) stimulate intestinal cal- cium absorption, a ketonic monosaccharide fructose found Intestinal calcium absorption can be afected by humoral in fruits, vegetables, and grains can produce an inhibitory agents acting from the serosal side. Although is efect [28, 85]. Both in vitro and in vivo studies revealed known as a hypocalcemic hormone, it does not have any that fructose diminished intestinal calcium absorption, in direct action on the intestinal epithelial cells. It induces part by reducing circulating 1,25(OH)2D3 levels, TRPV6 mainly by inhibiting -mediated and calbindin-D9K expression, and thus transcellular calcium bone resorption and enhancing renal calcium excretion [57]. transport [28]. Furthermore, high fructose intake was found So the importance of calcitonin in the day-to-day calcium to diminish calcium absorption in growing and lactating rats homeostasis is negligible. Recently, bone-derived FGF-23 through disrupting 1,25(OH)2D3 , i.e., enhanc- was proposed as a novel inhibitory regulator of intestinal ing 1,25(OH)2D3 catabolism and impairing 1,25(OH)2D3 calcium absorption [53, 54, 105]. synthesis [27, 28]. FGF‑23 Natural substances and byproducts of nutrient digestion In general, calcium in the plasma is the major factor mod- A number of naturally occurring substances in many green erate the intestinal calcium absorption through the long leafy vegetables (e.g., spinach, beet greens, and tea), for loop negative feedback by inhibiting PTH production and example, oxalate, phytate, and tannin, can block the intesti- release. Specifcally, a slight increase in the plasma-free ion- nal calcium absorption by binding to calcium, thereby ren- ized calcium leads to activation of CaSR in the parathyroid dering calcium insoluble and unavailable for absorption [2, chief cells [82], thereby reducing PTH release. A reduc- 69]. However, their physiological signifcance and the fnal tion in serum PTH results in decreases in the circulating efect on calcium absorption remain controversial. For exam- 1,25(OH)2D3 level and fnally the intestinal calcium absorp- ple, calcium bioavailability of green leafy vegetables such tion. However, some factors can also reduce the plasma cal- as centella, quinoa, and roselle, all of which contain high cium by acting at the absorption site. amounts of inhibitory factors, was not changed by cook- As mentioned earlier, FGF-23 is primarily known as a ing process or phytase digestion [2, 69]. On the other hand, bone-derived phosphaturic hormone that mainly enhances

1 3 The Journal of Physiological Sciences (2019) 69:683–696 691

renal phosphate excretion and 1,25(OH)2D3 catabolism [39, remains elusive, several signaling proteins, such as MAPK/ 51]. Besides being produced by bone cells, it is also abun- ERK, p38 MAPK, and PKC might be involved in the process dantly expressed in other cells, such as kidney, brain, lung, [53, 54]. liver, spleen, and intestinal enterocytes [53, 56]. Normally, FGF-23 production is not only stimulated by an increase in serum phosphate stimulates the secretion of 1,25(OH)2D3, it is also enhanced by other calciotropic hor- PTH and FGF-23, both of which enhance urinary phosphate mones, such as prolactin [105]. In mammalian, prolactin is excretion to prevent undesirable clinical problems due to known as a milk-producing hormone that acts as a major hyperphosphatemic spikes, such as ectopic calcifcation [43, calcium-regulating hormone during pregnancy and lactation, 49]. FGF-23 enhances phosphate excretion directly through the reproductive periods of high calcium demand [15]. Since inactivation of sodium/phosphate cotransporter (NaPi)-2a prolactin markedly stimulates the transcellular and solvent and -2c in the proximal renal tubules and indirectly by sup- drag-induced paracellular calcium absorption across the pressing 1,25(OH)2D3 synthesis and promoting 1,25(OH)2D3 intestinal epithelium [48, 91], the local production of FGF- conversion to inactive 24,25(OH)2D3, thereby reducing the 23 may be a counterregulatory mechanism to restrict exces- circulating 1,25(OH)2D3 levels and also intestinal phosphate sive calcium absorption during these reproductive periods. [51, 56, 90]. These complete the negative feedback loop for phosphorus homeostasis. However, an experiment in VDR knockout mice sug- Stanniocalcin gested that elevation of plasma calcium was a potent stimu- lator of FGF-23 production in a VDR-independent manner Previously recognized as a hypocalcemic hormone from the [90]. Later evidence supported the notion that FGF-23 not corpuscles of Stannius in osteichthyan fsh, stanniocalcin-1 only controls phosphorus metabolism but also acts as a cal- was also identifed in the kidney of several mammal species, cium-regulating hormone that directly controls the intestinal including rodent, cow, and human [44, 45, 99]. Regulation calcium absorption through both systemic and local (parac- of stanniocalcin production and release in mammals remain rine) mechanisms. The systemic efect was demonstrated by elusive, but 1,25(OH)2D3 may be a part of its regulatory complete abolishment of 1,25(OH)2D3-enhanced duodenal loop [44], similar to the negative feedback loop of FGF- calcium transport by intravenous FGF-23 injection [53]. Fur- 23. In opossum kidney proximal tubular cells, 1,25(OH)2D3 thermore, the fnding that FGF-23 in the intestine, ex vivo exposure was found to enhance stanniocalcin expression studies in murine duodenum demonstrated that FGF-23 in a dose-dependent manner [42, 44]. Once released from directly diminished the 1,25(OH)2D3-enhanced transcellular the kidney, the circulating stanniocalcin is thought to acti- and paracellular calcium absorption in ex vivo murine duo- vate its receptor on the basolateral membrane, but not the denum evinced that FGF-23 could exert direct action on the apical membrane, of enterocytes, to enhance the intestinal intestine [53, 54]. Moreover, other studies reported that FGF phosphate absorption, while suppressing calcium absorp- receptors (FGFRs) express in both small and large intestinal tion [45, 65]. In the gut, stanniocalcin has been reported to cells [70, 93, 105]. These fndings confrmed that FGF-23 express in stomach, small intestine, and colon of neonatal may have direct actions on the intestinal cells. Although and mature rats [55]. Functional study by Madsen and col- there were a number of reports in Fgfr1−/−, Fgfr3−/−, and leagues reported the negative efect of stanniocalcin-1 on net Fgfr4−/− mice [33, 34], the intestinal calcium fux in FGFRs calcium absorption across the swine duodenum, consistent knockout animals has never been investigated. with its hypocalcemic action observed in fsh [65]. Interestingly, FGF-23 not only elicits action from the Furthermore, stanniocalcin-1 not only functions as an basolateral side but also exerts a negative feedback regu- endocrine factor but is also produced locally in the intestine lation on calcium transport across the Caco-2 monolayer to control calcium absorption. Xiang et al. [107] demon- from the apical side [84], consistent with the presence of strated in intestinal epithelium-like Caco-2 cells that over- FGF receptors in both apical and basolateral membranes of expression of stanniocalcin-1 could downregulate TRPV5 the duodenal enterocytes [53]. The expression of FGF-23 and TRPV6 protein expression, suggesting that stannioc- protein in the enterocytes is upregulated by 1,25(OH)2D3 alcin-1—as a paracrine/autocrine factor—might directly as well as high apical calcium in a concentration-dependent and locally fine-tune calcium flux across the intestinal manner [84]. As mentioned earlier, CaSR on both apical epithelium. Exposure to recombinant stanniocalcin-1 also and basolateral membrane of the enterocytes have a role directly suppressed TRPV5 and TRPV6 expression in stan- in the monitoring of calcium transport across the entero- niocalcin-1-knockdown Caco-2 cell without having an efect cytes. It was interesting to note that activation of CaSR led on PMCA­ 1b, NCX1, or VDR expression [107]. Although to FGF-23 production and suppression of calcium transport. stanniocalcin-2 with ~ 80% homology to stanniocalcin-1 Although the exact molecular mechanism and signaling has been identifed in mammals, it is unclear whether this pathway by which FGF-23 reduces the calcium transport peptide can locally modulate intestinal calcium absorption.

1 3 692 The Journal of Physiological Sciences (2019) 69:683–696

Neural regulation of calcium transport responses, including inhibition of calcium absorption [23, 62]. Furthermore, there are certain molecules such Enterocytes are innervated by neurons from myenteric as FGF-23 that is produced by the enterocytes and nega- and submucosal plexuses in the enteric nervous system tively control calcium absorption in a paracrine/autocrine (ENS), which are in turn controlled by the autonomic neu- manner. Specifcally, bone-derived FGF-23 is probably an rons from both sympathetic and parasympathetic systems important part of the negative feedback loop, as depicted [67]. The ENS secretomotor neurons release a number of in Fig. 1, to help prevent excess calcium input or calcium neurotransmitters [67], such as serotonin, galanin, neuro- overload. Nevertheless, our knowledge on the negative peptide Y, , and vasoactive intestinal pep- regulators and their actions in the maintenance of calcium tide (VIP), the last of which was found to modulate cal- homeostasis is far from adequate, especially at organ level. cium absorption. However, there were few investigations For instance, the cellular and molecular mechanism(s) for regarding possible neural control on calcium absorption. local and systemic FGF-23 actions in the feedback regula- An in vitro study in Caco-2 and HT29.19A monolayers tion of calcium absorption are not completely understood, reported that transcellular calcium uptake was increased neither its contribution to body calcium homeostasis. Nev- by carbachol (a cholinomimetic drug) and VIP, whereas ertheless, most investigations are from the large intestine, paracellular calcium transport was decreased by VIP [6]. therefore, more investigations are required for the small In vivo study in rats supported the possibility of this neural intestine in order to confrm its physiological signifcance. control of calcium absorption since blockage of neural Moreover, most reports were from animal study, therefore transmission by a potent voltage-dependent ­Na+ channel its role in human is worth exploring. inhibitor, tetrodotoxin (TTX), diminished the leucine- Besides the humoral factors, luminal factors, particularly induced transcellular calcium transport (Thammayon, and iron and negatively charged molecules, can also hinder calcium Charoenphandhu, unpublished observation). Indeed, the absorption. For example, when iron absorption is upregulated, sympathetic and VIP-ergic neurons may be essential for calcium absorption is suppressed presumably as result of iron- calcium and anion transport in other organs, e.g., the feline induced oxidative stress and inhibition of vesicular calcium gallbladder mucosa. It was evident that noradrenergic transport [25, 58]. Meanwhile, negatively charged molecules, stimulation enhanced net absorption of calcium, bicarbo- e.g., phytate and oxalate, are able to physically trap calcium in nate, and water, whereas VIP induced an opposite response the lumen, thus reducing calcium uptake. Since it is possible to [73]. Therefore, it is possible that neurons could regulate alter the luminal content in the intestine by dietary manipula- intestinal calcium absorption. However, more investigation tion, better understanding of how luminal factors and nutrients is required to demonstrate how ENS controls the intestinal fne-tune or interfere with calcium absorption would provide calcium transport as well as the fnal outcome(s) when a strong foundation for dietary recommendation and further multiple types of neurotransmitters are simultaneously development of safe and efective calcium-fortifed products released from the ENS neurons. for calcium-defcient osteoporotic patients and those with high calcium demand, such as pregnant and lactating mothers.

Perspective and concluding remarks Funding This work was supported by grants from Mahidol University (to NC), the Thailand Research Fund (TRF) through the TRF Senior Research Scholar Grant (RTA6080007 to NC), the Faculty of Science, We have elaborated herein that to maintain appropriate Mahidol University (to NC), TRF International Research Network calcium balance besides being stimulated by the well Program (IRN60W0001 to KW and NC), Research Grant for New established PTH-1,25(OH) D hormonal axis, calcium Scholar from TRF and Ofce of the Higher Education Commission 2 3 (MRG6280198 to JT), and TRF through the Royal Golden Jubilee transport across the intestinal epithelium can be restricted Ph.D. Program (PHD/0105/2557 to MR). by a number of mechanisms. At the molecular level, when calcium concentration reaches a certain threshold level, Compliance with ethical standards binding of calcium ions to calcium-transporting proteins, channels, or tight junction proteins can inactivate these Conflict of interest The authors declare that they have no conficts of transporters and diminish transcellular and paracellular interest. calcium transport [23, 52, 62]. Enterocytes also express Ethical approval This article does not contain any studies with human CaSR, which detects free-ionized calcium on both luminal participants or animals performed by any of the authors. and pericellular vicinity, thereby initiating various cellular

1 3 The Journal of Physiological Sciences (2019) 69:683–696 693

References epithelium: evidence for modulation of fuid secretion. Am J Physiol Gastrointest Liver Physiol 283:G240–G250 19. Cheng SX, Lightfoot YL, Yang T, Zadeh M, Tang L, Sahay 1. Abrams SA, Grifn IJ, Davila PM (2002) Calcium and B, Wang GP, Owen JL, Mohamadzadeh M (2014) Epithelial absorption from lactose-containing and lactose-free infant for- CaSR defciency alters intestinal integrity and promotes pro- mulas. Am J Clin Nutr 76:442–446 infammatory immune responses. FEBS Lett 588:4158–4166 2. Amalraj A, Pius A (2015) Bioavailability of calcium and its 20. Christakos S (2012) Mechanism of action of 1,25-dihydroxyvi- absorption inhibitors in raw and cooked green leafy vegetables tamin ­D on intestinal calcium absorption. Rev Endocr Metab commonly consumed in India: an in vitro study. Food Chem 3 Disord 13:39–44 170:430–436 21. Colegio OR, Van Itallie C, Rahner C, Anderson JM (2003) 3. Armbrecht HJ, Zenser TV, Gross CJ, Davis BB (1980) Adapta- Claudin extracellular domains determine paracellular charge tion to dietary calcium and phosphorus restriction changes with selectivity and resistance but not tight junction fbril architec- age in the rat. Am J Physiol 239:E322–E327 ture. Am J Physiol Cell Physiol 284:C1346–C1354 4. Benn BS, Ajibade D, Porta A, Dhawan P, Hediger M, Peng JB, 22. Cook AE, Mistry SN, Gregory KJ, Furness SG, Sexton PM, Jiang Y, Oh GT, Jeung EB, Lieben L, Bouillon R, Carmeliet Scammells PJ, Conigrave AD, Christopoulos A, Leach K G, Christakos S (2008) Active intestinal calcium transport in (2015) Biased allosteric modulation at the CaS receptor engen- the absence of transient receptor potential vanilloid type 6 and dered by structurally diverse calcimimetics. Br J Pharmacol calbindin-D . 149:3196–3205 9k 172:185–200 5. Binder HJ, Singh SK, Geibel JP, Rajendran VM (1997) Novel 23. Derler I, Hofbauer M, Kahr H, Fritsch R, Muik M, Kepplinger transport properties of colonic crypt cells: fuid absorption and K, Hack ME, Moritz S, Schindl R, Groschner K, Romanin C Cl-dependent Na–H exchange. Comp Biochem Physiol A Physiol (2006) Dynamic but not constitutive association of calmodulin 118:265–269 with rat TRPV6 channels enables fne tuning of ­Ca2+-dependent 6. Blais A, Aymard P, Lacour B (1997) Paracellular calcium trans- inactivation. J Physiol 577:31–44 port across Caco-2 and HT29 cell monolayers. Pfugers Arch 24. Diaz de Barboza G, Guizzardi S, Tolosa de Talamoni N (2015) 434:300–305 Molecular aspects of intestinal calcium absorption. World J Gas- 7. Bronner F (2003) Mechanisms of intestinal calcium absorption. troenterol 21:7142–7154 J Cell Biochem 88:387–393 25. Diaz de Barboza G, Guizzardi S, Moine L, Tolosa de Talamoni 8. Brown AJ, Krits I, Armbrecht HJ (2005) Efect of age, vitamin D, N (2017) Oxidative stress, antioxidants and intestinal calcium and calcium on the regulation of rat intestinal epithelial calcium absorption. World J Gastroenterol 23:2841–2853 channels. Arch Biochem Biophys 437:51–58 26. Doroudi M, Schwartz Z, Boyan BD (2015) Membrane-medi- 9. Brun L, Brance M, Rigalli A (2012) Luminal calcium concen- ated actions of 1,25-dihydroxy vitamin D­ : a review of the tration controls intestinal calcium absorption by modifcation of 3 roles of phospholipase A­ activating protein and ­Ca2+/calmo- intestinal alkaline phosphatase activity. Br J Nutr 108:229–233 2 dulin-dependent protein kinase II. J Steroid Biochem Mol Biol 10. Centeno VA, Díaz de Barboza GE, Marchionatti AM, Alisio AE, 147:81–84 Dallorso ME, Nasif R, Tolosa de Talamoni NG (2004) Dietary 27. Douard V, Asgerally A, Sabbagh Y, Sugiura S, Shapses SA, calcium defciency increases ­Ca2+ uptake and ­Ca2+ extrusion Casirola D, Ferraris RP (2010) Dietary fructose inhibits intes- mechanisms in chick enterocytes. Comp Biochem Physiol A Mol tinal calcium absorption and induces vitamin D insufciency in Integr Physiol 139:133–141 CKD. J Am Soc Nephrol 21:261–271 11. Centeno V, Picotto G, Perez A, Alisio A, Tolosa de Talamoni N 28. Douard V, Sabbagh Y, Lee J, Patel C, Kemp FW, Bogden JD, (2011) Intestinal ­Na+/Ca2+ exchanger protein and gene expres- Lin S, Ferraris RP (2013) Excessive fructose intake causes sion are regulated by 1,25(OH) D in vitamin D-defcient chicks. 2 3 1,25-(OH) D -dependent inhibition of intestinal and renal cal- Arch Biochem Biophys 509:191–196 2 3 cium transport in growing rats. Am J Physiol Endocrinol Metab 12. Chang W, Shoback D (2004) Extracellular Ca­ 2+-sensing recep- 304:E1303–E1313 tors: an overview. Cell Calcium 35:183–196 29. Fleet JC (2004) Rapid, membrane-initiated actions of 1,25 dihy- 13. Charoenphandhu N, Krishnamra N (2007) Prolactin is an impor- droxyvitamin D: what are they and what do they mean? J Nutr tant regulator of intestinal calcium transport. Can J Physiol Phar- 134:3215–3218 macol 85:569–581 30. Fleet JC, Schoch RD (2010) Molecular mechanisms for regu- 14. Charoenphandhu N, Nakkrasae LI, Kraidith K, Teerapornpun- lation of intestinal calcium absorption by vitamin D and other takit J, Thongchote K, Thongon N, Krishnamra N (2009) Two- factors. Crit Rev Clin Lab Sci 47:181–195 step stimulation of intestinal Ca­ 2+ absorption during lactation 31. Fordtran JS, Locklear TW (1966) Ionic constituents and osmolal- by long-term prolactin exposure and suckling-induced prolactin ity of gastric and small-intestinal fuids after eating. Am J Dig surge. Am J Physiol Endocrinol Metab 297:E609–E619 Dis 11:503–521 15. Charoenphandhu N, Wongdee K, Krishnamra N (2010) Is prol- 32. Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, Uchiyama the cardinal calciotropic maternal hormone? Trends Endo- Y, Yamamoto Y, Wada T, Kojima T, Yokozaki H, Yamashita T, crinol Metab 21:395–401 Kato S, Sawada N, Chiba H (2008) Tight junction proteins clau- 16. Charoenphandhu N, Laohapitakworn S, Kraidith K, Nakkrasae din-2 and -12 are critical for vitamin D-dependent ­Ca2+ absorp- LI, Jongwattanapisan P, Tharabenjasin P, Krishnamra N (2011) tion between enterocytes. Mol Biol Cell 19:1912–1921 Electrogenic Na­ +/HCO − co-transporter-1 is essential for the 3 33. Gattineni J, Bates C, Twombley K, Dwarakanath V, Robinson parathyroid hormone-stimulated intestinal HCO­ − secretion. 3 ML, Goetz R, Mohammadi M, Baum M (2009) FGF23 decreases Biochem Biophys Res Commun 409:775–779 renal NaPi-2a and NaPi-2c expression and induces hypophos- 17. Chattopadhyay N, Cheng I, Rogers K, Riccardi D, Hall A, Diaz phatemia in vivo predominantly via FGF receptor 1. Am J Phys- R, Hebert SC, Soybel DI, Brown EM (1998) Identifcation and iol Renal Physiol 297:F282–F291 localization of extracellular ­Ca2+-sensing receptor in rat intes- 34. Gattineni J, Alphonse P, Zhang Q, Mathews N, Bates CM, Baum tine. Am J Physiol 274:G122–G130 M (2014) Regulation of renal phosphate transport by FGF23 is 18. Cheng SX, Okuda M, Hall AE, Geibel JP, Hebert SC (2002) mediated by FGFR1 and FGFR4. Am J Physiol Renal Physiol Expression of calcium-sensing receptor in rat colonic 306:F351–F358

1 3 694 The Journal of Physiological Sciences (2019) 69:683–696

35. Geibel J, Sritharan K, Geibel R, Geibel P, Persing JS, Seeger N, Charoenphandhu N (2012) Fibroblast growth factor-23 A, Roepke TK, Deichstetter M, Prinz C, Cheng SX, Martin D, abolishes 1,25-dihydroxyvitamin D­ 3-enhanced duodenal cal- Hebert SC (2006) Calcium-sensing receptor abrogates secre- cium transport in male mice. Am J Physiol Endocrinol Metab tagogue-induced increases in intestinal net fuid secretion by 302:E903–E913 enhancing cyclic nucleotide destruction. Proc Natl Acad Sci USA 54. Khuituan P, Wongdee K, Jantarajit W, Suntornsaratoon P, 103:9390–9397 Krishnamra N, Charoenphandhu N (2013) Fibroblast growth fac- 36. Gentili C, Morelli S, de Boland AR (2003) Characterization of tor-23 negates 1,25(OH)2D3-induced intestinal calcium transport PTH/PTHrP receptor in rat duodenum: efects of ageing. J Cell by reducing the transcellular and paracellular calcium fuxes. Biochem 88:1157–1167 Arch Biochem Biophys 536:46–52 37. Goltzman D (2018) Physiology of parathyroid hormone. Endo- 55. Kobayashi R, Nakagomi Y, Shimura Y, Mochizuki M, Kobayashi crinol Metab Clin North Am 47:743–758 K, Sugita K, Ohyama K (2009) Expression of stanniocalcin-1 38. Gulec S, Anderson GJ, Collins JF (2014) Mechanistic and regula- in gastrointestinal tracts of neonatal and mature rats. Biochem tory aspects of intestinal iron absorption. Am J Physiol Gastro- Biophys Res Commun 389:478–483 intest Liver Physiol 307:G397–G409 56. Kolek OI, Hines ER, Jones MD, LeSueur LK, Lipko MA, 39. Guo YC, Yuan Q (2015) Fibroblast growth factor 23 and bone Kiela PR, Collins JF, Haussler MR, Ghishan FK (2005) 1α,25- mineralisation. Int J Oral Sci 7:8–13 dihydroxyvitamin ­D3 upregulates FGF23 gene expression in 40. Haussler MR, Jurutka PW, Mizwicki M, Norman AW bone: the fnal link in a renal-gastrointestinal-skeletal axis that (2011) Vitamin D receptor (VDR)-mediated actions of controls phosphate transport. Am J Physiol Gastrointest Liver 1α,25(OH)2vitamin ­D3: genomic and non-genomic mechanisms. Physiol 289:G1036–G1042 Best Pract Res Clin Endocrinol Metab 25:543–559 57. Kopic S, Geibel JP (2013) Gastric acid, calcium absorption, and 41. Hebert SC, Cheng S, Geibel J (2004) Functions and roles of the their impact on bone health. Physiol Rev 93:189–268 extracellular ­Ca2+-sensing receptor in the . 58. Kraidith K, Svasti S, Teerapornpuntakit J, Vadolas J, Chaimana Cell Calcium 35:239–247 R, Lapmanee S, Suntornsaratoon P, Krishnamra N, Fucharoen 42. Honda S, Kashiwagi M, Ookata K, Tojo A, Hirose S (1999) S, Charoenphandhu N (2016) Hepcidin and 1,25(OH)2D3 efec- 2+ Regulation by 1α,25-dihydroxyvitamin ­D3 of expression of tively restore Ca­ transport in β-thalassemic mice: reciprocal stanniocalcin messages in the rat kidney and ovary. FEBS Lett phenomenon of Fe­ 2+ and ­Ca2+ absorption. Am J Physiol Endo- 459:119–122 crinol Metab 311:E214–E223 43. Houston J, Smith K, Isakova T, Sowden N, Wolf M, Gutierrez 59. Kumar V, Abbas AK, Aster JC (2015) Cellular responses to stress OM (2013) Associations of dietary phosphorus intake, urinary and toxic insults: adaptation, injury, and death. In: Kumar V, phosphate excretion, and fbroblast growth factor 23 with vascu- Abbas AK, Aster JC (eds) Robbins and Cotran pathologic basis lar stifness in . J Ren Nutr 23:12–20 of disease, 9th edn. Elsevier Saunders, Philadelphia, pp 31–68 44. Hung NT, Yamamoto H, Takei Y, Masuda M, Otani A, Kozai M, 60. Lamireau T, Zoltowska M, Levy E, Yousef I, Rosenbaum J, Ikeda S, Nakahashi O, Tanaka S, Taketani Y, Takeda E (2012) Tuchweber B, Desmouliere A (2003) Efects of bile acids on Up-regulation of stanniocalcin 1 expression by 1,25-dihydroxy biliary epithelial cells: proliferation, cytotoxicity, and vitamin ­D3 and parathyroid hormone in renal proximal tubular secretion. Life Sci 72:1401–1411 cells. J Clin Biochem Nutr 50:227–233 61. Laohapitakworn S, Thongbunchoo J, Nakkrasae LI, Krishnamra 45. Ishibashi K, Imai M (2002) Prospect of a stanniocalcin endo- N, Charoenphandhu N (2011) Parathyroid hormone (PTH) rap- − crine/paracrine system in mammals. Am J Physiol Renal Physiol idly enhances CFTR-mediated HCO­ 3 secretion in intestinal 282:F367–F375 epithelium-like Caco-2 monolayer: a novel ion regulatory action 46. Ishizawa M, Akagi D, Makishima M (2018) Lithocholic acid is of PTH. Am J Physiol Cell Physiol 301:C137–C149 a vitamin D receptor ligand that acts preferentially in the . 62. Lee D, Obukhov AG, Shen Q, Liu Y, Dhawan P, Nowycky MC, Int J Mol Sci 19:1975 Christakos S (2006) Calbindin-D28k decreases L-type calcium 47. Jando J, Camargo SMR, Herzog B, Verrey F (2017) Expression channel activity and modulates intracellular calcium homeostasis and regulation of the neutral amino acid transporter B­ 0AT1 in rat in response to K­ + depolarization in a rat beta cell line RINr1046- small intestine. PLoS One 12:e0184845 38. Cell Calcium 39:475–485 48. Jantarajit W, Thongon N, Pandaranandaka J, Teerapornpuntakit 63. Lertsuwan K, Wongdee K, Teerapornpuntakit J, Charoenphandhu J, Krishnamra N, Charoenphandhu N (2007) Prolactin-stimulated N (2018) Intestinal calcium transport and its regulation in thalas- transepithelial calcium transport in duodenum and Caco-2 mon- semia: interaction between calcium and iron metabolism. J Phys- olayer are mediated by the phosphoinositide 3-kinase pathway. iol Sci 68:221–232 Am J Physiol Endocrinol Metab 293:E372–E384 64. Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, Quarles LD 49. Jimbo R, Kawakami-Mori F, Mu S, Hirohama D, Majtan B, (2006) Fibroblast growth factor 23 is a counter-regulatory Shimizu Y, Yatomi Y, Fukumoto S, Fujita T, Shimosawa T phosphaturic hormone for vitamin D. J Am Soc Nephrol (2014) Fibroblast growth factor 23 accelerates phosphate- 17:1305–1315 induced vascular calcifcation in the absence of klotho defciency. 65. Madsen KL, Tavernini MM, Yachimec C, Mendrick DL, Kidney Int 85:1103–1111 Alfonso PJ, Buergin M, Olsen HS, Antonaccio MJ, Thomson 50. Jongwattanapisan P, Suntornsaratoon P, Wongdee K, Dorkkam AB, Fedorak RN (1998) Stanniocalcin: a novel protein regulating N, Krishnamra N, Charoenphandhu N (2012) Impaired body calcium and phosphate transport across mammalian intestine. calcium metabolism with low bone density and compensatory Am J Physiol 274:G96–G102 colonic calcium absorption in cecectomized rats. Am J Physiol 66. Marchionatti A, Rivoira M, Rodriguez V, Perez A, Tolosa Endocrinol Metab 302:E852–E863 de Talamoni N (2018) Molecular mechanisms triggered by 51. Jüppner H (2011) Phosphate and FGF-23. Kidney Int 79:S24–S27 bile acids on intestinal Ca­ 2+ absorption. Curr Med Chem 52. Kellett GL (2011) Alternative perspective on intestinal calcium 25:2122–2132 absorption: proposed complementary actions of Ca­ v1.3 and 67. Mittal R, Debs LH, Patel AP, Nguyen D, Patel K, O’Connor G, TRPV6. Nutr Rev 69:347–370 Grati M, Mittal J, Yan D, Eshraghi AA, Deo SK, Daunert S, Liu 53. Khuituan P, Teerapornpuntakit J, Wongdee K, Suntornsara- XZ (2017) Neurotransmitters: the critical modulators regulating toon P, Konthapakdee N, Sangsaksri J, Sripong C, Krishnamra gut-brain axis. J Cell Physiol 232:2359–2372

1 3 The Journal of Physiological Sciences (2019) 69:683–696 695

68. Morgan EL, Mace OJ, Affleck J, Kellett GL (2007) Apical 85. Rodriguez V, Rivoira M, Guizzardi S, Tolosa de Talamoni N 2+ GLUT2 and Ca­ v1.3: regulation of rat intestinal glucose and cal- (2017) Naringin prevents the inhibition of intestinal Ca­ absorp- cium absorption. J Physiol 580:593–604 tion induced by a fructose rich diet. Arch Biochem Biophys 69. Mosha TC, Gaga HE, Pace RD, Laswai HS, Mtebe K (1995) 636:1–10 Efect of blanching on the content of antinutritional factors in 86. Schröder B, Schlumbohm C, Kaune R, Breves G (1996) Role of 2+ selected vegetables. Plant Foods Hum Nutr 47:361–367 calbindin-D9k in bufering cytosolic free Ca­ ions in pig duo- 70. Murgue B, Tsunekawa S, Rosenberg I, deBeaumont M, Podolsky denal enterocytes. J Physiol 492:715–722 DK (1994) Identifcation of a novel variant form of fbroblast 87. Schwartz Z, Graham EJ, Wang L, Lossdorfer S, Gay I, Johnson- growth factor receptor 3 (FGFR3 IIIb) in human colonic epithe- Pais TL, Carnes DL, Sylvia VL, Boyan BD (2005) Phospholipase lium. Cancer Res 54:5206–5211 ­A2 activating protein (PLAA) is required for 1α,25(OH)2D3 sign- 71. Nemere I, Norman AW (1986) Parathyroid hormone stimulates aling in growth plate chondrocytes. J Cell Physiol 203:54–70 calcium transport in perfused duodena from normal chicks: com- 88. Sharma V, O’Halloran DM (2014) Recent structural and func- parison with the rapid (transcaltachic) efect of 1,25-dihydroxy- tional insights into the family of sodium calcium exchangers. vitamin ­D3. Endocrinology 119:1406–1408 Genesis 52:93–109 72. Nemere I, Garbi N, Hammerling GJ, Khanal RC (2010) Intestinal 89. Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, Takeuchi cell calcium uptake and the targeted knockout of the 1,25D3- Y, Fujita T, Nakahara K, Fukumoto S, Yamashita T (2004) FGF- MARRS (membrane-associated, rapid response steroid-binding) 23 is a potent regulator of vitamin D metabolism and phosphate receptor/PDIA3/Erp57. J Biol Chem 285:31859–31866 homeostasis. J Bone Miner Res 19:429–435 73. Nilsson B, Delbro D, Friman S, Thune A, Svanvik J (1996) 90. Shimada T, Yamazaki Y, Takahashi M, Hasegawa H, Urakawa I, Sympathetic and VIP-ergic control of calcium and bicarbonate Oshima T, Ono K, Kakitani M, Tomizuka K, Fujita T, Fukumoto transport in the feline gall bladder mucosa in vivo. J Auton Nerv S, Yamashita T (2005) Vitamin D receptor-independent FGF23 Syst 60:49–55 actions in regulating phosphate and vitamin D metabolism. Am 74. Noonan WT, Woo AL, Nieman ML, Prasad V, Schultheis PJ, J Physiol Renal Physiol 289:F1088–F1095 Shull GE, Lorenz JN (2005) Blood pressure maintenance in 91. Suntornsaratoon P, Kraidith K, Teerapornpuntakit J, Dorkkam NHE3-deficient mice with transgenic expression of NHE3 N, Wongdee K, Krishnamra N, Charoenphandhu N (2014) Pre- in small intestine. Am J Physiol Regul Integr Comp Physiol suckling calcium supplementation efectively prevents lactation- 288:R685–R691 induced osteopenia in rats. Am J Physiol Endocrinol Metab 75. Norman AW (2006) Minireview: vitamin D receptor: new 306:E177–E188 assignments for an already busy receptor. Endocrinology 92. Tahiri M, Tressol JC, Arnaud J, Bornet FR, Bouteloup-Demange 147:5542–5548 C, Feillet-Coudray C, Brandolini M, Ducros V, Pepin D, Brouns 76. O’Grady SM, Jiang X, Maniak PJ, Birmachu W, Scribner LR, F, Roussel AM, Rayssiguier Y, Coudray C (2003) Efect of short- Bulbulian B, Gullikson GW (2002) Cyclic AMP-dependent Cl chain fructooligosaccharides on intestinal calcium absorption secretion is regulated by multiple phosphodiesterase subtypes in and calcium status in postmenopausal women: a stable-isotope human colonic epithelial cells. J Membr Biol 185:137–144 study. Am J Clin Nutr 77:449–457 77. Owen JL, Cheng SX, Ge Y, Sahay B, Mohamadzadeh M (2016) 93. Takaishi S, Sawada M, Morita Y, Seno H, Fukuzawa H, Chiba The role of the calcium-sensing receptor in gastrointestinal T (2000) Identifcation of a novel alternative splicing of human infammation. Semin Cell Dev Biol 49:44–51 FGF receptor 4: soluble-form splice variant expressed in human 78. Quarles LD (2012) Skeletal secretion of FGF-23 regulates phos- gastrointestinal epithelial cells. Biochem Biophys Res Commun phate and vitamin D metabolism. Nat Rev Endocrinol 8:276–286 267:658–662 79. Quinn SJ, Thomsen AR, Pang JL, Kantham L, Brauner-Osborne 94. Tang L, Cheng CY, Sun X, Pedicone AJ, Mohamadzadeh M, H, Pollak M, Goltzman D, Brown EM (2013) Interactions Cheng SX (2016) The extracellular calcium-sensing receptor between calcium and phosphorus in the regulation of the pro- in the intestine: evidence for regulation of colonic absorption, duction of fbroblast growth factor 23 in vivo. Am J Physiol secretion, motility, and immunity. Front Physiol 7:245 Endocrinol Metab 304:E310–E320 95. Thacher TD, Aliu O, Grifn IJ, Pam SD, O’Brien KO, Imade 80. Raschka L, Daniel H (2005) Mechanisms underlying the efects GE, Abrams SA (2009) Meals and dephytinization afect cal- of inulin-type fructans on calcium absorption in the large intes- cium and zinc absorption in Nigerian children with . J Nutr tine of rats. Bone 37:728–735 139:926–932 81. Rievaj J, Pan W, Cordat E, Alexander RT (2013) The ­Na+/H+ 96. Thammayon N, Wongdee K, Lertsuwan K, Suntornsaratoon P, exchanger isoform 3 is required for active paracellular and tran- Thongbunchoo J, Krishnamra N, Charoenphandhu N (2017) ­Na+/ scellular Ca­ 2+ transport across murine cecum. Am J Physiol Gas- H+ exchanger 3 inhibitor diminishes the amino-acid-enhanced trointest Liver Physiol 305:G303–G313 transepithelial calcium transport across the rat duodenum. Amino 82. Ritter CS, Haughey BH, Miller B, Brown AJ (2012) Diferential Acids 49:725–734 gene expression by oxyphil and chief cells of human parathyroid 97. Thwaites DT, Anderson CM (2007) H­ +-coupled nutrient, micro- glands. J Clin Endocrinol Metab 97:E1499–E1505 nutrient and drug transporters in the mammalian small intestine. 83. Rivoira MA, Marchionatti AM, Centeno VA, Díaz de Barboza Exp Physiol 92:603–619 GE, Peralta López ME, Tolosa de Talamoni NG (2012) Sodium 98. Toka HR (2014) New functional aspects of the extracellu- deoxycholate inhibits chick duodenal calcium absorption through lar calcium-sensing receptor. Curr Opin Nephrol Hypertens oxidative stress and apoptosis. Comp Biochem Physiol A Mol 23:352–360 Integr Physiol 162:397–405 99. Tremblay G, Delbecchi L, Loiselle MC, Ster C, Wagner GF, 84. Rodrat M, Wongdee K, Panupinthu N, Thongbunchoo J, Teera- Talbot BG, Lacasse P (2009) Serum levels of stanniocalcin-1 in pornpuntakit J, Krishnamra N, Charoenphandhu N (2018) Holstein heifers and cows. Domest Anim Endocrinol 36:105–109 Prolonged exposure to 1,25(OH)2D3 and high ionized calcium 100. Vervloet M (2019) Renal and extrarenal efects of fbroblast induces FGF-23 production in intestinal epithelium-like Caco-2 growth factor 23. Nat Rev Nephrol 15:109–120 monolayer: a local negative feedback for preventing excessive 101. Wagner CA (2013) The calcium-sensing receptor directly regu- calcium transport. Arch Biochem Biophys 640:10–16 lates proximal tubular functions. Kidney Int 84:228–230

1 3 696 The Journal of Physiological Sciences (2019) 69:683–696

102. Wang CY, Liu S, Xie XN, Tan ZR (2017) Regulation profle 109. Younoszai MK, Nathan R (1985) Intestinal calcium absorption of the intestinal peptide transporter 1 (PepT1). Drug Des Devel is enhanced by d-glucose in diabetic and control rats. Gastroen- Ther 11:3511–3517 terology 88:933–938 103. Wasserman RH (2004) Vitamin D and the dual processes of 110. Yu AS, Cheng MH, Angelow S, Gunzel D, Kanzawa SA, Sch- intestinal calcium absorption. J Nutr 134:3137–3139 neeberger EE, Fromm M, Coalson RD (2009) Molecular basis 104. Wongdee K, Charoenphandhu N (2015) Vitamin D-enhanced for cation selectivity in claudin-2-based paracellular pores: duodenal calcium transport. Vitam Horm 98:407–440 identifcation of an electrostatic interaction site. J Gen Physiol 105. Wongdee K, Teerapornpuntakit J, Sripong C, Longkunan A, 133:111–127 Chankamngoen W, Keadsai C, Kraidith K, Krishnamra N, 111. Zierold C, Mings JA, DeLuca HF (2003) Regulation of Charoenphandhu N (2016) Intestinal mucosal changes and 25-hydroxyvitamin ­D3-24-hydroxylase mRNA by 1,25-dihy- upregulated calcium transporter and FGF-23 expression during droxyvitamin D­ 3 and parathyroid hormone. J Cell Biochem lactation: contribution of lactogenic hormone prolactin. Arch 88:234–237 Biochem Biophys 590:109–117 112. Zihni C, Mills C, Matter K, Balda MS (2016) Tight junctions: 106. Wongdee K, Krishnamra N, Charoenphandhu N (2017) Derange- from simple barriers to multifunctional molecular gates. Nat Rev ment of calcium metabolism in diabetes mellitus: negative out- Mol Cell Biol 17:564–580 come from the synergy between impaired bone turnover and intestinal calcium absorption. J Physiol Sci 67:71–81 Publisher’s Note Springer Nature remains neutral with regard to 107. Xiang J, Guo R, Wan C, Wu L, Yang S, Guo D (2016) Regulation jurisdictional claims in published maps and institutional afliations. of intestinal epithelial calcium transport proteins by stanniocal- cin-1 in Caco2 cells. Int J Mol Sci 17:1095 108. Yanahira S, Morita M, Aoe S, Suguri T, Takada Y, Miura S, Nakajima I (1997) Efects of lactitol-oligosaccharides on calcium and magnesium absorption in rats. J Nutr Sci Vitaminol (Tokyo) 43:123–132

1 3