Hostname: page-component-8448b6f56d-42gr6 Total loading time: 0 Render date: 2024-04-19T04:54:37.942Z Has data issue: false hasContentIssue false

Dietary and pharmacological compounds altering intestinal calcium absorption in humans and animals

Published online by Cambridge University Press:  15 October 2015

Vanessa Areco
Affiliation:
Laboratorio ‘Dr. Cañas’, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
María Angélica Rivoira
Affiliation:
Laboratorio ‘Dr. Cañas’, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
Valeria Rodriguez
Affiliation:
Laboratorio ‘Dr. Cañas’, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
Ana María Marchionatti
Affiliation:
Laboratorio ‘Dr. Cañas’, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
Agata Carpentieri
Affiliation:
Cátedra de Química Biológica, Facultad de Odontología, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
Nori Tolosa de Talamoni*
Affiliation:
Laboratorio ‘Dr. Cañas’, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
*
*Corresponding author: Professor Dr Nori Tolosa de Talamoni, fax +54 3543 435000, email ntolosa@biomed.fcm.unc.edu.ar, ntolosatalamoni@yahoo.com.ar
Rights & Permissions [Opens in a new window]

Abstract

The intestine is the only gate for the entry of Ca to the body in humans and mammals. The entrance of Ca occurs via paracellular and intracellular pathways. All steps of the latter pathway are regulated by calcitriol and by other hormones. Dietary and pharmacological compounds also modulate the intestinal Ca absorption process. Among them, dietary Ca and P are known to alter the lipid and protein composition of the brush-border and basolateral membranes and, consequently, Ca transport. Ca intakes are below the requirements recommended by health professionals in most countries, triggering important health problems. Chronic low Ca intake has been related to illness conditions such as osteoporosis, hypertension, renal lithiasis and incidences of human cancer. Carbohydrates, mainly lactose, and prebiotics have been described as positive modulators of intestinal Ca absorption. Apparently, high meat proteins increase intestinal Ca absorption while the effect of dietary lipids remains unclear. Pharmacological compounds such as menadione, dl-butionine-S,R-sulfoximine and ursodeoxycholic acid also modify intestinal Ca absorption as a consequence of altering the redox state of the epithelial cells. The paracellular pathway of intestinal Ca absorption is poorly known and is under present study in some laboratories. Another field that needs to be explored more intensively is the influence of the gene × diet interaction on intestinal Ca absorption. Health professionals should be aware of this knowledge in order to develop nutritional or medical strategies to stimulate the efficiency of intestinal Ca absorption and to prevent diseases.

Type
Review Article
Copyright
Copyright © The Authors 2015 

Introduction

Ca is the main mineral component of bone and, hence, is essential for achieving optimal peak bone mass in the first decades of life and for maintaining bone mass, later in life(Reference Ma, Johns and Stafford1). It also plays an important role in many physiological processes(Reference Eisner, Csordás and Hajnóczky2Reference Chaigne-Delalande and Lenardo5). The dysregulation of Ca homeostasis is not only associated with bone disorders, but also with hypertension, insulin resistance, obesity and the metabolic syndrome(Reference Zemel6Reference Bartlett, Gaspers and Pierobon9). Epidemiological and experimental studies have shown an inverse relationship between dietary Ca and risk of breast, colon, prostate and ovarian cancer(Reference Lin, Manson and Lee10Reference Merritt, Cramer and Vitonis13). Therefore, an appropriate Ca homeostasis preserves bone integrity, metabolic balance and avoids epithelial cancers.

Ca metabolism is predominantly regulated by the intestine, kidney, bone and parathyroid glands. Because of their coordinated work, serum Ca concentration is maintained within a narrow range(Reference Fleet and Schoch14). Intestinal Ca absorption is an essential process that occurs through an active transcellular pathway and a passive non-saturable route, named the paracellular pathway(Reference Bronner15). Both routes are regulated by hormones, nutrients and many other factors.

The transcellular pathway is a saturable process, which is prevalent in the proximal small intestine (duodenum and jejunum), vitamin D being the main modulator. This mechanism is energy dependent and implicates Ca movement from the mucosal to serosal side of the intestinal barrier occurring against a concentration gradient. In contrast, the paracellular mechanism occurs throughout the length of the intestine. It is a non-saturable and passive transport and is a linear function of Ca concentration in the lumen(Reference Wasserman16).

Ca2+ ions are absorbed mainly in the small intestine, which is responsible for about 90 % of overall Ca absorption. The longer residence time in the ileum as compared with the other segments of the small intestine favours Ca absorption in that segment(Reference Wasserman16). In rat ileum the transit half-time is about 100–120 min, whereas in the duodenum it is about 2–6 min(Reference Marcus and Lengemann17). The colon is responsible for less than 10 % of the total Ca absorbed; minor amounts of Ca2+ ions are absorbed from the stomach and large intestine(Reference Wasserman16). The major contributors to the amount of Ca absorbed are the residence time and the absorption rate in each intestinal segment. The order of Ca absorption rate is: duodenum > jejunum> ileum(Reference Wasserman16). Ca absorption in the colon is probably very important in pathological conditions such as short bowel syndrome(Reference Wali, Baum and Sitrin18).

Intestinal Ca absorption also depends on the physiological needs of Ca. When the requirements increase and/or the intakes are low, there is an improvement in the efficiency of Ca absorption(Reference Tolosa de Talamoni19). Ageing occurs with a decrease in intestinal Ca absorption(Reference Nordin, Need and Morris20), while growth, pregnancy and lactation promote cation absorption(Reference O’Brien, Nathanson and Mancini21Reference Liesegang, Riner and Boos24). There is little information about the mechanism of lactation-induced intestinal hyperabsorption. Increases have been found in the villous height, villous width and crypt depth with an expansion of the absorptive surface area in the duodenum of 21 d lactating rats. An enhancement in claudin 15 has been also demonstrated in the same animal model(Reference Wongdee, Teerapornpuntakit and Siangpro25). Recently, Teerapornpuntakit et al. (Reference Teerapornpuntakit, Klanchui and Karoonuthaisiri26) have developed a custom-designed cDNA microarray (CalGene Array) to study the expression of genes related to duodenal nutrient transport, among them those related to bone and Ca metabolism. They have determined the transcriptome responses of duodenal epithelial cells in pregnant and lactating rats; data were subsequently validated by quantitative real-time PCR. They have found that pregnancy and late lactation alter the expression of several transcripts, among them those belonging to Ca transporters.

Transcellular pathway

Epithelial calcium channels

TRPV6 (previously named ECaC2 or CaT1) and TRPV5 (previously named ECaC1 or CaT2) are the two epithelial Ca channels involved in Ca entry to the enterocytes. These channels are homologous members of the transient receptor potential (TRP) superfamily, belonging to the vanilloid subfamily (TRPV), which is different from the canonical (TRPC) and melastatin (TRPM) subfamilies(Reference van Abel, Hoenderop and Bindels27). TRPV6 and TRPV5 are co-expressed in the human kidney and intestine, but the first one is highly expressed in the intestine and the latter is the major isoform in the kidney. TRPV6 seems to be a major contributor to apical, intestinal Ca absorption, as suggested by a significant reduction in Ca absorption and serum Ca shown in TRPV6 knockout (KO) mice(Reference Bianco, Peng and Takanaga28, Reference Cui, Li and Johnson29). Both channels are also expressed in the pancreas, prostate, and mammary, sweat and salivary glands(Reference van Abel, Hoenderop and Bindels27). They present a similar structure to other members of the TRP family: six transmembrane domains, a short hydrophobic stretch between segments 5 and 6 involved in the Ca pore and large intracellular N and C terminal tails. The intracellular segments contain phosphorylation sites, postsynaptic density protein motifs and ankyrin repeat domains; all of them are involved in the regulation of channel activity and trafficking(Reference Den Dekker, Hoenderop and Nilius30). It has been demonstrated that the tetrameric structure of TRPV6 and TRPV5 can be combined with each other to form different heterotetrameric channel complexes(Reference Hoenderop, Voets and Hoefs31). Both channels have 75 % homology, share several properties, but have different N and C terminal tails. They are regulated by calcitriol, oestrogen and dietary Ca. Both are inactivated by intracellular Ca, but with a different kinetics. In addition, the affinity of TRPV5 for the inhibitor ruthenium red is 100-fold that of TRPV6(Reference Hoenderop, Vennekens and Müller32).

TRPV6 transcripts have been found in duodenum, but not in ileum, human biopsies. The duodenal expression of TRPV6 in men was detected to be vitamin D dependent, whereas in elderly women the TRPV6 and vitamin D receptor (VDR) expressions were low and not vitamin D dependent. This finding could explain, at least in part, the lower intestinal Ca absorption in elderly postmenopausal women(Reference Walters, Balesaria and Chavele33). In rats, the basal mRNA expression of TRPV6 has been found to be the highest in the duodenum, followed by the colon (46 % of duodenum), and negligible in the jejunum and ileum. The rank order of the basal levels of TRVP6 protein was duodenum > colon (72 % of duodenum) > ileum (25 % of duodenum)(Reference Chow, Quach and Vieth34).

Calbindins

These proteins appear to be responsible for carrying Ca2+ from the apical side of the enterocyte to the basal region of the cell. Calbindin (CB) CB9k is present in the intestine of mammals and CB28k in that from avian species(Reference Tolosa de Talamoni, Pérz and Alisio35). CB9k has four α-helical regions forming an EF-hand pair consisting of a canonical and a non-canonical/pseudo EF-hand domain, which are joined by a linker region. These EF-hands organised in tandem domains are the physiological relevant structures and two Ca2+ ions bind with positive cooperativity(Reference Schwaller36). CB28k has six EF-hand domains, four of which bind Ca2+ with medium/high affinity(Reference Cheung, Richards and Rogers37). EF-hand 2 is non-functional and under physiological conditions EF6 most probably is as well. The four medium/high-affinity sites(Reference Nägerl, Novo and Mody38) are considered Ca specific.

CB also buffer Ca2+ ions by keeping intracellular Ca2+ concentrations below 10–7m, which contribute to the prevention of premature cell death by apoptosis. When there is a down-regulation of CB, an excess of Ca2+ is provoked that may trigger apoptosis in the epithelial cells(Reference Choi, Cho and Kim39). Furthermore, it has been reported that CB28k also inhibits apoptosis in osteoblastic cells(Reference Bellido, Huening and Raval-Pandya40) and in germ cells from Robertsonian mice(Reference Merico, de Barboza and Vasco41, Reference Rodriguez, Diaz de Barboza and Ponce42). In addition, it has been shown in kidney that CB28k regulates the Ca2+ concentration in the vicinity of the TRPV5 pore by a direct association with the channel(Reference Lambers, Mahieu and Oancea43). This might occur in the intestine and in other tissues with important movements in intracellular Ca2+ concentrations.

Genetic studies have provided information that confused the understanding of CB on Ca homeostasis. Mice with ablation of the CB28k gene do not exhibit calcemic abnormalities(Reference Airaksinen, Eilers and Garaschuk44). In CB9k-null mutant mice as well as mice lacking the epithelial Ca channel TRPV6 it has been demonstrated that the regulation of active intestinal Ca absorption is independent of CB9k and TRPV6. The authors think that in the KO mice there is compensation by another Ca2+ channel or protein and that other novel factors are involved in intestinal Ca absorption(Reference Christakos, Dhawan and Ajibade45). It has been found that an ablation of CB9k alters the expression of paracellular tight junction genes. The compensatory expression of paracellular tight junction genes in the duodenum was associated with CB9k, but not with CB28kReference Hwang, Yang and Kang(46). This interaction between the transcellular and paracellular pathways might partially explain the variety of gut responses to absorb Ca under different pathophysiological conditions.

Calcium pump and Na+/Ca2+ exchanger

Plasma membrane Ca2+-ATPase (PMAC) 1 is an ATP-dependent transporter that pumps Ca out of the cytosol. This protein was detected in erythrocyte membranes and found to have a high Ca2+ affinity(Reference Schatzmann47). It presents four isoforms (PMCA1–4), which are divided into several subtypes by alternative splicing. PMCA1 is considered as the housekeeping isoform because its mRNA is in all tissues. The correlation between the regulation of Ca homeostasis by TRPV6 and PMCA1 and duodenal and renal function is not well known. Nevertheless, some reports have described a role for TRPV6 and PMCA1 in the uterus, duodenum, kidney and brain(Reference Barley, Howard and O’Callaghan48Reference Tribe, Moriarty and Poston52). PMCA has a relative molecular mass (Mr) of 130 kDa and a K m for Ca2+ of 0·2 µm in the presence of calmodulin(Reference Ghijsen, De Jong and Van Os53). In the intestine, PMCA is located in the caveolae, which can exist in open and closed forms that control Ca2+ efflux from the cell(Reference Anderson54). The predominant form in the intestine is the isoform PMCA1b. We have found that its expression and activity are higher in enterocytes from the villus tip than in those from the villus crypt, supporting the idea that mature enterocytes have the greatest capacity for transcellular Ca2+ movement(Reference Centeno, Díaz de Barboza and Marchionatti55).

Another novel protein seems to be crucial in the transcellular Ca2+ pathway. This is the protein 4.1R, which was also first identified in the erythrocyte membrane skeleton and is expressed in the epithelia of the intestine. So far, its physiological function remains unknown. Liu et al. (Reference Liu, Weng and Chen56) have detected that 4.1R co-localises with PMCA1b. These authors have shown that 4.1R KO mice exhibit deterioration in intestinal Ca absorption. In 4.1R KO mice, the expression of PMCA1b in enterocytes was decreased. This finding that the deficiency in the adaptor protein 4.1R produces an impaired intestinal Ca absorption suggests that many yet to be defined molecules might also play important functions in epithelial Ca transport.

Apparently, the intestinal Na+/Ca2+ exchanger (NCX1) is responsible for about 20 % of Ca exit. However, this protein has received little attention and many recent reviews ignore it as another molecule involved in the Ca2+ exit from the intestine. The activity of the intestinal Na+/Ca2+ exchanger depends on the gradient created by Na+/K+-ATPase(Reference Ghijsen, De Jong and Van Os57). There are several isoforms that result from three different genes(Reference Philipson, Nicoll and Matsuoka58), but in the intestine NCX1 is present mainly in the enterocytes. It has been detected in rats(Reference Ghijsen, De Jong and Van Os57), mice(Reference Dong, Sellers and Smith59), chicks(Reference Centeno, Díaz de Barboza and Marchionatti55), horses(Reference Hwang, Jung and Yang60) and dogs(Reference Kim, Yang and Hwang61), but not in rabbits(Reference Hoenderop, Hartog and Stuiver62). NCX1 has a stoichiometry of 3 Na+:1 Ca2+ and can function in either a forward mode (Ca2+ extrusion) or in a reversed mode (Ca2+ entry), depending on the Na+ and Ca gradients and the membrane potential(Reference Blaustein and Lederer63). We have found that the expression and activity of NCX1 are quite similar between mature and immature enterocytes from chick duodenum, but are slightly higher in the villus tip cells(Reference Centeno, Díaz de Barboza and Marchionatti55). Recently, it has been found that the gene expression of NCX1, PMCA1b and CB9k was down-regulated, whereas NCX1 expression was unchanged in the duodenum of a model of hypoxia in pregnant rats that shares clinical similarities with humans suffering from pre-eclampsia or other metabolic diseases. The authors have also found alterations in Ca2+ transporters from the placenta and kidney and showed that these changes caused Ca deficiencies associated with pre-eclampsia(Reference Yang, Lei and Cheng64). As they pointed out, it is quite possible that this study may contribute to a better understanding of the interrelationship between Ca imbalances and metabolic disturbances during pre-eclampsia pathogenesis.

Paracellular pathway

The intestinal epithelium is formed by a continuous layer of individual cells with very narrow spaces between them through which small molecules and ions diffuse(Reference Hoenderop, Nilius and Bindels65). The epithelium must regulate this paracellular pathway for the maintenance of selective permeability. The movement of molecules and ions through this pathway is regulated by tight junctions. They are intercellular structures where plasma membranes of adjacent enterocytes have very close contact. The tight junction proteins are synthesised in the adjacent cells and they include occludin (Ocln) and claudins (Cldns). The latter is a protein family with more than twenty members. Both Ocln and Cldns are integral proteins having the capability of interacting adhesively with complementary molecules on adjacent cells and of co-polymerising laterally(Reference González-Mariscal, Betanzos and Nava66). Ca2+ movement through the tight junctions is a passive process that depends on the concentration and the electric gradient across the epithelium. This transport is non-saturable and mainly occurs in the jejunum and ileum under conditions of adequate or high Ca intake(Reference Bronner and Pansu67). When Ca intake is high, the sojourn time in the intestine is short and there is down-regulation of proteins involved in the transcellular pathway, which switches on the paracellular route(Reference Bronner68) (see Fig. 1).

Fig. 1 Schematic model of transepithelial and paracellular calcium transport in the small intestine. The paracellular calcium pathway is carried out through tight junctions (TJ) by an electrochemical gradient (long arrow between cells). The transcellular calcium pathway consists of three steps: (1) apical entry of calcium through epithelial calcium channels TRPV5 and TRPV6 (the second one is the most abundant in intestine); (2) cytosolic diffusion bound to calbindins (CB); and (3) extrusion across the basolateral membranes by plasma membrane Ca2+-ATPase (PMCA1b) and Na+/Ca2+ exchanger (NCX1). Calcitriol (1,25-D3) stimulates the individual steps of transcellular calcium transport. Calcitriol molecules bind to their nuclear receptors (vitamin D receptors; VDR), and the complex 1,25-D3–VDR interacts with specific DNA sequences inducing transcription and increasing the expression levels of PMCA1b, NCX1, TRPV6 and CB. The real role of the intestinal alkaline phosphatase (AP) enzyme inintestinal calcium absorption has not been elucidated yet. Cldn, claudin.

Hormonal effects

Calcitriol

Calcitriol or 1,25-dihydroxycholecalciferol (1,25(OH)2D3) is the main stimulus for intestinal Ca absorption. It induces changes in the structure and function of intestinal epithelial cells, which enhance Ca transport across the intestine. Calcitriol acts through genomic and non-genomic pathways, after binding to a VDR. Most of the studies have been focused on the effect of calcitriol on the intestinal transcellular Ca pathway. It has been found that the expression or the activity of all molecules presumably involved in this route is increased by calcitriol in experimental animals and even in human subjects(Reference Bouillon, Lieben and Mathieu69Reference Balesaria, Sangha and Walters72). Recently, it has been shown in mice that a single administration of 1,25(OH)2D3 produced a 30-fold maximal increase in the ileal TRPV6 mRNA at 9 h. Multiple dosing of 1,25(OH)2D3 increased the ileal TRPV6 to 200- to 600-fold, being the highest changes observed at the 3rd and 4th doses. TRPV6 protein levels were increased 1·5-fold throughout the duodenum and ileum after the 3rd and 4th doses, while levels in the colon were increased after the 4th dose. These changes in protein correlate with a high TRPV6 mRNA induction in the ileum at the same period(Reference Chow, Quach and Vieth34). The 1,25(OH)2D3-enhanced Ca transport in mice was reported to be inhibited by FGF-23 as well as Ca transport in the colon cancer Caco-2 cells. Fibroblast growth factor-23 (FGF-23) produced an abolishment of the enhanced transcellular active Ca fluxes in both models. Despite the Arrhenius plot indicating that FGF-23 decreased the potential barrier of paracellular Ca movement, FGF-23 was found to modestly down-regulate the 1,25(OH)2D3-enhanced paracellular Ca transport(Reference Khuituan, Wongdee and Jantarajit73). VDR-null mice adapt to pregnancy by up-regulating duodenal TRPV6 and intestinal Ca absorption, which enables a rapid normalisation of bone mineral content. These mice lactate normally and fully restore bone mineral content after weaning. In other words, VDR seems not to be required for skeletal adaptation during pregnancy, lactation and after weaning(Reference Fudge and Kovacs74). In the elderly, there is an age-related decrease in Ca absorption and a higher Ca intake is needed. It seems that increasing Ca intake from dairy products and Ca-fortified foods is much better than supplements. The combination of vitamin D intake to 800 IU (20 μg) daily together with a total Ca intake of 1000 mg daily is a simple and inexpensive strategy that could reduce fractures in aged individuals by 30 %(Reference Gallagher75).

Two studies have demonstrated that calcitriol increases paracellular fluxes across the intestine, mainly in the jejunum and ileum(Reference Sheikh, Schiller and Fordtran76, Reference Karbach77). Fujita et al. (Reference Fujita, Sugimoto and Inatomi78) have demonstrated that 1,25(OH)2D3 significantly increases claudin 2 and claudin 12 mRNA levels in Caco-2 cells. They have also shown that mRNA and protein levels for these proteins were lower at 12 weeks in the jejunum of VDR KO mice in comparison with wild-type mice, and small interfering RNA against these claudins decreased Ca permeability in the Caco-2 cells.

An intestinal calcistat has been hypothesised in order to explain vitamin D deficiency (VDD) with and without the clinical disease. Not all individuals with varying degree of VDD present secondary hyperparathyroidism and decreased bone mineral density (BMD). The intestinal calcistat would control Ca absorption, independently of parathyroid hormone (PTH) levels. A protein called Ca receptor (CaR) would dampen the production of active vitamin D metabolites in intestinal cells and diminish transcellular Ca transport, but would increase the paracellular Ca pathway. This local adaptation would adjust the fractional Ca absorption (FCA) according to the body’s needs. When this local adaptation fails due to decreased Ca intake, decreased 25-hydroxyvitamin D3 (25(OH)D3), CaR mutation, the systemic adaptation comes into play. The systemic adaptations are an increase in PTH and in active vitamin D metabolites. A rise in PTH is the first indication of VDD with a decrease in BMD depending on the duration of VDD. Therefore, individuals with VDD with normal PTH and BMD should be called subclinical VDD(Reference Garg, Kalra and Mahalle79). The beneficial effect of vitamin D supplementation on this group of patients needs to be explored.

Parathyroid hormone

The action of PTH on intestinal Ca absorption occurs by the stimulation of renal CYP27B1 and, hence, increases 1,25(OH)2D3-dependent intestinal Ca absorption. Direct effects of PTH on Ca uptake by enterocytes from rat duodenum have been shown. PTH stimulates enterocyte Ca influx, which could be blocked by the Ca2+ channel antagonists verapamil and nitrendipine(Reference Picotto, Massheimer and Boland80). PTH/PTHrP receptors have been localised in intestinal epithelial cells along the villus(Reference Gentili, Morelli and de Boland81). PTH promotes nuclear effects such as a regulation of gene transcription and cell proliferation of enterocytes(Reference Nemere and Larsson82). So far, a direct effect of PTH on the global process of intestinal Ca absorption has not been reported yet.

Thyroid hormones

Cross et al. (Reference Cross, Debiec and Peterlik83) have demonstrated that thyroid hormone and vitamin D have a cooperative effect on intestinal Ca transport. They have also observed that thyroid hormones increase the genomic action of calcitriol in the intestine. Kumar et al. (Reference Kumar and Prasad84) have reported that hyperthyroid rats show larger Ca uptake by brush-border membrane vesicles and Ca efflux from the basolateral membranes of enterocytes than hypothyroid rats. The authors have also found that the Ca2+-ATPase activity is not altered by thyroid hormones, while NCX1 activity is highly increased.

Growth hormone and insulin-like growth factor-1

Growth hormone (GH) can promote intestinal Ca absorption, which would occur indirectly mediated through an activation of renal CYP27B1 and the increase of serum 1,25(OH)2D3 concentration(Reference Zoidis, Gosteli-Peter and Ghirlanda-Keller85). Fleet et al. (Reference Fleet, Bruns and Hock86) have shown that GH treatment increases intestinal Ca absorption and duodenal CB9k levels in aged rats without increasing serum 1,25(OH)2D3 levels. The effect of GH on Ca absorption is mediated through insulin-like growth factor-1 (IGF-1) and there is evidence that this effect does not depend on vitamin D signalling. Intestinal Ca absorption in adult men has been shown to be positively correlated with IGF-1 and the age-related declines in IGF-1 have a negative impact on Ca absorption, which could not be explained by a decrease in serum 1,25(OH)2D3(Reference Fatayerji, Mawer and Eastell87). Since the vitamin D-independent mechanism by which the GH/IGF-1 axis may regulate intestinal Ca absorption is not clear, this issue needs to be investigated.

Oestrogen

Oestrogen corrects the decline in the efficiency of intestinal Ca absorption at the onset of menopause, as suggested by cell culture studies(Reference Cotter and Cashman88), but the mechanisms that underlie this effect remain unknown. Colin et al. (Reference Colin, Van Den Bemd and Van Aken89) have shown that oestrogen acts independently of 1,25(OH)2D3 in the intestine, while others suggest that oestrogen alters intestinal Ca absorption through the vitamin D endocrine system(Reference Cotter and Cashman88). Most of the oestrogen studies have been done in ovariectomised (OVX) animals. This ablation decreases endogenous oestrogen, but not totally since the adrenal androgens can be aromatised to oestrogen(Reference Bouillon, Carmeliet and Van Cromphaut90). Oestrogen receptor (ER)α KO mice showed a decrease in duodenal TRPV6 mRNA expression, without changes in CB9k, PMCA1b and VDR levels. In contrast, ERβ KO mice did not alter the genes for intestinal Ca absorption. It seems that the genomic effects of oestrogen in mice are mainly mediated by ERα. This idea should not be extrapolated to humans because it has been shown that in normal colon and cancer colon cells the subtype β is the predominant form of the ER(Reference Campbell-Thompson, Lynch and Bhardwaj91). In OVX rats treated with oestradiol, van Abel et al. (Reference van Abel, Hoenderop and van der Kemp92) have found increased duodenal gene expression of TRPV5, TRPV6, CB9k and PMCA1b. They used CYP27B1 KO mice to analyse the calcitriol dependency of the stimulatory effects of oestradiol on intestinal Ca absorption and demonstrated that the oestradiol treatment increased mRNA levels of duodenal TRPV6.

The effect of two dietary phyto-oestrogens (coumestrol and apigenin) as well as ipriflavone, a synthetic phyto-oestrogen, on Ca absorption has been studied in the human Caco-2 cell line(Reference Cotter and Cashman93). A direct effect of these compounds on intestinal Ca absorption was not observed. These controversial results indicate that the mechanism(s) triggered by oestrogen in the intestine requires further investigation(Reference Park and Weaver94).

Glucocorticoids

Osteoporosis is one of the most important side effects after long-term glucocorticoid (GC) treatments. Despite a reduced intestinal Ca absorption being part of the pathogenesis of GC-induced osteoporosis(Reference Reid95), the mechanisms triggered by GC on the intestine are not clear. A short-term GC treatment in young animals does not affect the expression of genes involved in intestinal Ca absorption(Reference Van Cromphaut, Stockmans and Torrekens96), but a sustained dexamethasone suppresses mouse duodenal CB9k expression through the GC receptor pathway(Reference Lee, Choi and Jeung97). It has been also reported that prednisolone for 10 d diminishes rat intestinal Ca absorption through a decreased expression of the active Ca transporters, which occurs independently of 1,25(OH)2D3(Reference Huybers, Naber and Bindels98).

Nutritional factors affecting intestinal calcium absorption

Dietary calcium

Dietary Ca affects the composition of intestinal plasma membranes and alters intestinal Ca transport. All the genes involved in the transcellular pathway are enhanced by a low-Ca diet, which occurs by the activation of the vitamin D endocrine system(Reference Tolosa de Talamoni19, Reference Centeno, Díaz de Barboza and Marchionatti55, Reference Christakos, Dhawan and Liu99, Reference Brown, Krits and Armbrecht100). The increase in the expression and activity of the intestinal Ca pump and NCX1 caused by a Ca-deficient diet occurs in both mature and immature enterocytes. However, VDR levels are decreased by low-Ca diets, independently of the degree of cell differentiation(Reference Centeno, Díaz de Barboza and Marchionatti55). We think that high levels of serum calcitriol provoked by low-Ca diets promote differentiation, which would produce cells more capable of expressing vitamin D-dependent genes required for Ca absorption. The activity of intestinal alkaline phosphatase (AP) is concomitantly increased by dietary Ca restriction, but a real role of this protein in intestinal Ca absorption cannot be discarded(Reference Centeno, Díaz de Barboza and Marchionatti55). Benn et al. (Reference Benn, Ajibade and Porta101) have found that a low-Ca diet increases intestinal Ca absorption in wild-type, TRPV6 KO and CB9k KO mice. This study indicates that the active intestinal Ca absorption occurs in the absence of TRPV6 and CB9k, which challenges the dogma that both proteins are necessary for vitamin D-induced active intestinal Ca transport. Probably, TRPV6 is not the rate-limiting factor in the transcellular pathway or another factor/s is/are involved in its absence to compensate partially its function.

We have also observed that a low-Ca diet increases the reactivity and availability of sulfhydryl groups from intestinal brush-border membrane proteins of chicks(Reference Tolosa de Talamoni, Mykkanen and Wasserman102). It is quite possible that the sulfhydryl status of the brush-border membrane proteins is involved in the vitamin D-dependent intestinal Ca absorption. With regard to the lipid composition, we have detected minor changes in the fatty acid content of the basolateral membrane, but the lipid fluidity of these membranes is highly increased by dietary Ca restriction(Reference Tolosa de Talamoni19).

Recently, it has been reported that luminal Ca controls the intestinal Ca absorption through the modification of intestinal AP activity(Reference Brun, Brance and Rigalli103). Authors have found that luminal Ca concentration increases the activity of AP and simultaneously decreases percentage Ca absorption, acting as a minute-to-minute regulatory mechanism of Ca entry.

Some studies have found an inverse association between Ca intake and adiposity(Reference Zemel, Shi and Greer104Reference Huang and Qi106). However, not all studies have observed this relationship(Reference Barr107). The mechanisms underlying these responses are not quite clear.

Most patients with idiopathic hypercalciuria show an increased intestinal Ca absorption. This has been demonstrated in studies using radiolabelled Ca and comparing the intestinal Ca absorption in patients with idiopathic hypercalciuria with that in normal subjects(Reference Coe, Favus and Asplin108). One possible mechanism for the increased Ca absorption might be the highest levels of serum calcitriol found in these patients as compared with normal individuals(Reference Hoenderop, Nilius and Bindels65). Apparently, this response is independent of Ca intake and reflects an enhancement in active Ca transport by the intestine(Reference Lemann109).

With regard to the risk of kidney calculi formation in postmenopausal women, it is not clear if it is associated with Ca intake and vitamin D supplements. Haghighi et al. (Reference Haghighi, Samimagham and Gohardehi110) have shown that the administration of 1000 mg/d of dietary Ca and vitamin D had a weak association with the formation of kidney calculi (only 1·9 % of fifty-three patients). Jackson et al. (Reference Jackson, LaCroix and Gass111) have reported that in a study with 36282 postmenopausal women treated with 1000 mg of Ca and 400 IU (10 μg) of vitamin D/d, a 17 % higher rate of kidney calculi was shown in the treated group after 7 years of follow-up. In contrast, another study contradicted this result, suggesting no association between Ca and vitamin D consumption and kidney calculi formation(Reference Diaz-Lopez and Cannata-Andia112). A low-Ca diet enhances the absorption of oxalate in the gut from normal individuals and kidney stone formers leading to an increase in urinary oxalate excretion, which has an important role in Ca stone formation(Reference Curhan, Willett and Rimm113, Reference Borghi, Schianchi and Meschi114).

There is evidence that dietary Ca restriction is a risk factor for cancer incidence, particularly for colorectal cancer(Reference Park, Leitzmann and Subar115). By contrast, a high intake of dairy products and Ca intake protects against the distal colon and rectal tumours as compared with the proximal colon and reduces the risk of colon cancer(Reference Tárraga López, Albero and Rodríguez-Montes116). It is also quite possible that a low Ca intake contributes to the development of renal, gastric, pancreatic, ovarian, endometrial and lung cancer as well as multiple myeloma, but there is no conclusive evidence(Reference Peterlik, Grant and Cross117).

Several studies have shown that a poor Ca intake is associated with an increased risk of breast cancer(Reference McCullough, Rodriguez and Diver118Reference Chen, Hu and Xie120). Vergne et al. (Reference Vergne, Matta and Morales121) have recently demonstrated that Ca intake is associated with a high DNA repair capacity level, which in turn decreases the development of breast cancer.

Observational studies have identified an inverse relationship between maternal Ca intake and the incidence of pre-eclampsia(Reference Belizan and Villar122, Reference Belizan, Villar and Repke123). When Ca supplements are administered during pregnancy, the incidence of pre-eclampsia and its consequences are reduced, including severe maternal morbidity and death(Reference Camargo, Moraes and Souza124).

Several meta-analyses of randomised controlled trials have also indicated that Ca supplementation can lead to a small reduction in systolic and diastolic blood pressure; therefore, Ca supplements might be beneficial in patients with hypertension(Reference Griffith, Guyatt and Cook125, Reference van Mierlo, Arends and Streppel126). Varenna et al. (Reference Varenna, Manara and Galli127) have confirmed an association between hypertension and osteoporosis. There is a higher prevalence of hypertension in women with osteoporosis and a higher prevalence of osteoporosis in women with hypertension. The authors suggest that a low dairy Ca intake could be associated with an increased risk of both diseases and be a possible pathogenic link between the two conditions.

It is well known that a reduced intestinal Ca absorption is a risk factor for osteoporosis. There is evidence that the use of Ca supplements decreases bone turnover by about 20 %, and this is associated with a reduction in bone loss in postmenopausal women(Reference Reid, Mason and Horne128). Low Ca intake was significantly associated with low BMD and increased risk of osteoporosis. However, the association between Ca and BMD was not consistently linear, and a sufficient vitamin D level might compensate for the negative influence of low Ca intake on bone(Reference Kim, Choi and Lim129). Zhou et al. (Reference Zhou, Langsetmo and Berger130) have observed that a higher cumulative Ca and vitamin D intake in adult women was associated with better bone health, as indicated by BMD at multiple sites.

Other ions

Aluminium

Several authors have reported that aluminium ions (Al), at pharmacological doses, are able to reduce intestinal Ca absorption via the vitamin D-dependent transcellular pathway in the small intestine of humans and animals(Reference Adler and Berlyne131Reference Orihuela, Meichtry and Pizarro135). Orihuela et al. (Reference Orihuela, Meichtry and Pizarro135) have suggested that Al might interfere with Ca uptake by enterocytes through an effect on cell membranes. In addition, Al would decrease the intestinal glutathione (GSH) level affecting CB function and/or synthesis, which would lead to a reduced transcellular Ca absorption. Apparently, the inhibitory effect of Al varies according to thyroid hormone status(Reference Orihuela136). In late pregnancy and mainly during the middle lactation of rats, Al has also been shown to reduce transcellular Ca absorption in the duodenum by interfering with the mechanisms of Ca transport partially mediated by a high serum level of oestrogen and prolactin(Reference Orihuela137).

Phosphate

Severe dietary P deficiency is rare in humans and occurs only in conditions of severe starvation. Nevertheless, P deficiency can occur in alcoholics, patients with malabsorption syndromes, and those taking excessive amounts of Ca supplements(Reference Heaney and Nordin138). Dietary P deficiency affects intestinal Ca absorption. Under this deficiency, serum 1,25(OH)2D3 content increases(Reference Ribovich and DeLuca139, Reference Gray and Napoli140) as well as the levels of CB and CB mRNA synthesis(Reference Meyer, Tenenhouse and Meyer141). However, in some fashion, a low-P diet can stimulate CB formation and intestinal Ca absorption in the absence of an increased production of 1,25(OH)2D3(Reference Bar and Wasserman142). The reactivity and availability of sulfhydryl groups of chick intestinal brush-border membranes have been also shown to increase by a low-P diet(Reference Tolosa de Talamoni, Mykkanen and Wasserman102), but it remains unknown whether this response is related to increased intestinal Ca absorption. Meyer et al. (Reference Meyer, Fullmer and Wasserman143) have demonstrated that chickens fed a low-P diet displayed an increase in intestinal VDR mRNA. The authors have reported that a complex regulation of VDR expression occurs in that low-P restriction enhances VDR mRNA levels, possibly via increased serum 1,25(OH)2D3.

Magnesium

For several decades, it has been known that there is interaction between Ca2+ and Mg2+ in the intestine, which varies throughout the intestine. Mg2+ has been found to inhibit Ca absorption primarily in the duodenum, whereas Ca inhibits Mg2+ transport in the ileum but not in the duodenum(Reference Hendrix, Alcock and Archibald144). In studies of short-term uptake in rat duodenal mucosa, O’Donnell & Smith(Reference O’Donnell and Smith145) have demonstrated that Mg2+ inhibited the time-dependent uptake of Ca2+, but Ca2+ did not alter Mg2+ uptake. Increasing the Mg2+ concentration to 1·25 mmol/l decreased the mucosal-to-serosal flux of Ca by 50 % and abolished net Ca absorption, mainly due to a depression in the paracellular pathway(Reference Hardwick, Jones and Brautbar146). In patients with idiopathic hypercalciuria and renal Ca stone disease, oral supplementation of Mg2+ has been shown to be favourable because it decreases Ca absorption and increases Mg2+ absorption, which may reduce risk factors for renal Ca stone formation(Reference de Swart, Sokole and Wilmink147). By contrast, Mg2+ deficiency significantly increased enterocyte content of Ca(Reference Planells, Sánchez-Morito and Montellano148). The mechanism is difficult to understand because Mg2+ deprivation has been associated with a low production of 1,25(OH)2D3, the main stimulator of intestinal Ca absorption(Reference Dimai, Porta and Wirnsberger149). However, some studies contradict the previous data. Fine et al. (Reference Fine, Santa Ana and Porter150) have found that increasing dietary Mg2+ had no effect on Ca absorption. Kosakai et al. (Reference Kozakai, Uozumi and Katoh151) have demonstrated in sheep that the apparent Ca absorption tended to increase when the dietary Mg2+ content was increased, which was accompanied without an alteration in the plasma Ca concentration and increased urinary Ca excretion. Bae et al. (Reference Bae, Bu and Kim152) have shown that the consumption of seaweed Ca extract or inorganic calcium carbonate with Mg2+ oxide in OVX rats produced a similar intestinal Ca absorption, but only the seaweed Ca extract caused an increase in femoral BMD and strength in OVX rats. They concluded that seaweed Ca extract is a good Ca and Mg2+ source for improving bone health as compared with synthetic Ca2+ and Mg2+ supplementation.

Lipids

At present, the effects of dietary lipids on intestinal Ca absorption are not clear. Hessov et al. (Reference Hessov, Andersson and Isaksson153) have demonstrated that low-fat-diets increase intestinal Ca absorption in nine patients with fat malabsorption. Steatorrhoea has been associated with an inhibition of intestinal Ca absorption(Reference Haderslev, Jeppesen and Mortensen154). Jewell et al. (Reference Jewell, Cusack and Cashman155) have found that both the paracellular Ca transport and transcellular Ca transport across monolayers of Caco-2 cells were significantly increased after exposure to conjugated linoleic acid. Murphy et al. (Reference Murphy, Jewell and Hooiveld156) have found that zona occludens-1, Ocldn, and Cldn-4 were all up-regulated while Cldn-1 was down-regulated by trans-10, cis-12-conjugated linoleic acid, which explains the increase in the paracellular route. However, they did not find any effect on genes involved in the transcellular pathway, which should be further explored. Fatty acids directly contribute to increasing intestinal Ca absorption via a cation exchange mechanism between cellular H+ for luminal Ca2+ favoured by exchanger activity 2H+/Ca2+(Reference Coxam157). Fatty acids would increase the proliferation of colonic epithelial cells, producing a trophic effect on the mucosa; they contribute to increasing the absorptive surface(Reference Raschka and Daniel158, Reference Lobo, Cocato and Jorgetti159).

Recently, in high-fat diet-fed mice, Xiao et al. (Reference Xiao, Cui and Shi160) have found a marked decrease in the intestinal Ca absorption, which is accompanied by redox imbalance and increased duodenal oxidative damage, an effect that was avoided by lipoic acid or Ca supplementation. In addition, they have also shown that a high-fat diet down-regulates the gene expression of molecules involved in the transcellular pathway of intestinal Ca absorption, independently of calcitriol regulation. The authors think that the main cause for high-fat-diet-induced inhibitory intestinal Ca absorption is not Ca soap but duodenal oxidative stress.

Carbohydrates

Lactose is a disaccharide found in milk and dairy products that enhances Ca homeostasis, which should be beneficial for bone health(Reference Buchowski and Miller161). It is well established that lactose stimulates the intestinal Ca absorption that seems to occur by passive transport in the small intestine(Reference Dupuis, Tardivel and Porembska162). Apparently, lactose promotes intestinal Ca absorption, independently of the vitamin D endocrine system. Natsuko et al. (Reference Sogabe, Mizoi and Asahi163) have found that lactose alters intestinal AP in rats not only in a direct way, but also indirectly through a regulation of intestinal AP expression, mainly in the jejunum. Epilactose (a rare disaccharide in cows’ milk) has been demonstrated to increase Ca transport in everted small-intestinal sacs by promoting the generation of SCFA and other organic acids(Reference Nishimukai, Watanabe and Taguchi164). Later on, the same group of investigators has found that the epilactose-mediated promotion of intestinal Ca absorption involves the paracellular route in the rat small intestine through the induction of myosin regulatory light chain phosphorylation via myosin light chain kinase and Rho-associated kinase(Reference Suzuki, Nishimukai and Shinoki165). They have also shown that epilactose improves intestinal Ca absorption in gastrectomised rats. They think that the resulting SCFA production by intestinal microbes is responsible for this effect, as well as the increase in the caecal mucosa area and the soluble Ca concentration(Reference Suzuki, Nishimukai and Takechi166).

Some sugar alcohols such as erythritol, xylitol, sorbitol, maltitol, palatinit or lactitol have been found to enhance Ca transport from rat small and large intestine epithelium in vitro. Differences in Ca transport were shown in different segments of the intestine, but not between the sugar alcohols tested(Reference Mineo, Hara and Tomita167). Recently, Xiao et al. (Reference Xiao, Li and Min168) have found that mannitol improves the absorption and retention of Ca2+ and Mg2+ in growing rats, an effect that occurs through the fermentation of mannitol in the caecum.

Prebiotics

The fortification of milk with milk Ca or Ca salts is among the strategies suggested to increase Ca intake or absorption or both, but the availability of Ca salts in milk has not been well characterised(Reference López-Huertas, Teucher and Boza169). In addition, several food ingredients such as fructo-oligosaccharides and caseinophosphopeptides (CPP) have been proposed as enhancers of the absorption of Ca from milk or other foods. Fructo-oligosaccharides belong to the group of non-digestible oligosaccharides (NDOs), which includes inulin, oligofructose and galacto-oligosaccharides. They can be digested by the colonic microflora, which produces SCFA that decreases intestinal pH and increases Ca solubility leading to an enhancement of paracellular and transcellular Ca transport(Reference Scholz-Ahrens, Schaafsma and van den Heuvel170, Reference Suzuki and Hara171). It has also been suggested that NDOs increase active Ca transport by the activation of CB9k(Reference Takasaki, Inaba and Ohta172). Fukushima et al. (Reference Fukushima, Aizaki and Sakuma173) have demonstrated in rats that fructo-oligosaccharide consumption increases the gene expression of TRPV6 and CB9k through the SCFA formed in the fermentation. The fibres formed mainly by inulin have positive chronic effects on Ca metabolism related to changes in the intestine, resulting in improvement of bone health(Reference Legette, Lee and Martin174). In addition, galacto-oligosaccharides have also potential for improving mineral balance and bone properties(Reference Weaver, Martin and Nakatsu175). With regard to CPP, originating from casein digestion, it has been demonstrated that their addition to Ca-fortified milk increases intestinal Ca absorption in growing rats(Reference Tsuchita, Suzuki and Kuwata176), but the mechanisms involved in this response were not elucidated. Erba et al. (Reference Erba, Ciappellano and Testolin177) have studied the influence of different four CPP/Ca ratios and three mineral concentrations on the amount of passive Ca absorbed across the everted distal small intestine of rats. The positive effect was dependent on the relative amount of both species in the intestinal lumen, the ratio 15 being the most efficient at increasing mineral transport. Nevertheless, in a randomised cross-over trial undertaken in fifteen adults, no effect of CPP was found on intestinal Ca absorption(Reference Teucher, Majsak-Newman and Dainty178). Cosentino et al. (Reference Cosentino, Gravaghi and Donetti179) have demonstrated that both intestinal human HT-29 and Caco-2 cells have the ability to take up extracellular Ca under CPP stimulation. Recently, Colombini et al. (Reference Colombini, Perego and Ardoino180) did not find effects of CPP on paracellular Ca absorption and on TRPV6 mRNA expression in intestinal human HT-29 and Caco-2 cell lines.

A recent study has shown that the daily intake of soluble maize fibre, a well-tolerated prebiotic fibre, increases short-term Ca absorption in adolescents consuming less than the recommended amounts of Ca(Reference Whisner, Martin and Nakatsu181).

Probiotics

Probiotics are viable microbes that alter the microflora in a compartment of the host exerting beneficial health effects in this host(Reference Schrezenmeir and de Vrese182). Most probiotic products contain lactic acid-producing bacteria, which mainly belong to the genera Lactobacillus and Bifidobacterium. In growing rats, it has been demonstrated that probiotic yoghurt containing strains of Lactobacillus casei, L. reuteri and L. gasseri increase intestinal Ca absorption and bone mineral content(Reference Ghanem, Badawy and Abdel-Samam183). Besides, it has been observed that in Caco-2 cells, the probiotic L. salivarius causes an increase in Ca2+ uptake(Reference Gilman and Cashman184).

Synbiotics

Synbiotics are defined as products containing prebiotics and probiotics, in which the prebiotic compound favours the probiotic compound(Reference Schrezenmeir and de Vrese182). It has been shown in OVX rats that intestinal Ca absorption tended to be higher in the synbiotic group and was significantly higher in the prebiotic group in comparison with the control group(Reference Scholz-Ahrens, Ade and Marten185).

Proteins

Proteins are essential to bone, but the Ca-wasting effect of a high protein intake constitutes a point of debate. It has been known for many years that increasing dietary protein enhances urinary Ca either in human subjects or in rats(Reference Kerstetter, O’Brien and Insogna186). The idea was that the additional Ca excretion was of skeletal origin as a result of buffering in bones the metabolic acid load imposed by higher protein intake(Reference Johnson, Alcantara and Linkswiler187). The theory was that a high-protein diet, mainly meat, creates a higher acid load due to the high content of amino acids containing sulfur. This acid load cannot be neutralised by the kidneys and the body pulls Ca2+ from the skeleton to balance pH at the expense of bone, causing an enhancement of urinary Ca(Reference Darling, Millward and Torgerson188). However, clinical studies have demonstrated that a short-term high-protein diet (2·1 g/kg) significantly increases the intestinal Ca absorption as compared with a medium-protein diet (1 g/kg) and the increment in urinary Ca is quantitatively explained by an increase in intestinal Ca absorption efficiency(Reference Kerstetter, O’Brien and Caseria189). The transcellular route, the paracellular pathway or a combination of both mechanisms of intestinal Ca absorption might be involved in response to high dietary protein. By using duodenal brush-border membrane vesicles, Gaffney-Stomberg et al. (Reference Gaffney-Stomberg, Sun and Cucchi190) have demonstrated that the transcellular component of Ca absorption was accelerated in rats fed a high-protein diet, which was due to an enhancement in maximum velocity, without affecting the Michaelis–Menten constant. However, they did not study whether the gene or protein expression of the molecules involved in the transcellular pathway was modified. They did not find increased bone resorption or changes in serum PTH and calcitriol levels.

Since more research is necessary to resolve the protein debate, it has been suggested not to reduce the protein intake below the dietary reference intake because it could be detrimental to bone health, especially in old individuals(Reference Skipper191), and protein intakes and balance of different protein sources with a variety of different foods constitutes appropriate dietary advice(Reference Marcason192).

Black tea

Black tea (Camellia sinensis) is a medicinal plant with a rich flavonoid content and a plethora of health-promoting effects(Reference Das, Das and Mukherjee193, Reference Sharma and Rao194). Das et al. (Reference Das, Banerjee and Das195) have studied the ability of black tea extract as a suitable alternative adjunct for Ca supplementation in treating an OVX rat model of early osteoporosis. The results suggest that black tea could stimulate intestinal Ca absorption, which is associated with an increased activity of AP and Ca2+-ATPase. Black tea’s effectiveness in maintaining bone health was detected to be similar to 17β-oestradiol. Therefore, this study suggests that a simultaneous use of black tea is promising as a prospective candidate for adjunctive therapies for Ca supplementation in the early stage of menopausal bone changes.

Coffee

Coffee drinking is a popular habit worldwide. It is consumed in considerable amounts every day. Caffeine, a methylxanthine present in coffee, has been considered to be responsible for an increased risk of osteoporosis in coffee drinkers(Reference Hernandez-Avila, Colditz and Stampfer196, Reference Welch, Bingham and Reeve197). At present, data are inconsistent(Reference Cooper, Atkinson and Wahner198Reference Hallström, Byberg and Glynn200), hence, the effect of caffeine on intestinal Ca absorption is not well established. It has been demonstrated in rats that intestinal Ca absorption is stimulated by the increase in 1,25(OH)2D3 production after chronic administration of caffeine(Reference Yeh and Aloia201). In addition, urinary and faecal excretion is also increased. In postmenopausal osteoporotic women, a coffee intake in excess of 1000 ml could induce an extra Ca loss of 1·6 mmol Ca/d, while 1–2 cups of coffee/d would have little impact on Ca balance(Reference Hasling, Søndergaard and Charles202). Metabolic balance studies show a weak negative effect of caffeine on the efficiency of intestinal Ca absorption. However, the effect of caffeine is small enough to be fully offset by 1–2 tablespoons (15–30 ml) of milk(Reference Heaney and Nordin138). A recent study in OVX rats has shown that low to moderate caffeine intake may exert some beneficial effects on the skeleton, increasing bone mineralisation, and improving the strength and structure of cancellous bone and the mechanical properties of compact bone; however, it did not cause any significant effect in rats with normal oestrogen levels(Reference Folwarczna, Pytlik and Zych203). The continuous debate has weakened interest in the study of coffee as a risk factor for osteoporosis, which has been reinforced by the non-inclusion of coffee in the list of risk factors in the predictive scale for fracture informed by the WHO(Reference Cano-Marquina, Tarín and Cano204). The understanding of physiological effects of coffee consumption is difficult because of the vast array of components included in the brewed product and the varied effects of each compound. Recently, an unfavourable effect of trigonelline, an alkaloid present in coffee, has been demonstrated on bone mechanical properties in oestrogen-deficient rats, but not in control rats(Reference Folwarczna, Zych and Nowińska205). It is quite possible that the effects on intestinal Ca absorption and the Ca economy by caffeine or other bioactive compounds present in coffee depend on the amount and frequency of coffee intake. This is another issue that merits to be more investigated due to the considerable number of coffee drinkers and the rising life expectancy that will increase bone disorders in the next years.

Pharmacological compounds altering intestinal calcium absorption

Almost two decades ago, we reported that the intactness of the steady-state levels of intestinal glutathione (GSH) seemed to be critical for Ca absorption. By using dl-buthionine-(S,R)-sulfoximine (BSO), a specific inhibitor of γ-glutamylcysteine synthetase, we have shown that the Ca transfer from lumen to blood in vitamin D-supplemented chicks was inhibited, an effect that did not occur in vitamin D-deficient chicks. At that time we concluded that the effects of BSO on intestinal Ca absorption were dependent on the vitamin D status of the animal. One explanation that we gave was that GSH depletion might increase the reactive oxygen species and other substances that could deteriorate intestinal Ca absorption(Reference Tolosa de Talamoni19). Since intestinal AP was one of the best candidates to suffer oxidative stress(Reference Lowe, Strauss and Alpers206), we later studied the effect of BSO on the activity of this enzyme in chicks fed a commercial diet. In fact, the AP activity declined after BSO treatment, which was dose and time dependent. The effect occurred either in vivo or in vitro but was not direct; it was first necessary to deplete GSH in order to produce free hydroxyl radicals and an increment in the protein carbonyl content. The reversibility of the BSO effect was proved by the addition of GSH monoester to the duodenal loop(Reference Marchionatti, Alisio and Díaz de Barboza207).

Menadione (MEN) is another pharmacological compound that alters chick intestinal Ca absorption. It is a quinone that is clinically relevant because of its anti-tumour properties(Reference Chiou and Tzeng208) and its use in the treatment of osteoporosis(Reference Shiraki, Shiraki and Aoki209). MEN metabolism involves redox cycling, resulting in the release of various reactive oxygen species including free hydroxyl radicals(Reference Sata, Klonowski-Stumpe and Han210). We have demonstrated 30 min after a single large dose of MEN that the intestinal Ca absorption was inhibited, which lasted for 9 h. The inhibition affected the transcellular pathway as judged by the inhibition of Ca2+ pump activity, the main protein involved in Ca2+ extrusion from the enterocyte to the lamina propria. Intestinal AP activity was also inhibited, but not that from other brush-border membrane enzymes. GSH depletion, enhancement in the protein carbonyl content as well as the appearance of free hydroxyl radicals were indications that MEN caused oxidative stress provoking deleterious consequences on intestinal Ca absorption. The oral administration of GSH monoester prevented the inhibition of intestinal Ca absorption and the GSH deprivation produced by MEN(Reference Marchionatti, Díaz de Barboza and Centeno211). As mitochondria are the major source of reactive oxygen species(Reference Higuchi212), we have investigated the role of these organelles in the inhibition of the intestinal Ca absorption caused by MEN. The quinone produced mitochondrial dysfunction as shown by inhibition of enzymes from Krebs’ cycle, DNA fragmentation, release of cytochrome c, alteration of membrane potential and enhancement of Mn2+-superoxide dismutase activity. The mitochondrial dysfunction would be a consequence of mitochondrial GSH depletion, which would alter the membrane permeability triggering the release of apoptotic molecules leading to DNA fragmentation. The oxidant effects would alter the transcellular Ca pathway affecting the global process of intestinal Ca absorption(Reference Marchionatti, Perez and Diaz de Barboza213). Since the flavonol quercetin has antioxidant properties(Reference Suzuki and Hara214), we have studied the ability of quercetin to protect the chick intestine against the inhibition of intestinal Ca absorption caused by MEN. Effectively, quercetin abrogated the inhibitory effect of MEN on chick intestinal Ca absorption through the restoration of intestinal redox state, blockage of alterations in the mitochondrial membrane permeability, and abolition of the FasL/Fas/caspase-3 signalling pathway activation(Reference Marchionatti, Pacciaroni and Tolosa de Talamoni215). In addition, the hormone melatonin (MEL) was also able to restore chick intestinal Ca absorption inhibited by MEN. MEL is known as a direct scavenger of free radicals with the ability to remove singlet oxygen, the superoxide anion radical and hydroperoxide. It has also an indirect antioxidant action through a modulation of antioxidant enzyme activities. MEL by itself did not alter intestinal Ca absorption and other variables influencing that process. The MEL protective mechanism seems to be switched on under oxidative stress conditions produced by MEN, leading cells to the normal redox status. MEL administration after MEN injection returned rapidly the intestinal GSH and protein carbonyl contents to control values as well as the SOD and CAT activities. Concomitantly, intestinal Ca absorption went up to normal values, suggesting that the restoration of redox status of the gut by MEL allowed the recovering of the intestinal capability to absorb the cation properly. MEL not only normalised the redox status of the enterocytes but also rescued the epithelial cells from MEN-induced apoptosis(Reference Carpentieri, Marchionatti and Areco216). Therefore, MEL could be a potential drug of choice for the treatment of impaired intestinal Ca absorption caused by oxidative stress and exacerbated apoptosis, which occurs in certain pathophysiological conditions (ageing, coeliac disease, intestinal bowel disease, cancer and others) or after intake of drugs causing oxidation.

We have also shown that a single high concentration of sodium deoxycholate (NaDOC) inhibits intestinal Ca absorption through a down-regulation of proteins involved in the transcellular pathway, as a consequence of triggering oxidative stress and mitochondria-mediated apoptosis(Reference Rivoira, Marchionatti and Centeno217). This inhibitory effect on intestinal Ca absorption produced by NaDOC has been shown to be abolished by the concomitant use of the antioxidant quercetin, which clearly indicates that the response of NaDOC was mediated by the oxidative stress. Deoxycholic acid or its salt, NaDOC, is the major secondary bile acid in humans and is toxic in high concentrations causing liver damage during cholestasis and acting as a promoter of colon cancer in experimental animals(Reference Lamireau, Zoltowska and Levy218). It is well known that its concentration varies according to the diet; a high-fat diet is associated with an increased secretion of NaDOC(Reference Kawano, Ishikawa and Kamano219). This bile salt perturbs the membrane structures by alteration of membrane microdomains and decreases the transepithelial electrical resistance in the Caco-2 cell line through reactive oxygen species generation and other signalling mechanisms(Reference Jean-Louis, Akare and Ali220, Reference Araki, Katoh and Ogawa221). Therefore, the tight junctions constitute another target of NaDOC in the intestine, suggesting that the paracellular pathway of intestinal Ca absorption might be also affected by this bile salt. Based on the knowledge that a minor bile acid, ursodeoxycholic acid (UDCA), has beneficial effects of protection against cytotoxicity due to more toxic bile acids, we have tried to ascertain the potentiality of UDCA to prevent the inhibition of intestinal Ca absorption caused by NaDOC. In addition, we have studied the effects of UDCA alone on intestinal Ca absorption either in chicks or rats. The data have shown that UDCA not only prevented the inhibition of intestinal Ca absorption caused by NaDOC either in chicks or rats, but also UDCA alone enhanced that process. This was an unpredictable finding, which together with the previous data indicate that NaDOC is a bad whereas UDCA is a good bile acid for intestinal Ca absorption. The interesting point is that the combination of both bile acids neutralises the response of each other, probably because UDCA protects the intestine against the GSH depletion and protein carbonyl increment produced by NaDOC. Both NaDOC and UDCA altered protein and gene expression of molecules involved in the transcellular pathway of intestinal Ca absorption, but in the opposite way. NaDOC decreased the protein expression of PMCA1b, NCX1and CB, whereas UDCA increased the protein expression of all of them. The expression of these molecules was identical to those from the control group when the combined treatment was used. The gene expression of pmca1b, ncx1 and cb was increased by UDCA and UDCA + NaDOC. In contrast, NaDOC decreased the gene expression of pmca1b and cb without modifying that of ncx1. UDCA also increased the protein and gene expression of VDR, which suggests that VDR is involved in the enhancement of intestinal Ca absorption produced by UDCA(Reference Rodríguez, Rivoira and Marchionatti222). The relationship between UDCA and VDR is not surprising because it has been demonstrated that VDR also binds bile acids(Reference Makishima, Lu and Xie223, Reference Krasowski, Ni and Hagey224) and the increase in the cathelicidin expression in biliary epithelial cells from human liver caused by UDCA is mediated by VDR activation, an effect that is blunted by a small interfering RNA strategy(Reference D’Aldebert, Biyeyeme Bi Mve and Mergey225).

There are conflicting data with regard to the effects of proton pump inhibitors and osteoporotic fracture risk. Presumably, they increase the risk through hypochlorhydria and decreased FCA. Hansen et al. (Reference Hansen, Jones and Lindstrom226) have evaluated the effect of proton pump inhibitor therapy on FCA using the dual stable isotope method. Participants underwent three 24 h FCA studies; two of them were accomplished 1 month apart to establish the baseline of FCA, the third one was after taking omeprazole (40 mg/d for 30 d). The data revealed that age, gastric pH, serum omeprazole levels, adherence to omeprazole and 25-hydroxyvitamin D levels were not related to changes in FCA between visits 2 and 3. The level of serum 1,25(OH)2D3 was the only variable associated with the change in FCA between visits 2 and 3. More studies are necessary to elucidate the mechanisms by which proton pump inhibitors increase osteoporotic fracture risk.

Wahl et al. (Reference Wahl, Gobuty and Lukert227) have demonstrated reduced FCA in patients under anticonvulsant treatment. The possible mechanism underlying this process is complex. It has been demonstrated that phenytoin and carbamazepine inhibit active Ca transport from the apical to the basolateral side of Caco-2 cells under physiological Ca conditions and vitamin D improves the anti-epileptic drug-induced decrease in Ca permeability(Reference von Borstel Smith, Crofoot and Rodriguez-Proteau228).

Restraint stress significantly down-regulates the mRNA expressions of TRPV6 and Cav1.3, CB-D9k, and PMCA1b, but not the expression of TRPV5 or NCX1. In contrast, the mRNA expressions of paracellular genes, ZO-1, occludin and claudin-3, are not modified by restraint stress. Since several antidepressant or anxiolytic drugs alleviate stress-induced depressive and anxiety symptoms, Charoenphandhu et al. (Reference Charoenphandhu, Teerapornpuntakit and Lapmanee229) have hypothesised that these drugs might also enhance Ca transporter gene expression in stressed rats. In fact, a 4-week daily administration of 10 mg/kg fluoxetine, 10 mg/kg reboxetine or 10 mg/kg venlafaxine differentially increased the duodenal Ca transporter genes in stressed rats, whereas 2 mg/kg diazepam had no such effect. These findings might be applied to help ameliorate the stress-induced bone loss and osteoporosis by restoring intestinal Ca absorption.

Octyphenol, a degradative product used to produce rubber, pesticides and paints, and bisphenol A (BPA), an organic compound used for manufacturing polycarbonate plastic and epoxy resins, are known as endocrine disruptors. The effect of both on serum Ca levels and expressions of Ca transport genes in the duodenum and kidney was studied in pregnant mice. Either octyphenol or BPA decreased serum Ca levels. Both drugs decreased the levels of TRPV5 and CB9k in the kidney and the levels of TRPV6 in the duodenum. Gene expression and protein expression of CB9k were decreased in the duodenum by BPA but increased by octyphenol at high doses. These results indicate that decreased serum Ca levels caused by these disruptors might be a consequence of the alteration in the expression of genes related to Ca transport(Reference Kim, An and Yang230).

Gene × diet interactions influence intestinal calcium absorption

Dietary Ca restriction increases the efficiency of intestinal Ca absorption, but the impact of genetics on this adaptive response is not clear. In humans, the efficiency of intestinal Ca absorption varies from 7 to 75 %(Reference Alevizaki, Ikkos and Singhelakis231). The large variation is probably owing to the influence of multiple physiological factors (for example, growth, pregnancy, lactation, ageing) and environmental variables (for example, dietary Ca intake, vitamin D). Little information is available for the impact of genetics on the efficiency of intestinal Ca absorption and the adaptive up-regulation of Ca absorption to a low dietary Ca intake. Two laboratories have studied the efficiency of intestinal Ca absorption in different mice and have found that it is higher in C3H/HeJ mice in comparison with C57BL/6J mice, which suggests that genetic background might influence this trait(Reference Chen and Kalu232). In addition, racial differences in the ability of adolescent girls to increase Ca absorption efficiency during a low Ca intake also indicate that this adaptive response has a genetic component(Reference Wu, Martin and Braun233, Reference Weaver, McCabe and McCabe234) In agreement with this concept, adolescent black girls have been shown to exhibit higher intestinal Ca absorption as compared with white girls(Reference Bryant, Wastney and Martin235), and this may contribute to the higher bone deposition found in black girls(Reference Braun, Palacios and Wigertz236).

Replogle et al. (Reference Replogle, Li and Wang237) have examined eleven inbred lines of mice fed on defined diets containing either high or low Ca concentration from weaning to 12 weeks of age. The authors have shown that genetic variation and gene × diet interactions affect not only the active intestinal Ca absorption, but also its relationship to bone. These interactions are partially explained by variations in the traditional cellular mediators (i.e. TRPV6, CB9k, PMCA1b mRNA) and in the main hormonal regulator, 1,25(OH)2D3, of intestinal Ca absorption. This field is relatively new, hence many efforts are required to bring more light for the understanding of this knowledge.

Conclusions

Ca2+ is an ion involved in multiple physiological functions. Absorption through the intestinal epithelium is a complex process regulated by an intricate network of hormones and nutritional factors. At present, the Western diet model adopted is poor in Ca content and at the same time interferes with the proper absorption of the cation. Some diseases such as osteoporosis, hypertension and cancer are associated with dietary Ca restriction. Current recommendations are to obtain Ca from the diet in preference to supplements since dietary Ca intake has not been associated with the adverse effects of supplements, probably because Ca is provided in smaller boluses absorbed more slowly(Reference Reid238). Milk and dairy products are the best sources of Ca. There have been advances in food industry attempts to compensate for the Ca shortage through the introduction of prebiotics and probiotics in the basic diet. Certain dietary habits such as an increased protein intake remain a point of debate. It is important to take into account that alterations in the redox state of the intestinal epithelium produced by some medications such as MEN, BSO and UDCA also modify the intestinal Ca absorption. Therefore, intestinal Ca absorption must be carefully attended, so it is necessary to consider not only the intake, but also possible interactions with other ions, the genetic background, the effect of diet and the use of certain medications. Health professionals should be aware of this knowledge in order to develop nutritional or medical strategies to stimulate the efficiency of intestinal Ca absorption and to prevent diseases.

Acknowledgements

This work was supported by grants from the Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET; PIP 2013–15) and SECYT (UNC), Argentina. N. T. T., A. C. and V. R. are members of Investigator Career from CONICET. V. A. is a recipient of a fellowship from CONICET.

There are no conflicts of interest to declare.

References

1.Ma, J, Johns, RA & Stafford, RS (2007) Americans are not meeting current calcium recommendations. Am J Clin Nutr 85, 13611366.CrossRefGoogle Scholar
2.Eisner, V, Csordás, G & Hajnóczky, G (2013) Interactions between sarco-endoplasmic reticulum and mitochondria in cardiac and skeletal muscle – pivotal roles in Ca2+ and reactive oxygen species signaling. J Cell Sci 15, 29652978.Google Scholar
3.Szadujkis-Szadurska, K, Szadujkis-Szadurski, R, Szadujkis-Szadurski, L, et al. (2010) The role of calcium in modulating the reactivity of the smooth muscle cells during ischemia/reperfusion. Part 1. Postepy Hig Med Dosw 15, 188194.Google Scholar
4.Koklic, T, Majumder, R & Lentz, BR (2014) Ca2+ switches the effect of PS-containing membranes on factor Xa from activating to inhibiting: implications for initiation of blood coagulation. Biochem J 462, 591601.CrossRefGoogle ScholarPubMed
5.Chaigne-Delalande, B & Lenardo, MJ (2014) Divalent cation signaling in immune cells. Trends Immunol 35, 332344.CrossRefGoogle ScholarPubMed
6.Zemel, MB (2001) Calcium modulation of hypertension and obesity: mechanisms and implications. J Am Coll Nutr 20, 428S435S.CrossRefGoogle ScholarPubMed
7.Appel, LJ, Brands, MW, Daniels, SR, et al. (2006) Dietary approaches to prevent and treat hypertension: a scientific statement from the American Heart Association. Hypertension 47, 296308.CrossRefGoogle ScholarPubMed
8.Laraichi, S, Parra, P, Zamanillo, R, et al. (2013) Dietary supplementation of calcium may counteract obesity in mice mediated by changes in plasma fatty acids. Lipids 48, 817826.CrossRefGoogle ScholarPubMed
9.Bartlett, PJ, Gaspers, LD, Pierobon, N, et al. (2014) Calcium-dependent regulation of glucose homeostasis in the liver. Cell Calcium 55, 306316.CrossRefGoogle ScholarPubMed
10.Lin, J, Manson, JE, Lee, IM, et al. (2007) Intakes of calcium and vitamin D and breast cancer risk in women. Arch Intern Med 167, 10501059.CrossRefGoogle ScholarPubMed
11.Ju, J, Kwak, Y, Hao, X, et al. (2012) Inhibitory effects of calcium against intestinal cancer in human colon cancer cells and Apc Min/+ mice. Nutr Res Pract 6, 396404.CrossRefGoogle Scholar
12.Williams, CD, Whitley, BM, Hoyo, C, et al. (2012) Dietary calcium and risk for prostate cancer: a case–control study among US veterans. Prev Chronic Dis 9, 110125.CrossRefGoogle ScholarPubMed
13.Merritt, MA, Cramer, DW, Vitonis, AF, et al. (2013) Dairy foods and nutrients in relation to risk of ovarian cancer and major histological subtypes. Int J Cancer 132, 11141124.CrossRefGoogle ScholarPubMed
14.Fleet, JC & Schoch, RD (2010) Molecular mechanisms for regulation of intestinal calcium absorption by vitamin D and other factors. Crit Rev Clin Lab Sci 47, 181195.CrossRefGoogle ScholarPubMed
15.Bronner, F (1998) Calcium absorption – a paradigm for mineral absorption. J Nutr 128, 917920.CrossRefGoogle ScholarPubMed
16.Wasserman, RH (2004) Vitamin D and the dual processes of intestinal calcium absorption. J Nutr 134, 31373139.CrossRefGoogle ScholarPubMed
17.Marcus, CS & Lengemann, FW (1962) Absorption of Ca45 and Sr85 from solid and liquid food at various levels of the alimentary tract of the rat. J Nutr 77, 155160.CrossRefGoogle ScholarPubMed
18.Wali, RK, Baum, CL, Sitrin, MD, et al. (1990) 1,25(OH)2 vitamin D3 stimulates membrane phosphoinositide turnover, activates protein kinase C, and increases cytosolic calcium in rat colonic epithelium. J Clin Invest 85, 12961303.CrossRefGoogle Scholar
19.Tolosa de Talamoni, N (1996) Calcium and phosphorous deficiencies alter the lipid composition and fluidity of intestinal basolateral membranes. Comp Biochem Physiol A Physiol 115, 309315.CrossRefGoogle ScholarPubMed
20.Nordin, BE, Need, AG, Morris, HA, et al. (2004) Effect of age on calcium absorption in postmenopausal women. Am J Clin Nutr 80, 9981002.CrossRefGoogle ScholarPubMed
21.O’Brien, KO, Nathanson, MS, Mancini, J, et al. (2003) Calcium absorption is significantly higher in adolescents during pregnancy than in the early postpartum period. Am J Clin Nutr 78, 11881193.CrossRefGoogle ScholarPubMed
22.Prentice, A (2000) Maternal calcium metabolism and bone mineral status. Am J Clin Nutr 71, 1312S1316S.CrossRefGoogle ScholarPubMed
23.Zhu, Y, Goff, JP, Reinhardt, TA, et al. (1998) Pregnancy and lactation increase vitamin D-dependent intestinal membrane calcium adenosine triphosphatase and calcium binding protein messenger ribonucleic acid expression. Endocrinology 139, 35203524.CrossRefGoogle ScholarPubMed
24.Liesegang, A, Riner, K & Boos, A (2007) Effects of gestation and lactation on vitamin D receptor amounts in goats and sheep. Domest Anim Endocrinol 33, 190202.CrossRefGoogle ScholarPubMed
25.Wongdee, K, Teerapornpuntakit, J, Siangpro, C, et al. (2013) Duodenal villous hypertrophy and upregulation of claudin-15 protein expression in lactating rats. J Mol Histol 44, 103109.CrossRefGoogle ScholarPubMed
26.Teerapornpuntakit, J, Klanchui, A, Karoonuthaisiri, N, et al. (2014) Expression of transcripts related to intestinal ion and nutrient absorption in pregnant and lactating rats as determined by custom-designed cDNA microarray. Mol Cell Biochem 391, 103116.CrossRefGoogle ScholarPubMed
27.van Abel, M, Hoenderop, JG & Bindels, RJ (2005) The epithelial calcium channels TRPV5 and TRPV6: regulation and implications for disease. Naunyn Schmiedebergs Arch Pharmacol 371, 295306.CrossRefGoogle ScholarPubMed
28.Bianco, SD, Peng, JB & Takanaga, H (2007) Marked disturbance of calcium homeostasis in mice with targeted disruption of the Trpv6 calcium channel gene. J Bone Miner Res 22, 274285.CrossRefGoogle ScholarPubMed
29.Cui, M, Li, Q, Johnson, R, et al. (2012) Villin promoter-mediated transgenic expression of transient receptor potential cation channel, subfamily V, member 6 (TRPV6) increases intestinal calcium absorption in wild-type and vitamin D receptor knockout mice. J Bone Miner Res 27, 20972107.CrossRefGoogle ScholarPubMed
30.Den Dekker, E, Hoenderop, JG, Nilius, B, et al. (2003) The epithelial calcium channels, TRPV5 & TRPV6: from identification towards regulation. Cell Calcium 33, 497507.CrossRefGoogle ScholarPubMed
31.Hoenderop, JG, Voets, T, Hoefs, S, et al. (2003) Homo- and heterotetrameric architecture of the epithelial Ca2+ channels TRPV5 and TRPV6. EMBO J 17, 776785.CrossRefGoogle Scholar
32.Hoenderop, JG, Vennekens, R & Müller, D (2001) Function and expression of the epithelial Ca2+ channel family: comparison of mammalian ECaC1 and 2. J Physiol 15, 747761.CrossRefGoogle Scholar
33.Walters, JR, Balesaria, S, Chavele, KM, et al. (2006) Calcium channel TRPV6 expression in human duodenum: different relationships to the vitamin D system and aging in men and women. J Bone Miner Res 21, 17701777.CrossRefGoogle Scholar
34.Chow, EC, Quach, HP, Vieth, R, et al. (2013) Temporal changes in tissue 1α,25-dihydroxyvitamin D3, vitamin D receptor target genes, and calcium and PTH levels after 1,25(OH)2D3 treatment in mice. Am J Physiol Endocrinol Metab 304, E977E989.CrossRefGoogle ScholarPubMed
35.Tolosa de Talamoni, N, Pérz, A & Alisio, A (1998) Effect of cholecalciferol on intestinal epithelial cells. Trends Comp Biochem Physiol 5, 179185.Google Scholar
36.Schwaller, B (2010) Cytosolic Ca2+ buffers. Cold Spring Harb Perspect Biol 2, a004051.CrossRefGoogle ScholarPubMed
37.Cheung, WT, Richards, DE & Rogers, JH (1993) Calcium binding by chick calretinin and rat calbindin D28k synthesised in bacteria. Eur J Biochem 15, 401410.CrossRefGoogle Scholar
38.Nägerl, UV, Novo, D, Mody, I, et al. (2000) Binding kinetics of calbindin-D28k determined by flash photolysis of caged Ca2+. Biophys J 79, 30093018.CrossRefGoogle Scholar
39.Choi, KJ, Cho, DS, Kim, JY, et al. (2011) Ca-induced Ca release from internal stores in INS-1 rat insulinoma cells. Korean J Physiol Pharmacol 15, 5359.CrossRefGoogle ScholarPubMed
40.Bellido, T, Huening, M, Raval-Pandya, M, et al. (2000) Calbindin-D28k is expressed in osteoblastic cells and suppresses their apoptosis by inhibiting caspase-3 activity. J Biol Chem 275, 2632826332.CrossRefGoogle ScholarPubMed
41.Merico, V, de Barboza, GD, Vasco, C, et al. (2008) A mitochondrial mechanism is involved in apoptosis of Robertsonian mouse male germ cells. Reproduction 135, 797804.CrossRefGoogle ScholarPubMed
42.Rodriguez, V, Diaz de Barboza, G, Ponce, R, et al. (2010) Spermatocyte apoptosis, which involves both intrinsic and extrinsic pathways, explains the sterility of Graomys griseoflavus × Graomys centralis male hybrids. Reprod Fertil Dev 22, 478488.CrossRefGoogle ScholarPubMed
43.Lambers, TT, Mahieu, F, Oancea, E, et al. (2006) Calbindin-D28K dynamically controls TRPV5-mediated Ca2+ transport. EMBO J 12, 29782988.CrossRefGoogle Scholar
44.Airaksinen, MS, Eilers, J & Garaschuk, O (1997) Ataxia and altered dendritic calcium signaling in mice carrying a targeted null mutation of the calbindin D28k gene. Proc Natl Acad Sci U S A 18, 14881493.CrossRefGoogle Scholar
45.Christakos, S, Dhawan, P, Ajibade, D, et al. (2010) Mechanisms involved in vitamin D mediated intestinal calcium absorption and in non-classical actions of vitamin D. J Steroid Biochem Mol Biol 121, 183187.CrossRefGoogle ScholarPubMed
46.Hwang, I, Yang, H, Kang, HS, et al. (2013) Alteration of tight junction gene expression by calcium- and vitamin D-deficient diet in the duodenum of calbindin-null mice. Int J Mol Sci 14, 2299723010.CrossRefGoogle ScholarPubMed
47.Schatzmann, HJ (1966) ATP-dependent Ca++-extrusion from human red cells. Experientia 15, 364365.CrossRefGoogle Scholar
48.Barley, NF, Howard, A, O’Callaghan, D, et al. (2001) Epithelial calcium transporter expression in human duodenum. Am J Physiol Gastrointest Liver Physiol 280, G285G290.CrossRefGoogle ScholarPubMed
49.Peng, JB, Chen, XZ, Berger, UV, et al. (1999) Molecular cloning and characterization of a channel-like transporter mediating intestinal calcium absorption. J Biol Chem 274, 2273922746.CrossRefGoogle ScholarPubMed
50.Kim, HJ, Lee, GS, Ji, YK, et al. (2006) Differential expression of uterine calcium transporter 1 and plasma membrane Ca2+ ATPase 1b during rat estrous cycle. Am J Physiol Endocrinol Metab 291, E234E241.CrossRefGoogle ScholarPubMed
51.Stauffer, TP, Guerini, D, Celio, MR, et al. (1997) Immunolocalization of the plasma membrane Ca2+ pump isoforms in the rat brain. Brain Res 748, 2129.CrossRefGoogle ScholarPubMed
52.Tribe, RM, Moriarty, P & Poston, L (2000) Calcium homeostatic pathways change with gestation in human myometrium. Biol Reprod 63, 748755.CrossRefGoogle ScholarPubMed
53.Ghijsen, WE, De Jong, MD & Van Os, CH (1982) ATP-dependent calcium transport and its correlation with Ca2+-ATPase activity in basolateral plasma membranes of rat duodenum. Biochim Biophys Acta 28, 327336.CrossRefGoogle Scholar
54.Anderson, RG (1993) Caveolae: where incoming and outgoing messengers meet. Proc Natl Acad Sci U S A 90, 1090910913.CrossRefGoogle ScholarPubMed
55.Centeno, VA, Díaz de Barboza, GE, Marchionatti, AM, et al. (2004) Dietary calcium deficiency increases Ca2+ uptake and Ca2+ extrusion mechanisms in chick enterocytes. Comp Biochem Physiol A Mol Integr Physiol 139, 133141.CrossRefGoogle ScholarPubMed
56.Liu, C, Weng, H, Chen, L, et al. (2013) Impaired intestinal calcium absorption in protein 4.1R-deficient mice due to altered expression of plasma membrane calcium ATPase1b (PMCA1b). J Biol Chem 19, 1140711415.CrossRefGoogle Scholar
57.Ghijsen, WE, De Jong, MD & Van Os, CH (1983) Kinetic properties of Na+/Ca2+ exchange in basolateral plasma membranes of rat small intestine. Biochim Biophys Acta 730, 8594.CrossRefGoogle ScholarPubMed
58.Philipson, KD, Nicoll, DA, Matsuoka, S, et al. (1996) Molecular regulation of the Na+-Ca2+ exchanger. Ann N Y Acad Sci 779, 2028.CrossRefGoogle ScholarPubMed
59.Dong, H, Sellers, ZM, Smith, A, et al. (2005) Na+/Ca2+ exchange regulates Ca2+-dependent duodenal mucosal ion transport and HCO3– secretion in mice. Am J Physiol Gastrointest Liver Physiol 288, G457G465.CrossRefGoogle Scholar
60.Hwang, I, Jung, EM, Yang, H, et al. (2011) Tissue-specific expression of the calcium transporter genes TRPV5, TRPV6, NCX1, and PMCA1b in the duodenum, kidney and heart of Equus caballus. J Vet Med Sci 73, 14371444.CrossRefGoogle ScholarPubMed
61.Kim, JA, Yang, H, Hwang, I, et al. (2011) Expression patterns and potential action of the calcium transport genes Trpv5, Trpv6, Ncx1 and Pmca 1b in the canine duodenum, kidney and uterus. In Vivo 25, 773780.Google ScholarPubMed
62.Hoenderop, JG, Hartog, A, Stuiver, M, et al. (2000) Localization of the epithelial Ca2+ channel in rabbit kidney and intestine. J Am Soc Nephrol 11, 11711178.CrossRefGoogle ScholarPubMed
63.Blaustein, MP & Lederer, WJ (1999) Sodium/calcium exchange: its physiological implications. Physiol Rev 79, 763854.CrossRefGoogle ScholarPubMed
64.Yang, H, Lei, C, Cheng, C, et al. (2012) The antiapoptotic effect of galectin-3 in human endometrial cells under the regulation of estrogen and progesterone. Biol Reprod 87, 39.CrossRefGoogle ScholarPubMed
65.Hoenderop, JG, Nilius, B & Bindels, RJ (2005) Calcium absorption across epithelia. Physiol Rev 85, 373422.CrossRefGoogle ScholarPubMed
66.González-Mariscal, L, Betanzos, A, Nava, P, et al. (2003) Tight junction proteins. Prog Biophys Mol Biol 81, 144.CrossRefGoogle ScholarPubMed
67.Bronner, F & Pansu, D (1999) Nutritional aspects of calcium absorption. J Nutr 129, 912.CrossRefGoogle ScholarPubMed
68.Bronner, F (2003) Mechanisms of intestinal calcium absorption. J Cell Biochem 88, 387393.CrossRefGoogle ScholarPubMed
69.Bouillon, R, Lieben, L, Mathieu, C, et al. (2013) Vitamin D action: lessons from VDR and Cyp27b1 null mice. Pediatr Endocrinol Rev 10, 354366.Google ScholarPubMed
70.Wasserman, RH, Smith, CA, Brindak, ME, et al. (1992) Vitamin D and mineral deficiencies increase the plasma membrane calcium pump of chicken intestine. Gastroenterology 102, 886894.CrossRefGoogle ScholarPubMed
71.Centeno, V, Picotto, G & Pérez, A (2011) Intestinal Na+/Ca2+ exchanger protein and gene expression are regulated by 1,25(OH)2D3 in vitamin D-deficient chicks. Arch Biochem Biophys 15, 191196.CrossRefGoogle Scholar
72.Balesaria, S, Sangha, S & Walters, JR (2009) Human duodenum responses to vitamin D metabolites of TRPV6 and other genes involved in calcium absorption. Am J Physiol Gastrointest Liver Physiol 297, G1193G1197.CrossRefGoogle ScholarPubMed
73.Khuituan, P, Wongdee, K, Jantarajit, W, et al. (2013) Fibroblast growth factor-23 negates 1,25(OH)2D3-induced intestinal calcium transport by reducing the transcellular and paracellular calcium fluxes. Arch Biochem Biophys 536, 4652.CrossRefGoogle Scholar
74.Fudge, NJ & Kovacs, CS (2010) Pregnancy up-regulates intestinal calcium absorption and skeletal mineralization independently of the vitamin D receptor. Endocrinology 151, 886895.CrossRefGoogle ScholarPubMed
75.Gallagher, JC (2013) Vitamin D and aging. Endocrinol Metab Clin North Am 42, 319332.CrossRefGoogle ScholarPubMed
76.Sheikh, MS, Schiller, LR, Fordtran, JS, et al. (1990) In vivo intestinal absorption of calcium in humans. Miner Electrolyte Metab 16, 130146.Google ScholarPubMed
77.Karbach, U (1992) Paracellular calcium transport across the small intestine. J Nutr 122, 672677.CrossRefGoogle ScholarPubMed
78.Fujita, H, Sugimoto, K, Inatomi, S, et al. (2008) Tight junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2+ absorption between enterocytes. Mol Biol Cell 19, 19121921.CrossRefGoogle ScholarPubMed
79.Garg, MK, Kalra, S & Mahalle, N (2013) Defining vitamin D deficiency using surrogate markers. Indian J Endocrinol Metab 17, 784786.CrossRefGoogle ScholarPubMed
80.Picotto, G, Massheimer, V & Boland, R (1997) Parathyroid hormone stimulates calcium influx and the cAMP messenger system in rat enterocytes. Am J Physiol 273, C1349C1353.CrossRefGoogle ScholarPubMed
81.Gentili, C, Morelli, S & de Boland, AR (2003) Characterization of PTH/PTHrP receptor in rat duodenum: effects of ageing. J Cell Biochem 88, 11571167.CrossRefGoogle ScholarPubMed
82.Nemere, I & Larsson, D (2002) Does PTH have a direct effect on intestine? J Cell Biochem 86, 2934.CrossRefGoogle ScholarPubMed
83.Cross, HS, Debiec, H & Peterlik, M (1990) Thyroid hormone enhances the genomic action of calcitriol in the small intestine. Prog Clin Biol Res 332, 163180.Google ScholarPubMed
84.Kumar, V & Prasad, R (2003) Thyroid hormones stimulate calcium transport systems in rat intestine. Biochim Biophys Acta 1639, 185194.CrossRefGoogle ScholarPubMed
85.Zoidis, E, Gosteli-Peter, M, Ghirlanda-Keller, C, et al. (2002) IGF-I and GH stimulate Phex mRNA expression in lungs and bones and 1,25-dihydroxyvitamin D3 production in hypophysectomized rats. Eur J Endocrinol 146, 97105.CrossRefGoogle ScholarPubMed
86.Fleet, JC, Bruns, ME, Hock, JM, et al. (1994) Growth hormone and parathyroid hormone stimulate intestinal calcium absorption in aged female rats. Endocrinology 134, 17551760.CrossRefGoogle ScholarPubMed
87.Fatayerji, D, Mawer, EB & Eastell, R (2000) The role of insulin-like growth factor I in age-related changes in calcium homeostasis in men. J Clin Endocrinol Metab 85, 46574662.Google ScholarPubMed
88.Cotter, AA & Cashman, KD (2006) Effect of 17β-oestradiol on transepithelial calcium transport in human intestinal-like Caco-2 cells and its interactions with 1,25-dihydroxycholecalciferol and 9-cis retinoic acid. Eur J Nutr 45, 234241.CrossRefGoogle ScholarPubMed
89.Colin, EM, Van Den Bemd, GJ, Van Aken, M, et al. (1999) Evidence for involvement of 17β-estradiol in intestinal calcium absorption independent of 1,25-dihydroxyvitamin D3 level in the rat. J Bone Miner Res 14, 5764.CrossRefGoogle ScholarPubMed
90.Bouillon, R, Carmeliet, G & Van Cromphaut, S (2005) Intestinal calcium absorption: lessons from knockout mice and men. In Vitamin D, 2nd ed., pp. 429452 [D Feldman, FH Glorieux and JW Pike, editors]. San Diego, CA: Academic Press.CrossRefGoogle Scholar
91.Campbell-Thompson, M, Lynch, IJ, Bhardwaj, B, et al. (2001) Expression of estrogen receptor (ER) subtypes and ERβ isoforms in colon cancer. Cancer Res 61, 632640.Google ScholarPubMed
92.van Abel, M, Hoenderop, JG, van der Kemp, AW, et al. (2003) Regulation of the epithelial Ca2+ channels in small intestine as studied by quantitative mRNA detection. Am J Physiol Gastrointest Liver Physiol 285, G78G85.CrossRefGoogle ScholarPubMed
93.Cotter, AA & Cashman, KD (2005) The effect of two dietary and a synthetic phytoestrogen on transepithelial calcium transport in human intestinal-like Caco-2 cells. Eur J Nutr 44, 7278.CrossRefGoogle Scholar
94.Park, CY & Weaver, CM (2012) Vitamin D interactions with soy isoflavones on bone after menopause: a review. Nutrients 4, 16101621.CrossRefGoogle Scholar
95.Reid, IR (1997) Glucocorticoid osteoporosis – mechanisms and management. Eur J Endocrinol 137, 209217.CrossRefGoogle ScholarPubMed
96.Van Cromphaut, SJ, Stockmans, I, Torrekens, S, et al. (2007) Duodenal calcium absorption in dexamethasone-treated mice: functional and molecular aspects. Arch Biochem Biophys 460, 300305.CrossRefGoogle ScholarPubMed
97.Lee, GS, Choi, KC, Jeung, EB, et al. (2006) Glucocorticoids differentially regulate expression of duodenal and renal calbindin-D9k through glucocorticoid receptor-mediated pathway in mouse model. Am J Physiol Endocrinol Metab 290, E299E307.CrossRefGoogle ScholarPubMed
98.Huybers, S, Naber, TH, Bindels, RJ, et al. (2007) Prednisolone-induced Ca2+ malabsorption is caused by diminished expression of the epithelial Ca2+ channel TRPV6. Am J Physiol Gastrointest Liver Physiol 292, G92G97.CrossRefGoogle ScholarPubMed
99.Christakos, S, Dhawan, P & Liu, Y (2003) New insights into the mechanisms of vitamin D action. J Cell Biochem 88, 695705.CrossRefGoogle ScholarPubMed
100.Brown, AJ, Krits, I & Armbrecht, HJ (2005) Effect of age, vitamin D, and calcium on the regulation of rat intestinal epithelial calcium channels. Arch Biochem Biophys 437, 5158.CrossRefGoogle ScholarPubMed
101.Benn, BS, Ajibade, D & Porta, A (2008) Active intestinal calcium transport in the absence of transient receptor potential vanilloid type 6 and calbindin-D9k. Endocrinology 149, 31963205.CrossRefGoogle ScholarPubMed
102.Tolosa de Talamoni, N, Mykkanen, H & Wasserman, RH (1990) Enhancement of sulfhydryl group availability in the intestinal brush border membrane by deficiencies of dietary calcium and phosphorus in chicks. J Nutr 120, 11981204.CrossRefGoogle ScholarPubMed
103.Brun, LR, Brance, ML & Rigalli, A (2012) Luminal calcium concentration controls intestinal calcium absorption by modification of intestinal alkaline phosphatase activity. Br J Nutr 108, 229233.CrossRefGoogle ScholarPubMed
104.Zemel, MB, Shi, H, Greer, B, et al. (2000) Regulation of adiposity by dietary calcium. FASEB J 14, 11321138.CrossRefGoogle ScholarPubMed
105.Zemel, MB (2005) The role of dairy foods in weight management. J Am Coll Nutr 24, 537S546S.CrossRefGoogle ScholarPubMed
106.Huang, JY & Qi, SJ (2015) Childhood obesity and food intake. World J Pediatr 11, 101107.CrossRefGoogle ScholarPubMed
107.Barr, S (2003) Increased dairy product or calcium intake: is body weight or composition affected in humans? J Nutr 133, 245S248S.CrossRefGoogle ScholarPubMed
108.Coe, FL, Favus, MJ & Asplin, JR (2004) Nephrolithiasis. In The Kidney, 7th ed., pp. 18191866 [BM Brenner and FC Rector, editors]. Philadelphia, PA: Elsevier.Google Scholar
109.Lemann, J (2002) Idiopathic hypercalciuria. In Disorders of Bone and Mineral Metabolism, pp. 673697 [FL Coe and M Favus, editors]. Philadelphia, PA: Lippincott.Google Scholar
110.Haghighi, A, Samimagham, H & Gohardehi, G (2013) Calcium and vitamin D supplementation and risk of kidney stone formation in postmenopausal women. Iran J Kidney Dis 7, 210213.Google ScholarPubMed
111.Jackson, RD, LaCroix, AZ, Gass, M, et al. (2006) Calcium plus vitamin D supplementation and the risk of fractures. N Engl J Med 354, 669683.CrossRefGoogle ScholarPubMed
112.Diaz-Lopez, B & Cannata-Andia, JB (2006) Supplementation of vitamin D and calcium: advantages and risks. Nephrol Dial Transplant 21, 23752377.Google ScholarPubMed
113.Curhan, GC, Willett, WC, Rimm, EB, et al. (1993) A prospective study of dietary calcium and other nutrients and the risk of symptomatic kidney stones. N Engl J Med 328, 833838.CrossRefGoogle ScholarPubMed
114.Borghi, L, Schianchi, T, Meschi, T, et al. (2002) Comparison of two diets for the prevention of recurrent stones in idiopathic hypercalciuria. N Engl J Med 346, 7784.CrossRefGoogle ScholarPubMed
115.Park, Y, Leitzmann, MF, Subar, A, et al. (2009) Dairy food, calcium, and risk of cancer in the NIH-AARP diet and health study. Arch Intern Med 169, 391401.CrossRefGoogle ScholarPubMed
116.Tárraga López, PJ, Albero, JS & Rodríguez-Montes, JA (2014) Primary and secondary prevention of colorectal cancer. Clin Med Insights Gastroenterol 7, 3346.Google ScholarPubMed
117.Peterlik, M, Grant, WB & Cross, HS (2009) Calcium, vitamin D and cancer. Anticancer Res 29, 36873698.Google ScholarPubMed
118.McCullough, ML, Rodriguez, C, Diver, WR, et al. (2005) Dairy, calcium, and vitamin D intake and postmenopausal breast cancer risk in the Cancer Prevention Study II Nutrition Cohort. Cancer Epidemiol Biomarkers Prev 14, 28982904.CrossRefGoogle ScholarPubMed
119.Hong, Z, Tian, C & Zhang, X (2012) Dietary calcium intake, vitamin D levels, and breast cancer risk: a dose–response analysis of observational studies. Breast Cancer Res Treat 136, 309312.CrossRefGoogle Scholar
120.Chen, P, Hu, P, Xie, D, et al. (2010) Meta-analysis of vitamin D, calcium and the prevention of breast cancer. Breast Cancer Res Treat 121, 469477.CrossRefGoogle ScholarPubMed
121.Vergne, Y, Matta, J, Morales, L, et al. (2007) Intakes of calcium and vitamin D and breast cancer risk in women. Arch Intern Med 167, 10501059.Google Scholar
122.Belizan, JM & Villar, J (1980) The relationship between calcium intake and edema-, proteinuria-, and hypertension-getosis: an hypothesis. Am J Clin Nutr 33, 22022210.CrossRefGoogle ScholarPubMed
123.Belizan, JM, Villar, J & Repke, J (1988) The relationship between calcium intake and pregnancy-induced hypertension: up-to-date evidence. Am J Obstet Gynecol 158, 898902.CrossRefGoogle ScholarPubMed
124.Camargo, EB, Moraes, LF, Souza, CM, et al. (2013) Survey of calcium supplementation to prevent preeclampsia: the gap between evidence and practice in Brazil. BMC Pregnancy Childbirth 13, 206.CrossRefGoogle Scholar
125.Griffith, LE, Guyatt, GH & Cook, RJ (1999) The influence of dietary and nondietary calcium supplementation on blood pressure: an updated metaanalysis of randomized controlled trials. Am J Hypertens 12, 8492.CrossRefGoogle ScholarPubMed
126.van Mierlo, LA, Arends, LR, Streppel, MT, et al. (2006) Blood pressure response to calcium supplementation: a meta-analysis of randomized controlled trials. J Hum Hypertens 20, 571580.CrossRefGoogle ScholarPubMed
127.Varenna, M, Manara, M, Galli, L, et al. (2013) The association between osteoporosis and hypertension: the role of a low dairy intake. Calcif Tissue Int 93, 8692.CrossRefGoogle ScholarPubMed
128.Reid, IR, Mason, B, Horne, A, et al. (2006) Randomized controlled trial of calcium in healthy older women. Am J Med 119, 777785.CrossRefGoogle ScholarPubMed
129.Kim, KM, Choi, SH, Lim, S, et al. (2014) Interactions between dietary calcium intake and bone mineral density or bone geometry in a low calcium intake population. J Clin Endocrinol Metab 99, 24092417.CrossRefGoogle ScholarPubMed
130.Zhou, W, Langsetmo, L, Berger, C, et al. (2013) Longitudinal changes in calcium and vitamin D intakes and relationship to bone mineral density in a prospective population-based study: the Canadian Multicentre Osteoporosis Study (CaMos). J Musculoskelet Neuronal Interact 13, 470479.Google Scholar
131.Adler, AJ & Berlyne, GM (1985) Duodenal aluminum absorption in the rat: effect of vitamin D. Am J Physiol 249, G209G213.Google ScholarPubMed
132.Dunn, MA, Johnson, NE, Liew, MY, et al. (1993) Dietary aluminum chloride reduces the amount of intestinal calbindin D-28K in chicks fed low calcium or low phosphorus diets. J Nutr 123, 17861793.CrossRefGoogle ScholarPubMed
133.Cox, KA & Dunn, MA (2001) Aluminum toxicity alters the regulation of calbindin-D28k protein and mRNA expression in chick intestine. J Nutr 131, 20072013.CrossRefGoogle ScholarPubMed
134.Orihuela, D, Meichtry, V, Pregi, N, et al. (2005) Short-term oral exposure to aluminium decreases glutathione intestinal levels and changes enzyme activities involved in its metabolism. J Inorg Biochem 99, 18711878.CrossRefGoogle ScholarPubMed
135.Orihuela, D, Meichtry, V & Pizarro, M (2005) Aluminium-induced impairment of transcellular calcium absorption in the small intestine: calcium uptake and glutathione influence. J Inorg Biochem 99, 18791886.CrossRefGoogle ScholarPubMed
136.Orihuela, D (2009) Inhibitory effect of aluminium on calcium absorption in small intestine of rats with different thyroid hormone status. J Inorg Biochem 103, 15421547.CrossRefGoogle ScholarPubMed
137.Orihuela, D (2007) Effect of aluminium on duodenal calcium transport in pregnant and lactating rats treated with bromocriptine. J Inorg Biochem 101, 12701274.CrossRefGoogle ScholarPubMed
138.Heaney, RP & Nordin, BE (2002) Calcium effects on phosphorus absorption: implications for the prevention and co-therapy of osteoporosis. J Am Coll Nutr 21, 239244.CrossRefGoogle ScholarPubMed
139.Ribovich, ML & DeLuca, HF (1978) Effect of dietary calcium and phosphorus on intestinal calcium absorption and vitamin D metabolism. Arch Biochem Biophys 188, 145156.CrossRefGoogle ScholarPubMed
140.Gray, RW & Napoli, JL (1983) Dietary phosphate deprivation increases 1,25-dihyroxyvitamin D3 synthesis in rat kidney in vitro. J Biol Chem 258, 11521155.CrossRefGoogle ScholarPubMed
141.Meyer, RA Jr, Tenenhouse, HS, Meyer, MH, et al. (1989) The renal phosphate transport defect in normal mice parabiosed to X-linked hypophosphatemic mice persists after parathyroidectomy. J Bone Miner Res 4, 523532.CrossRefGoogle ScholarPubMed
142.Bar, A & Wasserman, RH (1973) Control of calcium absorption and intestinal calcium-binding protein synthesis. Biochem Biophys Res Commun 54, 191196.CrossRefGoogle ScholarPubMed
143.Meyer, J, Fullmer, CS, Wasserman, RH, et al. (1992) Dietary restriction of calcium, phosphorus, and vitamin D elicits differential regulation of the mRNAs for avian intestinal calbindin-D28k and the 1,25-dihydroxyvitamin D3 receptor. J Bone Miner Res 7, 441448.CrossRefGoogle Scholar
144.Hendrix, ZJ, Alcock, NW & Archibald, RM (1963) competition between calcium, strontium, and magnesium for absorption in the isolated rat intestine. Clin Chem 12, 734744.CrossRefGoogle ScholarPubMed
145.O’Donnell, JM & Smith, MW (1973) Uptake of calcium and magnesium by rat duodenal mucosa analysed by means of competing metals. J Physiol 229, 733749.CrossRefGoogle ScholarPubMed
146.Hardwick, LL, Jones, MR, Brautbar, N, et al. (1991) Magnesium absorption: mechanisms and the influence of vitamin D, calcium and phosphate. J Nutr 121, 1323.CrossRefGoogle ScholarPubMed
147.de Swart, PM, Sokole, EB & Wilmink, JM (1998) The interrelationship of calcium and magnesium absorption in idiopathic hypercalciuria and renal calcium stone disease. J Urol 159, 669672.CrossRefGoogle ScholarPubMed
148.Planells, E, Sánchez-Morito, N, Montellano, MA, et al. (2000) Effect of magnesium deficiency on enterocyte Ca, Fe, Cu, Zn, Mn and Se content. J Physiol Biochem 56, 217222.CrossRefGoogle ScholarPubMed
149.Dimai, H-P, Porta, S, Wirnsberger, G, et al. (1998) Daily oral magnesium supplementation suppresses bone turnover in young adult males. J Clin Endocrinol Metab 83, 27422748.Google ScholarPubMed
150.Fine, KD, Santa Ana, CA, Porter, JL, et al. (1991) Intestinal absorption of magnesium from food and supplements. J Clin Invest 88, 396402.CrossRefGoogle ScholarPubMed
151.Kozakai, T, Uozumi, N, Katoh, K, et al. (2002) Dietary magnesium increases calcium absorption of ovine small intestine in vivo and in vitro. Reprod Nutr Dev 42, 2533.CrossRefGoogle ScholarPubMed
152.Bae, YJ, Bu, SY, Kim, JY, et al. (2011) Magnesium supplementation through seaweed calcium extract rather than synthetic magnesium oxide improves femur bone mineral density and strength in ovariectomized rats. Biol Trace Elem Res 144, 9921002.CrossRefGoogle ScholarPubMed
153.Hessov, I, Andersson, H & Isaksson, B (1983) Effects of a low-fat diet on mineral absorption in small-bowel disease. Scand J Gastroenterol 18, 551554.CrossRefGoogle ScholarPubMed
154.Haderslev, KV, Jeppesen, PB, Mortensen, PB, et al. (2000) Absorption of calcium and magnesium in patients with intestinal resections treated with medium chain fatty acids. Gut 46, 819823.CrossRefGoogle ScholarPubMed
155.Jewell, C, Cusack, S & Cashman, KD (2005) The effect of conjugated linoleic acid on transepithelial calcium transport and mediators of paracellular permeability in human intestinal-like Caco-2 cells. Prostaglandins Leukot Essent Fatty Acids 72, 163171.CrossRefGoogle ScholarPubMed
156.Murphy, EF, Jewell, C, Hooiveld, GJ, et al. (2006) Conjugated linoleic acid enhances transepithelial calcium transport in human intestinal-like Caco-2 cells: an insight into molecular changes. Prostaglandins Leukot Essent Fatty Acids 74, 295301.CrossRefGoogle ScholarPubMed
157.Coxam, V (2007) Current data with inulin-type fructans and calcium, targeting bone health in adults. J Nutr 137, 2527S2533S.CrossRefGoogle ScholarPubMed
158.Raschka, L & Daniel, H (2005) Diet composition and age determine the effects of inulin-type fructans on intestinal calcium absorption in rat. Eur J Nutr 44, 360364.CrossRefGoogle ScholarPubMed
159.Lobo, AR, Cocato, ML & Jorgetti, V (2009) Changes in bone mass, biomechanical properties, and microarchitecture of calcium- and iron-deficient rats fed diets supplemented with inulin-type fructans. Nutr Res 29, 873881.CrossRefGoogle ScholarPubMed
160.Xiao, Y, Cui, J, Shi, YH, et al. (2010) Effects of duodenal redox status on calcium absorption and related genes expression in high-fat diet-fed mice. Nutrition 26, 11881194.CrossRefGoogle ScholarPubMed
161.Buchowski, MS & Miller, DD (1991) Lactose, calcium source and age affect calcium bioavailability in rats. J Nutr 121, 17461754.CrossRefGoogle ScholarPubMed
162.Dupuis, Y, Tardivel, S, Porembska, Z, et al. (1991) Effect of some alkaline phosphatase inhibitors on intestinal calcium transfer. Int J Biochem 23, 175180.CrossRefGoogle ScholarPubMed
163.Sogabe, N, Mizoi, L, Asahi, K, et al. (2004) Enhancement by lactose of intestinal alkaline phosphatase expression in rats. Bone 35, 249255.CrossRefGoogle ScholarPubMed
164.Nishimukai, M, Watanabe, J, Taguchi, H, et al. (2008) Effects of epilactose on calcium absorption and serum lipid metabolism in rats. J Agric Food Chem 56, 1034010345.CrossRefGoogle ScholarPubMed
165.Suzuki, T, Nishimukai, M, Shinoki, A, et al. (2010) Ingestion of epilactose, a non-digestible disaccharide, improves postgastrectomy osteopenia and anemia in rats through the promotion of intestinal calcium and iron absorption. J Agric Food Chem 58, 1078710792.CrossRefGoogle ScholarPubMed
166.Suzuki, T, Nishimukai, M, Takechi, M, et al. (2010) The nondigestible disaccharide epilactose increases paracellular Ca absorption via rho-associated kinase- and myosin light chain kinase-dependent mechanisms in rat small intestines. J Agric Food Chem 58, 19271932.CrossRefGoogle ScholarPubMed
167.Mineo, H, Hara, H & Tomita, F (2002) Sugar alcohols enhance calcium transport from rat small and large intestine epithelium in vitro. Dig Dis Sci 47, 13261333.CrossRefGoogle ScholarPubMed
168.Xiao, J, Li, X, Min, X, et al. (2013) Mannitol improves absorption and retention of calcium and magnesium in growing rats. Nutrition 29, 325331.CrossRefGoogle ScholarPubMed
169.López-Huertas, E, Teucher, B, Boza, JJ, et al. (2006) Absorption of calcium from milks enriched with fructo-oligosaccharides, caseinophosphopeptides, tricalcium phosphate, and milk solids. Am J Clin Nutr 83, 310316.CrossRefGoogle ScholarPubMed
170.Scholz-Ahrens, KE, Schaafsma, G, van den Heuvel, EG, et al. (2001) Effects of prebiotics on mineral metabolism. Am J Clin Nutr 73, 459S464S.CrossRefGoogle ScholarPubMed
171.Suzuki, T & Hara, H (2004) Various non-digestible saccharides increase intracellular calcium ion concentration in rat small-intestinal enterocytes. Br J Nutr 92, 751755.CrossRefGoogle ScholarPubMed
172.Takasaki, M, Inaba, H, Ohta, A, et al. (2000) Dietary short-chain fructooligosaccharides increase calbindin-D9k levels only in the large intestine in rats independent of dietary calcium deficiency or serum 1,25 dihydroxy vitamin D levels. Int J Vitam Nutr Res 70, 206213.CrossRefGoogle ScholarPubMed
173.Fukushima, A, Aizaki, Y & Sakuma, K (2009) Short-chain fatty acids induce intestinal transient receptor potential vanilloid type 6 expression in rats and Caco-2 cells. J Nutr 139, 2025.CrossRefGoogle ScholarPubMed
174.Legette, LL, Lee, W, Martin, BR, et al. (2012) Prebiotics enhance magnesium absorption and inulin-based fibers exert chronic effects on calcium utilization in a postmenopausal rodent model. J Food Sci 77, H88H94.CrossRefGoogle Scholar
175.Weaver, CM, Martin, BR, Nakatsu, CH, et al. (2011) Galactooligosaccharides improve mineral absorption and bone properties in growing rats through gut fermentation. J Agric Food Chem 59, 65016510.CrossRefGoogle ScholarPubMed
176.Tsuchita, H, Suzuki, T & Kuwata, T (2001) The effect of casein phosphopeptides on calcium absorption from calcium-fortified milk in growing rats. Br J Nutr 85, 510.CrossRefGoogle ScholarPubMed
177.Erba, D, Ciappellano, S & Testolin, G (2002) Effect of the ratio of casein phosphopeptides to calcium (w/w) on passive calcium transport in the distal small intestine of rats. Nutrition 18, 743746.CrossRefGoogle ScholarPubMed
178.Teucher, B, Majsak-Newman, G, Dainty, JR, et al. (2006) Calcium absorption is not increased by caseinophosphopeptides. Am J Clin Nutr 84, 162166.CrossRefGoogle Scholar
179.Cosentino, S, Gravaghi, C, Donetti, E, et al. (2010) Caseinphosphopeptide-induced calcium uptake in human intestinal cell lines HT-29 and Caco2 is correlated to cellular differentiation. J Nutr Biochem 21, 247254.CrossRefGoogle ScholarPubMed
180.Colombini, A, Perego, S, Ardoino, I, et al. (2013) Evaluation of a possible direct effect by casein phosphopeptides on paracellular and vitamin D controlled transcellular calcium transport mechanisms in intestinal human HT-29 and Caco2 cell lines. Food Funct 4, 11951203.CrossRefGoogle ScholarPubMed
181.Whisner, CM, Martin, BR & Nakatsu, CH (2014) Soluble maize fibre affects short-term calcium absorption in adolescent boys and girls: a randomised controlled trial using dual stable isotopic tracers. Br J Nutr 112, 446456.CrossRefGoogle ScholarPubMed
182.Schrezenmeir, J & de Vrese, M (2001) Probiotics, prebiotics, and synbiotics – approaching a definition. Am J Clin Nutr 73, 361S364S.CrossRefGoogle ScholarPubMed
183.Ghanem, KZ, Badawy, IH & Abdel-Samam, AM (2004) Influence of yogourt and probiotic yogourt on the absorption of calcium, magnesium, iron and bone mineralization in rats. Milchwissenschaft 59, 472475.Google Scholar
184.Gilman, J & Cashman, KD (2006) The effect of probiotic bacteria on transepithelial calcium transport and calcium uptake in human intestinal-like Caco-2 cells. Curr Issues Intest Microbiol 7, 15.Google ScholarPubMed
185.Scholz-Ahrens, K, Ade, P, Marten, B, et al. (2007) Prebiotics, probiotics, and synbiotics affect mineral absorption, bone mineral content, and bone structure. J Nutr 137, 838S846S.CrossRefGoogle ScholarPubMed
186.Kerstetter, JE, O’Brien, KO & Insogna, KL (2003) Low protein intake: the impact on calcium and bone homeostasis in humans. J Nutr 133, 855S861S.CrossRefGoogle ScholarPubMed
187.Johnson, NE, Alcantara, EN & Linkswiler, H (1970) Effect of level of protein intake on urinary and fecal calcium and calcium retention of young adult males. J Nutr 100, 14251430.CrossRefGoogle ScholarPubMed
188.Darling, AL, Millward, DJ, Torgerson, DJ, et al. (2009) Dietary protein and bone health: a systematic review and meta-analysis. Am J Clin Nutr 90, 16741692.CrossRefGoogle Scholar
189.Kerstetter, JE, O’Brien, KO, Caseria, DM, et al. (2005) The impact of dietary protein on calcium absorption and kinetic measures of bone turnover in women. J Clin Endocrinol Metab 90, 2631.CrossRefGoogle ScholarPubMed
190.Gaffney-Stomberg, E, Sun, BH, Cucchi, CE, et al. (2010) The effect of dietary protein on intestinal calcium absorption in rats. Endocrinology 151, 10711078.CrossRefGoogle ScholarPubMed
191.Skipper, A (2010) Nutrition Care Manual. Chicago, IL. American Dietetic Association, American Dietetic Association Nutrition Care Manual. http://www.nutritioncaremanual.org (accessed September 2015).Google Scholar
192.Marcason, W (2010) What is the effect of a high-protein diet on bone health? J Am Diet Assoc 110, 812.CrossRefGoogle ScholarPubMed
193.Das, AS, Das, D, Mukherjee, M, et al. (2005) Phytoestrogenic effects of black tea extract (Camellia sinensis) in an oophorectomized rat (Rattus norvegicus) model of osteoporosis. Life Sci 77, 30493057.CrossRefGoogle Scholar
194.Sharma, V & Rao, LJ (2009) A thought on the biological activities of black tea. Crit Rev Food Sci Nutr 49, 379404.CrossRefGoogle ScholarPubMed
195.Das, AS, Banerjee, M, Das, D, et al. (2013) Black tea may be a prospective adjunct for calcium supplementation to prevent early menopausal bone loss in a rat model of osteoporosis. J Osteoporos 2013, 760586.CrossRefGoogle Scholar
196.Hernandez-Avila, M, Colditz, GA, Stampfer, MJ, et al. (1991) Caffeine, moderate alcohol intake, and risk of fractures of the hip and forearm in middle-aged women. Am J Clin Nutr 54, 157163.CrossRefGoogle ScholarPubMed
197.Welch, AA, Bingham, SA, Reeve, J, et al. (2007) More acidic dietary acid–base load is associated with reduced calcaneal broadband ultrasound attenuation in women but not in men: results from the EPIC-Norfolk cohort study. Am J Clin Nutr 85, 11341141.CrossRefGoogle Scholar
198.Cooper, C, Atkinson, EJ, Wahner, HW, et al. (1992) Is caffeine consumption a risk factor for osteoporosis? J Bone Miner Res 7, 465471.CrossRefGoogle ScholarPubMed
199.Huopio, J, Kröger, H, Honkanen, R, et al. (2000) Risk factors for perimenopausal fractures: a prospective study. Osteoporos Int 11, 219227.CrossRefGoogle ScholarPubMed
200.Hallström, H, Byberg, L, Glynn, A, et al. (2013) Long-term coffee consumption in relation to fracture risk and bone mineral density in women. Am J Epidemiol 178, 898909.CrossRefGoogle ScholarPubMed
201.Yeh, JK & Aloia, JF (1986) Differential effect of caffeine administration on calcium and vitamin D metabolism in young and adult rats. J Bone Miner Res 1, 251258.CrossRefGoogle ScholarPubMed
202.Hasling, C, Søndergaard, K, Charles, P, et al. (1992) Calcium metabolism in postmenopausal osteoporotic women is determined by dietary calcium and coffee intake. J Nutr 122, 11191126.CrossRefGoogle ScholarPubMed
203.Folwarczna, J, Pytlik, M, Zych, M, et al. (2013) Favorable effect of moderate dose caffeine on the skeletal system in ovariectomized rats. Mol Nutr Food Res 57, 17721784.CrossRefGoogle ScholarPubMed
204.Cano-Marquina, A, Tarín, JJ & Cano, A (2013) The impact of coffee on health. Maturitas 75, 721.CrossRefGoogle ScholarPubMed
205.Folwarczna, J, Zych, M, Nowińska, B, et al. (2014) Unfavorable effect of trigonelline, an alkaloid present in coffee and fenugreek, on bone mechanical properties in estrogen-deficient rats. Mol Nutr Food Res 58, 14571464.CrossRefGoogle Scholar
206.Lowe, M, Strauss, AW, Alpers, R, et al. (1990) Molecular cloning and expression of a cDNA encoding the membrane-associated rat intestinal alkaline phosphatase. Biochim Biophys Acta 1037, 170177.CrossRefGoogle ScholarPubMed
207.Marchionatti, A, Alisio, A, Díaz de Barboza, G, et al. (2001) dl-Buthionine-S,R-sulfoximine affects intestinal alkaline phosphatase activity. Comp Biochem Physiol C Toxicol Pharmacol 129, 8591.CrossRefGoogle ScholarPubMed
208.Chiou, TJ & Tzeng, WF (2000) The roles of glutathione and antioxidant enzymes in menadione-induced oxidative stress. Toxicology 154, 7584.CrossRefGoogle ScholarPubMed
209.Shiraki, M, Shiraki, Y, Aoki, C, et al. (2000) Vitamin K2 (menatetrenone) effectively prevents fractures and sustains lumbar bone mineral density in osteoporosis. J Bone Miner Res 15, 515521.CrossRefGoogle ScholarPubMed
210.Sata, N, Klonowski-Stumpe, H, Han, B, et al. (1997) Menadione induces both necrosis and apoptosis in rat pancreatic acinar AR4-2J cells. Free Radic Biol Med 23, 844850.CrossRefGoogle ScholarPubMed
211.Marchionatti, AM, Díaz de Barboza, GE, Centeno, VA, et al. (2003) Effects of a single dose of menadione on the intestinal calcium absorption and associated variables. J Nutr Biochem 14, 466472.CrossRefGoogle ScholarPubMed
212.Higuchi, Y (2004) Glutathione depletion-induced chromosomal DNA fragmentation associated with apoptosis and necrosis. J Cell Mol Med 8, 455464.CrossRefGoogle ScholarPubMed
213.Marchionatti, AM, Perez, AV, Diaz de Barboza, GE, et al. (2008) Mitochondrial dysfunction is responsible for the intestinal calcium absorption inhibition induced by menadione. Biochim Biophys Acta 1780, 101107.CrossRefGoogle ScholarPubMed
214.Suzuki, T & Hara, H (2009) Quercetin enhances intestinal barrier function through the assembly of zonula [corrected] occludens-2, occludin, and claudin-1 and the expression of claudin-4 in Caco-2 cells. J Nutr 139, 965974.CrossRefGoogle ScholarPubMed
215.Marchionatti, AM, Pacciaroni, A & Tolosa de Talamoni, NG (2013) Effects of quercetin and menadione on intestinal calcium absorption and the underlying mechanisms. Comp Biochem Physiol A Mol Integr Physiol 164, 215220.CrossRefGoogle ScholarPubMed
216.Carpentieri, A, Marchionatti, A, Areco, V, et al. (2014) Antioxidant and antiapoptotic properties of melatonin restore intestinal calcium absorption altered by menadione. Mol Cell Biochem 387, 197205.CrossRefGoogle ScholarPubMed
217.Rivoira, MA, Marchionatti, AM, Centeno, VA, et al. (2012) Sodium deoxycholate inhibits chick duodenal calcium absorption through oxidative stress and apoptosis. Comp Biochem Physiol A Mol Integr Physiol 162, 397405.CrossRefGoogle ScholarPubMed
218.Lamireau, T, Zoltowska, M, Levy, E, et al. (2003) Effects of bile acids on biliary epithelial cells: proliferation, cytotoxicity, and cytokine secretion. Life Sci 72, 14011411.CrossRefGoogle ScholarPubMed
219.Kawano, A, Ishikawa, H, Kamano, T, et al. (2010) Significance of fecal deoxycholic acid concentration for colorectal tumor enlargement. Asian Pac J Cancer Prev 11, 15411546.Google ScholarPubMed
220.Jean-Louis, S, Akare, S, Ali, MA, et al. (2006) Deoxycholic acid induces intracellular signaling through membrane perturbations. J Biol Chem 281, 1494814960.CrossRefGoogle ScholarPubMed
221.Araki, Y, Katoh, T, Ogawa, A, et al. (2005) Bile acid modulates transepithelial permeability via the generation of reactive oxygen species in the Caco-2 cell line. Free Radic Biol Med 39, 769780.CrossRefGoogle ScholarPubMed
222.Rodríguez, V, Rivoira, M, Marchionatti, A, et al. (2013) Ursodeoxycholic and deoxycholic acids: a good and a bad bile acid for intestinal calcium absorption. Arch Biochem Biophys 540, 1925.CrossRefGoogle Scholar
223.Makishima, M, Lu, TT, Xie, W, et al. (2002) Vitamin D receptor as an intestinal bile acid sensor. Science 296, 13131316.CrossRefGoogle ScholarPubMed
224.Krasowski, MD, Ni, A, Hagey, LR, et al. (2011) Evolution of promiscuous nuclear hormone receptors: LXR, FXR, VDR, PXR, and CAR. Mol Cell Endocrinol 334, 3948.CrossRefGoogle ScholarPubMed
225.D’Aldebert, E, Biyeyeme Bi Mve, MJ, Mergey, M, et al. (2009) Bile salts control the antimicrobial peptide cathelicidin through nuclear receptors in the human biliary epithelium. Gastroenterology 136, 14351443.CrossRefGoogle ScholarPubMed
226.Hansen, KE, Jones, AN, Lindstrom, MJ, et al. (2010) Do proton pump inhibitors decrease calcium absorption? J Bone Miner Res 25, 27862789.CrossRefGoogle ScholarPubMed
227.Wahl, TO, Gobuty, AH, Lukert, BP, et al. (1981) Long-term anticonvulsant therapy and intestinal calcium absorption. Clin Pharmacol Ther 30, 506512.CrossRefGoogle ScholarPubMed
228.von Borstel Smith, M, Crofoot, K, Rodriguez-Proteau, R, et al. (2007) Effects of phenytoin and carbamazepine on calcium transport in Caco-2 cells. Toxicol In Vitro 21, 855862.CrossRefGoogle ScholarPubMed
229.Charoenphandhu, N, Teerapornpuntakit, J, Lapmanee, S, et al. (2012) Duodenal calcium transporter mRNA expression in stressed male rats treated with diazepam, fluoxetine, reboxetine, or venlafaxine. Mol Cell Biochem 369, 8794.CrossRefGoogle ScholarPubMed
230.Kim, S, An, BS, Yang, H, et al. (2013) Effects of octylphenol and bisphenol A on the expression of calcium transport genes in the mouse duodenum and kidney during pregnancy. Toxicology 303, 99106.CrossRefGoogle ScholarPubMed
231.Alevizaki, CC, Ikkos, DG & Singhelakis, P (1973) Progressive decrease of true intestinal calcium absorption with age in normal man. J Nucl Med 14, 760762.Google ScholarPubMed
232.Chen, C & Kalu, DN (1999) Strain differences in bone density and calcium metabolism between C3H/HeJ and C57BL/6J mice. Bone 25, 413420.CrossRefGoogle ScholarPubMed
233.Wu, L, Martin, BR, Braun, MM, et al. (2010) Calcium requirements and metabolism in Chinese-American boys and girls. J Bone Miner Res 25, 18421849.CrossRefGoogle ScholarPubMed
234.Weaver, CM, McCabe, LD, McCabe, GP, et al. (2008) Vitamin D status and calcium metabolism in adolescent black and white girls on a range of controlled calcium intakes. J Clin Endocrinol Metab 93, 39073914.CrossRefGoogle ScholarPubMed
235.Bryant, RJ, Wastney, ME, Martin, BR, et al. (2003) Racial differences in bone turnover and calcium metabolism in adolescent females. J Clin Endocrinol Metab 88, 10431047.CrossRefGoogle ScholarPubMed
236.Braun, M, Palacios, C, Wigertz, K, et al. (2007) Racial differences in skeletal calcium retention in adolescent girls with varied controlled calcium intakes. Am J Clin Nutr 85, 16571663.CrossRefGoogle ScholarPubMed
237.Replogle, RA, Li, Q, Wang, L, et al. (2014) Gene-by-diet interactions influence calcium absorption and bone density in mice. J Bone Miner Res 29, 657665.CrossRefGoogle ScholarPubMed
238.Reid, IR (2014) Should we prescribe calcium supplements for osteoporosis prevention? J Bone Metab 21, 2128.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Schematic model of transepithelial and paracellular calcium transport in the small intestine. The paracellular calcium pathway is carried out through tight junctions (TJ) by an electrochemical gradient (long arrow between cells). The transcellular calcium pathway consists of three steps: (1) apical entry of calcium through epithelial calcium channels TRPV5 and TRPV6 (the second one is the most abundant in intestine); (2) cytosolic diffusion bound to calbindins (CB); and (3) extrusion across the basolateral membranes by plasma membrane Ca2+-ATPase (PMCA1b) and Na+/Ca2+ exchanger (NCX1). Calcitriol (1,25-D3) stimulates the individual steps of transcellular calcium transport. Calcitriol molecules bind to their nuclear receptors (vitamin D receptors; VDR), and the complex 1,25-D3–VDR interacts with specific DNA sequences inducing transcription and increasing the expression levels of PMCA1b, NCX1, TRPV6 and CB. The real role of the intestinal alkaline phosphatase (AP) enzyme inintestinal calcium absorption has not been elucidated yet. Cldn, claudin.