Next Article in Journal
Phytochemical Profile and Antioxidant Activity of Aerial and Underground Parts of Salvia bulleyana Diels. Plants
Next Article in Special Issue
Vitamin D Metabolism and Its Role in Mineral and Bone Disorders in Chronic Kidney Disease in Humans, Dogs and Cats
Previous Article in Journal
Single-Step Extraction Coupled with Targeted HILIC-MS/MS Approach for Comprehensive Analysis of Human Plasma Lipidome and Polar Metabolome
Previous Article in Special Issue
Protective Role of Vitamin D in Renal Tubulopathies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vitamin D in Small Animal Bone Metabolism

by
Rafael Vessecchi Amorim Zafalon
1,
Bruna Ruberti
2,
Mariana Fragoso Rentas
1,
Andressa Rodrigues Amaral
3,
Thiago Henrique Annibale Vendramini
1,
Fernanda Chicharo Chacar
4,
Marcia Mery Kogika
2 and
Marcio Antonio Brunetto
1,3,*
1
Pet Nutrology Research Center, Nutrition and Production Department, School of Veterinary Medicine and Animal Science, University of São Paulo, Jardim Elite, Pirassununga 13635-900, Brazil
2
Small Animal Internal Medicine Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, Cidade Universitária, São Paulo 05508-270, Brazil
3
Veterinary Nutrology Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, Cidade Universitária, São Paulo 05508-270, Brazil
4
Department of Internal Medicine, Federal Institute of Education, Science and Technology of South of Minas Gerais, IFSULDEMINAS, Muzambinho 37890-000, Brazil
*
Author to whom correspondence should be addressed.
Submission received: 30 September 2020 / Revised: 9 November 2020 / Accepted: 10 November 2020 / Published: 3 December 2020
(This article belongs to the Special Issue Vitamin D and Bone Metabolism)

Abstract

:
Dogs and cats have differences in vitamin D metabolism compared to other mammalian species, as they are unable to perform vitamin D cutaneous synthesis through sun exposure. Therefore, they are dependent on the dietary intake of this nutrient. The classic functions of vitamin D are to stimulate intestinal calcium and phosphate absorption, renal calcium and phosphate reabsorption and regulate bone mineral metabolism. Thus, it is an important nutrient for calcium and phosphorus homeostasis. This review highlights the evidence of the direct and indirect actions of vitamin D on bone mineral metabolism, the consequences of nutritional imbalances of this nutrient in small animals, as well as differences in vitamin D metabolism between different size dogs.

1. Introduction

Vitamin D is an essential nutrient for dogs and cats since they are unable to synthesize vitamin D3 through skin sun exposure; thus, it is essential that they receive this nutrient in their diet [1,2]. vitamin D has two nonactive forms in nature: cholecalciferol or vitamin D3, and ergocalciferol or vitamin D2. After ingestion, vitamin D undergoes a multistep enzymatic conversion until conversion to an active vitamin known as 1,25-dyhydroxyvitamin D (1,25(OH)2D3) [3]. After activation, vitamin D binds to the vitamin D receptor (VDR), a representative receptor for the steroid receptors family, which mediates its biological functions [4] that go beyond the traditional role of calcium and phosphate homeostasis [5].
Regarding bone metabolism vitamin D functions, calcitriol target organs are intestine, kidneys, bone tissue and the parathyroid gland [6,7]. In the intestine, calcitriol acts on transcellular and passive paracellular calcium transport besides phosphate absorption [8,9]. In the kidneys, calcitriol stimulates renal calcium and phosphate reabsorption. Finally, calcitriol acts in the parathyroid gland regulating parathyroid hormone (PTH) secretion. All these functions can indirectly regulate bone growth and mineralization. In addition, it is known that both osteoblasts and chondrocytes express the enzyme 1α-hydroxylase and VDR [10] and it has been demonstrated that vitamin D can act directly on bone growth and mineralization [11,12,13], as well as in bone remodeling [14].
Although the importance of vitamin D in bone metabolism is well established in humans, its deficiency is prevalent worldwide [15]. In dogs and cats, with the wide use of complete and balanced commercial foods, there was a decrease in vitamin D deficiency cases diagnosis [16], which was very common in the past. However with the growing interest in homemade pet foods, there is a growing risk of hypovitaminosis D [17,18].
It is known that vitamin D deficiency can result in hypocalcemia and hypophosphatemia and, consequently, can result in bone disease characterized by impaired bone mineralization associated either with rickets or osteomalacia [7,16]. In addition, dietary vitamin D deficiency can result in both direct and indirect nutritional secondary hyperparathyroidism (NSHP) [7,19,20]. Excessive vitamin D is also harmful and causes of hypervitaminosis D in small animals. This can be due to commercial food formulation miscalculation [21,22,23,24], accidental consumption of rodenticides [25,26,27,28,29], consumption of certain plants that may contain calcitriol glycosides (such as jessamine [30]) and the use of topical ointments for psoriasis treatment, which are based on vitamin D analogs such as calcipotriol and maxacalcitol [31,32,33,34]. Acute vitamin D intoxication results in hypercalcemia, which causes clinical signs such as polydipsia, polyuria, anorexia, vomiting, constipation, seizures, etc. In the case of chronic intoxication, osteochondrosis and delayed bone remodeling can occur mostly in growing giant breed dogs [35,36,37].
Therefore, this review aims to summarize the current knowledge about the direct and indirect role of vitamin D in bone metabolism, as well as discussing the negative effects of vitamin D deficiency and excess in dogs’ and cats’ bone health.

2. Vitamin D Metabolism in Small Animals

Two forms of vitamin D are found in nature, cholecalciferol or vitamin D3, and ergocalciferol or vitamin D2. While ergocalciferol is synthesized from ergosterol found in fungi and plants through exposure to UVB ultraviolet rays (290–315 nm) [38], cholecalciferol is synthesized in the skin of several mammalian animals after sun exposure [39,40]. In the deep layers of the epidermis (spinous and basal strata) the precursor of cholecalciferol, 7-dihydrocholesterol (a derivative of cholesterol), is located in the cell membranes’ phospholipid bilayer [41]. The 7-dihydrocholesterol absorbs a UVB photon and this allows the photolytic break of the bond between carbons nine and 10 of ring B of the pentanoperhydrophenanthrene cycle, forming previtamin D3, which has two broken rings. This substance is thermostable and undergoes heat-induced isomerization giving rise to vitamin D3, which has a special, more stable, configuration. Vitamin D3 gains circulation due to the steric energy of this new three-dimensional conformation of the molecule [41].
Dogs and cats are unable to perform cutaneous synthesis of Vitamin D3 through sun exposure, due to the high activity of 7-dihydrocholesterol-Δ7-reductase enzyme that converts 7-dehydrocholesterol into cholesterol. Therefore, a significant decrease in skin 7-dehydrocholesterol concentrations occurs [3,39,40]. In an in-vitro study conducted by How et al. [39], total 7-dehydrocholesterol concentrations in the skin of dogs and cats before sun exposure, were, on average, almost 10 times lower when compared to rats. After sun exposure, there was no change in vitamin D3 concentrations in dog and cat skin extracts, while there was a 40-fold increase in vitamin D3 concentrations in rat skin. In cats, an in-vivo study demonstrated that kittens with low vitamin D status upon receiving a 7-dihydrocholesterol-Δ7-reductase enzyme inhibitor had a progressive increase in vitamin D status when exposed to ultraviolet light for 2 h/day, even if receiving a purified vitamin D-free diet, while a group that did not receive the enzyme inhibitor had no change in vitamin D status [40]. These authors demonstrated that the high activity of 7-dihydrocholesterol-Δ7-reductase prevents adequate vitamin D3 skin synthesis.
The inability to synthesize cutaneous vitamin D3 justifies the fact that no seasonal variation in vitamin D status was observed in dogs in a study conducted in Australia [42]. It is believed that the inability to synthesize vitamin D3 in the skin in dogs and cats is related to the evolutionary aspect that once, in nature, these animals consumed preys which stores vitamin D3 in the liver and adipose tissue. Therefore, when prey was consumed regularly, their vitamin D requirements were met. Consequently these animals did not need to develop the ability of cutaneous vitamin D3 synthesis [3,43].
As a consequence, dogs and cats are totally dependent on their dietary intake. After ingestion, both cholecalciferol and ergocalciferol are absorbed in a process that depends on digestive enzymes, bile acids and chylomicrons [44]. After absorption, cholecalciferol and/or ergocalciferol are transported through intestinal lymphatic vessels and the portal system, linked to a glycoprotein vitamin D binding protein (VDBP), to the liver where they undergo a hydroxylation process on Carbon 25, which can occur through enzymes of the cytochrome P450 family such as CYP27A1, CYP3A4, CYP2R1 and CYP2J3 [45], also known as 25-hydroxylases, forming 25-hydroxyvitamin D (25(OH)D) or calcidiol. If the animal has high circulating calcidiol concentrations, the absorbed cholecalciferol can be stored in adipose tissue and, to a lesser extent, in muscle tissue, instead of being transported for hydroxylation in the liver [46].
Dogs convert ergocalciferol to calcidiol as well as cholecalciferol [44]. However, cats use cholecalciferol more efficiently [47]. This may be due to the type of food that these animals had in nature during the evolutionary process. Dogs have a more omnivorous nature, so that in nature they consumed ergocalciferol and then developed the ability to use this form of vitamin D to synthesize calcidiol in the liver. In contrast, cats are considered strict carnivores, and in nature they did not consume ergocalciferol, a fact that may justify their lesser ability to use this substance for hepatic calcidiol synthesis [3]. After hepatic synthesis, calcidiol is transported by VDBP to the proximal renal tubules where it undergoes a hydroxylation process in carbon 1 through a mitochondrial enzyme of the CYP450 family, 1α-hydroxylase (CYP27B1), giving rise to 1,25 dihydroxyvitamin D [1,25(OH)2D] or calcitriol, the active metabolite of vitamin D which performs all its known hormonal actions. It is worth mentioning that renal synthesis is largely responsible for circulating calcitriol. However, it has been demonstrated in humans that several other tissues express 1α-hydroxylase, including the breast, parathyroid gland, colon, prostate, beta-pancreatic cells, placenta, brain, immune cells, endothelial cells and keratinocytes [48].
Although calcitriol is the active metabolite, vitamin D status is assessed by measuring circulating calcidiol concentrations as it has a long biological half-life, ranging from 10 days to three weeks, whereas the biological half-life of calcitriol varies from three to six hours [49]. Besides, calcidiol circulates in concentrations around 1000 times higher compared to calcitriol. It is also worth mentioning that calcitriol has a more hydrophobic nature, which makes it difficult to measure [3,50], in addition to it being less stable [7,51].

3. Regulation of Calcitriol Synthesis and Catabolism

Calcitriol synthesis regulation is influenced by several factors, such as plasma calcium and phosphate concentrations, parathyroid hormone (PTH), fibroblast growth factor (FGF-23), klotho gene and calcitonin [3,52,53,54,55]. In hypocalcemic conditions, there is an increase in PTH release, which can directly stimulate the activity of 1α-hydroxylase by stimulation of the 1α-hydroxylase promoter gene, or indirectly by stimulating 1α-hydroxylase mRNA expression and then the activity of 1α-hydroxylase via cAMP-dependent pathway in order to increase calcitriol synthesis [7,56,57,58]. Under normocalcemic conditions, calcitonin acts to maintain serum calcitriol concentrations, by inducing 1α-hydroxylase transcription in renal cells, which seems to occur due to positive regulation of the transcription factor C/EBPβ [55].
The 24-hydroxylase enzyme is responsible for vitamin D catabolism. This mitochondrial enzyme is found mainly in proximal renal tubules but it is also expressed in intestine, keratinocytes, fibroblasts, lymphocytes and macrophages [59]. The 24-hydroxylase enzyme acts through two metabolic pathways and promotes hydroxylation in the side carbon chain, both in calcidiol and calcitriol. It promotes hydroxylation on carbon 23 of calcitriol, producing 1α,23,25-trihydroxyvitamin D3 (1α,23,25(OH)3D3) which, in turn, undergoes an oxidation process giving rise to 1α,25-dihydroxyvitamin D3 -26,23-lactone.
Through a second metabolic pathway, 24-hydroxylase promotes hydroxylation on carbon 24 of calcidiol and calcitriol, resulting in the formation of 24,25–dihydroxyvitamin D3 (24,25(OH)2D3) and 1α,24,25-trihydroxyvitamin D3 (1α,24,25(OH)3D3), respectively. The 1α,24,25(OH)3D3 still undergoes an oxidation process, forming calcitroic acid, a metabolite excreted through bile [60,61,62]. These metabolites functions are not yet well known, but 24,25(OH)2D3 is believed to be related to the bone mineralization process [63].
Calcitriol stimulates 24-hydroxylase activity, playing a protective role against hypercalcemia [61]. In situations of high serum phosphate concentrations, there is an increase in synthesis and release of FGF-23, an important peptide produced by osteoblasts and mainly by osteocytes, with hypophosphatemic action, so that it stimulates renal phosphorus excretion, increases 24-hydroxylase activity, decreases 1α-hydroxylase activity and, therefore, decreases calcitriol synthesis, since this hormone has the function of intestinal phosphorus absorption and tubular reabsorption of inorganic phosphate [61,64,65]. Calcitriol synthesis regulation is also affected by a transmembrane protein called Klotho, which is a mandatory and necessary coreceptor for FGF-23 to bind to its receptor [66].

4. Differences in Vitamin D Metabolism between Miniature and Giant Dog Breeds

It is known that dogs of giant breeds are prone to develop skeletal disorders when compared to dogs of small breeds fed with low or excessive calcium diets [67]. Among other things, this seems to be related to differences in vitamin D metabolism. In the study by Hazewinkel and Tryfonidou [68], the plasma concentrations of the vitamin D main metabolites were measured in giant breed dogs (adult weight: 60 kg) and small breed dogs (adult weight: 6 kg) at 14 weeks of age managed under the same conditions. There was no difference in concentrations of 25(OH)D and 1,25(OH)2D3. However giant breed dogs had lower 24,25(OH)2D concentrations, with values which were about 10 times lower than small breed dogs.
In another study, carried out by Tryfonidou et al. [67], Great Dane puppies showed higher plasma 1,25(OH)2D concentrations and lower plasma 24,25(OH)2D concentrations compared to miniature poodle puppies. According to Hazewinkel and Tryfonidou [68], the lower plasma 24,25(OH)2D concentrations may be related to disturbances in endochondral ossification during the rapid growth phase observed in large and giant breed dogs. Osteochondrosis is a disorder of maturation that results in a delay of cartilage mineralization. It is known that both 24,25(OH)2D and 1,25(OH)2D play an important role in differentiating cartilage cells and matrix mineralization [68,69]. In order to avoid disturbances in endochondral ossification, the physiological balance between 24,25(OH)2D and 1,25(OH)2D is of major importance [70]. Therefore, the imbalance between these vitamin D dihydroxylated metabolites in large and giant breeds dogs seems to be related to a greater disposition for osteochondrosis during growth.
The reason why giant breed dogs have lower plasma 24,25(OH)2D concentrations is not yet fully elucidated, but it is believed that this can be explained by higher circulating growth factor concentrations that these animals present, such as growth hormone (GH) and insulin-like growth factor I (IGF-I) [68]. Even in the study by Tryfonidou et al. [67], higher plasma GH and IGF-1 concentrations were observed in Great Dane puppies than in miniature poodle puppies. In humans, an increase in circulating 1,25(OH)2D concentrations has been reported after GH therapy in children with GH deficiency [71], normal adults [72] and elderly people [73], and Condamine et al. [74] observed that IGF-1 stimulates the 1,25(OH)2D synthesis in mammalian renal cells under conditions of hypophosphatemia. In addition, an association has been demonstrated between high plasma GH and IGF-I concentrations and low plasma 24,25(OH)2D concentrations [75], as well as showing that GH and IGF-I decrease 24-hydroxylase activity [76], which may justify this association.

5. The VDR Receptor

Brumbaugh and Haussler [77] first described the vitamin D receptor, a nuclear transcription factor belonging to a nuclear receptor superfamily for steroid hormones [3,78,79]. For the biological activity of vitamin D to occur, circulating calcitriol bound to VDBP crosses the cell membrane and binds to VDR. After this binding, a calcitriol-VDR complex is formed which interacts with retinoic acid receptor (RXR) forming a calcitriol-VDR-RXR heterodimer that acts on promoter regions of target genes, known as vitamin D response elements (VDRE) [80,81]. Consequently, there is a cascade of molecular interactions evolving nuclear coactivators and transcription factors, positive and negative, which interact with calcitriol-VDR-RXR regulating the transcription and/or suppression of specific genes [6,79,82]. Under gene induction or repression, changes in the expression of proteins required for a biological response occurs [83,84]. This vitamin D genomic response process may involve regulation of about 2000 genes, which represent almost 10% of the human genome [85].
The VDR can also show nongenomic responses, which occurs when calcitriol interacts with VDR located in sphingolipid and cholesterol rich cell membranes [86]. In response to binding of calcitriol to these receptors, there is an activation of guanylate cyclase, phosphatidylinositol-3-kinase (PIK 3) [87], cyclic adenosine monophosphate (cAMP), mitogen-activated protein kinase (MAPK) [88]; protein kinase (PK) A and phospholipase C (PLC) pathways [89], the opening of voltage-dependent calcium and chlorine channels [89,90] and mobilization of calcium from the sarcoplasmic reticulum [89,91]. According to Norman et al. [89], these mechanisms cause the migration of smooth muscle cells from the endothelium, rapid absorption of calcium by the duodenal epithelium and exocytosis of insulin by β-pancreatic cells.

6. The Functions of Vitamin D Related to Bone Metabolism

Regarding vitamin D functions related to bone metabolism, the target organs of calcitriol are intestine, kidneys, bone tissue and parathyroid gland [6,7].

6.1. Effects of Vitamin D on the Intestine

In the intestine, calcitriol acts on both transcellular and passive paracellular calcium transport [8,9]. Transcellular calcium transport consists of three stages: calcium entry into the enterocyte through the calcium epithelial channel transient receptor potential vanilloid type 6 (TRPV6); the cytoplasmic transfer of calcium linked to the calbindin-D9k protein, which facilitates the diffusion of calcium through the cell and the extrusion of calcium through the basolateral membrane via Ca2+–ATPase (PMCA 1b) and/or a Na-Ca exchanger [8], which results in calcium entering the circulation. Vitamin D positively regulates TRPV6 and calbindine-D9k and, consequently, there is an increase in calcium absorption [92,93]. The importance of vitamin D in regulation of TRPV6 and calbindin-D9k was proven in studies with VDR knockout mice in which a reduction of 50% in intestinal calbindin-D9k mRNA was observed, as well as a reduction of up to 95% in TRPV6 mRNA, when compared to wild type mice [94,95].
Paracellular transport occurs mainly in the most distal regions of the intestine, but it can happen over the entire length of the intestine [96]. This pathway consists of a luminal electrochemical gradient and the integrity of the intercellular junctions [96,97]. The tight junctions are located between apical and basolateral membranes of the enterocyte, providing cell-cell adhesion in epithelial and endothelial cell layers. These junctions provide a primary barrier to the passage of ions, proteins, cytoskeleton molecules and cytoplasmic signaling molecules [97]. There are several transmembrane components at the tight junctions, such as sodium/potassium ATPase, claudin 3, aquaporin 8, cadherin 17 and RhoA, which can be suppressed by calcitriol and suggests that vitamin D regulates the permeability of tight junctions and, consequently, paracellular calcium transport [98]. In addition, calcitriol stimulates the expression of paracellins, intercellular proteins that form channels where calcium is passively transferred due to differences in concentration gradient [92].
In addition to calcium absorption, in the intestine, vitamin D also acts on phosphate absorption. Calcitriol stimulates phosphate absorption via the Naþ phosphate transporter (NaPi-IIb) pathway so that calcitriol increases Na-Pi transporter expression, which corresponds to a family of SLC34 solute carriers (SLC34A2) [99].

6.2. Effects of Vitamin D on the Kidneys

Calcitriol stimulates renal calcium reabsorption so that calcium transport through renal epithelial cells is similar to calcium transport through intestinal epithelial cells. In the apical cell membrane of the distal convoluted tubules and collecting ducts there is an epithelial calcium channel (TRPV5) that is responsible for transporting calcium into the cells. Calcium transport occurs through renal tubular cells through the calbindin-D28K protein, and in the basolateral cell membrane there are calcium transporters PMCA1b and NCX responsible for transporting calcium into the bloodstream [100]. In addition, a calcium influx protein called epithelial Ca21 channel (ECaC) was discovered. ECaC acts in the active reabsorption of calcium in the distal portion of the nephron [101,102,103]. Calcitriol upregulates TRPV5, calbindin-D28K, PMCA1b, NCX1 and ECaC gene expression [7,104,105,106].

6.3. Effects of Vitamin D on the Parathyroid Gland

It is known that calcitriol receptor mRNA has high expression in the parathyroid gland [107,108]. After the discovery of the active form of vitamin D, the effects of this hormone on the parathyroid gland began to be investigated and it was demonstrated in-vitro that calcitriol decreased PTH production in primary bovine parathyroid cell culture by decreasing the PTH gene transcription [109]. Subsequently, calcitriol was shown to decrease levels of PTH mRNA in rats [90]. Because of the effect of reducing PTH synthesis in humans with chronic kidney disease (CKD), calcitriol or analogues have been used to treat secondary renal hyperparathyroidism [110]. In contrast, in a study conducted by Hostutler et al. [111] with healthy and CKD cats, no difference in serum PTH concentrations was observed after a 14-day oral treatment with calcitriol (2.5 ng/kg every 24 h or 8.75 ng/kg every 84 h), and no difference in serum ionized calcium concentrations was noticed. It is worth mentioning that further studies are needed to investigate whether the dosages used in that study were insufficient to reduce PTH synthesis. According to International Renal Interest Society (IRIS) [112], there is evidence to suggest that the rational use of calcitriol (1.5 to 3.5 ng/kg) prolongs the survival of dogs with CKD in stages three and four, where phosphate is controlled and ionized calcium and PTH are monitored. As for cats, according to IRIS [113], the beneficial effects of an ultralow dose of calcitriol have not yet been established, so its use is not currently recommended.
In humans, the treatment with calcitriol in CKD patients promotes beneficial effects on the kidneys unrelated to PTH and mineral metabolism, including activity of renin-angiotensin-aldosterone system suppression, reduction of proteinuria, reduction of podocyte loss associated with glomerular hypertrophy and antiproliferative and antifibrotic effects [114,115,116,117]. These effects seem to be important to explain the benefits demonstrated by calcitriol therapy in CKD human patients, where it has been shown to limit the progression of CKD, improve survival and be independently associated with preservation of renal function [116,118].
The mechanisms by which vitamin D decreases PTH synthesis are well described, so that it suppresses the parathyroid gland cells growth gland by decreasing growth factors (transforming growth factor—epidermal growth factor receptor growth loop) and through the increase in inhibitors of cell growth (cyclin-dependent kinase inhibitors p21 and p27) [7,19]. In addition, calcitriol has been shown to positively regulate the transcription of gene encoding the calcium-sensing receptor (CaSR) in the parathyroid gland, which provides greater sensitivity of this gland to plasma ionized calcium concentration resulting in decreased PTH secretion [119].

6.4. Effects of Vitamin D on Bone Remodeling

In bone tissue, under conditions of hypocalcemia, calcitriol works in conjunction with PTH to mobilize calcium from bones for circulation to increase circulating ionized calcium concentrations [12,120]. This occurs through the RANKL/osteoprotegerin (OPG) regulatory system, so that in immature osteoblastic cells calcitriol stimulates the activator receptor for NF-kB ligand (RANKL), increasing osteoclastogenesis [14]. RANKL binds to osteoclasts via the activator receptor for NF-kB (RANK) or via the osteoprotegerin receptor [121]. The binding of RANKL to RANK induces differentiation and maturation of osteoclast progenitor cells to osteoclasts. On the other hand, the binding of RANKL to osteoprotegerin inhibits osteoclastogenesis and bone resorption. Calcitriol inhibits osteoprotegerin production, thus allowing bone resorption [122,123,124]. Therefore, VDR signaling negatively regulates bone mass, which was demonstrated in a study with mice with inactivated VDR in which an increase in bone mass was observed [125].

6.5. Effects of Vtamin D on Bone Growth and Mineralization

In the past, it was believed that calcitriol influenced bone growth and mineralization only indirectly, that is, by increasing the calcium and phosphorus concentrations in the circulation through stimulating intestinal absorption and renal reabsorption of these minerals [126]. Later, it was noticed that both osteoblasts and chondrocytes express 1α-hydroxylase and VDR [10]. In addition, an aberrant growth plaque development was observed in VDR-deficient mice before the appearance of hypocalcemia, which reinforces the possible defined role by 1,25(OH)2D-mediated signaling in the growth of endochondral bone [11].
As previously discussed, in negative calcium balance, VDR signaling is directed towards maintaining circulating calcium concentrations, that is, there is an increase in resorption and a decrease in bone mineralization [127]. In contrast, in situations of positive calcium balance, the role of VDR signaling in bone tissue is still unclear. However, it is believed that specific functions depend on osteoblast differentiation stages [127]. In mature osteoblasts, calcitriol, via VDR signaling, has been shown to perform anabolic and anticatabolic activity, consequently promoting an increase in bone mass as demonstrated in a study with mice with transgenetically increased VDR expression [12]. In this study, it was observed that the decrease in bone resorption occurred due to increased expression of osteoprotegerin, which resulted in a decrease in the RANKL/osteoprotegerin ratio. In addition, it was shown that calcitriol can induce Lrp5 mRNA transcripts (Lrp5 acts as a coreceptor in Wnt signaling [13]) so it is important in bone formation and mineralization.

7. Vitamin D Recommendations for Small Animals

Minimum and maximum recommendations of cholecalciferol for dogs and cats according to National Research Council (NRC) [1], European Pet Food Industry Federation (FEDIAF) [128] and Association of American Feed Control Officials (AAFCO) [129] are presented in Table 1 and Table 2.
Few studies have investigated vitamin D requirements for dogs and cats, and all of these studies were performed in puppies and kittens since the demand for skeletal calcification is higher at this life stage and growing animals are more sensitive to vitamin D deficient diets. As there are no studies that investigated vitamin D requirements for adult dogs and cats, as well as breeding females, the recommendations are based on studies of growing animals.
A pioneering study carried out in 1918 by Mellanby [130] observed that the use of cod liver oil and butter included in food for dogs with rickets improved clinical condition of these animals. The first study to work with doses was published in 1939 by Arnold and Elvehjem [131]. These authors performed two experiments with Great Dane puppies, suggesting a dosage of 20 IU vitamin D per 100 g food of about 3.8 kcal g−1 (212 IU kg−1 diet of 4 kcal g−1) for growing puppies consuming a diet with 9.0 g of calcium and 7.5 g of phosphorus per kg. However, it was only in 1944 that the first vitamin D recommendation for dogs was proposed by Michaud and Elvehjem [132], in which the authors recommended an intake of 10 IU to 20 IU kg BW−1·d−1. Finally, Tryfonidou [133] provided a diet with 500 IU of cholecalciferol.kg−1 to Great Dane puppies and did not observe any adverse bone disease.
Regarding cats, few studies have evaluated different concentrations of vitamin D in feline nutrition. The first study was published by Gershoff et al. [134] in which the authors performed oral supplementation on kittens with 250 IU of cholecalciferol per animal twice a week, and prevention of rickets was observed in these animals. In addition, the authors suggested that adult cats have a lower vitamin D requirement since they observed remission of clinical signs in cats older than 12 months who were fed diets with vitamin D inadequacies.
Morris et al. [135] evaluated six groups of kittens and each group received a different cholecalciferol concentrations diet (0.0, 120, 125, 250, 500, 750 or 1000 IU kg−1 of diet). In this study, the authors provided low cholecalciferol concentration diets to the kittens’ mothers during pregnancy and lactation, thus inducing a reduction in the vitamin D kitten reserves. The kittens were fed from nine to 22 weeks of age with the diets and it was noticed that the plasma 25(OH)D concentrations grew linearly with dietary intake of cholecalciferol. In addition, animals fed 0.0 and 125 IU cholecalciferol kg−1 of diet, consumed the food until 34 weeks of age. In this part of the study, the authors observed a sharp drop in the plasma 25(OH)D concentrations of the animals that consumed the food without the addition of vitamin D. However, the kittens that consumed the food with 125 IU cholecalciferol kg−1 of diet had higher plasma 25(OH)D concentrations at 22 and 34 weeks than at nine weeks of age, which indicates that the inclusion of 125 IU cholecalciferol kg−1 of diet would be suitable for kittens. However, the authors recommended a dose of 250 IU cholecalciferol kg−1 of diet to maintain a safety margin for both kittens, pregnant and lactating cats.
Morris and Earle [136] published another study with growing kittens, using a concentration of 125 IU cholecalciferol kg−1 of purified diet, with calcium concentrations ranging from 3.8 to 8.1 g/kg of diet and constant Ca:P ratio. In this study, the authors also observed plasma 25(OH)D concentrations within the normal range.

8. Vitamin D Deficiency in Dogs and Cats

Vitamin D deficiency can be caused by abnormal vitamin D metabolism related to congenital abnormalities due to genetic mutations [137,138], as well as by the intake of unbalanced diets [7,18]. Currently, in the face of the growing production of commercial complete foods, dietary vitamin D deficiency has become less common in dogs and cats [16]. However, the recent increased use of unbalanced diets, that may include homemade and raw foods or inappropriate feeding strategies used by veterinarians or pet owners, may increase nutritional disorders in animals, including vitamin D deficiency [17,18].
As previously discussed, vitamin D requirements are not well established for dogs and cats, which are dependent on vitamin D dietary intake. Vitamin D deficiency results in hypocalcemia and hypophosphatemia, which can result in clinical manifestation of bone disease characterized by impaired bone mineralization associated either with rickets or osteomalacia [7,16]. Rickets is a disease characterized by delayed mineralization of growth cartilage and a failure to remodel freshly deposited bone [7,139], while osteomalacia is abnormal osteoid mineralization that affects bone during its remodeling so unmineralized bone matrix accumulates in all parts of the skeleton [139,140].
In humans, nutritional rickets continues to be reemerging as a disease problem across the world [140,141]. Human rickets is seen usually during infancy and at puberty; the periods of maximum growth [142]. Nutritional rickets has been reported in young dogs [139] and cats [134], although this disease is considered rare [7]. However, rickets can be caused by an inborn error in vitamin D metabolism in dogs and cats [138,143,144,145,146,147].
Vitamin D-dependent rickets type I (VDDR-I), also referred to as vitamin D 1α-hydroxylase deficiency, is an autosomal recessive disorder caused by a deficiency of the renal enzyme 1α-hydroxylase due to a defect in the gene encoding it. Affected animals have normal serum calcidiol concentrations and low calcitriol concentrations [137,147]. Vitamin D-dependent rickets type II (VDDR-II) can also occur in animals as a result of an end-organ resistance to calcitriol and it is usually a secondary defect in the gene encoding the VDR [137,138,145].
A case of VDDR-I in a cat was characterized by hypocalcemia, hyperphosphatemia and skeletal abnormalities such as abnormal bone calcification, delayed teething and hyperparathyroidism [147]. The authors reported low calcitriol concentrations, and a genetic analysis proved the VDDR-I diagnostic. Variations of VDDR-I was reported after that. Grahn et al. [148] related a case by CYP27B1 mutation that caused a feline vitamin D-dependent rickets type 1A (VDDR-IA), and Teshima et al. [149] identified and reported a genetic variant of CYP2R1 identified in a cat with vitamin D-dependent rickets type 1B (VDDR-IB). This change caused low serum concentrations of ionized calcium and calcitriol, and both bone demineralization and abnormal growth plaques were observed on radiographic examination. In all cases, oral administration of calcitriol improved the cat’s wellbeing [147,149]. Although VDDR-I has been reported in cats, there is little research in its prevalence. However, such studies are hard to do as the diagnosis requires genetic analysis.
VDDR-II has been reported in dogs and cats and is a disease characterized by hypocalcemia, secondary hyperparathyroidism, low bone mineralization, rickets and, in some cases, alopecia [138,145]. The affected animals received higher doses of calcitriol associated with calcium supplementation for treatment because it is more difficult to increase circulating calcium concentrations compared to cases of VDDR-I [138,145]. Despite the association of calcitriol and calcium, the animals affected by VDDR-II generally showed persistent hypocalcemia. Godfrey et al. [145] observed no change in plasma calcium concentrations in a cat with VDDR-II after the administration of 110 ng/kg/day of calcitriol for five months and 3410 mg/kg/day of calcium carbonate. Likewise, Levine et al. [138] found no increase in serum calcium concentrations in a dog diagnosed with VDDR-II after supplementation of 64 ng/kg/day of calcitriol and 64 mg/kg/day of calcium carbonate.
Osteomalacia, another metabolic bone disease, rarely causes clinical signs in dogs or cats. However, the inability to detect bone pain and the insensitivity of radiographs of domestic animals, suggest that this condition may be underdiagnosed. In humans, the incidence of this disease remains largely underestimated across the globe as well [7]. Osteomalacia is an adult disease that occurs after cartilage growth is ceased. In animals, the clinical signs are dependent, and vary, according to age, growth rate and activity levels. In mammals, the lesions are most prominent in the long bones at the sites of rapid growth. They yield under the weight of the adult body, twist and bend, and deformities are presented. Fractures are extremely common, besides muscle weakness in elderly patients [150]. The treatment should be reversed by vitamin D supplementation. Calcium is also helpful particularly in those with a poor calcium intake [115,125].

9. Nutritional Secondary Hyperparathyroidism

Nutritional secondary hyperparathyroidism (NSHP) is a common consequence of a nutritional deficiency in pets consuming unbalanced diets [18,151,152]. It can be induced by low calcium concentrations, excess dietary phosphorus even in the presence of optimal dietary calcium, or in association with vitamin D deficiency [20,153]. Dietary vitamin D deficiency can result in NSHP both directly and indirectly. As previously discussed, the calcitriol receptor mRNA is highly expressed in the parathyroid gland [107,108] so that vitamin D also directly influences PTH secretion. Low vitamin D intake results in a decrease in circulating concentrations of calcitriol, which causes an increase in PTH synthesis, since one of the functions of calcitriol is to decrease PTH synthesis by suppressing the growth of parathyroid gland cells due to decrease in growth factors (transforming growth factor—epidermal growth factor receptor growth loop), and through the increase of inhibitors of cell growth (cyclin-dependent kinase inhibitors p21 and p27) [7,19].
Moreover, when low vitamin D dietary concentrations are available, there is a decrease in calcium absorption, which may result in hypocalcemia [154,155]. Low circulating calcium concentrations can stimulate the calcium receptors in the parathyroid gland leading to the production and secretion of PTH [20], which characterizes an indirect role of vitamin D in the development of NSHP. As a result of an excess of circulating PTH the number of osteocytes and osteoclasts increases, and calcium mobilization from bone to the extracellular fluid compartment occurs [154,156].
Few studies have reported NSHP in dogs and cats. Usually, the development of this change has been associated with the consumption of full meat-based diets by young animals due to the high calcium demand for bone growth [20,152,157]. In a report of six kittens affected by NSHP, they received a diet that had low calcium and phosphorus concentrations. Four of the six cats had been fed a full meat-based diet and the other two animals were fed a homemade diet containing only beef/chicken and rice. Some of them had severe osteopenia, which resulted in spontaneous fractures or excitation, muscle twitching or seizure. Four cats had clinical recovery after consuming a balanced diet, parenteral calcium gluconate and cage rest, while the other two cats had to be euthanized because of progressive neurological deficits secondary to spinal fractures [157]. More recently, a case was reported in which a six-month-old female dog with a history of lameness and difficulty in walking, and fed a nutritionally unbalanced homemade diet, developed NSHP. The authors reported that switching to suitable food that met nutritional recommendations was sufficient to promote the animal’s recovery [152].
Morphological changes in bones due to calcium mobilization makes the bones poriferous and prone to fracture. Mature bone replacement by fibrous tissue can lead the animal to develop fibrous osteodystrophy, which affects especially the jaw bones [7,151,152]. In a case report by de Fornel-Thibaud et al. [151], a six-year-old, spayed female rottweiler who consumed a homemade diet deficient in calcium and vitamin D developed facial enlargement from swelling of the maxilla and mandible, characterizing the rubber jaw syndrome. On radiographic and tomodensitometric examinations of the skull, severe osteopenia of the skull bones was observed. The animal had extremely low serum calcidiol concentrations (<0.7 nmol/L; reference range 19 to 90 nmol/L) and high plasma PTH concentrations (104.6 pmol/L; reference range 2 to 13 pmol/L). There was clinical improvement when switching to an adequate diet, with increased skull mineralization.
The treatment of animals with NSHP includes a complete and balanced diet, but sometimes it is necessary for the addition of calcium supplements. Cage rest is also important for the first few weeks of treatment in order to decrease the risk of bone fractures. The prognosis for noncomplicated cases is good and usually dietary correction normalizes body levels of calcium, phosphorus and PTH. On the other hand, complicated cases evolving spinal fractures, or more severe bony changes, support a bad prognosis [20,157].

10. Vitamin D Toxicosis in Small Animals

Signs of acute vitamin D toxicosis are related to the development of hypercalcemia when serum total calcium concentrations exceed 18 mg/dL, and serum ionized calcium concentrations are greater than 1.40 mmol/L. Clinical signs include polydipsia and polyuria due to difficulty in concentrating urine and stimulation of the thirst center, anorexia, vomiting and constipation as a consequence of decreased excitability of gastrointestinal smooth muscle and, finally, decreased neuromuscular excitability may cause weakness, depression, muscle twitching and seizures.
On the other hand, smaller but chronic intake of higher quantities of vitamin D (exceeding by 100 times de NRC recommended) are known as a cause of osteochondrosis and retarded bone remodeling in growing giant breed puppies like Great Danes, who do not have tight control of mineral deposition in large bones compared to smaller breeds [35,36,37].
Until now, cases of hypervitaminosis D are apparently rare and experimentally-induced, since the inclusion of this nutrient is controlled during pet food fabrication, although, in 2005, Mellanby et al. [21] reported two clinical cases of accidental and nutritional hypervitaminosis in dogs consuming a specific diet which was found at the manufacturing plant to be due to a human error that caused the over supplementation of vitamin D in the diet in which it should have been 80 IU/100 g but was 92.2 IU/g instead; over 100 times higher than the stated in manufacturer’s datasheet. The food was recalled from the market.
In both cases, dogs presented at the veterinary hospital with pyrexia, inappetence, polydipsia, polyuria, lethargy, a stiff gait, marked ionized calcium and total hypercalcemia. Palpation of both carpi demonstrated marked soft tissue swelling in one of them and flexion of both carpi caused considerable discomfort to the dog whose radiographs of the left and right carpi, elbows and stifles revealed multiple joint effusions. The consumption of the same diet indicated nutritional involvement. The dog from case one was treated only with intravenous saline solution for 24 h and sent home. The owners considered the dog to be generally free of clinical signs by day 180. On the other hand, the dog from case two received home oral steroid treatment associated with antibiotics and the owners reported that its condition was much improved and exercising normally by day 16.
In both cases, the food with vitamin D excess was withdrawn, ionized calcium was close or within the reference range by the second week after the first visit to the hospital. However, 180 days after, plasma concentrations of 25(OH)D were above the reference range, possibly due to its long half-life (29 days approximately), and may be why hypercalcemia signs persist for a while even when vitamin D was promptly taken out from diet [158].
As the etiology of vitamin D toxicosis is related to hypercalcemia, methods that inhibit bone resorption are the best alternative for treatment. In this scenario, intravenous injections of bisphosphonates are the best alternative, already described for humans and dogs [158,159]. It is important to consider that the approach to treating vitamin D toxicosis depends not only on the severity of hypercalcemia but also on the period of development (acute vs. chronic). Asymptomatic patients with mild hypercalcemia and with normal Ca × P product may not require immediate treatment, while individuals with severe acute increase in Ca levels may require more aggressive treatment. Accidental nutritional vitamin D toxicosis was reported in cats [22,23,24] and in those cases, respiratory discomfort was an important clinical sign among them due to soft tissue calcinosis. All of them were treated according to their symptoms, but only in a study by Crossley et al. [24], was bisphosphonate used and had a good clinical outcome. It is worth mentioning that in the study by Crossley et al. [24], of the three cats intoxicated with vitamin D and with hypercalcemia, pamidronate was administered in only one of them, since conventional therapy for hypercalcemia (fluid therapy, furosemide, prednisone) had no effect on circulating calcium concentrations, unlike in the others two cats, who had a good response to conventional therapy. An important observation of the before mentioned study is the fact that the cats were not made ill by a single food with inadequate amounts of vitamin D, but they received, as part of their diet, complimentary food in addition to dry extruded food, such as canned food and cat milk daily, and these were responsible for the reported vitamin D toxicosis. In this study, analysis was performed of cholecalciferol and ergocalciferol concentrations in the three foods consumed by the cats and all of them had vitamin D concentrations above the declared amount of added vitamin D. The canned food had vitamin D concentrations about 11 times higher than nutritional limit, and both dry food and kitten milk had vitamin D concentrations above the legal limit for declared additives. This is clearly striking and of great importance since these foods administered to patients were commercial foods.
The causes of hypervitaminosis D are not always nutritional, and the most common one is cholecalciferol rodenticide described in dogs [25,26,27,28]. Granulomatous diseases like sarcoidosis and Wegener granulomatosis can also produce hypercalcemia through the unregulated production of calcitriol, which is called metabolite-mediated hypercalcemia [160]. In addition, there are other possible causes of vitamin D toxicity, such as accidental consumption of plants containing calcitriol glycosides such as jessamine [30] and topical ointments based on vitamin D analogues such as calcipotriol and maxacalcitol, which are used for psoriasis treatment [31,32,33,34].

11. Vitamin D and Its Nonclassical Actions

The nonclassical actions of vitamin D were not a focus of this review. However it is important to remember that vitamin D is also related to numerous functions not associated with bone metabolism [3,44]. From 1983, when the presence of vitamin D receptors in human immune cells was discovered, research on the unrelated effects on bone metabolism of vitamin D intensified, and it was discovered that cells in almost all body tissues express vitamin D receptors (3).
In small animals has been demonstrated an association between low vitamin D circulating concentrations and diseases not associated with bone metabolism, such as: cancer [161,162,163,164], congestive heart failure [165,166], gastrointestinal diseases [167,168,169,170,171], acute pancreatitis [172], acute polyradiculoneuritis [173], CKD [161,174,175,176,177] and infectious diseases [178,179,180,181]. It has also been demonstrated in hospitalized cats that when 25(OH)D was represented as a categorical variable, cats with 25(OH)D circulating concentrations in the lower tertile had a higher risk of mortality compared to cats in the reference category and the middle tertile [182]. In a study with hospitalized dogs in a critical condition, it was observed that 25(OH)D serum concentrations at the time of diagnosis was a predictor of mortality rate for dogs with chronic enteropathy [171].
In addition, in-vitro antineoplastic effects of calcitriol have been shown in various types of canine tumors, including osteosarcoma [183], squamous cell carcinoma [184], prostatic epithelial tumor [185], anal sac adenocarcinoma [186], mammary gland cancer [187] and mastocytoma [188]; as well as in vivo antineoplastic effects in dogs with mastocytoma [188].
It is not yet clear whether low vitamin D status is the cause or consequence of the diseases mentioned above, with evidence supporting both hypotheses [3].

12. Conclusions

It is concluded that dogs and cats have different vitamin D metabolisms compared to other mammal species. Little is known about the vitamin D requirements for dogs and cats, especially concerning adult animals and females in the reproductive phase. New trends in dog and cat nutrition, such as a homemade diet, may increase the risk of developing vitamin D deficiency. Vitamin D poisoning is not common in dogs and cats, but it can occur as a result of errors in the formulation of commercial foods, accidental consumption of rodenticides, plants containing calcitriol glycosides and topical ointments based on vitamin D analogues.

Author Contributions

Writing—original draft, R.V.A.Z., B.R., M.F.R. and A.R.A.; Writing—review and editing, T.H.A.V., F.C.C., M.M.K. and M.A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

All authors declare no conflict of interest.

References

  1. National Research Council (NRC). Nutrient Requirements of Dogs and Cats; National Academies Press: Washington, DC, USA, 2006; ISBN 0309086280. [Google Scholar]
  2. Battault, S.; Whiting, S.J.; Peltier, S.L.; Sadrin, S.; Gerber, G.; Maixent, J.M. Vitamin D metabolism, functions and needs: From science to health claims. Eur. J. Nutr. 2013, 52, 429–441. [Google Scholar] [CrossRef] [PubMed]
  3. Zafalon, R.V.; Risolia, L.W.; Pedrinelli, V.; Vendramini, T.H.; Rodrigues, R.B.; Amaral, A.R.; Kogika, M.M.; Brunetto, M.A. Vitamin D metabolism in dogs and cats and its relation to diseases not associated with bone metabolism. J. Anim. Physiol. Anim. Nutr. 2020, 104, 322–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Pike, J.W.; Christakos, S. Biology and mechanisms of action of the vitamin D hormone. Endocrin. Metab. Clin. 2017, 46, 815–843. [Google Scholar] [CrossRef] [PubMed]
  5. Goltzman, D. Functions of vitamin D in bone. Histochem. Cell Biol. 2018, 149, 305–312. [Google Scholar] [CrossRef]
  6. Dusso, A.S.; Brown, A.J.; Slatopolsky, E. Vitamin D. Am. J. Physiol. Renal Physiol. 2005, 289, F8–F28. [Google Scholar] [CrossRef]
  7. Dittmer, K.E.; Thompson, K.G. Vitamin D metabolism and rickets in domestic animals: A review. Vet. Pathol. 2011, 48, 389–407. [Google Scholar] [CrossRef]
  8. Wasserman, R.H. Vitamin D and intestinal absorption of calcium: A view and overview. In Vitamin D; Feldman, J.W.P.D., Glorieux, F., Eds.; Academic Press: San Diego, CA, USA, 2005; p. 411. ISBN 9780080543642. [Google Scholar]
  9. Fujita, H.; Sugimoto, K.; Inatomi, S.; Maeda, T.; Osanai, M.; Uchiyama, Y.; Yamamoto, Y.; Wada, T.; Kojima, T.; Yokozaki, H.; et al. Tight junction proteins claudin-2 and -12 are critical for vitamin D dependent Ca2+ absorption between enterocytes. Mol. Biol. Cell. 2008, 19, 1912–1921. [Google Scholar] [CrossRef] [Green Version]
  10. St-Arnaud, R. The direct role of vitamin D on bone homeostasis. Arch. Biochem. Biophys. 2008, 473, 225–230. [Google Scholar] [CrossRef]
  11. Li, Y.C.; Pirro, A.E.; Amling, M.; Delling, G.; Baron, R.; Bronson, R.; Demay, M.B. Targeted ablation of the vitamin D receptor: An animal model of vitamin D-dependent rickets type II with alopecia. Proc. Natl. Acad. Sci. USA 1997, 94, 9831–9835. [Google Scholar] [CrossRef] [Green Version]
  12. Baldock, P.A.; Thomas, G.P.; Hodge, J.M.; Baker, S.U.; Dressel, U.; O’Loughlin, P.D.; Nicholson, G.C.; Briffa, K.H.; Eisman, J.A.; Gardiner, E.M. Vitamin D action and regulation of bone remodeling: Suppression of osteoclastogenesis by the mature osteoblast. J. Bone Miner. Res. 2006, 21, 1618–1626. [Google Scholar] [CrossRef]
  13. Fretz, J.A.; Zella, L.A.; Kim, S.; Shevde, N.K.; Pike, J.W. 1, 25-Dihydroxyvitamin D3 regulates the expression of low-density lipoprotein receptor-related protein 5 via deoxyribonucleic acid sequence elements located downstream of the start site of transcription. J. Mol. Endocrinol. 2006, 20, 2215–2230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Anderson, P.H.; Atkins, G.J. The skeleton as an intracrine organ for vitamin D metabolism. Mol. Asp. Med. 2008, 29, 397–406. [Google Scholar] [CrossRef] [PubMed]
  15. Hurst, E.A.; Homer, N.Z.; Mellanby, R.J. Vitamin D metabolism and profiling in veterinary species. Metabolites 2020, 10, 371. [Google Scholar] [CrossRef]
  16. Uhl, E.W. The pathology of vitamin D deficiency in domesticated animals: An evolutionary and comparative overview. Int. J. Paleopathol. 2018, 23, 100–109. [Google Scholar] [CrossRef] [PubMed]
  17. Laflamme, D.P.; Abood, S.K.; Fascetti, A.J.; Fleeman, L.M.; Freeman, L.M.; Michel, K.E.; Bauer, C.; Kemp, B.L.E.; Van Doren, J.R.; Willoughby, K.N. Pet feeding practices of dog and cat owners in the United States and Australia. J. Am. Vet. Med. Assoc. 2018, 232, 687–694. [Google Scholar] [CrossRef]
  18. Verbrugghe, A.D.; Paepe, L.; Verhaert, J.; Saunders, J.; Fritz, J.; Janssens, G.P.J.; Hesta, M. Metabolic bone disease and hyperparathyroidism in an adult dog fed an unbalanced homemade diet. Vlaams Diergeneeskd. Tijdschr. 2011, 80, 61–68. [Google Scholar]
  19. Dusso, A.S.; Thadhani, R.; Slatopolsky, E. Vitamin D receptor and analogs. Semin. Nephrol. 2004, 24, 10–16. [Google Scholar] [CrossRef] [Green Version]
  20. Parker, V.J.; Gilor, C.; Chew, D.J. Feline hyperparathyroidism: Pathophysiology, diagnosis and treatment of primary and secondary disease. J. Feline Med. Surg. 2015, 17, 427–439. [Google Scholar] [CrossRef]
  21. Mellanby, R.J.; Mee, A.P.; Berry, J.L.; Herrtage, M.E. Hypercalcaemia in two dogs caused by excessive dietary supplementation of vitamin D. J. Small Anim. Pract. 2005, 46, 334–338. [Google Scholar] [CrossRef]
  22. Morita, T.; Awakura, T.; Shimada, A.; Umemura, T.; Nagai, T.; Haruna, A. Vitamin D toxicosis in cats: Natural outbreak and experimental study. J. Vet. Med. Sci. 1995, 57, 831–837. [Google Scholar] [CrossRef] [Green Version]
  23. Wehner, A.; Katzenberger, J.; Groth, A.; Dorsch, R.; Koelle, P.; Hartmann, K.; Weber, K. Vitamin D intoxication caused by ingestion of commercial cat food in three kittens. J. Feline Med. Surg. 2013, 15, 730–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Crossley, V.J.; Bovens, C.P.; Pineda, C.; Hibbert, A.; Finch, N.C. Vitamin D toxicity of dietary origin in cats fed a natural complementary kitten food. J. Feline Med. Surg. 2017, 3, 2055116917743613. [Google Scholar] [CrossRef] [PubMed]
  25. Gunther, R.; Felice, L.J.; Nelson, R.K.; Franson, A.M. Toxicity of a vitamin D3 rodenticide to dogs. J. Am. Vet. Med. Assoc. 1988, 193, 211–214. [Google Scholar]
  26. Fooshee, S.K.; Forrester, S.D. Hypercalcemia secondary to cholecalciferol rodenticide toxicosis in two dogs. J. Am. Vet. Med. Assoc. 1990, 196, 1265–1268. [Google Scholar] [PubMed]
  27. Livezey, K.L.; Dorman, D.C.; Hooser, S.B.; Buck, W.B. Hypercalcemia induced by vitamin-d3 toxicosis in 2 dogs. Canine Pract. 1991, 16, 26–32. [Google Scholar]
  28. Berry, C.R.; Gallaway, A.; Thrall, D.E.; Carlisle, C. Rodenticide toxicity in fourteen dogs. Vet. Radiol. Ultrasound 1993, 34, 391–396. [Google Scholar] [CrossRef]
  29. Gerhard, C.; Jaffey, J.A. Persistent increase in serum 25-hydroxyvitamin D concentration in a dog following cholecalciferol intoxication. Front. Vet. Sci. 2020, 6, 472–477. [Google Scholar] [CrossRef] [Green Version]
  30. Finch, N.C. Hypercalcaemia in cats: The complexities of calcium regulation and associated clinical challenges. J. Feline Med. Surg. 2016, 18, 387–399. [Google Scholar] [CrossRef] [Green Version]
  31. Durtnell, R.E. Canine vitamin D toxicosis. J. Small Anim. Pract. 1999, 40, 550. [Google Scholar] [CrossRef]
  32. Hilbe, M.; Sydler, T.; Fischer, L.; Naegeli, H. Metastatic calcification in a dog attributable to ingestion of a tacalcitol ointment. Vet. Pathol. 2000, 37, 490–492. [Google Scholar] [CrossRef] [Green Version]
  33. Torley, D.; Drummond, A.; Bilsland, D.J. Calcipotriol toxicity in dogs. Br. J. Dermatol. 2002, 147, 1270. [Google Scholar] [CrossRef] [PubMed]
  34. Nakamura, K.; Tohyama, N.; Yamasaki, M.; Ohta, H.; Morishita, K.; Takiguchi, M. Hypercalcemia in a dog with chronic ingestion of maxacalcitol ointment. J. Am. Anim. Hosp. Assoc. 2016, 52, 256–258. [Google Scholar] [CrossRef] [PubMed]
  35. Schoenmakers, I.; Hazewinkel, H.A.W.; Voorhout, G.; Carlson, C.S.; Richardson, D. Effect of diets with different calcium and phosphorus contents on the skeletal development and blood chemistry of growing great danes. Vet. Rec. 2000, 147, 652–660. [Google Scholar] [PubMed]
  36. Tryfonidou, M.A.; Holl, M.S.; Stevenhagen, J.J.; Buurman, C.J.; Deluca, H.F.; Oosterlaken-Dijksterhuis, M.A.; Van Den Brom, W.E.; Van Leeuwen, J.P.; Hazewinkel, H.A. Dietary 135-fold cholecalciferol supplementation severely disturbs the endochondral ossification in growing dogs. Domest. Anim. Endocrinol. 2003, 24, 265–285. [Google Scholar] [CrossRef]
  37. Cline, J. Calcium and vitamin D metabolism, deficiency, and excess. Top. Companion Anim. Med. 2012, 27, 159–164. [Google Scholar] [CrossRef]
  38. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  39. How, K.L.; Hazewinkel, H.A.W.; Mol, J.A. Dietary vitamin D dependence of cat and dog due to inadequate cutaneous synthesis of vitamin D. Gen. Comp. Endocrinol. 1994, 96, 12–18. [Google Scholar] [CrossRef]
  40. Morris, J.G. Ineffective vitamin D synthesis in cats is reversed by an inhibitor of 7-dehydrocholestrol-Δ7-reductase. J. Nutr. 1999, 129, 903–908. [Google Scholar] [CrossRef] [Green Version]
  41. Norman, A. Vitamin D; Elsevier: Amsterdam, The Netherlands, 2012. [Google Scholar]
  42. Laing, C.J.; Malik, R.; Wigney, D.I.; Fraser, D.R. Seasonal vitamin D status of greyhounds in Sydney. Aust. Vet. J. 1999, 77, 35–38. [Google Scholar] [CrossRef]
  43. Morris, J.G. Idiosyncratic nutrient requirements of cats appear to be diet-induced evolutionary adaptations. Nutr. Res. Rev. 2002, 15, 153–168. [Google Scholar] [CrossRef] [Green Version]
  44. Parker, V.J.; Rudinsky, A.J.; Chew, D.J. Vitamin D metabolism in canine and feline medicine. J. Am. Vet. Med. Assoc. 2017, 250, 1259–1269. [Google Scholar] [CrossRef] [PubMed]
  45. Prosser, D.E.; Jones, G. Enzymes involved in the activation and inactivation of vitamin D. Trends Biochem. Sci. 2004, 29, 664–673. [Google Scholar] [CrossRef] [PubMed]
  46. Heaney, R.P.; Horst, R.L.; Cullen, D.M.; Armas, L.A. Vitamin D3 distribution and status in the body. J. Am. Coll. Nutr. 2009, 28, 252–256. [Google Scholar] [CrossRef] [PubMed]
  47. Morris, J.G. Cats discriminate between cholecalciferol and ergocalciferol. J. Anim. Physiol. Anim. Nutr. 2002, 86, 229–238. [Google Scholar] [CrossRef]
  48. Norman, A.W. From vitamin D to hormone D: Fundamentals of the vitamin D endocrine system essential for good health. Am. J. Clin. Nutr. 2008, 88, 491S–499S. [Google Scholar] [CrossRef] [Green Version]
  49. Dechant, K.L.; Goa, K.L. Calcitriol. Drugs Aging 1994, 5, 300–317. [Google Scholar] [CrossRef]
  50. Souberbielle, J.C.; Cavalier, E.; Delanaye, P.; Massart, C.; Brailly-Tabard, S.; Cormier, C.; Chanson, P. Serum calcitriol concentrations measured with a new direct automated assay in a large population of adult healthy subjects and in various clinical situations. Clin. Chim. Acta 2015, 451, 149–153. [Google Scholar] [CrossRef]
  51. Jones, G.; Strugnell, S.A.; DeLuca, H.F. Current understanding of the molecular actions of vitamin D. Physiol. Ver. 1998, 78, 1193–1231. [Google Scholar] [CrossRef] [Green Version]
  52. Azam, N.; Zhang, M.Y.H.; Wang, X.M.; Tenenhouse, H.S.; Portale, A.A. Disordered regulation of renal 25-hydroxyvitamin D-1a-hydroxylase gene expression by phosphorus in X-linked hypophosphatemic (Hyp) mice. Endocrinology 2003, 144, 3463–3468. [Google Scholar] [CrossRef] [Green Version]
  53. Fujiwara, I.; Aravindan, R.; Horst, R.L.; Drezner, M.K. Abnormal regulation of renal 25-hydroxyvitamin D-1 alpha-hydroxylase activity in X-linked hypophosphatemia: A translational or posttranslational defect. J. Bone Miner. Res. 2003, 18, 434–442. [Google Scholar] [CrossRef]
  54. Shimada, T.; Hasegawa, H.; Yamazaki, Y.; Muto, T.; Hino, R.; Takeuchi, Y.; Yamashita, T. FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J. Bone Miner. Res. 2004, 19, 429–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhong, Y.; Armbrecht, H.J.; Christakos, S. Calcitonin, a regulator of the 25-hydroxyvitamin D3 1α-hydroxylase gene. J. Biol. Chem. 2009, 284, 11059–11069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Brenza, H.L.; Kimmel-Jehan, C.; Jehan, F.; Shinki, T.; Wakino, S.; Anazawa, H.; Suda, T.; DeLuca, H.F. Parathyroid hormone activation of the 25-hydroxyvitamin D3-1a-hydroxylase gene promoter. Proc. Natl. Acad. Sci. USA 1998, 95, 1387–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Murayama, A.; Takeyama, K.; Kitanaka, S.; Kodera, Y.; Hosoya, T.; Kato, S. The promoter of the human 25-hydroxyvitamin D3 1ahydroxylase gene confers postive and negative responsiveness to PTH, calcitonin, and 1a,25(OH)2D3. Biochem. Biophys. Res. Commun. 1998, 249, 11–16. [Google Scholar] [CrossRef] [PubMed]
  58. Rost, C.R.; Bikle, D.D.; Kaplan, R.A. In vitro stimulation of 25- hydroxycholecalciferol 1a-hydroxylation by parathyroid hormone in chick kidney slices: Evidence for a role for adenosine 30,50-monophoshate. Endocrinology 1981, 108, 1002–1006. [Google Scholar] [CrossRef] [PubMed]
  59. Zierold, C.; Mings, J.A.; Deluca, H.F. Regulation of 25-hydroxyvitamin D3-24-hydroxylase mRNA by 1,25-dihydroxyvitamin D3 and parathyroid hormone. J. Cell. Biochem. 2003, 88, 234–237. [Google Scholar] [CrossRef] [PubMed]
  60. Christakos, S.; Ajibade, D.V.; Dhawan, P.; Fechner, A.J.; Mady, L.J. Vitamin D: Metabolism. Rheum. Dis. Clin. 2012, 38, 1–11. [Google Scholar] [CrossRef]
  61. de Brito Galvao, J.F.; Nagode, L.A.; Schenck, P.A.; Chew, D.J. Calcitriol, calcidiol, parathyroid hormone, and fibroblast growth factor-23 interactions in chronic kidney disease. J. Vet. Emerg. Crit. Care 2013, 23, 134–162. [Google Scholar] [CrossRef]
  62. Jones, G.; Prosser, D.E.; Kaufmann, M. Cytochrome P450-mediated metabolism of vitamin D. J. Lipid Res. 2014, 55, 13–31. [Google Scholar] [CrossRef] [Green Version]
  63. Norman, A.W.; Okamura, W.H.; Bishop, J.E.; Henry, H.L. Update on biological actions of 1α, 25 (OH) 2-vitamin D3 (rapid effects) and 24R, 25 (OH) 2-vitamin D3. Mol. Cell. Endocrinol. 2002, 197, 1–13. [Google Scholar] [CrossRef]
  64. Seiler, S.; Heine, G.H.; Fliser, D. Clinical relevance of FGF -23 in chronic kidney disease. Kidney Int. 2009, 76, S34–S42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dusso, A.S.; Tokumoto, M. Defective renal maintenance of the vitamin D endocrine system impairs vitamin D renoprotection: A downward spiral in kidney disease. Kidney Int. 2011, 79, 715–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hu, M.C.; Kuro, O.M.; Moe, O.W. Secreted klotho and chronic kidney disease. Adv. Exp. Med. Biol. 2012, 728, 126–157. [Google Scholar] [PubMed] [Green Version]
  67. Tryfonidou, M.A.; Holl, M.S.; Vastenburg, M.; Oosterlaken-Dijksterhuis, M.A.; Birkenhager-Frenkel, D.H.; Van Den Brom, W.E.; Hazewinkel, H.A. Hormonal regulation of calcium homeostasis in two breeds of dogs during growth at different rates. Anim. Sci. J. 2003, 81, 1568–1580. [Google Scholar] [CrossRef]
  68. Hazewinkel, H.A.; Tryfonidou, M.A. Vitamin D3 metabolism in dogs. Mol. Cell. Endocrinol. 2002, 197, 23–33. [Google Scholar] [CrossRef]
  69. Planchot, J.J.; DuBois, M.B.; Halpern, S.; Cournot-Witmer, G.; Garabedian, M.; Balsan, S. In vitro action of 1,25-dihydroxycholecalciferol and 24,25-dihydroxycholecalciferol on matrix organization and mineral distribution in rabbit growth plate. Metab. Bone Dis. Related Res. 1982, 4, 135–142. [Google Scholar] [CrossRef]
  70. Boyan, B.D.; Sylvia, V.L.; Dean, D.D.; Schwartz, Z. 24,25-(OH)2D3 regulates cartilage and bone via autocrine and endocrine mechanisms. Steroids 2001, 66, 363–374. [Google Scholar] [CrossRef]
  71. Burstein, S.; Chen, I.W.; Tsang, R.C. Effects of growth hormone replacement therapy on 1.2 5-dihydroxyvitamin D and calcium metabolism. J. Clin. Endocrinol. Metab. 1983, 56, 1246–1251. [Google Scholar] [CrossRef]
  72. Brixen, K.; Nielsen, H.K.; Bouillon, R.; Flyvbjerg, A.; Mosekilde, L. Effects of short-term growth hormone treatment on PTH, calcitriol, thyroid hormone, insulin and glucagon. Acta Endocrinol. 1992, 127, 331–336. [Google Scholar] [CrossRef]
  73. Marcus, R.; Butterfield, G.; Holloway, L.; Gilliland, L.; Baylink, D.J.; Hintz, R.L.; Sherman, B.M. Effects of short term administration of recombinant human growth hormone to elderly people. J. Clin. Endocrinol. Metab. 1990, 70, 519–527. [Google Scholar] [CrossRef]
  74. Condamine, L.; Vztovsnik, F.; Friedlander, G.; Menaa, C.; Garabedian, M. Local action of phosphate depletion and insulin-like growth factor I on in vitro production of 1,25-dihydroxyvitamin D by cultured mammalian kidney cells. J. Clin. Investig. 1994, 94, 1673–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lund, B.; Eskildsen, P.C.; Lund, B.; Norman, A.W.O.H. Sorensen calcium and vitamin D metabolism in acromegaly. Acta Endocrinol. 1981, 96, 444–450. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, S.; Grieff, M.; Brown, A.J. Regulation of renal vitamin D-24-hydroxylase by phosphate: Effects of hypophysectomy, growth hormone and insulin-like growth factor I. Biochem. Biophys. Res. Commun. 1997, 233, 813–817. [Google Scholar] [CrossRef] [PubMed]
  77. Brumbaugh, P.F.; Haussler, M.R. lα, 25-dihydroxyvitamin D3 receptor: Competitive binding of vitamin D analogs. Life Sci. 1973, 13, 1737–1746. [Google Scholar] [CrossRef]
  78. Haddad, J.G.J. Transport of vitamin D metabolites. Clin. Orthop. Relat. Res. 1979, 142, 249–261. [Google Scholar] [CrossRef]
  79. Carrilo-Lopez, N.; Fernández-Martín, J.L.; Cannata-Andía, J.B. The role of calcium, calcitriol and its receptors in parathyroid regulation. Nefrología 2009, 29, 103–108. [Google Scholar]
  80. Haussler, M.R.; Whitfield, G.K.; Haussler, C.A.; Hsieh, J.C.; Thompson, P.D.; Selznick, S.H.; Dominguez, C.E.; Jurutka, P.W. The nuclear vitamin D receptor: Biological and molecular regulatory properties revealed. J. Bone Miner. Res. 1998, 13, 325–349. [Google Scholar] [CrossRef]
  81. Jehan-Kimmel, C.; Darwish, H.M.; Strugnell, S.A.; Jehan, F.; Wiefling, B.; DeLuca, H.F. DNA bending is induced by binding of vitamin D receptor-retinoid X receptor heterodimers to vitamin D responsive elements. J. Cell. Biochem. 1999, 74, 220–228. [Google Scholar] [CrossRef]
  82. Rachez, C.; Freedman, L.P. Mechanisms of gene regulation by vitamin D3 receptor: A network of coactivator interactions. Gene 2000, 246, 9–21. [Google Scholar] [CrossRef]
  83. Rochel, N.; Wurtz, J.M.; Mitschler, A.; Klaholz, B.; Moras, D. The crystal structure of the nuclear receptor for vitamin D bound to its natural ligand. Mol. Cell. 2000, 5, 173–179. [Google Scholar] [CrossRef]
  84. McKenna, N.J.; O’Malley, B.W. Combinatorial control of gene expression by nuclear receptors and coregulators. Cell 2002, 108, 465–474. [Google Scholar] [CrossRef] [Green Version]
  85. Ramagopalan, S.V.; Heger, A.; Berlanga, A.J.; Maugeri, N.J.; Lincoln, M.R.; Burrell, A.; Watson, C.T. A ChIP-seq defined genome-wide map of vitamin D receptor binding: Associations with disease and evolution. Genome Res. 2010, 20, 1352–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Huhtakangas, J.A.; Olivera, C.J.; Bishop, J.E.; Zanello, L.P.; Norman, A.W. The vitamin D receptor is present in caveolae-enriched plasma membranes and binds 1,25(OH)2-vitamin D3 in vivo and in vitro. Mol. Endocrinol. 2004, 18, 2660–2671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Massheimer, V.; Boland, R.; De Boland, A.R. Rapid 1,25(OH)2-vitamin D3 stimulation of calcium uptake by rat intestinal cells involves a dihydropyridine-sensitive cAMPdependent pathway. Cell. Signal. 1994, 6, 299–304. [Google Scholar] [CrossRef]
  88. Feldman, D.; Pike, J.W.; Glorieux, F.H. Vitamin D, 2nd ed.; Elsevier Academic Press: San Diego, CA, USA, 2005. [Google Scholar]
  89. Norman, A.W. Minireview: Vitamin D receptor: New assignments for an already busy receptor. Endocrinology 2006, 147, 5542–5548. [Google Scholar] [CrossRef] [Green Version]
  90. Zanello, L.; Norman, A.W. Stimulation by 1,25(OH)2-vitamin D3 of whole cell chloride currents in osteoblastic ROS 17/2.8 cells: A structure-function study. J. Biol. Chem. 1997, 272, 22617–22622. [Google Scholar] [CrossRef] [Green Version]
  91. Le Mellay, V.; Grosse, B.; Lieberherr, M. Phospholipase C beta and membrane action of calcitriol and estradiol. J. Biol. Chem. 1997, 272, 11902–11907. [Google Scholar] [CrossRef] [Green Version]
  92. Hoenderop, J.G.; Nilius, B.; Bindels, R.J. Calcium absorption across epithelia. Physiol. Rev. 2005, 85, 373–422. [Google Scholar] [CrossRef] [Green Version]
  93. Meyer, M.B.; Watanuki, M.; Kim, S.; Shevde, N.K.; Pike, J.W. The human transient receptor potential vanilloid type 6 distal promoter contains multiple vitamin D receptor binding sites that mediate activation by 1,25-dihydroxyvitamin D3 in intestinal cells. Mol. Endocrinol. 2006, 20, 1447–1461. [Google Scholar] [CrossRef] [Green Version]
  94. Li, Y.C.; Amling, M.; Pirro, A.E.; Priemel, M.; Meuse, J.; Baron, R.; Delling, G.; Demay, M.B. Normalization of mineral ion homeostasis by dietary means prevents hyperparathyroidism, rickets and osteomalacia, but not alopecia in vitamin D receptor-ablated mice. Endocrinology 1998, 139, 4391–4396. [Google Scholar] [CrossRef]
  95. Van Cromphaut, S.J.; Dewerchin, M.; Hoenderop, J.G.; Stockmans, I.; Van Herck, E.; Kato, S.; Bindels, R.J.; Collen, D.; Carmeliet, P.; Bouillon, R.; et al. Duodenal calcium absorption in vitamin D receptor-knockout mice: Functional and molecular aspects. Proc. Natl. Acad. Sci. USA 2001, 98, 13324–13329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Christakos, S.; Dhawan, P.; Porta, A.; Mady, L.J.; Seth, T. Vitamin D and intestinal calcium absorption. Mol. Cell. Endocrinol. 2011, 347, 9–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Tsukita, S.; Furuse, M.; Itoh, M. Multifunctional strands in tight junctions. Nat. Rev. Mol. Cell. Biol. 2001, 2, 285–293. [Google Scholar] [CrossRef] [PubMed]
  98. Kutuzova, G.D.; DeLuca, H.F. Gene expression profiles in rat intestine identify pathways for 1, 25-dihydroxyvitamin D3 stimulated calcium absorption and clarify its immunomodulatory properties. Arch. Biochem. Biophys. 2004, 432, 152–166. [Google Scholar] [CrossRef] [PubMed]
  99. Kiela, P.R.; Ghishan, F.K. Physiology of intestinal absorption and secretion. Best. Pract. Res. Clin. Gastroenterol. 2016, 30, 145–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Boros, S.; Bindels, R.; Hoenderop, J. Active Ca2þ reabsorption in the connecting tubule. Pflugers Arch. 2009, 458, 99–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Hoenderop, J.G.J.; Willems, P.H.G.M.; Bindels, R.J.M. Toward a comprehensive molecular model of active calcium reabsorption. Am. J. Physiol. 2000, 278, F352–F360. [Google Scholar] [CrossRef]
  102. Hoenderop, J.G.J.; Muller, D.; Suzuki, M.; Van Os, C.H.; Bindels, R.J.M. The epithelial calcium channel: Gatekeeper of active calcium reabsorption. Curr. Opin. Nephrol. Hypertens. 2000, 9, 335–340. [Google Scholar] [CrossRef]
  103. Hoenderop, J.G.J.; Van der Kemp, A.W.C.M.; Hartog, A.; Van de Graaf, S.F.J.; Van Os, C.H.; Willems, P.H.G.M.; Bindels, R.J.M. Molecular identification of the apical Ca21 channel in 1,25-dihydroxyvitamin D-responsive epithelia. J. Biol. Chem. 1999, 274, 8375–8378. [Google Scholar] [CrossRef] [Green Version]
  104. Hoenderop, J.G.J.; Dardenne, O.; Van Abel, M.; Van Der Kemp, A.W.C.M.; Van Os, C.H.; St-Arnaud, R.; Bindels, R.J.M. Modulation of renal Ca2þ transport protein genes by dietary Ca2þ and 1,25- dihydroxyvitamin D3 in 25-hydroxyvitamin D3-1a-hydroxylase knockout mice. FASEB J. 2002, 16, 1398–1406. [Google Scholar] [CrossRef]
  105. Hoenderop, J.G.J.; Muller, D.; Van Der Kemp, A.W.; Hartog, A.; Suzuki, M.; Ishibashi, K.; Imai, M.; Sweep, F.; Willems, P.H.; Van Os, C.H.; et al. Calcitriol controls the epithelial calcium channel in kidney. J. Am. Soc. Nephrol. 2001, 12, 1342–1349. [Google Scholar] [PubMed]
  106. Van Abel, M.; Hoenderop, J.G.J.; Dardenne, O.; St Arnaud, R.; Van Os, C.H.; Van Leeuwen, H.J.P.T.M.; Bindels, R.J.M. 1,25- dihydroxyvitamin D3-independent stimulatory effect of estrogen on the expression of ECaC1 in the kidney. J. Am. Soc. Nephrol. 2002, 13, 2102–2109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Naveh-Many, T.; Marx, R.; Keshet, E.; Pike, J.W.; Silver, J. Regulation of 1,25-dihydroxyvitamin D3 receptor gene expression by 1,25-dihydroxyvitamin D3 in the parathyroid in vivo. J. Clin. Investig. 1990, 86, 1968–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Jacquillet, G.; Unwin, R.J. Physiological regulation of phosphate by vitamin D, parathyroid hormone (PTH) and phosphate (Pi). Pflügers Arch. Eur. J. Phy. 2019, 471, 83–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Silver, J.; Russell, J.; Sherwood, L.M. Regulation by vitamin D metabolites of messenger ribonucleic acid for preproparathyroid hormone in isolated bovine parathyroid cells. Proc. Natl. Acad. Sci. USA 1985, 82, 4270–4273. [Google Scholar] [CrossRef] [Green Version]
  110. Yuen, N.K.; Ananthakrishnan, S.; Campbell, M.J. Hyperparathyroidism of renal disease. Perm. J. 2016, 20, 78–83. [Google Scholar] [CrossRef] [Green Version]
  111. Hostutler, R.A.; DiBartola, S.P.; Chew, D.J.; Nagode, L.A.; Schenck, P.A.; Rajala-Schultz, P.J.; Drost, W.T. Comparison of the effects of daily and intermittent-dose calcitriol on serum parathyroid hormone and ionized calcium concentrations in normal cats and cats with chronic renal failure. J. Vet. Intern. Med. 2006, 20, 1307–1313. [Google Scholar] [CrossRef]
  112. IRIS. Treatment recommendations for CKD in dogs. Int. Ren. Interest Soc. 2019, 1–16. [Google Scholar]
  113. IRIS. Treatment recommendations for CKD in cats. Int. Ren. Interest Soc. 2019, 1–16. [Google Scholar]
  114. De Borst, M.H.; Hajhosseiny, R.; Tamez, H.; Wenger, J.; Thadhani, R.; Goldsmith, D.J. Active vitamin D treatment for reduction of residual proteinuria: A systematic review. J. Am. Soc. Nephrol. 2013, 24, 1863–1871. [Google Scholar] [CrossRef]
  115. Li, X.H.; Feng, L.; Yang, Z.H.; Liao, Y.H. Effect of active vitamin D on cardiovascular outcomes in predialysis chronic kidney diseases: A systematic review and meta-analysis. Nephrology 2015, 20, 706–714. [Google Scholar] [CrossRef] [PubMed]
  116. Toussaint, N.D.; Damasiewicz, M.J. Do the benefits of using calcitriol and other vitamin D receptor activators in patients with chronic kidney disease outweigh the harms? Nephrology 2017, 22, 51–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Wu, C.C.; Liao, M.T.; Hsiao, P.J.; Lu, C.L.; Hsu, Y.J.; Lu, K.C.; Chu, P. Antiproteinuria effect of calcitriol in patients with chronic kidney disease and vitamin D deficiency: A randomized controlled study. J. Ren. Nutr. 2020, 30, 200–207. [Google Scholar] [CrossRef] [PubMed]
  118. Arai, Y.; Kanda, E.; Iimori, S.; Naito, S.; Noda, Y.; Kawasaki, T.; Sato, H.; Ando, R.; Sasaki, S.; Sohara, E.; et al. The use of vitamin D analogs is independently associated with the favorable renal prognosis in chronic kidney disease stages 4–5: The CKD-route study. Clin. Exp. Nephrol. 2017, 21, 481–487. [Google Scholar] [CrossRef] [PubMed]
  119. Canaff, L.; Hendy, G.N. Human calcium-sensing receptor gene. Vitamin D response elements in promoters P1 and P2 confer transcriptional responsiveness to 1,25-dihydroxyvitamin D. J. Biol. Chem. 2002, 277, 30337–30350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Suda, T.; Takahashi, N.; Udagawa, N.; Jimi, E.; Gillespie, M.T.; Martin, T.J. Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr. Ver. 1999, 20, 345–357. [Google Scholar] [CrossRef]
  121. Suda, T.; Ueno, Y.; Fujii, K.; Shinki, T. Vitamin D and bone. J. Cell. Biochem. 2003, 88, 259–266. [Google Scholar] [CrossRef]
  122. Kitazawa, S.; Kajimoto, K.; Kondo, T.; Kitazawa, R. Vitamin D3 supports osteoclastogenesis via functional vitamin D response element of human RANKL gene promoter. J. Cell. Biochem. 2003, 89, 771–777. [Google Scholar] [CrossRef]
  123. Kondo, T.; Kitazawa, R.; Maeda, S.; Kitazawa, S. 1a,25 Dihydroxyvitamin D3 rapidly regulates the mouse osteoprotegerin gene through dual pathways. J. Bone Miner. Res. 2004, 19, 1411–1419. [Google Scholar] [CrossRef]
  124. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch. Biochem. Biophys. 2008, 473, 139–146. [Google Scholar] [CrossRef] [Green Version]
  125. Yamamoto, Y.; Yoshizawa, T.; Fukuda, T.; Shirode-Fukuda, Y.; Yu, T.; Sekine, K.; Sato, T.; Kawano, H.; Aihara, K.; Nakamichi, Y.; et al. Vitamin D receptor in osteoblasts is a negative regulator of bone mass control. Endocrinology 2013, 154, 1008–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Naja, R.P.; Dardenne, O.; Arabian, A.; St. Arnaud, R. Chondrocyte-specific modulation of Cyp27b1 expression supports a role for local synthesis of 1,25-dihydroxyvitamin D3 in growth plate development. Endocrinology 2009, 150, 4024–4032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, molecular mechanism of action, and pleiotropic effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [PubMed]
  128. European Pet Food Industry Federation (FEDIAF). Nutritional Guidelines for Complete and Complementary Pet Food for Cats and Dogs; FEDIAF: Bruxelles, Belgium, 2020; 96p. [Google Scholar]
  129. Association of American Feed Control Officials. AAFCO Dog and Cat Food Nutrient Profiles; 2018 official publication; Association of American Feed Control Officials: Oxford, UK, 2018; ISBN 978-0-9983250-3-3. [Google Scholar]
  130. Mellanby, E. A further demonstration of the part played by accessory food factors in the aetiology of rickets. J. Physiol. 1918, 52, 53–54. [Google Scholar]
  131. Arnold, A.; Elvehjem, C.A. Nutritional requirements of dogs. J. Am. Vet. Med. Assoc. 1939, 95, 187–194. [Google Scholar]
  132. Michaud, L.; Elvehjem, E.A. The nutritional requirements of the dog. N. Am. Vet. 1944, 25, 657–666. [Google Scholar]
  133. Tryfonidou, M.A.; Stevenhagen, J.J.; Van Den Bemd, G.J.; Oosterlaken-Dijksterhuis, M.A.; DeLuca, H.F.; Mol, J.A.; Van Den Brom, W.E.; Van Leeuwen, J.P.; Hazewinkel, H.A. Moderate cholecalciferol supplementation depresses intestinal calcium absorption in growing dogs. J. Nutr. 2002, 132, 2644–2650. [Google Scholar] [CrossRef] [Green Version]
  134. Gershoff, S.N.; Legg, M.A.; O’connor, F.J.; Hegsted, D.M. The effect of vitamin D-deficient diets containing various Ca: P ratios on cats. J. Nutr. 1957, 63, 79–93. [Google Scholar] [CrossRef]
  135. Morris, J.G.; Earle, K.E.; Anderson, P.A. Plasma 25-hydroxyvitamin D in growing kittens is related to dietary intake of cholecalciferol. J. Nutr. 1999, 129, 909–912. [Google Scholar] [CrossRef] [Green Version]
  136. Morris, J.G.; Earle, K.E. Growing kittens require less dietary calcium than current allowances. J. Nutr. 1999, 129, 1698–1704. [Google Scholar] [CrossRef] [Green Version]
  137. Takeda, E.; Yamamoto, H.; Taketani, Y.; Miyamoto, K.I. Vitamin D-dependent rickets type I and type II. Pediatr. Int. 1997, 39, 508–513. [Google Scholar] [CrossRef] [PubMed]
  138. LeVine, D.N.; Zhou, Y.; Ghiloni, R.J.; Fields, E.L.; Birkenheuer, A.J.; Gookin, J.L.; Roberston, I.D.; Malloy, P.J.; Feldman, D. Hereditary 1,25-Dihydroxyvitamin D-Resistant Rickets in a Pomeranian Dog Caused by a Novel Mutation in the Vitamin D Receptor Gene. J. Vet. Intern. Med. 2009, 23, 1278–1283. [Google Scholar] [CrossRef] [PubMed]
  139. Malik, R.; Laing, C.; Davis, P.E.; Allan, G.S.; Wigney, D.I. Rickets in a litter of racing greyhounds. J. Small Anim. Pract. 1997, 38, 109–114. [Google Scholar] [CrossRef] [PubMed]
  140. Thacher, T.D.; Clarke, B.L. Vitamin D insufficiency. Mayo Clin. Proc. 2011, 86, 50–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Hatun, S.; Ozkan, B.; Orbak, Z.; Doneray, H.; Cizmecioglu, F.; Toprak, D.; Calikoglu, A.S. Vitamin D deficiency in early infancy. J. Nutr. 2005, 135, 279–282. [Google Scholar] [CrossRef] [PubMed]
  142. Wharton, B.; Bishop, N. Rickets. Lancet 2003, 362, 1389–1400. [Google Scholar] [CrossRef]
  143. Johnson, K.A.; Church, D.B.; Barton, R.J.; Wood, A.K.W. Vitamin D-dependent rickets in a Saint Bernard dog. J. Small Anim. Pract. 1988, 29, 657–666. [Google Scholar] [CrossRef]
  144. Schreiner, C.A.; Nagode, L.A. Vitamin D-dependent rickets type 2 in a four-month-old cat. J. Am. Vet. Med. Assoc. 2003, 222, 337–339. [Google Scholar] [CrossRef]
  145. Godfrey, D.R.; Anderson, R.M.; Barber, P.J.; Hewison, M. Vitamin D-dependent rickets type II in a cat. J. Small Anim. Pract. 2005, 46, 440–444. [Google Scholar] [CrossRef]
  146. Tanner, E.; Langley-Hobbs, S.J. Vitamin D-dependent rickets type 2 with characteristic radiographic changes in a 4-month-old kitten. J. Feline Med. Surg. 2005, 7, 307–311. [Google Scholar] [CrossRef]
  147. Geisen, V.; Weber, K.; Hartmann, K. Vitamin D-Dependent Hereditary Rickets Type I in a Cat. J. Vet. Intern. Med. 2009, 23, 196–199. [Google Scholar] [CrossRef] [PubMed]
  148. Grahn, R.A.; Ellis, M.R.; Grahn, J.C.; Lyons, L.A. A novel CYP27B1 mutation causes a feline vitamin D-dependent rickets type IA. J. Feline Med. Surg. 2012, 14, 587–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Teshima, T.; Kurita, S.; Sasaki, T.; Matsumoto, H.; Niina, A.; Abe, D.; Kanno, N.; Koyama, H. A genetic variant of CYP2R1 identified in a cat with type 1B vitamin D-dependent rickets: A case report. BMC Vet. Res. 2019, 15, 62. [Google Scholar] [CrossRef]
  150. Berry, J.L.; Davies, M.; Mee, A.P. Vitamin D metabolism, rickets, and osteomalacia. Semin. Musculoskelet. Radiol. 2002, 6, 173–181. [Google Scholar] [CrossRef] [PubMed]
  151. de Fornel-Thibaud, P.; Blanchard, G.; Escoffier-Chateau, L.; Segond, S.; Guetta, F.; Begon, D.; Delisle, F.; Rosenberg, D. Unusual case of osteopenia associated with nutritional calcium and vitamin D deficiency in an adult dog. J. Am. Anim. Hosp. Assoc. 2007, 43, 52–60. [Google Scholar] [CrossRef]
  152. Tal, M.; Parr, J.M.; MacKenzie, S.; Verbrugghe, A. Dietary imbalances in a large breed puppy, leading to compression fractures, vitamin D deficiency, and suspected nutritional secondary hyperparathyroidism. Can. Vet. J. La Rev. Vet. Can. 2018, 59, 36–42. [Google Scholar]
  153. Cloutier, M.; Gascon-Barré, M.; Amour, P.D. Chronic adaptation of dog parathyroid function to a low-calcium-high-sodium-vitamin D-deficient diet. J. Bone Miner. Res. 1992, 7, 1021–1028. [Google Scholar] [CrossRef]
  154. Garabedian, M.; Tanaka, Y.; Holick, M.F.; Deluca, H.F. Response of intestinal calcium transport and bone calcium mobilization to 1,25-Dihydroxyvitamin D3 in thyroparathyroidectomized rats. Endocrinology 1974, 94, 1022–1027. [Google Scholar] [CrossRef]
  155. Veldurthy, V.; Wei, R.; Oz, L.; Dhawan, P.; Jeon, Y.H.; Christakos, S. Vitamin D, calcium homeostasis and aging. Bone Res. 2016, 4, 1–7. [Google Scholar] [CrossRef] [Green Version]
  156. Brown, E.M. Role of the calcium-sensing receptor in extracellular calcium homeostasis. Best. Pract. Res. Clin. Endocrinol. Metab. 2013, 27, 333–343. [Google Scholar] [CrossRef]
  157. Tomsa, K.; Glaus, T.; Hauser, B.; Flückiger, M.; Arnold, P.; Wess, G.; Reusch, C. Nutritional secondary hyperparathyroidism in six cats. J. Small Anim. Pract. 1999, 40, 533–539. [Google Scholar] [CrossRef] [PubMed]
  158. Rumbeiha, W.K.; Kruger, J.M.; Fitzgerald, S.F.; Nachreiner, R.F.; Kaneene, J.B.; Braselton, W.E.; Chiapuzio, C.L. Use of pamidronate to reverse vitamin D3-induced toxicosis in dogs. Am. J. Vet. Res. 1999, 60, 1092–1097. [Google Scholar] [PubMed]
  159. Selby, P.L.; Davies, M.; Marks, J.S.; Mawer, E.B. Vitamin D intoxication causes hypercalcaemia by increased bone resorption which responds to pamidronate. Clin. Endocrinol. 1995, 43, 531–536. [Google Scholar] [CrossRef] [PubMed]
  160. Adams, J.S. Vitamin D metabolite-mediated hypercalcemia. Endocrin. Metab. Clin. 1989, 18, 768–778. [Google Scholar] [CrossRef]
  161. Gerber, B.; Hauser, B.; Reusch, C.E. Serum levels of 25-hydroxycholecalciferol and 1,25dihydroxycholecalciferol in dogs with hypercalcaemia. Vet. Res. Commun. 2004, 28, 669–680. [Google Scholar] [CrossRef] [PubMed]
  162. Selting, K.A.; Sharp, C.R.; Ringold, R.; Thamm, D.H.; Backus, R. Serum 25-hydroxyvitamin D concentrations in dogs–correlation with health and cancer risk. Vet. Comp. Oncol. 2016, 14, 295–305. [Google Scholar] [CrossRef] [Green Version]
  163. Wakshlag, J.J.; Rassnick, K.M.; Malone, E.K.; Struble, A.M.; Vachhani, P.; Trump, D.L.; Tian, L. Cross-sectional study to investigate the association between vitamin D status and cutaneous mast cell tumours in Labrador retrievers. Br. J. Nutr. 2011, 106, S60–S63. [Google Scholar] [CrossRef] [Green Version]
  164. Weidner, N.; Woods, J.P.; Conlon, P.; Meckling, K.A.; Atkinson, J.L.; Bayle, J.; Verbrugghe, A. Influence of various factors on circulating 25 (OH) vitamin D concentrations in dogs with cancer and healthy dogs. J. Vet. Intern. Med. 2017, 31, 1796–1803. [Google Scholar] [CrossRef]
  165. Kraus, M.S.; Rassnick, K.M.; Wakshlag, J.J.; Gelzer, A.R.M.; Waxman, A.S.; Struble, A.M.; Refsal, K. Relation of vitamin D status to congestive heart failure and cardiovascular events in dogs. J. Vet. Intern. Med. 2014, 28, 109–115. [Google Scholar] [CrossRef] [Green Version]
  166. Osuga, T.; Nakamura, K.; Morita, T.; Lim, S.Y.; Nisa, K.; Yokoyama, N.; Takiguchi, M. Vitamin D status in different stages of disease severity in dogs with chronic valvular heart disease. J. Vet. Intern. Med. 2015, 29, 1518–1523. [Google Scholar] [CrossRef] [Green Version]
  167. Allenspach, K.; Rizzo, J.; Jergens, A.E.; Chang, Y.M. Hypovitaminosis D is associated with negative outcome in dogs with protein losing enteropathy: A retrospective study of 43 cases. BMC Vet. Res. 2017, 13, 96–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Gow, A.G.; Else, R.; Evans, H.; Berry, J.L.; Herrtage, M.E.; Mellanby, R.J. Hypovitaminosis D in dogs with inflammatory bowel disease and hypoalbuminaemia. J. Small Anim. Pract. 2011, 52, 411–418. [Google Scholar] [CrossRef] [PubMed]
  169. Lalor, S.; Schwartz, A.M.; Titmarsh, H.; Reed, N.; Tasker, S.; Boland, L.; Mellanby, R.J. Cats with inflammatory bowel disease and intestinal small cell lymphoma have low serum concentrations of 25-hydroxyvitamin D. J. Vet. Intern. Med. 2014, 28, 351–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Titmarsh, H.F.; Gow, A.G.; Kilpatrick, S.; Cartwright, J.A.; Milne, E.M.; Philbey, A.W.; Mellanby, R.J. Low vitamin D status is associated with systemic and gastrointestinal inflammation in dogs with a chronic enteropathy. PLoS ONE 2015, 10, e0137377. [Google Scholar] [CrossRef] [Green Version]
  171. Titmarsh, H.; Gow, A.G.; Kilpatrick, S.; Sinclair, J.; Hill, T.; Milne, E.; Philbey, A.; Berry, J.; Handel, I.; Mellanby, R.J. Association of vitamin D status and clinical outcome in dogs with a chronic enteropathy. J. Vet. Intern. Med. 2015, 29, 1473–1478. [Google Scholar] [CrossRef]
  172. Kim, D.I.; Kim, H.; Son, P.; Kang, J.H.; Kang, B.T.; Yang, M.P. Serum 25-hydroxyvitamin D concentrations in dogs with suspected acute pancreatitis. J. Vet. Med. Sci. 2017, 79, 1366–1373. [Google Scholar] [CrossRef] [Green Version]
  173. Laws, E.J.; Kathrani, A.; Harcourt-Brown, T.R.; Granger, N.; Rose, J.H. 25-Hydroxy vitamin D3 serum concentration in dogs with acute polyradiculoneuritis compared to matched controls. J. Small Anim. Pract. 2018, 59, 222–227. [Google Scholar] [CrossRef]
  174. Cortadellas, O.; Fernandez del Palacio, M.J.; Talavera, J.; Bayón, A. Calcium and phosphorus homeostasis in dogs with spontaneous chronic kidney disease at different stages of severity. J. Vet. Intern. Med. 2010, 24, 73–79. [Google Scholar] [CrossRef]
  175. Galler, A.; Tran, J.L.; Krammer-Lukas, S.; Höller, U.; Thalhammer, J.G.; Zentek, J.; Willmann, M. Blood vitamin levels in dogs with chronic kidney disease. Vet. J. 2012, 192, 226–231. [Google Scholar] [CrossRef]
  176. Gerber, B.; Hässig, M.; Reusch, C.E. Serum concentrations of 1,25-dihydroxycholecalciferol and 25-hydroxycholecalciferol in clinically normal dogs and dogs with acute and chronic renal failure. Am. J. Vet. Res. 2003, 64, 1161–1166. [Google Scholar] [CrossRef]
  177. Parker, V.J.; Harjes, L.M.; Dembek, K.; Young, G.S.; Chew, D.J.; Toribio, R.E. Association of vitamin D metabolites with parathyroid hormone, fibroblast growth factor-23, calcium, and phosphorus in dogs with various stages of chronic kidney disease. J. Vet. Intern. Med. 2017, 31, 791–798. [Google Scholar] [CrossRef] [PubMed]
  178. Lalor, S.M.; Mellanby, R.J.; Friend, E.J.; Bowlt, K.L.; Berry, J.; Gunn-Moore, D. Domesticated cats with active mycobacteria infections have low serum vitamin D (25 (OH) D) concentrations. Transbound. Emerg. Dis. 2012, 59, 279–281. [Google Scholar] [CrossRef] [PubMed]
  179. Rodriguez-Cortes, A.; Martori, C.; Martinez-Florez, A.; Clop, A.; Amills, M.; Kubejko, J.; Llull, J.; Nadal, J.M.; Alberola, J. Canine Leishmaniasis progression is associated with vitamin D deficiency. Sci. Rep. 2017, 7, 3346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Rosa, C.T.; Schoeman, J.P.; Berry, J.L.; Mellanby, R.J.; Dvir, E. Hypovitaminosis D in dogs with spirocercosis. J. Vet. Intern. Med. 2013, 27, 1159–1164. [Google Scholar] [CrossRef] [PubMed]
  181. Titmarsh, H.F.; Lalor, S.M.; Tasker, S.; Barker, E.N.; Berry, J.; Gunn-More, D.; Mellanby, R.J. Vitamin D status in cats with feline immunodeficiency virus. Vet. Med. Sci. 2015, 1, 72–78. [Google Scholar] [CrossRef] [Green Version]
  182. Titmarsh, H.; Kilpatrick, S.; Sinclair, J.; Boag, A.; Bode, E.F.; Lalor, S.M.; Gaylor, D.; Berry, J.; Bommer, N.X.; Gunn-Moore, D.; et al. Vitamin D status predicts 30 day mortality in hospitalised cats. PLoS ONE 2015, 10, e0125997. [Google Scholar] [CrossRef]
  183. Barroga, E.F.; Kadosawa, T.; Okumura, M.; Fujinaga, T. Effects of vitamin D and retinoids on the differentiation and growth in vitro of canine osteosarcoma and its clonal cell lines. Res. Vet. Sci. 1999, 66, 231–236. [Google Scholar] [CrossRef]
  184. Kunakornsawat, S.; Rosol, T.J.; Capen, C.C.; Middleton, R.P.; Hannah, S.S.; Inpanbutr, N. Effects of 1,25 (OH) 2D3, EB1089, and analog V on PTHrP production, PTHrP mRNA expression and cell growth in SCC 2/88. Anticancer Res. 2001, 21, 3355–3363. [Google Scholar]
  185. Kunakornsawat, S.; Rosol, T.J.; Capen, C.C.; Omdahl, J.L.; Leroy, B.E.; Inpanbutr, N. Effects of 1, 25 (OH) 2D3, 25OHD3, and EB1089 on cell growth and Vitamin D receptor mRNA and 1α-hydroxylase mRNA expression in primary cultures of the canine prostate. J. Steroid Biochem. Mol. Biol. 2004, 89, 409–412. [Google Scholar] [CrossRef]
  186. Kunakornsawat, S.; Rosol, T.J.; Capen, C.C.; Reddy, G.S.; Binderup, L.; Inpanbutr, N. Effects of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] and its analogues (EB1089 and analog V) on canine adenocarcinoma (CAC-8) in nude mice. Biol. Pharm. Bull. 2002, 25, 642–647. [Google Scholar] [CrossRef] [Green Version]
  187. Rassnick, K.M.; Muindi, J.R.; Johnson, C.S.; Balkman, C.E.; Ramnath, N.; Yu, W.D.; Engler, K.L.; Page, R.L.; Trump, D.L. In vitro and in vivo evaluation of combined calcitriol and cisplatin in dogs with spontaneously occurring tumors. Cancer Chemother. Pharmacol. 2008, 62, 881–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Malone, E.K.; Rassnick, K.M.; Wakshlag, J.J.; Russell, D.S.; Al-Sarraf, R.; Ruslander, D.M.; Johnson, C.S.; Trump, D.L. Calcitriol (1,25-dihydroxycholecalciferol) enhances mast cell tumour chemotherapy and receptor tyrosine kinase inhibitor activity in vitro and has single-agent activity against spontaneously occurring canine mast cell tumours. Vet. Comp. Oncol. 2010, 8, 209–220. [Google Scholar] [CrossRef] [PubMed]
Table 1. Minimum and maximum recommendations of dietary cholecalciferol for adult dogs and cats according to scientific literature.
Table 1. Minimum and maximum recommendations of dietary cholecalciferol for adult dogs and cats according to scientific literature.
MinimumMaximum
DogCatDogCat
NRC (2006) (IU/Mkcal)110 a/136 b56 a/70 b8007520
FEDIAF (2020) (IU/Mkcal)138 c/159 d62.5 e/83.3 f8007500
AAFCO (2018) (IU/Mkcal)125707507520
a Adequate intake; b recommended allowance; c 110 kcal × (kg of body weight)0.75 as fed; d 95 kcal × (kg of body weight)0.75 as fed; e 100 kcal × (kg of body weight)0.67 as fed; f 75 kcal × (kg of body weight)0.67 as fed. NRC = National Research Council; FEDIAF = European Pet Food Industry Federation; AAFCO = American Feed Control Officials; IU = international units; Mkcal = megacalories.
Table 2. Minimum and maximum recommendations of dietary cholecalciferol for growing puppies and kittens according to scientific literature.
Table 2. Minimum and maximum recommendations of dietary cholecalciferol for growing puppies and kittens according to scientific literature.
MinimumMaximum
DogsCatsDogsCats
NRC (2006) (IU/Mkcal)110 a–136 b- a/56 b8007520
FEDIAF (2020) (IU/Mkcal)125 c/138 d708007500
AAFCO (2018) (IU/Mkcal)125707507520
a Adequate intake; b recommended allowance; c Late growth (>14 weeks); d Early growth (<14 weeks). NRC = National Research Council; FEDIAF = European Pet Food Industry Federation; AAFCO = American Feed Control Officials; IU = international units; Mkcal = megacalories.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zafalon, R.V.A.; Ruberti, B.; Rentas, M.F.; Amaral, A.R.; Vendramini, T.H.A.; Chacar, F.C.; Kogika, M.M.; Brunetto, M.A. The Role of Vitamin D in Small Animal Bone Metabolism. Metabolites 2020, 10, 496. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo10120496

AMA Style

Zafalon RVA, Ruberti B, Rentas MF, Amaral AR, Vendramini THA, Chacar FC, Kogika MM, Brunetto MA. The Role of Vitamin D in Small Animal Bone Metabolism. Metabolites. 2020; 10(12):496. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo10120496

Chicago/Turabian Style

Zafalon, Rafael Vessecchi Amorim, Bruna Ruberti, Mariana Fragoso Rentas, Andressa Rodrigues Amaral, Thiago Henrique Annibale Vendramini, Fernanda Chicharo Chacar, Marcia Mery Kogika, and Marcio Antonio Brunetto. 2020. "The Role of Vitamin D in Small Animal Bone Metabolism" Metabolites 10, no. 12: 496. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo10120496

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop