Next Article in Journal
Association between Micronutrient-Related Dietary Pattern and Cognitive Function among Persons 55 Years and Older in China: A Longitudinal Study
Next Article in Special Issue
The One-Hundred-Year Anniversary of the Discovery of the Sunshine Vitamin D3: Historical, Personal Experience and Evidence-Based Perspectives
Previous Article in Journal
Nutritional Care in Children with Cystic Fibrosis
Previous Article in Special Issue
A Pleiotropic Nuclear Hormone Labelled Hundred Years Ago Vitamin D
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin D and Bone: A Story of Endocrine and Auto/Paracrine Action in Osteoblasts

Department of Internal Medicine, Room Ee585c, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(3), 480; https://doi.org/10.3390/nu15030480
Submission received: 2 December 2022 / Revised: 11 January 2023 / Accepted: 12 January 2023 / Published: 17 January 2023

Abstract

:
Despite its rigid structure, the bone is a dynamic organ, and is highly regulated by endocrine factors. One of the major bone regulatory hormones is vitamin D. Its renal metabolite 1α,25-OH2D3 has both direct and indirect effects on the maintenance of bone structure in health and disease. In this review, we describe the underlying processes that are directed by bone-forming cells, the osteoblasts. During the bone formation process, osteoblasts undergo different stages which play a central role in the signaling pathways that are activated via the vitamin D receptor. Vitamin D is involved in directing the osteoblasts towards proliferation or apoptosis, regulates their differentiation to bone matrix producing cells, and controls the subsequent mineralization of the bone matrix. The stage of differentiation/mineralization in osteoblasts is important for the vitamin D effect on gene transcription and the cellular response, and many genes are uniquely regulated either before or during mineralization. Moreover, osteoblasts contain the complete machinery to metabolize active 1α,25-OH2D3 to ensure a direct local effect. The enzyme 1α-hydroxylase (CYP27B1) that synthesizes the active 1α,25-OH2D3 metabolite is functional in osteoblasts, as well as the enzyme 24-hydroxylase (CYP24A1) that degrades 1α,25-OH2D3. This shows that in the past 100 years of vitamin D research, 1α,25-OH2D3 has evolved from an endocrine regulator into an autocrine/paracrine regulator of osteoblasts and bone formation.

1. Introduction

The skeleton plays a fundamental role in the human body by providing structural support and allowing movement. Moreover, it has a protective role for vital internal organs and stem cells, is a source for mineral and growth factors, and is the center of regulatory pathways. Bone is highly dynamic and undergoes continuous remodeling throughout life; it can repair itself. To illustrate this, damaged or (micro)fractured areas are removed by osteoclastic bone resorption, which is followed by new bone formation by osteoblasts (bone remodeling). Bone formation is characterized by secretion of an extracellular proteinaceous matrix, which is subsequently mineralized. Bone remodeling is tightly controlled by an interplay of local, bone and bone marrow-derived factors (e.g., cytokines, growth factors, chemokines) and endocrine factors. One of these endocrine factors is the seco-steroid 1α,25-dihydroxyvitamin D3 (1α,25-OH2D3). 1α,25-OH2D3 can affect bone in a direct as well as an indirect manner [1,2,3]. The indirect effect occurs via control of calcium reabsorption in the kidney and absorption in the intestine, as well as via control of parathyroid hormone production. Although rickets and osteomalacia were prevented in vitamin D receptor (VDR) knockout mice fed with a rescue diet that contained high levels of calcium and phosphorus, not all bone changes were rescued, indicating the importance of a direct role for 1α,25-OH2D3 in bone metabolism [4,5,6]. The presence of VDRs in cells of the osteoblast lineage [7,8] enables direct effects of 1α,25-OH2D3 on bone metabolism. VDR expression in osteoblasts can be regulated by 1α,25-OH2D3 itself, as well as by other factors including parathyroid hormone, glucocorticoids, transforming growth factor-β, and epidermal growth factor [9,10,11,12,13]. Transgenic mice specifically overexpressing the VDR in osteoblasts have increased trabecular bone volume and increased bone strength, supporting an anabolic effect of 1α,25-OH2D3 [14]. This observation was confirmed in a study using mice with a different genetic background [15]. Interestingly, a study with global VDR knockout mice [5] knockout mice reported a similar phenotype, with increased trabecular thickness and increased osteoid volume and osteoblast numbers, suggesting an inhibitory effect of 1α,25-OH2D3 on bone formation. This was supported by data from an osteoblast-specific VDR knockout mouse study [16]. In this latter study, the bone effect appeared to be via reduced bone resorption. The effects on bone may be related to overall levels of calcium intake [17], but whether this explains the apparent opposite effects in murine studies remains to be established. Nevertheless, these observations support a direct effect of 1α,25-OH2D3 on bone metabolism via osteoblasts. There is less consensus on VDR expression in osteoclasts. Genomic deletion of the VDR in osteoclasts did not impact the positive effect of a 1α,25-OH2D3 analog (eldecalcitol) on bone mass [7]. This is supported by Verlinden et al., who reported that VDRs in osteoclast precursors are not essential to maintain bone homeostasis [18]. It was concluded that 1α,25-OH2D3 regulates osteoclasts indirectly via cells of the osteoblast lineage. In the current review, we will focus on 1α,25-OH2D3 in osteoblast function and bone metabolism.

2. Literature Search Strategy

We built on our pre-existing literature database and expanded this with a new search from 2016 until October 2022. With the support of the Erasmus MC Medical Library Literature Search Service, the search strategy was developed and executed. Supplemental Figure S1 shows in detail the search strings used. In this way, we obtained a list of 2713 publications on vitamin D. From this dataset, we excluded 2583 clinical and (genetic) epidemiological association studies and focused on 128 bone-related molecular and cellular studies. Two publications appeared to be retracted after the search was performed.

3. Osteoblasts

Osteoblasts originate from mesenchymal stromal cells via a tightly controlled differentiation process. The eventual fate of osteoblasts is three-fold, either to become lining cells that cover the bone surface, or to become embedded in the extracellular matrix as osteocytes, or to die via apoptosis.

3.1. Proliferation and Apoptosis

The data on 1α,25-OH2D3 effects on osteoblast proliferation are variable. Inhibition [19,20,21,22,23,24,25,26,27], as well as stimulation [20,28] or no effect [29,30] on the proliferation of osteoblasts of mouse, rat, and human origins have been reported. Effects on cell viability [31] and apoptosis [32,33] have also been documented. Although different directions in effect have been observed, these data demonstrate direct effects of 1α,25-OH2D3 on osteoblast proliferation and survival. The direction of effect may depend on the timing of treatment, dosage, origin, and environment of the osteoblasts [27,34,35,36].

3.2. Differentiation

Immature mesenchymal stromal cells differentiate into osteoblasts that produce extracellular matrix proteins, enzymes, and matrix vesicles involved in the mineralization of the extracellular matrix produced (Figure 1). It has been demonstrated that 1α,25-OH2D3 impacts all of these processes [3,37,38]. 1α,25-OH2D3 stimulation of differentiation has been shown in all in vitro studies using human osteoblasts, human mesenchymal stem cells, and osteogenic-induced pluripotent stem cells [30,39,40,41,42,43,44,45,46]. Most studies with rat osteoblasts resemble these studies using human osteoblasts and show increased differentiation [29,47,48]. Studies with mouse osteoblasts are more diverse. These studies show inhibition [49,50], as well as stimulation of osteoblast differentiation by 1α,25-OH2D3 [51]. The definitive explanation for the discrepancies in 1α,25-OH2D3 effects between, on the one hand, mouse osteoblast cultures, and on the other hand, between mouse and human/rat osteoblast cultures, is absent; however, several explanations can be put forward. The source of osteoblasts may play a role. Different sites of the skeleton differ in origin and bone formation, such as enchondral (long bones) and intramembranous (calvaria) sites. 1α,25-OH2D3 did not affect osteoblasts from cortical bone, and inhibited differentiation of calvaria-derived cells [52,53]. Furthermore, within one skeletal element, differences in osteoblast regulation are observed. A recent study reported differences between periosteal- and bone-marrow-derived osteoblasts in cortical bone [54]. Whether this fully explains the diverse effects observed is not clear, but it shows the importance of origin for the eventual activity and regulation. This may also relate to stage of osteoblast differentiation, donor age, culture conditions, etc., which have been shown to relate to 1α,25-OH2D3 action [17,47,55,56]. Furthermore, differences may be species-intrinsic, and may have a genomic explanation. 1α,25-OH2D3 increases RUNX2 and BGLAP (osteocalcin) gene expressions in human osteoblasts, while in murine osteoblasts, 1α,25-OH2D3 treatment inhibits the gene expressions of RUNX2 and BGLAP [43,57,58,59,60,61].
A picture that emerges from all in vitro osteoblast data is that the osteoblast (micro)environment is a determinant of the eventual outcome of 1α,25-OH2D3 action. The extracellular milieu (growth factors, cytokines, matrix proteins, ions (calcium/phosphate), and other signaling molecules) and the intracellular milieu (e.g., the insulin-like growth factor binding protein-6) are important for the eventual effect of 1α,25-OH2D3 [62,63]. For example, interactions with transforming growth factor-β, insulin-like growth factor, bone morphogenetic proteins, and interferon have been demonstrated [64,65,66,67,68,69]. Consequently, the absence or presence of these, but potentially other factors as well, can modulate 1α,25-OH2D3 action and determine the eventual response. An example of interaction with other intracellular regulatory pathways is Wnt signaling. Wnt signaling plays an important role in osteoblast differentiation and bone formation. An interplay between 1α,25-OH2D3 and Wnt signaling has been described [70,71,72,73,74].
Osteoblast differentiation, bone matrix production, and mineralization, as part of bone formation, are high energy-demanding processes [75,76,77]. Regulation of energy metabolism impacts osteoblast differentiation and bone formation [78,79,80]. Vitamin D and energy metabolism have been discussed in relation to obesity and metabolic syndrome [81] and to cancer [82,83,84], but data on vitamin D and energy metabolism in the context of osteoblast differentiation remain limited. Forkhead Box O (FoxO) transcription factors are regulated by 1α,25-OH2D3 in murine MC3T3 osteoblasts. FoxO3a is upregulated, FoxO1 is downregulated, and FoxO4 is unchanged after 1α,25-OH2D3 treatment. si-RNA knockdown of the FoxOs did not change 1α,25-OH2D3 inhibition of proliferation [85]. Unfortunately, the effect on differentiation was not reported. Changes in FoxO expression were coupled to increase in reactive oxygen species accumulation, which may be linked to cellular metabolism and bone formation [75,80,86]. Glucose, insulin, and 1α,25-OH2D3 regulation of osteoblast proliferation, alkaline phosphatase activity, and production of (uncarboxylated) osteocalcin have been studied in isolated rat osteoblasts, but unfortunately, no coupling to mineralization was made [87]. Nevertheless, these data, together with those on interactions between vitamin D and PPARγ signaling in osteoblast differentiation [88], support that control of energy metabolism can be a vitamin D target in bone formation and mineralization.

3.3. Mineralization

Mineralization can be divided into two phases. In the first phase, formation of hydroxyapatite (HA) crystals takes place in nano-sized extracellular matrix vesicles produced by osteoblasts. In the second phase, HA is propagated outside these vesicles, with a resulting buildup of mineral in the extracellular matrix [89,90]. Calcium and phosphate concentrations increase inside matrix vesicles via involvement of specific proteins, and when the solubility product of calcium and phosphate is exceeded, mineral deposits are formed inside the extracellular vesicles and the second phase of mineralization starts with the release of the preformed HA crystals [90,91]. Proteomic analyses of extracellular matrix vesicles revealed many proteins with a potential role in mineralization [92,93]. Gene profiling studies also identified novel regulators of osteoblast matrix mineralization [94].
Mineralization is controlled by a balanced action of promoters and inhibitors. Alkaline phosphatase and bone sialoprotein are important promoters [95,96]. Alkaline phosphatase increases the phosphate concentration in matrix vesicles by hydrolyzing inorganic pyrophosphate. Pyrophosphate is an inhibitor of mineralization; consequently, alkaline phosphatase also decreases the level of this inhibitor. Pyrophosphatase phosphodiesterase 1 (NPP1, encoded by the gene ENPP1) and ankylosis protein (ANK) are involved in inhibiting mineralization. NPP1 generates pyrophosphate, and the transmembrane protein ANK allows pyrophosphate to pass through the plasma membrane to the extracellular matrix; thus, HA formation is inhibited in the extracellular vesicles [97,98]. 1α,25-OH2D3 stimulates mineralization via direct action on osteoblasts [68,88,99]. 1α,25-OH2D3 can influence the mineralization process via gene expression and matrix vesicle production. Gene expression profiling studies demonstrated that the 1α,25-OH2D3 effect is not likely primarily due to changes in the expression of extracellular matrix genes, and thereby to changes in composition of the extracellular matrix [99]. Studies on the expression and production of procollagen type I by human osteoblasts showed stimulation [100,101] as well as no effect [101,102,103,104], or inhibition [105].
Figure 1. Alizarin red staining of osteoblast culture exemplifying the pre-mineralization and mineralization phases. Red staining shows mineralization. Details on cell culture and Alizarin red staining procedures can be found in Woeckel et al. [99]. Adapted with permission from Eijken, M., Koedam, M., van Driel, M., Buurman, C.J., Pols, H.A.P., van Leeuwen J.P.T.M. The essential role of glucocorticoids for proper human osteoblast differentiation and matrix mineralization. Mol Cell Endocrinol 2006, 248(1–2):87–93. https://doi.org/10.1016/j.mce.2005.11.034. 2006, J.P.T.M. van Leeuwen.
Figure 1. Alizarin red staining of osteoblast culture exemplifying the pre-mineralization and mineralization phases. Red staining shows mineralization. Details on cell culture and Alizarin red staining procedures can be found in Woeckel et al. [99]. Adapted with permission from Eijken, M., Koedam, M., van Driel, M., Buurman, C.J., Pols, H.A.P., van Leeuwen J.P.T.M. The essential role of glucocorticoids for proper human osteoblast differentiation and matrix mineralization. Mol Cell Endocrinol 2006, 248(1–2):87–93. https://doi.org/10.1016/j.mce.2005.11.034. 2006, J.P.T.M. van Leeuwen.
Nutrients 15 00480 g001
It is postulated that vitamin D may enhance mineralization by stimulating both NPP1, generating pyrophosphate, and alkaline phosphatase, generating phosphate from pyrophosphate [106]. This involves acceleration of the production of alkaline phosphatase-positive matrix vesicles, leading to enhanced formation and deposition of HA crystals, and eventually mineralization. This direct effect of vitamin D occurred in the period prior to the onset of mineralization, and also involved accelerated extracellular matrix maturation [99]. Interestingly, treatment with vitamin D after initiation of mineralization did not affect mineralization. This supports the above-described osteoblast differentiation stage dependency of the 1α,25-OH2D3 effect. A study by Yajima et al. described the significance of 1α,25-OH2D3 for osteocytic perilacunar mineralization [107].
1α,25-OH2D3 also directly stimulates the production of inhibitors of mineralization. VDR-dependent 1α,25-OH2D3 expression of ENPP1 and ANK in murine osteoblasts led to an increase in the mineralization inhibitor pyrophosphate [108]. 1α,25-OH2D3 also stimulates activin A expression in human osteoblasts. Treatment with the activin A blocker follistatin enhanced vitamin-D-induced mineralization of human osteoblasts [109]. 1α,25-OH2D3 also increases the expression of osteopontin, which is shown to inhibit mineralization. These observations may provide a fine-tuning mechanism to prevent excessive mineralization of bone. 1α,25-OH2D3 induction of carboxylated osteocalcin may be in line with this. 1α,25-OH2D3-stimulated mineralization is enhanced by blocking osteocalcin carboxylation by warfarin [109]. The interaction of 1α,25-OH2D3 with other factors, as described above, also holds for mineralization, for example, the interaction with DKK1, the inhibitor of Wnt signaling [74].
The counterbalance of bone formation and mineralization by osteoblasts is bone resorption by osteoclasts. In the healthy skeleton, these processes are in balance, securing healthy and strong bones. The osteoblasts/osteocytes are the major regulators of osteoclast formation and action via production of the stimulating factor RANKL, and the RANKL inhibitor, osteoprotegerin (OPG). 1α,25-OH2D3 influences the RANKL/OPG ratio, and thereby also impacts bone resorption [110,111,112,113]. 1α,25-OH2D3 is involved at both the bone formation and the bone resorption sides of the balance, and is an important player in maintaining healthy bones via direct effects on bone, in addition to indirect effects via calcium and phosphate homeostasis [114].

3.4. Gene Expression

The basis of all cellular effects of 1α,25-OH2D3 involves VDR-mediated transcriptional regulation. The VDR is a member of the nuclear receptor family. Upon binding to 1α,25-OH2D3, the VDR heterodimerizes with the retinoic X receptor (RXR), and binds as a dimer to the vitamin D response element (VDRE) in the DNA to regulate gene expression [115]. Over the years, many studies have unraveled the molecular fundamentals of 1α,25-OH2D3 transcriptional regulation. Examples and information can be found in these publications and references therein [116,117,118]. In a previous publication, we discussed 1α,25-OH2D3 and gene transcription in osteoblasts [38]. This will not be repeated or discussed in detail in this review.
A factor that may determine the transcriptional effect of 1α,25-OH2D3 effect is not only the basal level of gene expression [51,119], but also the stage of osteoblast differentiation [99]. Studies with rat osteoblasts in the early 1990s showed already that effects of 1α,25-OH2D3 on osteoblasts may depend on the osteoblast differentiation phase [119]. An example is the 1α,25-OH2D3 stimulation of phosphaturic hormone fibroblast growth factor 23 (FGF23) only in late-stage differentiation osteoblasts and osteocytes [120,121]. FGF23 is a hormone that acts in the kidney to enhance phosphate excretion, and suppresses 1α,25-OH2D3 synthesis by inhibiting 1α-hydroxylase (CYP27B1), forming an important loop in the regulation of mineralization [122,123]. Vitamin D signaling in osteocytes [124] is further supported by the 1α,25-OH2D3 regulation of PHEX (phosphate-regulating neutral endopeptidase, X linked), which suppresses FGF23 transcription [125].
The various osteoblast differentiation stages actually reflect different functional stages of the osteoblast, e.g., proliferation, extracellular matrix production, mineralizing and mechanosensing. It is important to keep in mind the osteoblast differentiation stage when studying 1α,25-OH2D3 effects, as this may be an important determinant of the eventual effect (e.g., stimulation or inhibition) on gene transcription and subsequent cellular responses and bone formation. The relationship between the osteoblast differentiation stage and 1α,25-OH2D3 gene expression control was shown by Woeckel et al. [99]. 1α,25-OH2D3 changed the expression of different sets of genes in the phase before the onset of mineralization, and during the mineralization. For this review, we performed a reanalysis of this gene profiling study [99] with the 2022 updated annotation. Comparison of transcripts regulated (i.e., two-fold up or down) in the phase before and after the start of mineralization (Figure 1) demonstrated that only 2.5% (18 out of the 721 regulated transcripts) were regulated in both phases (Table 1). The gene symbols of the transcripts regulated in both phases are shown in Table 2. To focus in more detail on phase-specific gene expression, we next selected the transcripts that were uniquely regulated in either the pre-mineralization or in the mineralization phase [99]. In this regard, the transcripts should be at least two-fold up- or downregulated in one phase (either pre-mineralization or mineralization phase), and not regulated (fold change on average between 0.8 and 1.2) in the other phase (either the mineralization or pre-mineralization phase). Table 3 shows the number of transcripts uniquely regulated in either of these phases, and Table 4 reports the gene symbols belonging to these transcripts. This binary comparison of pre-mineralization and mineralization phases is not absolute and does not mean that further zooming in on specific phases of osteoblast differentiation will not reveal other sets of vitamin-D-regulated genes. However, it does support the notion that vitamin D gene regulation during osteoblast differentiation and mineralization displays temporal dynamics, and it does show that for proper interpretation of vitamin D effects, knowledge on the differentiation and functional stage of cells and tissues is important. This knowledge can explain the apparent differences in 1α,25-OH2D3 effects that have been reported.

4. Vitamin D Metabolism

Metabolism, synthesis of the active form of 1α,25-OH2D3 as well as its inactivation, has been an important research topic since the identification of vitamin D. This has contributed to the understanding of the initiation and termination actions of vitamin D and its endocrine function. Figure 2 shows the classical vitamin D metabolism pathway. Serum levels of 1α,25-OH2D3 are determined by the activity of the renal enzyme 1α-hydroxylase (CYP27B1). 24-Hydroxylase (CYP24A1) is the first step of a 1α,25-OH2D3 inactivation cascade present in all target tissues. In the next sections, we discuss CYP27B1 and CYP24A1 in osteoblasts.

4.1. CYP27B1

In the late 1970s and early 1980s, reports were already coming out that in tissues other than the kidney, 1α,25-OH2D3 can be synthesized. Cells isolated from chicken calvaria [126] and human osteosarcoma cells, as well as bone cells isolated from an ileac crest biopsy [127], can produce 1α,25-OH2D3.. Its functional significance in human osteoblasts was shown by the fact that inhibition of 1α-hydroxylase activity by ketoconazole blocked the 25(OH)D3 induction of CYP24A1 and osteocalcin expression [30]. This was supported by studies on siRNA silencing in human osteoblasts [19,46]. Additional evidence came from a study showing the importance of CYP27B1 for proliferation and osteogenic differentiation of human mesenchymal stromal cells (MSCs) [128,129]. MSCs of older donors had reduced CYP27B1 expression and resistance to 25(OH)D3 regulation of osteoblast differentiation [130]. Broader tissue distribution of extra renal CYP27B1 expression beyond bone was recently summarized by Bikle et al. [131].
However, renal synthesis is still considered the major contributor to circulating 1α,25-OH2D3 levels. Only in diseases such as sarcoidosis extra is renal synthesis sufficient to contribute to circulating levels. The presence of 1α,25-OH2D3 synthesis within bone provides a means to explain the associations of bone phenotypes and other parameters with circulating 25(OH)D3 and not with 1α,25-OH2D3, as discussed by Anderson and colleagues [132,133]. Pharmacokinetic differences between locally produced 1α,25-OH2D3 from 25(OH)D3 and added 1α,25-OH2D3 have been suggested from a cell culture study [134]. Further studies, in particular, in vivo studies, are needed for full appreciation of the impact of an autocrine/paracrine role of 1α,25-OH2D3.
Observations that the vitamin-D-binding protein receptors cubulin and megalin, as well as the vitamin D3 25-hydroxylase genes CYP2R1 and CYP3A4, are also expressed in human osteoblasts, supports an autocrine/paracrine role [19,30,131].
Renal CYP27B1 is tightly controlled by factors such as parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23), which are involved in calcium and phosphate homeostasis. Extrarenal CYP27B1 expression is differently regulated, and probably involves other factors and tissue specificity [135]. For example, PTH and ambient calcium do not regulate CYP27B1 in human osteoblasts [30], while 1α,25-OH2D3 reduces CYP27B1 expression in human MSCs similar as in the kidney [136]. Several growth factors and cytokines can regulate CYP27B1 expression. IGF-I increases CYP27B1 expression in human MSCs [136]. Interleukin-1 stimulates while interferon-β reduces CYP27B1 expression in human osteoblasts [30,69]. The earlier described impact of the osteoblast differentiation stage on 1α,25-OH2D3 action can also be translated to expression of CYP27B1. CYP27B1 expression is increased by 25(OH)D3 in human MSCs [136], but not in mature osteoblasts [30].

4.2. CYP24A1

The first step in the degradation cascade of 1α,25-OH2D3 is hydroxylation at the C-24 position by 24-hydroxylase (CYP24A1) [137]. CYP24A1 is expressed in all vitamin D target cells, and its expression is very rapidly and strongly increased after 1α,25-OH2D3 binding to VDRs [138,139,140,141]. The VDR level is tightly linked to the induction of CYP24A1 expression and 24-hydroxylase activity and, consequently, degradation of 1α,25-OH2D3. Thus, the homologous upregulation of VDRs concomitantly induces the inactivation of 1α,25-OH2D3, and thereby limits its effect [142,143]. Hydroxylation at the C-24 position of 1α,25-OH2D3 or 25(OH)D3 alone does not immediately lead to an inactive vitamin D molecule. Henry and Norman demonstrated in the 1970s the functional significance of 24,25(OH)2D3 for normal chicken egg hatchability and calcium and phosphorus homeostasis [144,145]. The effects of 24,25(OH)2D3 on bone metabolism were shown in human, chicken, rat, and mouse studies. 24,25(OH)2D3, synergistically with PTH, directly stimulates mineralization, and 24,25(OH)2D3 decreases the number and size of resorption sites on the bone surface [146,147]. 24,25(OH)2D3 restores and accelerates the bone mineral apposition rate in vitamin-D-deficient and in parathryoidectomized rats [147]. 24,25(OH)2D3 did not change bone histomorphometric parameters in ovariectomized rats [148], but 24,25(OH)2D3, and not 1α,25-OH2D3, increased bone strength [149].
Several studies focused on 24-hydroxylated vitamin D molecules and fracture healing. 24,25(OH)2D3 binds to fracture calluses [150], and improves fracture healing [151,152,153]. Serum 24,25(OH)2D3 levels were found to correlate with fracture healing in chicken [151], but not in a small human study in 1978 [154]. However, a study on pre-dialysis renal insufficiency patients supported a direct, i.e., PTH-independent, functional role of 24,25(OH)2D3 in human bone. 24,25(OH)2D3, together with 1α,25-(OH)2D3, preserved the osteoblast perimeter and improved mineralization, while 1α,25-(OH)2D3 alone was ineffective [155]. A direct effect on bone, in particular osteoblasts, is supported by in vitro studies showing that, similarly to 1α,25-OH2D3, 24,25(OH)2D3 has direct effects on human osteoblast differentiation [45]. Knowing that 24-hydroxylation per se does not lead to inactivation of vitamin D molecules, it is important to understand target tissue/target cell dynamics of the next steps in the degradation cascade. Control of the velocity of the subsequent steps in the degradation pathway can be a means to regulate vitamin D action in target tissues/cells. Together, these data on CYP24A1 and the biological activities of 24,25(OH)2D3 add to the notion of an auto/paracrine vitamin D regulatory system in bone. This system is most likely not restricted to bone and may also be present in other tissues.

5. Conclusions

This review revealed that the central role for vitamin D in bone physiology is directed via osteoblasts and depends on their stage of development. VDRs and the vitamin-D-metabolizing enzymes CYP27B1 and CYP24A1, known from the vitamin D endocrine system, are present and functional in osteoblasts. This uncovers a direct local role for 1α,25-OH2D3 vitamin D in osteoblast function, and expands the vitamin D action profile from endocrine regulation of calcium and phosphate homeostasis to an auto/paracrine regulatory network in bone. Several target-tissue-derived factors (growth factors, cytokines), intracellular signaling cascades (Wnt), and functional states of the osteoblast interact with this auto/paracrine network and determine the eventual response. In this way, vitamin D controls the proliferation, apoptosis, differentiation, and mineralization of osteoblasts, as well as their gene profile and interaction with other factors that maintain healthy bone. Moreover, even local degradation products of vitamin D metabolism (24,25(OH)2D3) have a beneficial contribution to osteoblast function. Together, these observations underscore the importance of contextual knowledge (molecular and cellular) in order to fully understand and appreciate the effects of vitamin D on bone cells.
This warrants research for the next 100 years: future studies may focus on assessing tissue levels of vitamin D metabolites in addition to circulating levels, and study functionality of the complete metabolic profile of vitamin D.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/nu15030480/s1, Figure S1: Literature search strategy performed October 2022.

Author Contributions

M.v.D.: writing—review and editing; J.P.T.M.v.L.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Jeroen van de Peppel from the Erasmus MC Department of Internal Medicine for support with reanalysis of gene expression data, and we are very grateful to Wichor Bramer from the Erasmus MC Medical Library for his support in developing and updating the literature search strategies.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Datta, H.K.; Ng, W.-F.; Walker, J.A.; Tuck, S.P.; Varanasi, S.S. The cell biology of bone metabolism. J. Clin. Pathol. 2008, 61, 577–587. [Google Scholar] [CrossRef] [PubMed]
  2. Bikle, D.D. Vitamin D and Bone. Curr. Osteoporos. Rep. 2012, 10, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Christakos, S.; Li, S.; DeLa Cruz, J.; Verlinden, L.; Carmeliet, G. Vitamin D and Bone. Handb. Exp. Pharmacol. 2020, 262, 47–63. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. Amling, M.; Priemel, M.; Holzmann, T.; Chapin, K.; Rueger, J.M.; Baron, R.; DeMay, M.B. Rescue of the Skeletal Phenotype of Vitamin D Receptor-Ablated Mice in the Setting of Normal Mineral Ion Homeostasis: Formal Histomorphometric and Biomechanical Analyses. Endocrinology 1999, 140, 4982–4987. [Google Scholar] [CrossRef]
  6. Panda, D.K.; Miao, D.; Bolivar, I.; Li, J.; Huo, R.; Hendy, G.N.; Goltzman, D. Inactivation of the 25-Hydroxyvitamin D 1α-Hydroxylase and Vitamin D Receptor Demonstrates Independent and Interdependent Effects of Calcium and Vitamin D on Skeletal and Mineral Homeostasis. J. Biol. Chem. 2004, 279, 16754–16766. [Google Scholar] [CrossRef] [Green Version]
  7. Nakamichi, Y.; Udagawa, N.; Horibe, K.; Mizoguchi, T.; Yamamoto, Y.; Nakamura, T.; Hosoya, A.; Kato, S.; Suda, T.; Takahashi, N. VDR in Osteoblast-Lineage Cells Primarily Mediates Vitamin D Treatment-Induced Increase in Bone Mass by Suppressing Bone Resorption. J. Bone Miner. Res. 2017, 32, 1297–1308. [Google Scholar] [CrossRef]
  8. Zarei, A.; Morovat, A.; Javaid, K.; Brown, C.P. Vitamin D receptor expression in human bone tissue and dose-dependent activation in resorbing osteoclasts. Bone Res. 2016, 4, 16030. [Google Scholar] [CrossRef]
  9. Pols, H.A.P.A.; van Leeuwen, J.P.T.M.P.; Schilte, J.P.P.; Visser, T.J.J.; Birkenhäger, J.C.C. Heterologous up-regulation of the 1,25-dihydroxyvitamin D3 receptor by parathyroid hormone (PTH) and PTH-like peptide in osteoblast-like cells. Biochem. Biophys. Res. Commun. 1988, 156, 588–594. [Google Scholar] [CrossRef]
  10. Van Leeuwen, J.P.; Birkenhäger, J.C.; Buurman, C.J.; Bemd, G.J.V.D.; Bos, M.P.; A Pols, H. Bidirectional regulation of the 1,25-dihydroxyvitamin D3 receptor by phorbol ester-activated protein kinase-C in osteoblast-like cells: Interaction with adenosine 3′,5′-monophosphate-induced up-regulation of the 1,25-dihydroxyvitamin D3 receptor. Endocrinology 1992, 130, 2259–2266. [Google Scholar] [CrossRef]
  11. van Leeuwen, J.P.; Pols, H.A.; Schilte, J.P.; Visser, T.J.; Birkenhäger, J.C. Modulation by epidermal growth factor of the basal 1,25(OH)2D3 receptor level and the heterologous up-regulation of the 1,25(OH)2D3 receptor in clonal osteoblast-like cells. Calcif. Tissue Int. 1991, 49, 35–42. [Google Scholar] [CrossRef] [Green Version]
  12. Godschalk, M.; Levy, J.R.; Downs, R.W., Jr. Glucocorticoids decrease vitamin D receptor number and gene expression in human osteosarcoma cells. J. Bone Miner. Res. 1992, 7, 21–27. [Google Scholar] [CrossRef]
  13. Reinhardt, T.A.; Horst, R.L. Parathyroid Hormone Down-Regulates 1,25-Dihydroxyvitamin D Receptors (VDR) and VDR Messenger Ribonucleic Acid in Vitro and Blocks Homologous Up-Regulation of VDR in Vivo. Endocrinology 1990, 127, 942–948. [Google Scholar] [CrossRef]
  14. Gardiner, E.M.; Baldock, P.A.; Thomas, G.P.; Sims, N.A.; Henderson, N.K.; Hollis, B.; White, C.P.; Sunn, K.L.; Morrison, N.A.; Walsh, W.R.; et al. Increased formation and decreased resorption of bone in mice with elevated vitamin D receptor in mature cells of the osteoblastic lineage. FASEB J. 2000, 14, 1908–1916. [Google Scholar] [CrossRef] [Green Version]
  15. Triliana, R.; Lam, N.N.; Sawyer, R.K.; Atkins, G.J.; Morris, H.A.; Anderson, P.H. Skeletal characterization of an osteoblast-specific vitamin D receptor transgenic (ObVDR-B6) mouse model. J. Steroid Biochem. Mol. Biol. 2016, 164, 331–336. [Google Scholar] [CrossRef]
  16. Yamamoto, Y.; Yoshizawa, T.; Fukuda, T.; Shirode-Fukuda, Y.; Yu, T.; Sekine, K.; Sato, T.; Kawano, H.; Aihara, K.-I.; 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] [Green Version]
  17. Lieben, L.; Carmeliet, G. The delicate balance between vitamin D, calcium and bone homeostasis: Lessons learned from intestinal- and osteocyte-specific VDR null mice. J. Steroid Biochem. Mol. Biol. 2013, 136, 102–106. [Google Scholar] [CrossRef]
  18. Verlinden, L.; Janssens, I.; Doms, S.; Vanhevel, J.; Carmeliet, G.; Verstuyf, A. Vdr expression in osteoclast precursors is not critical in bone homeostasis. J. Steroid Biochem. Mol. Biol. 2019, 195, 105478. [Google Scholar] [CrossRef]
  19. Atkins, G.J.; Anderson, P.; Findlay, D.M.; Welldon, K.J.; Vincent, C.; Zannettino, A.; O’Loughlin, P.D.; Morris, H.A. Metabolism of vitamin D3 in human osteoblasts: Evidence for autocrine and paracrine activities of 1α,25-dihydroxyvitamin D3. Bone 2007, 40, 1517–1528. [Google Scholar] [CrossRef]
  20. van den Bemd, G.J.; Pols, H.A.; Birkenhäger, J.C.; Kleinekoort, W.M.; van Leeuwen, J.P. Differential effects of 1,25-dihydroxyvitamin D3-analogs on osteoblast-like cells and on in vitro bone resorption. J. Steroid Biochem. Mol. Biol. 1995, 55, 337–346. [Google Scholar] [CrossRef]
  21. Döhla, J.; Kuuluvainen, E.; Gebert, N.; Amaral, A.; Englund, J.I.; Gopalakrishnan, S.; Konovalova, S.; Nieminen, A.I.; Salminen, E.S.; Torregrosa Muñumer, R.; et al. Metabolic determination of cell fate through selective inheritance of mitochondria. Nat. Cell Biol. 2022, 24, 148–154. [Google Scholar] [CrossRef] [PubMed]
  22. Eelen, G.; Verlinden, L.; Van Camp, M.; Mathieu, C.; Carmeliet, G.; Bouillon, R.; Verstuyf, A. Microarray analysis of 1α,25-dihydroxyvitamin D3-treated MC3T3-E1 cells. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 405–407. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, T.L.; Cone, C.M.; Feldman, D. Effects of 1α,25-dihydroxyvitamin D3 and glucocorticoids on the growth of rat and mouse osteoblast-like bone cells. Calcif. Tissue Int. 1983, 35, 806–811. [Google Scholar] [CrossRef] [PubMed]
  24. Murray, S.S.; A Glackin, C.; Murray, E.J.B. Variation in 1,25-dihydroxyvitamin D3 regulation of proliferation and alkaline phosphatase activity in late-passage rat osteoblastic cell lines. J. Steroid Biochem. Mol. Biol. 1993, 46, 227–233. [Google Scholar] [CrossRef] [PubMed]
  25. Kanatani, M.; Sugimoto, T.; Fukase, M.; Chihara, K. Effect of 1.25-Dihydroxyvitamin D3 on the Proliferation of Osteoblastic MC3T3-E1 Cells by Modulating the Release of Local Regulators from Monocytes. Biochem. Biophys. Res. Commun. 1993, 190, 529–535. [Google Scholar] [CrossRef]
  26. Rubin, J.; Fan, X.; Thornton, D.; Bryant, R.; Biskobing, D. Regulation of Murine Osteoblast Macrophage Colony-Stimulating Factor Production by 1,25(OH)2D3. Calcif. Tissue Int. 1996, 59, 291–296. [Google Scholar] [CrossRef]
  27. Maehata, Y.; Takamizawa, S.; Ozawa, S.; Kato, Y.; Sato, S.; Kubota, E.; Hata, R.-I. Both direct and collagen-mediated signals are required for active vitamin D3-elicited differentiation of human osteoblastic cells: Roles of osterix, an osteoblast-related transcription factor. Matrix Biol. 2006, 25, 47–58. [Google Scholar] [CrossRef]
  28. Ishida, H.; Bellows, C.G.; E Aubin, J.; Heersche, J.N. Characterization of the 1,25-(OH)2D3-induced inhibition of bone nodule formation in long-term cultures of fetal rat calvaria cells. Endocrinology 1993, 132, 61–66. [Google Scholar] [CrossRef]
  29. Wang, D.; Song, J.; Ma, H. An in vitro Experimental Insight into the Osteoblast Responses to Vitamin D3 and Its Metabolites. Pharmacology 2018, 101, 225–235. [Google Scholar] [CrossRef]
  30. van Driel, M.; Koedam, M.; Buurman, C.J.J.; Hewison, M.; Chiba, H.; Uitterlinden, A.G.; Pols, H.A.P.; Van Leeuwen, J.P.T.M. Evidence for auto/paracrine actions of vitamin D in bone: 1a-hydroxylase expression and activity in human bone cells. FASEB J. 2006, 20, 2417–2419. [Google Scholar] [CrossRef]
  31. Shi, Y.-C.; Worton, L.; Esteban, L.; Baldock, P.; Fong, C.; Eisman, J.A.; Gardiner, E.M. Effects of continuous activation of vitamin D and Wnt response pathways on osteoblastic proliferation and differentiation. Bone 2007, 41, 87–96. [Google Scholar] [CrossRef]
  32. Hansen, C.M.; Hansen, D.; Holm, P.K.; Binderup, L. Vitamin D compounds exert anti-apoptotic effects in human osteosarcoma cells in vitro. J. Steroid Biochem. Mol. Biol. 2001, 77, 1–11. [Google Scholar] [CrossRef]
  33. Thompson, L.; Wang, S.; Tawfik, O.; Templeton, K.; Tancabelic, J.; Pinson, D.; Anderson, H.C.; Keighley, J.; Garimella, R. Effect of 25-hydroxyvitamin D3 and 1 α,25 dihydroxyvitamin D3 on differentiation and apoptosis of human osteosarcoma cell lines. J. Orthop. Res. 2011, 30, 831–844. [Google Scholar] [CrossRef]
  34. Van Leeuwen, J.P.T.M.; van Driel, M.; van den Bemd, G.J.C.M.; Pols, H.A.P. Vitamin D Control of Osteoblast Function and Bone Extracellular Matrix Mineralization. Crit. Rev. Eukaryot. Gene Expr. 2001, 11, 199–226. [Google Scholar] [CrossRef]
  35. Skjødt, H.; Gallagher, J.A.; Beresford, J.N.; Couch, M.; Poser, J.W.; Russell, R.G.G. Vitamin D metabolites regulate osteocalcin synthesis and proliferation of human bone cells in vitro. J. Endocrinol. 1985, 105, 391–396. [Google Scholar] [CrossRef]
  36. Uranoa, T.; Hosoib, T.; Shirakic, M.; Toyoshimad, H.; Ouchia, Y.; Inoue, S. Possible Involvement of the p57Kip2 Gene in Bone Metabolism. Biochem. Biophys. Res. Commun. 2000, 269, 422–426. [Google Scholar] [CrossRef]
  37. van Driel, M.; van Leeuwen, J.P. Vitamin D endocrinology of bone mineralization. Mol. Cell. Endocrinol. 2017, 453, 46–51. [Google Scholar] [CrossRef]
  38. van de Peppel, J.; van Leeuwen, J.P.T.M. Vitamin D and gene networks in human osteoblasts. Front. Physiol. 2014, 5, 137. [Google Scholar] [CrossRef] [Green Version]
  39. Zhou, S.; Glowacki, J.; Kim, S.W.; Hahne, J.; Geng, S.; Mueller, S.M.; Shen, L.; Bleiberg, I.; LeBoff, M.S. Clinical characteristics influence in vitro action of 1,25-dihydroxyvitamin D3in human marrow stromal cells. J. Bone Miner. Res. 2012, 27, 1992–2000. [Google Scholar] [CrossRef] [Green Version]
  40. Geng, S.; Zhou, S.; Bi, Z.; Glowacki, J. Vitamin D metabolism in human bone marrow stromal (mesenchymal stem) cells. Metabolism 2013, 62, 768–777. [Google Scholar] [CrossRef]
  41. Piek, E.; Sleumer, L.S.; van Someren, E.P.; Heuver, L.; de Haan, J.R.; de Grijs, I.; Gilissen, C.; Hendriks, J.M.; van Ravestein-van Os, R.I.; Bauerschmidt, S.; et al. Osteo-transcriptomics of human mesenchymal stem cells: Accelerated gene expression and osteoblast differentiation induced by vitamin D reveals c-MYC as an enhancer of BMP2-induced osteogenesis. Bone 2010, 46, 613–627. [Google Scholar] [CrossRef] [PubMed]
  42. Kato, H.; Ochiai-Shino, H.; Onodera, S.; Saito, A.; Shibahara, T.; Azuma, T. Promoting effect of 1,25(OH)2 vitamin D3 in osteogenic differentiation from induced pluripotent stem cells to osteocyte-like cells. Open Biol. 2015, 5, 140201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Prince, M.; Banerjee, C.; Javed, A.; Green, J.; Lian, J.B.; Stein, G.S.; Bodine, P.V.; Komm, B.S. Expression and regulation of Runx2/Cbfa1 and osteoblast phenotypic markers during the growth and differentiation of human osteoblasts. J. Cell. Biochem. 2001, 80, 424–440. [Google Scholar] [CrossRef] [PubMed]
  44. Jorgensen, N.R.; Henriksen, Z.; Sorensen, O.H.; Civitelli, R. Dexamethasone, BMP-2, and 1,25-dihydroxyvitamin D enhance a more differentiated osteoblast phenotype: Validation of an in vitro model for human bone marrow-derived primary osteoblasts. Steroids 2004, 69, 219–226. [Google Scholar] [CrossRef] [PubMed]
  45. van Driel, M.; Koedam, M.; Buurman, C.J.J.; Roelse, M.; Weyts, F.; Chiba, H.; Uitterlinden, A.G.G.; Pols, H.A.P.; Van Leeuwen, J.P.T.M. Evidence that both 1α,25-dihydroxyvitamin D3 and 24-hydroxylated D3 enhance human osteoblast differentiation and mineralization. J. Cell. Biochem. 2006, 99, 922–935. [Google Scholar] [CrossRef]
  46. van der Meijden, K.; Lips, P.; van Driel, M.; Heijboer, A.C.; Schulten, E.A.J.M.; Heijer, M.D.; Bravenboer, N. Primary Human Osteoblasts in Response to 25-Hydroxyvitamin D3, 1,25-Dihydroxyvitamin D3 and 24R,25-Dihydroxyvitamin D3. PLoS ONE 2014, 9, e110283. [Google Scholar] [CrossRef] [Green Version]
  47. van Driel, M.; van Leeuwen, J.P.T.M. Vitamin D endocrine system and osteoblasts. BoneKEy Rep. 2014, 3, 493. [Google Scholar] [CrossRef] [Green Version]
  48. Siggelkow, H.; Rebenstorff, K.; Kurre, W.; Niedhart, C.; Engel, I.; Schulz, H.; Atkinson, M.J.; Hüfner, M. Development of the osteoblast phenotype in primary human osteoblasts in culture: Comparison with rat calvarial cells in osteoblast differentiation. J. Cell. Biochem. 1999, 75, 22–35. [Google Scholar] [CrossRef]
  49. Weitzmann, M.N.; Yamaguchi, M. High dose 1,25(OH)2D3 inhibits osteoblast mineralization in vitro. Int. J. Mol. Med. 2012, 29, 934–938. [Google Scholar] [CrossRef] [Green Version]
  50. Chen, Y.-C.; Ninomiya, T.; Hosoya, A.; Hiraga, T.; Miyazawa, H.; Nakamura, H. 1α,25-Dihydroxyvitamin D3 inhibits osteoblastic differentiation of mouse periodontal fibroblasts. Arch. Oral Biol. 2012, 57, 453–459. [Google Scholar] [CrossRef]
  51. Matsumoto, T.; Igarashi, C.; Takeuchi, Y.; Harada, S.; Kikuchi, T.; Yamato, H.; Ogata, E. Stimulation by 1,25-Dihydroxyvitamin D3 of in vitro mineralization induced by osteoblast-like MC3T3-E1 cells. Bone 1991, 12, 27–32. [Google Scholar] [CrossRef]
  52. Yang, D.; Atkins, G.J.; Turner, A.G.; Anderson, P.H.; Morris, H.A. Differential effects of 1,25-dihydroxyvitamin D on mineralisation and differentiation in two different types of osteoblast-like cultures. J. Steroid Biochem. Mol. Biol. 2012, 136, 166–170. [Google Scholar] [CrossRef]
  53. Shevde, N.K.; Plum, L.A.; Clagett-Dame, M.; Yamamoto, H.; Pike, J.W.; DeLuca, H.F. A potent analog of 1α,25-dihydroxyvitamin D3 selectively induces bone formation. Proc. Natl. Acad. Sci. USA 2002, 99, 13487–13491. [Google Scholar] [CrossRef] [Green Version]
  54. Jeffery, E.C.; Mann, T.L.; Pool, J.A.; Zhao, Z.; Morrison, S.J. Bone marrow and periosteal skeletal stem/progenitor cells make distinct contributions to bone maintenance and repair. Cell Stem Cell 2022, 29, 1547–1561.e6. [Google Scholar] [CrossRef]
  55. Eisman, J.A.; Bouillon, R. Vitamin D: Direct effects of vitamin D metabolites on bone: Lessons from genetically modified mice. BoneKEy Rep. 2014, 3, 499. [Google Scholar] [CrossRef] [Green Version]
  56. Yan, X.-Z.; Yang, W.; Yang, F.; Kersten-Niessen, M.; Jansen, J.A.; Both, S.K. Effects of Continuous Passaging on Mineralization of MC3T3-E1 Cells with Improved Osteogenic Culture Protocol. Tissue Eng. Part C Methods 2014, 20, 198–204. [Google Scholar] [CrossRef] [Green Version]
  57. Viereck, V.; Siggelkow, H.; Tauber, S.; Raddatz, D.; Schutze, N.; Hüfner, M. Differential regulation of Cbfa1/Runx2 and osteocalcin gene expression by vitamin-D3, dexamethasone, and local growth factors in primary human osteoblasts. J. Cell. Biochem. 2002, 86, 348–356. [Google Scholar] [CrossRef]
  58. Zhang, R.; Ducy, P.; Karsenty, G. 1,25-Dihydroxyvitamin D3 Inhibits Osteocalcin Expression in Mouse through an Indirect Mechanism. J. Biol. Chem. 1997, 272, 110–116. [Google Scholar] [CrossRef] [Green Version]
  59. Lian, J.B.; Shalhoub, V.; Aslam, F.; Frenkel, B.; Green, J.; Hamrah, M.; Stein, G.S.; Stein, J.L. Species-Specific Glucocorticoid and 1,25-Dihydroxyvitamin D Responsiveness in Mouse MC3T3-E1 Osteoblasts: Dexamethasone Inhibits Osteoblast Differentiation and Vitamin D Down-Regulates Osteocalcin Gene Expression. Endocrinology 1997, 138, 2117–2127. [Google Scholar] [CrossRef]
  60. Drissi, H.; Pouliot, A.; Koolloos, C.; Stein, J.L.; Lian, J.B.; Stein, G.S.; van Wijnen, A.J. 1,25-(OH)2-Vitamin D3 Suppresses the Bone-Related Runx2/Cbfa1 Gene Promoter. Exp. Cell Res. 2002, 274, 323–333. [Google Scholar] [CrossRef]
  61. Thomas, G.P.; Bourne, A.; Eisman, J.A.; Gardiner, E.M. Species-Divergent Regulation of Human and Mouse Osteocalcin Genes by Calciotropic Hormones. Exp. Cell Res. 2000, 258, 395–402. [Google Scholar] [CrossRef] [PubMed]
  62. Cui, J.; Ma, C.; Qiu, J.; Ma, X.; Wang, X.; Chen, H.; Huang, B. A novel interaction between insulin-like growth factor binding protein-6 and the vitamin D receptor inhibits the role of vitamin D3 in osteoblast differentiation. Mol. Cell. Endocrinol. 2011, 338, 84–92. [Google Scholar] [CrossRef] [PubMed]
  63. Ito, N.; Findlay, D.M.; Anderson, P.H.; Bonewald, L.F.; Atkins, G.J. Extracellular phosphate modulates the effect of 1α,25-dihydroxy vitamin D3 (1,25D) on osteocyte like cells. J. Steroid Biochem. Mol. Biol. 2013, 136, 183–186. [Google Scholar] [CrossRef] [PubMed]
  64. Staal, A.; Birkenhäger, J.C.; Pols, H.A.; Buurman, C.J.; Vink-van Wijngaarden, T.; Kleinekoort, W.M.; van den Bemd, G.J.; van Leeuwen, J.P. Transforming growth factor beta-induced dissociation between vitamin D receptor level and 1,25-dihydroxyvitamin D3 action in osteoblast-like cells. Bone Miner. 1994, 26, 27–42. [Google Scholar] [CrossRef] [PubMed]
  65. Staal, A.; Geertsma-Kleinekoort, W.M.C.; Van Den Bemd, G.J.C.M.; Buurman, C.J.; Birkenhäger, J.C.; Pols, H.A.P.; Van Leeuwen, J.P.T.M. Regulation of Osteocalcin Production and Bone Resorption by 1,25-Dihydroxyvitamin D3 in Mouse Long Bones: Interaction with the Bone-Derived Growth Factors TGF-β and IGF-I. J. Bone Miner. Res. 1998, 13, 36–43. [Google Scholar] [CrossRef] [PubMed]
  66. Staal, A.; VanWijnen, A.; Desai, R.; Pols, H.; Birkenhager, J.; Deluca, H.; Denhardt, D.; Stein, J.; Van Leeuwen, J.; Stein, G.; et al. Antagonistic effects of transforming growth factor-beta on vitamin D3 enhancement of osteocalcin and osteopontin transcription: Reduced interactions of vitamin D receptor/retinoid X receptor complexes with vitamin E response elements. Endocrinology 1996, 137, 2001–2011. [Google Scholar] [CrossRef] [Green Version]
  67. Wergedal, J.E.; Matsuyama, T.; Strong, D.D. Differentiation of normal human bone cells by transforming growth factor-β and 1,25(OH)2 vitamin D3. Metabolism 1992, 41, 42–48. [Google Scholar] [CrossRef]
  68. Chen, J.; Dosier, C.R.; Park, J.H.; De, S.; Guldberg, R.E.; Boyan, B.D.; Schwartz, Z. Mineralization of three-dimensional osteoblast cultures is enhanced by the interaction of 1α,25-dihydroxyvitamin D3 and BMP2 via two specific vitamin D receptors. J. Tissue Eng. Regen. Med. 2013, 10, 40–51. [Google Scholar] [CrossRef]
  69. Woeckel, V.J.J.; Koedam, M.; van de Peppel, J.; Chiba, H.; van der Eerden, B.C.J.; van Leeuwen, J.P.T.M. Evidence of vitamin D and interferon-β cross-talk in human osteoblasts with 1α,25-dihydroxyvitamin D3 being dominant over interferon-β in stimulating mineralization. J. Cell. Physiol. 2012, 227, 3258–3266. [Google Scholar] [CrossRef]
  70. Fretz, J.A.; Zella, L.A.; Kim, S.; Shevde, N.K.; Pike, J.W. 1,25-Dihydroxyvitamin D3 induces expression of the Wnt signaling co-regulator LRP5 via regulatory elements located significantly downstream of the gene’s transcriptional start site. J. Steroid Biochem. Mol. Biol. 2007, 103, 440–445. [Google Scholar] [CrossRef]
  71. Haussler, M.R.; Haussler, C.A.; Whitfield, G.K.; Hsieh, J.-C.; Thompson, P.D.; Barthel, T.K.; Bartik, L.; Egan, J.B.; Wu, Y.; Kubicek, J.L.; et al. The nuclear vitamin D receptor controls the expression of genes encoding factors which feed the “Fountain of Youth” to mediate healthful aging. J. Steroid Biochem. Mol. Biol. 2010, 121, 88–97. [Google Scholar] [CrossRef] [Green Version]
  72. Guler, E.; Baripoglu, Y.E.; Alenezi, H.; Arikan, A.; Babazade, R.; Unal, S.; Duruksu, G.; Alfares, F.S.; Yazir, Y.; Oktar, F.N.; et al. Vitamin D3/vitamin K2/magnesium-loaded polylactic acid/tricalcium phosphate/polycaprolactone composite nanofibers demonstrated osteoinductive effect by increasing Runx2 via Wnt/β-catenin pathway. Int. J. Biol. Macromol. 2021, 190, 244–258. [Google Scholar] [CrossRef]
  73. Doroudi, M.; Olivares-Navarrete, R.; Hyzy, S.L.; Boyan, B.D.; Schwartz, Z. Signaling components of the 1α,25(OH)2D3-dependent Pdia3 receptor complex are required for Wnt5a calcium-dependent signaling. Biochim. Biophys. Acta BBA Mol. Cell Res. 2014, 1843, 2365–2375. [Google Scholar] [CrossRef] [Green Version]
  74. Jo, S.; Yoon, S.; Lee, S.Y.; Kim, S.Y.; Park, H.; Han, J.; Choi, S.H.; Han, J.-S.; Yang, J.-H.; Kim, T.-H. DKK1 Induced by 1,25D3 Is Required for the Mineralization of Osteoblasts. Cells 2020, 9, 236. [Google Scholar] [CrossRef] [Green Version]
  75. Chen, C.-T.; Shih, Y.-R.V.; Kuo, T.K.; Lee, O.K.; Wei, Y.-H. Coordinated Changes of Mitochondrial Biogenesis and Antioxidant Enzymes during Osteogenic Differentiation of Human Mesenchymal Stem Cells. Stem Cells 2008, 26, 960–968. [Google Scholar] [CrossRef]
  76. Weivoda, M.M.; Chew, C.K.; Monroe, D.G.; Farr, J.N.; Atkinson, E.J.; Geske, J.R.; Eckhardt, B.; Thicke, B.; Ruan, M.; Tweed, A.J.; et al. Identification of osteoclast-osteoblast coupling factors in humans reveals links between bone and energy metabolism. Nat. Commun. 2020, 11, 87. [Google Scholar] [CrossRef] [Green Version]
  77. Shen, L.; Hu, G.; Karner, C.M. Bioenergetic Metabolism In Osteoblast Differentiation. Curr. Osteoporos. Rep. 2022, 20, 53–64. [Google Scholar] [CrossRef]
  78. Pal, S.; Singh, M.; Porwal, K.; Rajak, S.; Das, N.; Rajput, S.; Trivedi, A.K.; Maurya, R.; Sinha, R.A.; Siddiqi, M.I.; et al. Adiponectin receptors by increasing mitochondrial biogenesis and respiration promote osteoblast differentiation: Discovery of isovitexin as a new class of small molecule adiponectin receptor modulator with potential osteoanabolic function. Eur. J. Pharmacol. 2021, 913, 174634. [Google Scholar] [CrossRef]
  79. Bruedigam, C.; Eijken, M.; Koedam, M.; Pols, H.A.P.; van Leeuwen, J.P.T. New insights into peroxisome proliferatoractivated receptor gamma action: Stimulation of human osteoblast differentiation. Calcif. Tissue Int. 2007, 80, S73. [Google Scholar]
  80. Bruedigam, C.; Eijken, M.; Koedam, M.; van de Peppel, J.; Drabek, K.; Chiba, H.; van Leeuwen, J.P.T.M. A New Concept Underlying Stem Cell Lineage Skewing That Explains the Detrimental Effects of Thiazolidinediones on Bone. Stem Cells 2010, 28, 916–927. [Google Scholar] [CrossRef]
  81. Bouillon, R.; Carmeliet, G.; Lieben, L.; Watanabe, M.; Perino, A.; Auwerx, J.; Schoonjans, K.; Verstuyf, A. Vitamin D and energy homeostasis—Of mice and men. Nat. Rev. Endocrinol. 2013, 10, 79–87. [Google Scholar] [CrossRef] [PubMed]
  82. Abu el Maaty, M.A.; Wölfl, S. Vitamin D as a Novel Regulator of Tumor Metabolism: Insights on Potential Mechanisms and Implications for Anti-Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sheeley, M.P.; Andolino, C.; Kiesel, V.A.; Teegarden, D. Vitamin D regulation of energy metabolism in cancer. Br. J. Pharmacol. 2021, 179, 2890–2905. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, P.; Schatz, A.; Adeyemi, B.; Kozminski, D.; Welsh, J.; Tenniswood, M.; Wang, W.-L.W. Vitamin D and testosterone co-ordinately modulate intracellular zinc levels and energy metabolism in prostate cancer cells. J. Steroid Biochem. Mol. Biol. 2019, 189, 248–258. [Google Scholar] [CrossRef]
  85. Eelen, G.; Verlinden, L.; Meyer, M.B.; Gijsbers, R.; Pike, J.W.; Bouillon, R.; Verstuyf, A. 1,25-Dihydroxyvitamin D3 and the aging-related Forkhead Box O and Sestrin proteins in osteoblasts. J. Steroid Biochem. Mol. Biol. 2013, 136, 112–119. [Google Scholar] [CrossRef]
  86. Komarova, S.V.; Ataullakhanov, F.I.; Globus, R.K. Bioenergetics and mitochondrial transmembrane potential during differentiation of cultured osteoblasts. Am. J. Physiol. Cell. Physiol. 2000, 279, C1220–C1229. [Google Scholar] [CrossRef] [Green Version]
  87. Wu, Y.-Y.; Yu, T.; Zhang, X.-H.; Liu, Y.-S.; Li, F.; Wang, Y.-Y.; Wang, Y.-Y.; Gong, P. 1,25(OH)2D3 inhibits the deleterious effects induced by high glucose on osteoblasts through undercarboxylated osteocalcin and insulin signaling. J. Steroid Biochem. Mol. Biol. 2012, 132, 112–119. [Google Scholar] [CrossRef]
  88. Woeckel, V.J.; Bruedigam, C.; Koedam, M.; Chiba, H.; van der Eerden, B.C.; van Leeuwen, J.P. 1α,25-Dihydroxyvitamin D3 and rosiglitazone synergistically enhance osteoblast-mediated mineralization. Gene 2012, 512, 438–443. [Google Scholar] [CrossRef]
  89. Ali, S.Y.; Sajdera, S.W.; Anderson, H.C. Isolation and Characterization of Calcifying Matrix Vesicles from Epiphyseal Cartilage. Proc. Natl. Acad. Sci. USA 1970, 67, 1513–1520. [Google Scholar] [CrossRef] [Green Version]
  90. Anderson, H.C. Molecular biology of matrix vesicles. Clin. Orthop. Relat. Res. 1995, 314, 266–280. [Google Scholar] [CrossRef]
  91. Anderson, H.C. Matrix vesicles and calcification. Curr. Rheumatol. Rep. 2003, 5, 222–226. [Google Scholar] [CrossRef]
  92. Xiao, Z.; Camalier, C.E.; Nagashima, K.; Chan, K.C.; Lucas, D.A.; de la Cruz, M.J.; Gignac, M.; Lockett, S.; Issaq, H.J.; Veenstra, T.D.; et al. Analysis of the extracellular matrix vesicle proteome in mineralizing osteoblasts. J. Cell. Physiol. 2006, 210, 325–335. [Google Scholar] [CrossRef]
  93. Thouverey, C.; Malinowska, A.; Balcerzak, M.; Strzelecka-Kiliszek, A.; Buchet, R.; Dadlez, M.; Pikula, S. Proteomic characterization of biogenesis and functions of matrix vesicles released from mineralizing human osteoblast-like cells. J. Proteom. 2011, 74, 1123–1134. [Google Scholar] [CrossRef]
  94. Staines, K.A.; Zhu, D.; Farquharson, C.; MacRae, V.E. Identification of novel regulators of osteoblast matrix mineralization by time series transcriptional profiling. J. Bone Miner. Metab. 2013, 32, 240–251. [Google Scholar] [CrossRef] [Green Version]
  95. Tye, C.E.; Hunter, G.K.; Goldberg, H.A. Identification of the Type I Collagen-binding Domain of Bone Sialoprotein and Characterization of the Mechanism of Interaction. J. Biol. Chem. 2005, 280, 13487–13492. [Google Scholar] [CrossRef] [Green Version]
  96. Orimo, H. The Mechanism of Mineralization and the Role of Alkaline Phosphatase in Health and Disease. J. Nippon Med. Sch. 2010, 77, 4–12. [Google Scholar] [CrossRef] [Green Version]
  97. Kim, H.J.; Minashima, T.; McCarthy, E.F.; A Winkles, J.; Kirsch, T. Progressive ankylosis protein (ANK) in osteoblasts and osteoclasts controls bone formation and bone remodeling. J. Bone Miner. Res. 2010, 25, 1771–1783. [Google Scholar] [CrossRef] [Green Version]
  98. Millán, J.L. The Role of Phosphatases in the Initiation of Skeletal Mineralization. Calcif. Tissue Int. 2012, 93, 299–306. [Google Scholar] [CrossRef] [Green Version]
  99. Woeckel, V.J.; Alves, R.D.; Swagemakers, S.M.; Eijken, M.; Chiba, H.; van der Eerden, B.C.; van Leeuwen, J.P. 1α,25-(OH)2D3 acts in the early phase of osteoblast differentiation to enhance mineralization via accelerated production of mature matrix vesicles. J. Cell. Physiol. 2010, 225, 593–600. [Google Scholar] [CrossRef]
  100. Franceschi, R.T.; Romano, P.R.; Park, K.Y. Regulation of type I collagen synthesis by 1,25-dihydroxyvitamin D3 in human osteosarcoma cells. J. Biol. Chem. 1988, 263, 18938–18945. [Google Scholar] [CrossRef]
  101. Hicok, K.C.; Thomas, T.; Gori, F.; Rickard, D.J.; Spelsberg, T.C.; Riggs, B.L. Development and Characterization of Conditionally Immortalized Osteoblast Precursor Cell Lines from Human Bone Marrow Stroma. J. Bone Miner. Res. 1998, 13, 205–217. [Google Scholar] [CrossRef] [PubMed]
  102. Ingram, R.T.; Bonde, S.K.; Riggs, B.L.; Fitzpatrick, L.A. Effects of transforming growth factor beta (TGFβ) and 1,25 dihydroxyvitamin D3 on the function, cytochemistry and morphology of normal human osteoblast-like cells. Differentiation 1994, 55, 153–163. [Google Scholar] [CrossRef] [PubMed]
  103. Siggelkow, H.; Schulz, H.; Kaesler, S.; Benzler, K.; Atkinson, M.J.; Hüfner, M. 1,25 Dihydroxyvitamin-D3 Attenuates the Confluence-Dependent Differences in the Osteoblast Characteristic Proteins Alkaline Phosphatase, Procollagen I Peptide, and Osteocalcin. Calcif. Tissue Int. 1999, 64, 414–421. [Google Scholar] [CrossRef] [PubMed]
  104. Kim, H.T.; Chen, T.L. 1,25-Dihydroxyvitamin D3Interaction with Dexamethasone and Retinoic Acid: Effects on Procollagen Messenger Ribonucleic Acid Levels in Rat Osteoblast-Like Cells. Mol. Endocrinol. 1989, 3, 97–104. [Google Scholar] [CrossRef] [Green Version]
  105. Harrison, J.R.; Petersen, D.N.; Lichtler, A.C.; Mador, A.T.; Rowe, D.W.; Kream, B.E. 1,25-Dihydroxyvitamin D3Inhibits Transcription of Type I Collagen Genes in the Rat Osteosarcoma Cell Line ROS 17/2.8. Endocrinology 1989, 125, 327–333. [Google Scholar] [CrossRef]
  106. Yang, D.; Turner, A.G.; Wijenayaka, A.R.; Anderson, P.H.; Morris, H.A.; Atkins, G.J. 1,25-Dihydroxyvitamin D3 and extracellular calcium promote mineral deposition via NPP1 activity in a mature osteoblast cell line MLO-A5. Mol. Cell. Endocrinol. 2015, 412, 140–147. [Google Scholar] [CrossRef]
  107. Yajima, A.; Tsuchiya, K.; Burr, D.B.; Wallace, J.M.; Damrath, J.D.; Inaba, M.; Tominaga, Y.; Satoh, S.; Nakayama, T.; Tanizawa, T.; et al. The Importance of Biologically Active Vitamin D for Mineralization by Osteocytes After Parathyroidectomy for Renal Hyperparathyroidism. JBMR Plus 2019, 3, e10234. [Google Scholar] [CrossRef] [Green Version]
  108. Lieben, L.; Masuyama, R.; Torrekens, S.; Van Looveren, R.; Schrooten, J.; Baatsen, P.; Lafage-Proust, M.-H.; Dresselaers, T.; Feng, J.Q.; Bonewald, L.F.; et al. Normocalcemia is maintained in mice under conditions of calcium malabsorption by vitamin D–induced inhibition of bone mineralization. J. Clin. Investig. 2012, 122, 1803–1815. [Google Scholar] [CrossRef]
  109. Woeckel, V.J.; van der Eerden, B.C.; Schreuders-Koedam, M.; Eijken, M.; Van Leeuwen, J.P. 1α,25-dihydroxyvitamin D3stimulates activin A production to fine-tune osteoblast-induced mineralization. J. Cell. Physiol. 2013, 228, 2167–2174. [Google Scholar] [CrossRef]
  110. 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]
  111. Shymanskyi, I.; Lisakovska, O.; Mazanova, A.; Labudzynskyi, D.; Veliky, M. Vitamin D3 Modulates Impaired Crosstalk Between RANK and Glucocorticoid Receptor Signaling in Bone Marrow Cells After Chronic Prednisolone Administration. Front. Endocrinol. 2018, 9, 303. [Google Scholar] [CrossRef]
  112. Khalaf, R.M.; Almudhi, A.A. The effect of vitamin D deficiency on the RANKL/OPG ratio in rats. J. Oral Biol. Craniofacial Res. 2022, 12, 228–232. [Google Scholar] [CrossRef]
  113. Kim, S.; Yamazaki, M.; Zella, L.A.; Shevde, N.K.; Pike, J.W. Activation of Receptor Activator of NF-κB Ligand Gene Expression by 1,25-Dihydroxyvitamin D3 Is Mediated through Multiple Long-Range Enhancers. Mol. Cell. Biol. 2006, 26, 6469–6486. [Google Scholar] [CrossRef] [Green Version]
  114. Bouillon, R.; Carmeliet, G. Vitamin D and the skeleton. Curr. Opin. Endocr. Metab. Res. 2018, 3, 68–73. [Google Scholar] [CrossRef]
  115. Carlberg, C.; Muñoz, A. An update on vitamin D signaling and cancer. Semin. Cancer Biol. 2020, 79, 217–230. [Google Scholar] [CrossRef]
  116. Carlberg, C. Vitamin D and Its Target Genes. Nutrients 2022, 14, 1354. [Google Scholar] [CrossRef]
  117. Haussler, M.R.; Livingston, S.; Sabir, Z.L.; Haussler, C.A.; Jurutka, P.W. Vitamin D Receptor Mediates a Myriad of Biological Actions Dependent on Its 1,25-Dihydroxyvitamin D Ligand: Distinct Regulatory Themes Revealed by Induction of Klotho and Fibroblast Growth Factor-23. JBMR Plus 2021, 5, e10432. [Google Scholar] [CrossRef]
  118. Haussler, M.R.; Whitfield, G.K.; Kaneko, I.; Haussler, C.A.; Hsieh, D.; Hsieh, J.-C.; Jurutka, P.W. Molecular Mechanisms of Vitamin D Action. Calcif. Tissue Int. 2013, 92, 77–98. [Google Scholar] [CrossRef]
  119. Owen, T.A.; Aronow, M.S.; Barone, L.M.; Bettencourt, B.; Stein, G.S.; Lian, J.B. Pleiotropic Effects of Vitamin D on Osteoblast Gene Expression Are Related to the Proliferative and Differentiated State of the Bone Cell Phenotype: Dependency upon Basal Levels of Gene Expression, Duration of Exposure, and Bone Matrix Competency in Normal Rat Osteoblast Cultures. Endocrinology 1991, 128, 1496–1504. [Google Scholar] [CrossRef]
  120. Saji, F.; Shigematsu, T.; Sakaguchi, T.; Ohya, M.; Orita, H.; Maeda, Y.; Ooura, M.; Mima, T.; Negi, S. Fibroblast growth factor 23 production in bone is directly regulated by 1α,25-dihydroxyvitamin D, but not PTH. Am. J. Physiol. Physiol. 2010, 299, F1212–F1217. [Google Scholar] [CrossRef] [Green Version]
  121. Yamamoto, R.; Minamizaki, T.; Yoshiko, Y.; Yoshioka, H.; Tanne, K.; Aubin, J.E.; Maeda, N. 1,25-dihydroxyvitamin D3 acts predominately in mature osteoblasts under conditions of high extracellular phosphate to increase fibroblast growth factor 23 production in vitro. J. Endocrinol. 2010, 206, 279–286. [Google Scholar] [CrossRef] [PubMed]
  122. Quarles, L.D. Skeletal secretion of FGF-23 regulates phosphate and vitamin D metabolism. Nat. Rev. Endocrinol. 2012, 8, 276–286. [Google Scholar] [CrossRef] [PubMed]
  123. Razzaque, M.S. Interactions between FGF23 and vitamin D. Endocr. Connect. 2022, 11, e220239. [Google Scholar] [CrossRef] [PubMed]
  124. Lanske, B.; Densmore, M.J.; Erben, R.G. Vitamin D endocrine system and osteocytes. BoneKEy Rep. 2014, 3, 494. [Google Scholar] [CrossRef] [Green Version]
  125. Hines, E.R.; Kolek, O.I.; Jones, M.D.; Serey, S.H.; Sirjani, N.B.; Kiela, P.R.; Jurutka, P.W.; Haussler, M.R.; Collins, J.F.; Ghishan, F.K. 1,25-Dihydroxyvitamin D3 Down-regulation of PHEX Gene Expression Is Mediated by Apparent Repression of a 110 kDa Transfactor That Binds to a Polyadenine Element in the Promoter. J. Biol. Chem. 2004, 279, 46406–46414. [Google Scholar] [CrossRef] [Green Version]
  126. Turner, R.T.; Puzas, J.E.; Forte, M.D.; Lester, G.E.; Gray, T.K.; Howard, G.A.; Baylink, D.J. In vitro synthesis of 1 alpha,25-dihydroxycholecalciferol and 24,25-dihydroxycholecalciferol by isolated calvarial cells. Proc. Natl. Acad. Sci. USA 1980, 77, 5720–5724. [Google Scholar] [CrossRef] [Green Version]
  127. Howard, G.A.; Turner, R.T.; Sherrard, D.J.; Baylink, D.J. Human bone cells in culture metabolize 25-hydroxyvitamin D3 to 1,25-dihydroxyvitamin D3 and 24,25-dihydroxyvitamin D3. J. Biol. Chem. 1981, 256, 7738–7740. [Google Scholar] [CrossRef]
  128. Geng, S.; Zhou, S.; Glowacki, J. Effects of 25-hydroxyvitamin D3 on proliferation and osteoblast differentiation of human marrow stromal cells require CYP27B1/1α-hydroxylase. J. Bone Miner. Res. 2011, 26, 1145–1153. [Google Scholar] [CrossRef] [Green Version]
  129. Lou, Y.-R.; Toh, T.C.; Tee, Y.H.; Yu, H. 25-Hydroxyvitamin D3 induces osteogenic differentiation of human mesenchymal stem cells. Sci. Rep. 2017, 7, srep42816. [Google Scholar] [CrossRef] [Green Version]
  130. Geng, S.; Zhou, S.; Glowacki, J. Age-related decline in osteoblastogenesis and 1α-hydroxylase/CYP27B1 in human mesenchymal stem cells: Stimulation by parathyroid hormone. Aging Cell 2011, 10, 962–971. [Google Scholar] [CrossRef] [Green Version]
  131. Bikle, D.D.; Patzek, S.; Wang, Y. Physiologic and pathophysiologic roles of extra renal CYP27b1: Case report and review. Bone Rep. 2018, 8, 255–267. [Google Scholar] [CrossRef]
  132. Anderson, P.H.; Lam, N.N.; Turner, A.G.; Davey, R.A.; Kogawa, M.; Atkins, G.J.; Morris, H.A. The pleiotropic effects of vitamin D in bone. J. Steroid Biochem. Mol. Biol. 2013, 136, 190–194. [Google Scholar] [CrossRef]
  133. Hewison, M.; Zehnder, D.; Chakraverty, R.; Adams, J.S. Vitamin D and barrier function: A novel role for extra-renal 1α-hydroxylase. Mol. Cell. Endocrinol. 2004, 215, 31–38. [Google Scholar] [CrossRef]
  134. Yang, D.; Anderson, P.H.; Turner, A.G.; Morris, H.A.; Atkins, G.J. Comparison of the biological effects of exogenous and endogenous 1,25-dihydroxyvitamin D3 on the mature osteoblast cell line MLO-A5. J. Steroid Biochem. Mol. Biol. 2016, 164, 374–378. [Google Scholar] [CrossRef]
  135. Meyer, M.B.; Pike, J.W. Mechanistic homeostasis of vitamin D metabolism in the kidney through reciprocal modulation of Cyp27b1 and Cyp24a1 expression. J. Steroid Biochem. Mol. Biol. 2020, 196, 105500. [Google Scholar] [CrossRef]
  136. Zhou, S.; LeBoff, M.S.; Glowacki, J. Vitamin D Metabolism and Action in Human Bone Marrow Stromal Cells. Endocrinology 2010, 151, 14–22. [Google Scholar] [CrossRef] [Green Version]
  137. St-Arnaud, R.; Jones, G. Chapter 6—CYP24A1: Structure, Function, and Physiological Role. In Vitamin D, 4th ed.; Feldman, D., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 81–95. [Google Scholar]
  138. Van Leeuwen, J.P.T.M.; Van Den Bemd, G.J.C.M.; Van Driel, M.; Buurman, C.J.; Pols, H.A.P. 24,25-Dihydroxyvitamin D3 and bone metabolism. Steroids 2001, 66, 375–380. [Google Scholar] [CrossRef]
  139. Väisänen, S.; Dunlop, T.W.; Sinkkonen, L.; Frank, C.; Carlberg, C. Spatio-temporal Activation of Chromatin on the Human CYP24 Gene Promoter in the Presence of 1α,25-Dihydroxyvitamin D3. J. Mol. Biol. 2005, 350, 65–77. [Google Scholar] [CrossRef]
  140. Henry, H.L. The 25(OH)D3/1α,25(OH)2D3-24R-hydroxylase: A catabolic or biosynthetic enzyme? Steroids 2001, 66, 391–398. [Google Scholar] [CrossRef] [PubMed]
  141. Moena, D.; Nardocci, G.; Acevedo, E.; Lian, J.; Stein, G.; Stein, J.; Montecino, M. Ezh2-dependent H3K27me3 modification dynamically regulates vitamin D3-dependent epigenetic control of CYP24A1 gene expression in osteoblastic cells. J. Cell. Physiol. 2020, 235, 5404–5412. [Google Scholar] [CrossRef]
  142. Pols, H.A.; Birkenhager, J.C.; Schilte, J.P.; Visser, T.J. Evidence that the self-induced metabolism of 1,25-dihydroxyvitamin D-3 limits the homologous up-regulation of its receptor in rat osteosarcoma cells. Biochim. et Biophys. Acta (BBA) Mol. Cell Res. 1988, 970, 122–129. [Google Scholar] [CrossRef]
  143. Staal, A.; vandenBemd, G.; Birkenhager, J.; Pols, H.; van Leeuwen, J. Consequences of vitamin D receptor regulation for the 1,25-dihydroxyvitamin D3-induced 24-hydroxylase activity in osteoblast-like cells: Initiation of the C24-oxidation pathway. Bone 1997, 20, 237–243. [Google Scholar] [CrossRef] [PubMed]
  144. Henry, H.L.; Norman, A.W. Vitamin D: Two Dihydroxylated Metabolites Are Required for Normal Chicken Egg Hatchability. Science 1978, 201, 835–837. [Google Scholar] [CrossRef]
  145. Norman, A.W.; Henry, H.L.; Malluche, H.H. 24R,25-dihydroxyvitamin D3 and 1α,25-dihydroxyvitamin D3 are both indispensable for calcium and phosphorus homeostasis. Life Sci. 1980, 27, 229–237. [Google Scholar] [CrossRef]
  146. Endo, H.; Kiyoki, M.; Kawashima, K.; Naruchi, T.; Hashimoto, Y. Vitamin D3 metabolites and PTH synergistically stimulate bone formation of chick embryonic femur in vitro. Nature 1980, 286, 262–264. [Google Scholar] [CrossRef]
  147. Galus, K.; Szymendera, J.; Zaleski, A.; Schreyer, K. Effects of 1α-hydroxyvitamin D3 and 24R,25-dihydroxyvitamin D3 on bone remodeling. Calcif. Tissue Int. 1980, 31, 209–213. [Google Scholar] [CrossRef]
  148. Erben, R.G.; Weiser, H.; Sinowatz, F.; Rambeck, W.A.; Zucker, H. Vitamin D metabolites prevent vertebral osteopenia in ovariectomized rats. Calcif. Tissue Int. 1992, 50, 228–236. [Google Scholar] [CrossRef]
  149. Matsumoto, T.; Ezawa, I.; Morita, K.; Kawanobe, Y.; Ogata, E. Effect of Vitamin D Metabolites on Bone Metabolism in a Rat Model of Postmenopausal Osteoporosis. J. Nutr. Sci. Vitaminol. 1985, 31, S61–S65. [Google Scholar] [CrossRef]
  150. Kato, A.; Seo, E.G.; Einhorn, T.A.; Bishop, J.E.; Norman, A.W. Studies on 24R,25-dihydroxyvitamin D3: Evidence for a nonnuclear membrane receptor in the chick tibial fracture-healing callus. Bone 1998, 23, 141–146. [Google Scholar] [CrossRef]
  151. Seo, E.-G.; Einhorn, T.A.; Norman, A.W. 24R,25-Dihydroxyvitamin D3: An Essential Vitamin D3 Metabolite for Both Normal Bone Integrity and Healing of Tibial Fracture in Chicks. Endocrinology 1997, 138, 3864–3872. [Google Scholar] [CrossRef]
  152. Martineau, C.; Kaufmann, M.; Arabian, A.; Jones, G.; St-Arnaud, R. Preclinical safety and efficacy of 24R,25-dihydroxyvitamin D3 or lactosylceramide treatment to enhance fracture repair. J. Orthop. Transl. 2020, 23, 77–88. [Google Scholar] [CrossRef] [PubMed]
  153. St-Arnaud, R. CYP24A1-deficient mice as a tool to uncover a biological activity for vitamin D metabolites hydroxylated at position 24. J. Steroid Biochem. Mol. Biol. 2010, 121, 254–256. [Google Scholar] [CrossRef]
  154. Weisman, Y.; Salama, R.; Harell, A.; Edelstein, S. Serum 24,25-dihydroxyvitamin D and 25-hydroxyvitamin D concentrations in femoral neck fracture. BMJ 1978, 2, 1196–1197. [Google Scholar] [CrossRef] [Green Version]
  155. Birkenhager-Frenkel, D.H.; Pols, H.A.; Zeelenberg, J.; Eijgelsheim, J.J.; Schot, R.; Nigg, A.L.; Weimar, W.; Mulder, P.G.; Birkenhager, J.C. Effects of 24r,25-dihydroxyvitamin D3 in combination with 1α-hydroxyvitamin D3 in predialysis renal insufficiency: Biochemistry and histomorphometry of cancellous bone. J. Bone Miner. Res. 1995, 10, 197–204. [Google Scholar] [CrossRef]
Figure 2. Schematic representation of classic vitamin D metabolism and signaling pathway. Either from sunlight or food, vitamin D is converted via enzymatic reactions in the liver and kidney into its active metabolite, 1α,25-OH2D3, which binds to the VDR. Gene activation follows after binding of the vitamin D/receptor complex to vitamin D response elements (VDREs) in target genes.
Figure 2. Schematic representation of classic vitamin D metabolism and signaling pathway. Either from sunlight or food, vitamin D is converted via enzymatic reactions in the liver and kidney into its active metabolite, 1α,25-OH2D3, which binds to the VDR. Gene activation follows after binding of the vitamin D/receptor complex to vitamin D response elements (VDREs) in target genes.
Nutrients 15 00480 g002
Table 1. Number of transcripts on average that are 2-fold up- or downregulated in the pre-mineralization or mineralization phase of human osteoblasts *.
Table 1. Number of transcripts on average that are 2-fold up- or downregulated in the pre-mineralization or mineralization phase of human osteoblasts *.
Condition# of Genes UP# of Genes DOWN
Pre-mineralization phase 155164
Mineralization phase166236
In both phases108
* Experimental procedures and culture conditions of human osteoblasts (SV-HFO) are described in Woeckel et al. [99]. Two-fold change is based on the average expression at the timepoints in the pre-mineralization or mineralization period.
Table 2. Gene symbols of transcripts that are 2-fold upregulated or downregulated in both the pre-mineralization and mineralization phases of human osteoblasts (i.e., 10 and 8 in both phases in Table 1) *.
Table 2. Gene symbols of transcripts that are 2-fold upregulated or downregulated in both the pre-mineralization and mineralization phases of human osteoblasts (i.e., 10 and 8 in both phases in Table 1) *.
Upregulated Downregulated
ABCC3AGAP10
CYP24A1CCL20
MAGEE1DDIT3
RARRES2GRK4
RICH2LOC727869
SLC25A45NFE2L2
SULT1C2ODF1
THBDTSC22D2
TMEM180
TOX3
* Experimental procedures and culture conditions of human osteoblasts (SV-HFO) are described in Woeckel et al. [99]. Two-fold change is based on the average expression at the timepoints in the pre-mineralization or mineralization period.
Table 3. Number of transcripts uniquely 2-fold up- or downregulated in either the pre-mineralization or in the mineralization phase of human osteoblasts *.
Table 3. Number of transcripts uniquely 2-fold up- or downregulated in either the pre-mineralization or in the mineralization phase of human osteoblasts *.
Condition# of Genes UP# of Genes DOWN
Pre-mineralization phase 6566
Mineralization phase77100
* Experimental procedures and culture conditions of human osteoblasts (SV-HFO) are described in Woeckel et al. [99]. The 2-fold and 0.8–1.2-fold change is based on the average expression at the timepoints in the pre-mineralization or mineralization period.
Table 4. Overview of transcript gene names that are uniquely 2-fold up- or downregulated in either the pre-mineralization or mineralization phase of human osteoblasts *.
Table 4. Overview of transcript gene names that are uniquely 2-fold up- or downregulated in either the pre-mineralization or mineralization phase of human osteoblasts *.
Pre-Mineralization PhaseMineralization Phase
UpregulatedDownregulatedUpregulatedDownregulated
AQRRAB9BP1ADAM22RARAABCD4MYH11AASDHMOSPD1
ARHGEF7RLTPRADORA1RBMAKAP13NFIXABCD3MRPS23
ATAT1SARDHATF7IP2RIMKLBANKRD11ORC5LABT1MS4A1
ATG16L1SHISA8BAGESLC19A1APIPPCDHB3ACTR3CMTUS2
ATP1A4SLC38A11BRS3SLC26A7ARHGDIBPDLIM5ANUBL1NCRNA00188
BCL11ASZT2BRWD1SLC3A1ASH1LPDZRN4AP5S1NDRG2
BMFTEX9BST2SNRPNATMPGAP1B4GALNT2NDUFB7
BMP15TMEM120BC1orf68TBK1BNC2PLEKHG2C11orf65NRAP
C15orf48TMEM33CACNA1ATFAP4BPTFPPP4R4C14orf156NUDT14
C2orf27AUBE2G2CCDC144CTHPOBRD4PRPF18C14orf2OGFR
C3orf20UBXN10CSF2RATMPRSS15CAP1PTGESC17orf104PANK2
C8orf34UNC13CCTNSTRIB3CCDC67PTGS1C4orf36PAPPA
CCDC124ZC3H12A-DTDEFB132TRMT2ACCDC76RAB3IPCCL5PAX8
COL24A1ZNF668EDATTBK2CD14RASAL2CCT2PIP5K1A
CTU2ZNF703ERCC6L2ZNF396CLCN4RG9MTD2CNOT2PLCH1
DCTN2 FAM219AZNF93CROCCL1SERTAD4COX7CPMCH
DOCK6 FCGR2C DCLK3SMARCA4CSRP2BPPML
DST FLJ10213 DPP4SRGAP1DAZLPOLE4
DUSP28 FSD1L EGFRSRRM2DBIPOLR2K
EPG5 GAS2 EP300SULF1DCUN1D1PTPRA
EYA2 GLIPR1 FAM102ATBC1D13DNAH1RHEB
GABRB3 GPR155 FAM186ATBL1XDUSP16RPAIN
GNRHR HM13 FAM20CUGGT2EEF1DRPL13
HCRTR2 ICA1 FGF7VCANEGFL8RPL14
HIST1H4C KLHL36 FLJ11292ZNF397EHD1RPL34
HSPB7 KLK7 FLJ13773ZNF462ELP6RPS11
IL1RN LEKR1 FOXP2ZSWIM1ESPNLSEMA6D
KCNJ15 LELP1 GABRA5 EXOGSHLD1
LOC100131283 LIN28B H2AFY EXOSC2SLC10A7
LOC148987 LOC100286895 HMCN1 FABP4SLC9A5
LOC149351 LOC100287114 HOXA6 FAM126ASNAP23
LOC285205 LOC283854 HSPA12A FAM27ASNCAIP
LOC645591 LOC285692 IL17C FAXCSNTG1
LOC728903 LOC390595 INTS4 FUT7STEEP1
LOC780529 LOC440944 KCNAB1 GOSR1STK32A
LRRC46 MAN1A2 KCNG3 GPR39STMN3
LYZL6 MAPRE3 KRTAP3-3 GSNSUPT16H
MGC42157 MGC12916 LOC100127980 HCG4P6TAL1
MRS2 MRPL19 LOC100128640 IRGQTBC1D8
NCOR2 MSR1 LOC100131993 KCNIP3TEN1
NOX4 MYO10 LOC283682 KYTLK1
NTRK2 NR2E3 LOC285500 LOC100133109TWF1
OR1J4 NUP210L LOC388210 LOC100287911TXNIP
PDE1A OTX2 LOC441461 LOC100289246UHRF1BP1L
PENK PCLO MAGEB18 LOC338862UQCRB
PGM2L1 PKP2 MARK2 LOC643749UQCRQ
PHC3 PLXNA2 MEGF10 LPAR5VMA21
POU2F1 POU2F2 MGAT5B MATR3WFDC21P
PRRG2 PRLR MLXIP MMP16XAF1
PTCD3 RAD54L2 MS4A6A MMP17ZNF880
* Experimental procedures and culture conditions of human osteoblasts (SV-HFO) are described in Woeckel et al. [99].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

van Driel, M.; van Leeuwen, J.P.T.M. Vitamin D and Bone: A Story of Endocrine and Auto/Paracrine Action in Osteoblasts. Nutrients 2023, 15, 480. https://doi.org/10.3390/nu15030480

AMA Style

van Driel M, van Leeuwen JPTM. Vitamin D and Bone: A Story of Endocrine and Auto/Paracrine Action in Osteoblasts. Nutrients. 2023; 15(3):480. https://doi.org/10.3390/nu15030480

Chicago/Turabian Style

van Driel, Marjolein, and Johannes P. T. M. van Leeuwen. 2023. "Vitamin D and Bone: A Story of Endocrine and Auto/Paracrine Action in Osteoblasts" Nutrients 15, no. 3: 480. https://doi.org/10.3390/nu15030480

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