Skip to main content

Modulation of the vitamin D/vitamin D receptor system in osteoporosis pathogenesis: insights and therapeutic approaches

Abstract

Osteoporosis is a prevalent bone disorder characterized by low bone mineral density (BMD) and deteriorated bone microarchitecture, leading to an increased risk of fractures. Vitamin D (VD), an essential nutrient for skeletal health, plays a vital role in maintaining bone homeostasis. The biological effects of VD are primarily mediated through the vitamin D receptor (VDR), a nuclear receptor that regulates the transcription of target genes involved in calcium and phosphate metabolism, bone mineralization, and bone remodeling. In this review article, we conduct a thorough literature search of the PubMed and EMBASE databases, spanning from January 2000 to September 2023. Utilizing the keywords “vitamin D,” “vitamin D receptor,” “osteoporosis,” and “therapy,” we aim to provide an exhaustive overview of the role of the VD/VDR system in osteoporosis pathogenesis, highlighting the most recent findings in this field. We explore the molecular mechanisms underlying VDR’s effects on bone cells, including osteoblasts and osteoclasts, and discuss the impact of VDR polymorphisms on BMD and fracture risk. Additionally, we examine the interplay between VDR and other factors, such as hormonal regulation, genetic variants, and epigenetic modifications, that contribute to osteoporosis susceptibility. The therapeutic implications of targeting the VDR pathway for osteoporosis management are also discussed. By bringing together these diverse aspects, this review enhances our understanding of the VD/VDR system’s critical role in the pathogenesis of osteoporosis and highlights its significance as a potential therapeutic target.

Introduction

Osteoporosis is a significant and growing health concern worldwide, particularly among the aging population [1, 2]. It is characterized by reduced bone mineral density (BMD) and deteriorated bone microarchitecture, resulting in increased skeletal fragility and a heightened risk of fractures [1, 2]. According to the International Osteoporosis Foundation, osteoporosis affects an estimated 200 million individuals globally, and this number is projected to rise as the population continues to age [3]. The overall prevalence of osteoporosis among adults aged 50 and over is nearly 13% while it rises to approximately 49% among women over 50 ages [2, 3]. The present rapid growth of the elderly population has made osteoporosis a major healthcare burden that has resulted in enormous and growing costs to national healthcare systems [1,2,3].

The pathogenesis of osteoporosis is complex and multifactorial, involving a delicate interplay between genetic, hormonal, environmental, and lifestyle factors (Fig. 1A) [4, 5]. Importantly, a deficiency of vitamin D (VD) is an important cause that leads to osteoporosis [6]. It is crucial to understand the underlying mechanisms that contribute to bone loss and compromised bone quality in order to develop effective preventive and therapeutic strategies.

Fig. 1
figure 1

Factors involved in the pathogenesis of osteoporosis and chemical structures of vitamin D2/D3. A Factors contributing to pathogenesis of osteoporosis. Osteoporosis can be affected by multiple factors, such as gender, age, race, body size, family history, diet, changes in hormones, lifestyle, and special medicines. B Chemical structures of vitamin D2 (C28H44O) and D3 (C27H44O) are shown

VD, in particular VD2 and VD3 (Fig. 1B), is a group of fat-soluble secosteroids vital for skeletal health, and it plays a key role in maintaining bone homeostasis [7, 8]. Its biological effects are primarily mediated via the vitamin D receptor (VDR), a nuclear receptor that governs the transcription of target genes [7, 8]. These genes are involved in a wide array of processes, including calcium and phosphate metabolism, bone mineralization, and bone remodeling, all critical to the preservation of bone health [9]. Dysregulation of the VD/VDR system has been linked to several bone pathologies, including osteoporosis [10]. Recent advancements have shed light on how the VD/VDR system influences the behavior of bone cells, such as osteoblasts and osteoclasts, and how VDR polymorphisms might affect BMD and fracture risk [1, 11, 12]. Moreover, the interplay between the VDR and other factors, such as hormonal regulation, genetic variants, and epigenetic modifications, has become a topic of intense investigation [13, 14].

In this review, we aim to provide a comprehensive overview of the current understanding of the VD/VDR system's role in osteoporosis pathogenesis. By consolidating recent findings, we underscore the relevance of the VD/VDR system in the onset and progression of osteoporosis, while also highlighting potential therapeutic avenues for disease management.

Uptake, synthesis, and metabolism of VD

VD can be sourced from natural sun exposure and certain foods, including fatty fish, fish liver oils, beef liver, egg yolks, and mushrooms. Additionally, various foods, such as cheeses, milk, butter, cereal, and whole grains are often fortified with VD supplements to enhance dietary intake (Fig. 2) [7, 15]. The synthesis of VD begins in the skin, where exposure to ultraviolet B (UVB) radiation converts 7-dehydrocholesterol, a compound found in significant amounts in the skin, into previtamin D3 (Fig. 2) [7, 15]. Previtamin D3 is then transformed into vitamin D3 (VD3), or cholecalciferol, via a heat-dependent process (Fig. 2) [7, 15]. Following its synthesis, VD3 is metabolized in the liver by the enzyme 25-hydroxylase to 25-hydroxyvitamin D3, also known as calcifediol (Fig. 2) [7, 15].

Fig. 2
figure 2

Uptake, synthesis, and metabolism of vitamin D2 and D3. Vitamin D can be obtained through natural sun exposure and from dietary sources such as fish, mushrooms, cheeses, milk, butter, cereal, and whole grains. Vitamin D3 is synthesized in the skin when pro-vitamin D3 (7-dehydrocholesterol) is converted to pre-vitamin D3 in response to sunlight exposure (ultraviolet B radiation). Alternatively, Vitamin D3 can be obtained from natural or fortified foods and supplements from vitamin D2 (ergocalciferol) and vitamin D3. After entering the bloodstream, vitamin D2 and D3 bind to vitamin D-binding protein (DBP) and are transported to the liver where they are hydroxylated by liver 25-hydroxylases to form 25-hydroxycholecalciferol [25(OH)D] (calcifediol or calcidiol). 25(OH)D is then transported to the kidney where it is further hydroxylated by 1α-hydroxylase to produce the active secosteroid 1,25(OH)2D (calcitriol). The synthesis of 1,25(OH)2D is regulated by parathyroid hormone and is suppressed by calcium, phosphate, and 1,25(OH)2D itself

VD metabolism occurs primarily in the kidneys but also in other tissues, where the enzyme 1α-hydroxylase converts calcifediol into its active form, calcitriol (1,25-dihydroxyvitamin D3) (Fig. 2) [7, 15, 16]. This conversion is tightly regulated by several factors including serum levels of parathyroid hormone, calcium, and phosphate [11]. Recent studies have found that dysregulation in this final conversion step could lead to inadequate levels of active VD, contributing to the pathogenesis of various health conditions including osteoporosis [17].

In addition to endogenous synthesis, dietary intake also contributes to the body's VD status [7, 8]. The primary dietary forms of VD are vitamin D3, found in foods like fatty fish and egg yolks, and vitamin D2, or ergocalciferol, which is obtained from plant sources and fortified foods (Fig. 2) [7, 8]. Both forms are metabolized in the same manner within the body [7, 8].

Structure and functions of VDR

Molecular structure of VDR

VDR is a nuclear hormone receptor that regulates gene expression in response to VD [18]. The VDR protein consists of several domains that contribute to its structure and function [19, 20]. The N-terminal domain (NTD) contains transcriptional activation functions and interacts with coactivators and corepressors (Fig. 3A) [19, 20]. The DNA-binding domain (DBD) allows the VDR to bind to specific DNA sequences called vitamin D response elements (VDREs) (Fig. 3A) [19, 20]. The ligand-binding domain (LBD) is responsible for ligand recognition and dimerization with the retinoid X receptor (RXR) (Fig. 3A) [19, 20]. Together, these domains enable the VDR to interact with DNA and other proteins to modulate gene expression [19, 20].

Fig. 3
figure 3

Functional domains of VDR protein and activation of VDR. A Functional domains of VDR proteins. Different domains, including NTD, DBD, and LBD, are shown. B The classic VDR signaling. The VDR-ligand complex forms a heterodimer with RXR, and the complex translocates to the nucleus. In the nucleus, the VDR-RXR heterodimer binds to VDREs located in the promoter regions of target genes, leading to the recruitment of coactivators and the initiation of gene transcription

Ligand binding and activation of VDR

VDR is activated by binding to its ligand, calcitriol [21]. Upon ligand binding, the VDR undergoes conformational changes that facilitate its interaction with coactivator proteins [22]. The VDR-ligand complex then forms a heterodimer with RXR, and the complex translocates to the nucleus [22]. In the nucleus, the VDR-RXR heterodimer binds to VDREs located in the promoter regions of target genes, leading to the recruitment of coactivators and the initiation of gene transcription (Fig. 3B) [22]. The ligand-binding process is crucial for the activation of VDR and the regulation of target gene expression [22].

VDR signaling pathways

Activation of VDR can trigger a multitude of signaling pathways that contribute to its myriad physiological effects, ranging from maintaining bone homeostasis to modulating immune responses [23,24,25]. The binding of VDR–RXR heterodimer to VDREs on gene promoters represents the classic genomic pathway of VDR, which has been shown to regulate numerous genes [e.g., Calbindin (CaBP), Cytochrome P450, Family 27, Subfamily B, Polypeptide 1 (CYP27B1), nuclear factor kappa-Β ligand (RANKL), fibroblast growth factor 23 (FGF23)] involved in calcium and phosphate metabolism (Fig. 3B), crucial for bone health [9, 23, 26].

Besides the classic pathway, VDR can also initiate rapid non-genomic actions, which take effect within minutes to hours of VD administration [27, 28]. In these pathways, VDR interacts with and is phosphorylated by various protein kinases, such as protein kinase A (PKA), mitogen-activated protein kinase (MAPK), protein kinase C beta (PKC-β), and casein kinase 2 (CK2) or with phospholipase C to modulate intracellular calcium and phosphate levels (Fig. 4A) [27, 29, 30].

Fig. 4
figure 4

Phosphorylation sites of VDR by kinases and chromosomal positions of VDR polymorphisms. A Phosphorylation sites of VDR by kinases (PKC-β, PKA, and CK2). B Chromosomal positions of four VDR polymorphisms (rs7975232, rs1544410, rs228570, and rs731236) are shown

Moreover, VDR can interact with other signaling molecules, exemplified by its crosstalk with the Wnt/β-catenin signaling pathway, which is instrumental for bone formation [31, 32]. In addition, VDR has been found to interact with other nuclear receptors, such as estrogen receptor (ER) and peroxisome proliferator-activated receptor gamma (PPARγ), indicating complex interactions in regulating bone health and inflammation [33].

Emerging evidence indicates a pivotal role for the VD/VDR signaling pathway in modulating inflammatory responses as well. Some studies have demonstrated the inhibitory effect of VD/VDR signaling on the activation of the NF-κB pathway, a key player in inflammation, in specific cell types such as embryonic fibroblasts and intestinal epithelial cells [34,35,36,37]. Moreover, VD has been found to downregulate the expression of hypoxia-inducible factor-1α (HIF-1α) in osteoclasts, while tumor necrosis factor-alpha (TNFα) appears to reciprocally decrease VDR levels [38]. Additionally, VDR signaling has been implicated in immune regulation, cell proliferation, and differentiation [39, 40]. The VDR is expressed in immune cells, and its activation can modulate immune responses, such as the production of antimicrobial peptides and the regulation of immune cell differentiation [39, 40].

Regulation of VDR expression

The expression of VDR can be regulated at multiple levels. Transcriptional regulation plays a crucial role in determining VDR abundance. Factors such as VD status, calcium levels, and hormonal signals can influence VDR gene transcription [25, 41]. Additionally, posttranscriptional mechanisms, including mRNA stability and microRNA-mediated regulation, can impact VDR expression [25, 41]. Furthermore, epigenetic modifications, such as DNA methylation and histone modifications, can affect VDR expression by altering chromatin structure and accessibility [25, 41]. These modifications can be influenced by environmental factors, including diet, lifestyle, and exposure to sunlight [25, 41].

Effects of VD/VDR on osteoblasts and osteoclasts

VDR regulation of osteoblast function and mineralization

Osteoblasts, the cells responsible for bone formation, play a crucial role in maintaining skeletal integrity and bone remodeling [42]. Emerging evidence has highlighted the importance of VDR in regulating osteoblast function and differentiation [43, 44]. After binding of calcitriol, VDR activation promotes osteoblast differentiation, a tightly regulated process involving the transition of mesenchymal stem cells into mature osteoblasts [43, 44]. Studies have demonstrated that VDR activation enhances the expression of key osteogenic markers, such as runt-related transcription factor 2 (Runx2), osterix (Osx), and alkaline phosphatase (ALP) [45,46,47]. These factors are essential for osteoblast commitment and maturation, as well as the subsequent synthesis and mineralization of the bone matrix [48].

Beyond its role in osteoblast differentiation, VDR exerts profound effects on osteoblast function and mineralization [49]. VDR activation stimulates the production of various extracellular matrix proteins, including type I collagen, osteopontin, and osteocalcin, which are essential for bone formation and maintenance of bone strength [43, 44]. Moreover, VDR activation modulates the balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption by influencing the expression of receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) [50,51,52]. RANKL is a key osteoclastogenic factor, while OPG acts as a decoy receptor, inhibiting RANKL and preventing osteoclast activation [50,51,52]. VDR activation in osteoblasts leads to decreased RANKL expression and increased OPG expression, thereby suppressing osteoclastogenesis and maintaining bone homeostasis [50,51,52].

VDR regulation of osteoclast function and bone resorption

Osteoclasts, the cells responsible for bone resorption, play a crucial role in maintaining bone homeostasis through the removal of old or damaged bone tissue [53]. Emerging evidence suggests that VDR is involved in the regulation of osteoclast differentiation and activation. Studies have demonstrated that VDR activation inhibits osteoclast differentiation by suppressing the expression of key osteoclastogenic factors such as RANKL and macrophage colony-stimulating factor (M-CSF) [54]. VDR activation reduces the production of RANKL and M-CSF, thereby inhibiting osteoclast formation and subsequent bone resorption [54].

In addition to its effects on osteoclast differentiation, VDR has been shown to regulate the function of mature osteoclasts and the process of bone resorption [54, 55]. VDR activation has been reported to suppress the expression of genes involved in osteoclast activity, such as cathepsin K, matrix metalloproteinase-9 (MMP-9), and tartrate-resistant acid phosphatase (TRAP) [56]. Cathepsin K is a lysosomal protease involved in the degradation of the organic matrix of bone, while MMP-9 participates in the breakdown of the collagenous matrix [56]. TRAP is an enzyme associated with osteoclast function and is used as a marker for osteoclast activity [56]. By downregulating the expression of these genes, VDR activation reduces the resorptive capacity of osteoclasts [56]. By increasing the OPG-to-RANKL ratio and Wnt/β-catenin signaling, VDR activation helps maintain a balance between bone formation and resorption [32].

Genetic variations in the VDR gene and osteoporosis

Polymorphisms in the VDR gene and osteoporosis susceptibility

The human VDR gene is located on the short arm of chromosome 12q13.1 [57]. This gene is more than 100 kb in length and consists of 6 untranslated exons (exons 1a–1f) and 8 protein-coding exons (exons 2–9) [58]. More than 60 polymorphisms of the VDR gene have been reported, ranging from sites in the promoter, exons, and introns to the 3’-untranslated region (UTR) [59]. Genetic variations in the VDR gene have been extensively studied in relation to osteoporosis susceptibility. Several single nucleotide polymorphisms (SNPs) have been identified in the VDR gene, and these variations can influence VDR function and subsequent effects on bone health. Commonly studied VDR SNPs include FokI (rs2228570), ApaI (rs7975232), BsmI (rs1544410), and TaqI (rs731236). Of these polymorphisms, ApaI and BsmI are located in the intron between exon 8 and exon 9, FokI is present in exon 2, and TaqI is located in exon 9 (Fig. 4B) [60].

Extensive research has been carried out globally on the relationship between VDR gene polymorphisms, particularly ApaI, TaqI, and BsmI, and osteoporosis (Table 1) [61,62,63,64,65,66,67,68,69,70,71,72,73,74,75]. Although there are variations in study results, a significant association between these polymorphisms and osteoporosis risk has been predominantly reported (Table 1). Notably, studies conducted in various ethnic populations including Saudis, British, and Chinese revealed significant associations between these polymorphisms and BMD or osteoporosis risk, despite some discrepancies (Table 1) [61,62,63,64,65,66,67,68,69,70,71,72,73,74,75].

Table 1 Association of different VDR polymorphisms with osteoporosis risks

In addition to the mentioned polymorphisms, others like rs11568820, and genetic variants p.Gly14Ala and p.His305Gln have been identified as potential osteoporosis predictors in certain populations (Table 1) [12, 76, 77]. Nevertheless, their association with osteoporosis risk remains inconclusive, as results varied among different studies.

Influence of VDR polymorphisms on BMD

BMD is a key indicator of bone health and osteoporosis risk [2]. Multiple studies have reported that the FokI, BsmI, TaqI, and ApaI polymorphisms significantly impact BMD in different populations, thereby influencing osteoporosis risk. However, these findings have been inconsistent, with some studies revealing significant associations between these polymorphisms and BMD, while others show no such relationship [61,62,63,64,65,66,67,68,69,70,71,72,73,74,75]. This disparity has been attributed to factors such as ethnic diversity and genetic heterogeneity.

Recent advances in genomic research have further shed light on how VDR polymorphisms might interact with environmental factors to influence BMD. For instance, the impact of VDR polymorphisms on BMD may be modulated by VD levels, dietary calcium intake, and sun exposure [78, 79]. Several studies have reported that the association between VDR polymorphisms and BMD becomes more pronounced under conditions of low calcium intake or VD deficiency [80, 81]. Additionally, research has started to unravel the mechanisms through which these polymorphisms might influence BMD, with data suggesting that they can affect the efficiency of VD signaling, and therefore the regulation of calcium and phosphate homeostasis [79,80,81]. However, further research is needed to fully understand the complex interactions between VDR polymorphisms, BMD, and osteoporosis risk.

Interplay between VDR and other key players in bone metabolism

VDR and parathyroid hormone

The interplay between VDR and parathyroid hormone (PTH) is crucial for maintaining calcium homeostasis and bone health [82]. PTH plays a central role in regulating serum calcium levels, while VD and its receptor are involved in calcium absorption and utilization in the body [82]. The VDR is expressed in the parathyroid glands, where it modulates the production and secretion of PTH [83]. PTH acts on target tissues, such as the kidneys and bones, to increase calcium levels [83]. Through its interaction with the VDR, PTH regulates the expression of genes involved in calcium metabolism and bone remodeling [18, 82, 83].

VDR and calcium-sensing receptor

The calcium-sensing receptor (CaSR) is another key player in calcium homeostasis [84]. It is expressed in various tissues, including the parathyroid glands, kidneys, and bones [84]. The CaSR detects changes in extracellular calcium levels and regulates PTH secretion accordingly [85]. The VDR and CaSR have a complex interplay in maintaining calcium balance. VD, acting through the VDR, stimulates intestinal calcium absorption, which indirectly affects the activity of the CaSR [84, 85]. The CaSR, in turn, modulates PTH secretion and calcium reabsorption in the kidneys [84, 85]. Both the VDR and CaSR are involved in feedback mechanisms that regulate PTH production and calcium levels in the body [84, 85].

VDR and estrogen receptor

Estrogen plays a critical role in bone health, and its deficiency contributes to the development of osteoporosis [86]. The estrogen receptor (ER) and VDR have interactions that influence bone metabolism [87, 88]. Estrogen stimulates calcium absorption in the intestines and inhibits bone resorption by osteoclasts [89]. Estrogen and VD signaling pathways can cross-regulate each other, highlighting the interconnectedness between these receptors in maintaining bone health.

Understanding the interplay between the VDR, PTH, CaSR, and ER is crucial for unraveling the complex mechanisms involved in bone metabolism and calcium homeostasis. Dysregulation or disruption of these interactions can lead to imbalances in bone remodeling, impaired calcium absorption, and increased osteoporosis risk. Further research is needed to elucidate the intricate molecular pathways and regulatory mechanisms underlying the interplay between the VDR and other key players in bone metabolism. This knowledge may open new avenues for targeted therapeutic interventions for bone-related disorders, including osteoporosis.

Therapeutic implications of targeting the VD/VDR signaling

VD supplementation in osteoporosis

VD supplementation is a widely used therapeutic approach in the management of osteoporosis [90, 91]. As VD is essential for calcium absorption and bone mineralization, inadequate levels can contribute to bone loss and increased fracture risk [90, 91]. Supplementation with VD aims to optimize serum VD levels and enhance calcium absorption, thereby improving bone health [90, 91].

Clinical studies have shown that VD supplementation, especially when combined with calcium, can reduce the risk of fractures in osteoporotic individuals [90, 91]. The optimal dosage and duration of VD supplementation vary depending on individual factors such as age, sunlight exposure, and baseline VD levels [90, 91]. Regular monitoring of serum VD levels is recommended to ensure adequate supplementation [90, 91].

Selective VDR modulators

Selective vitamin D receptor modulators (VDRMs) (e.g., paricalcitol, eldecalcitol, maxacalcitol, seocalcitol, and lithocholic acid, Fig. 5) are a class of drugs that target the VDR pathway [92, 93]. These compounds interact with the VDR and selectively modulate its activity, offering the potential to enhance bone health without the side effects associated with systemic VD [92, 93].

Fig. 5
figure 5

Chemical structures of VDRMs. Chemical structures of dfferent VDRMs, such as paricalcitol, eldecalcitol, maxacalcitol, seocalcitol, and lithocholic acid, are shown

VDRMs have shown promise in preclinical and clinical studies for their ability to promote bone formation, reduce bone resorption, and improve BMD [94, 95]. These compounds exhibit tissue-selective effects, acting specifically on bone and reducing the risk of hypercalcemia and other adverse effects associated with high-dose VD supplementation [94,95,96]. However, further research is needed to evaluate their long-term efficacy, safety, and potential benefits compared to traditional VD supplementation.

Combination therapies targeting VDR pathway

Combination therapies that target the VDR pathway have gained attention as potential approaches to optimize bone health and reduce fracture risk in osteoporotic individuals. These therapies involve combining VD supplementation or VDRMs with other agents that enhance bone formation or inhibit bone resorption. For example, combining vitamin K, VD supplementation with calcium and bisphosphonates, which inhibit bone resorption, has been shown to have synergistic effects in improving BMD and reducing fracture risk [97, 98]. Other agents, such as denosumab (a monoclonal antibody targeting bone resorption) [99], teriparatide (a parathyroid hormone analog promoting bone formation) [100], and selective estrogen receptor modulators (SERMs) [101], can also be used in combination with VD-based therapies to optimize bone health.

The rationale behind combination therapies is to target multiple aspects of bone metabolism, including calcium absorption, bone formation, and bone resorption, to achieve a comprehensive and synergistic effect on bone health. However, further research is needed to determine the optimal combinations, dosages, and treatment durations, as well as to assess the long-term safety and efficacy of these approaches.

Discrepancies in findings: VD supplementation and fracture risk reduction

In recent years, randomized controlled trials in the general population examining the effects of supplemental VD on fracture outcomes have yielded inconsistent results. Some studies have demonstrated benefits, whereas others showed no significant effects [102,103,104,105,106,107]. Several factors might account for these inconsistent findings, including the use of bolus dosing, limited sample sizes in some studies, co-administration of calcium, and baseline VD levels [102,103,104,105,106,107]. A critical aspect yet to be fully addressed is the potential variability in VDR functionality among the participants. To more comprehensively understand the correlation between VD supplementation and fractures, forthcoming experimental designs should address certain limitations. These encompass the heterogeneity of study populations, ambiguities related to the optimal blood concentration of VD, the unavailability of data on participants’ baseline VD levels, and the absence of insights into genetic variations in the VDR gene.

Conclusion and future perspectives

In this review, we have explored the role of the VD/VDR system in the pathogenesis of osteoporosis. The VDR is a key regulator of bone metabolism, mediating the effects of VD on calcium absorption, bone mineralization, and bone remodeling. Genetic variations in the VDR gene have been associated with osteoporosis susceptibility and BMD. Furthermore, the VDR interacts with other key players in bone metabolism, including the parathyroid hormone, the calcium-sensing receptor, and the estrogen receptor, contributing to the complex regulation of bone homeostasis.

Therapeutically, targeting the VDR pathway has shown promise in the management of osteoporosis [90,91,92,93]. VD supplementation, particularly in combination with calcium, has been widely used to improve bone health and reduce fracture risk. Selective VDR modulators offer a more targeted approach, selectively modulating VDR activity to enhance bone formation and reduce bone resorption [90,91,92,93]. Combination therapies that combine VDR-targeting agents with other bone-modifying drugs have also shown potential synergistic effects [97, 98].

Despite significant advancements in our understanding of the VDR and its role in osteoporosis, several areas warrant further investigation. First, more studies are needed to elucidate the mechanisms underlying the interplay between the VDR and other key players in bone metabolism, such as PTH, CaSR, and ER. Understanding these interactions at a molecular level may uncover novel therapeutic targets for osteoporosis. Furthermore, the influence of genetic variations in the VDR gene on osteoporosis susceptibility and treatment response requires further exploration. Large-scale genetic studies, including genome-wide association studies and functional analyses, can provide insights into the specific VDR polymorphisms and their implications for personalized medicine in osteoporosis. In terms of therapeutics, future research should focus on optimizing the use of VDR-targeting agents, including selective VDR modulators and combination therapies. This includes identifying the most effective combinations, determining optimal dosages and treatment durations, and assessing long-term safety and efficacy in diverse patient populations. Additionally, the potential role of VDR-based therapies in other bone-related conditions, such as osteomalacia and secondary osteoporosis, should be explored. Lastly, the integration of emerging technologies, such as omics approaches and advanced imaging techniques, can enhance our understanding of the VDR pathway and its interactions with other molecular networks in bone metabolism. These technologies can provide a comprehensive and multidimensional view of the molecular mechanisms underlying osteoporosis, paving the way for personalized and precision medicine approaches.

In conclusion, the VDR pathway plays a critical role in the pathogenesis of osteoporosis, and targeting this pathway holds significant therapeutic potential. Continued research efforts are needed to unravel the complexities of the VDR network, explore genetic influences, optimize therapeutic strategies, and leverage emerging technologies. These advancements will contribute to the development of novel treatments and improve outcomes for individuals with osteoporosis in the future.

Availability of data and materials

The data underlying the present study are available on request (corresponding author).

References

  1. Foger-Samwald U, Dovjak P, Azizi-Semrad U, Kerschan-Schindl K, Pietschmann P. Osteoporosis: pathophysiology and therapeutic options. EXCLI J. 2020;19:1017–37.

    PubMed  PubMed Central  Google Scholar 

  2. Sozen T, Ozisik L, Basaran NC. An overview and management of osteoporosis. Eur J Rheumatol. 2017;4(1):46–56.

    Article  PubMed  Google Scholar 

  3. Salari N, Ghasemi H, Mohammadi L, Behzadi MH, Rabieenia E, Shohaimi S, Mohammadi M. The global prevalence of osteoporosis in the world: a comprehensive systematic review and meta-analysis. J Orthop Surg Res. 2021;16(1):609.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Pouresmaeili F, Kamalidehghan B, Kamarehei M, Goh YM. A comprehensive overview on osteoporosis and its risk factors. Ther Clin Risk Manag. 2018;14:2029–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Raterman HG, Bultink IE, Lems WF. Osteoporosis in patients with rheumatoid arthritis: an update in epidemiology, pathogenesis, and fracture prevention. Expert Opin Pharmacother. 2020;21(14):1725–37.

    Article  PubMed  Google Scholar 

  6. De Martinis M, Allegra A, Sirufo MM, Tonacci A, Pioggia G, Raggiunti M, Ginaldi L, Gangemi S. Vitamin D deficiency, osteoporosis and effect on autoimmune diseases and hematopoiesis: a review. Int J Mol Sci. 2021;22(16):8855.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Dominguez LJ, Farruggia M, Veronese N, Barbagallo M. Vitamin D sources, metabolism, and deficiency: available compounds and guidelines for its treatment. Metabolites. 2021;11(4):255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mungai LNW, Mohammed Z, Maina M, Anjumanara O. Vitamin D review: the low hanging fruit for human health. J Nutr Metab. 2021;2021:6335681.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Carlberg C. Vitamin D and its target genes. Nutrients. 2022;14(7):1354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bouillon R, Marcocci C, Carmeliet G, Bikle D, White JH, Dawson-Hughes B, Lips P, Munns CF, Lazaretti-Castro M, Giustina A, Bilezikian J. Skeletal and extraskeletal actions of vitamin D: current evidence and outstanding questions. Endocr Rev. 2019;40(4):1109–51.

    Article  PubMed  Google Scholar 

  11. Christakos S, Dhawan P, Verstuyf A, Verlinden L, Carmeliet G. Vitamin D: metabolism, molecular mechanism of action, and pleiotropic effects. Physiol Rev. 2016;96(1):365–408.

    Article  CAS  PubMed  Google Scholar 

  12. Mu YY, Liu B, Chen B, Zhu WF, Ye XH, Li HZ, He XF. Evaluation of association studies and an updated meta-analysis of VDR polymorphisms in osteoporotic fracture risk. Front Genet. 2021;12:791368.

    Article  CAS  PubMed  Google Scholar 

  13. Bahrami A, Sadeghnia HR, Tabatabaeizadeh SA, Bahrami-Taghanaki H, Behboodi N, Esmaeili H, Esmaeili H, Ferns GA, Mobarhan MG, Avan A. Genetic and epigenetic factors influencing vitamin D status. J Cell Physiol. 2018;233(5):4033–43.

    Article  CAS  PubMed  Google Scholar 

  14. Lee SM, Meyer MB, Benkusky NA, O’Brien CA, Pike JW. Mechanisms of enhancer-mediated hormonal control of vitamin D receptor gene expression in target cells. J Biol Chem. 2015;290(51):30573–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Al Mheid I, Patel RS, Tangpricha V, Quyyumi AA. Vitamin D and cardiovascular disease: Is the evidence solid? Eur Heart J. 2013;34(48):3691–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jeon SM, Shin EA. Exploring vitamin D metabolism and function in cancer. Exp Mol Med. 2018;50(4):1–14.

    PubMed  Google Scholar 

  17. Bikle DD. Vitamin D metabolism, mechanism of action, and clinical applications. Chem Biol. 2014;21(3):319–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Haussler MR, Haussler CA, Whitfield GK, Hsieh JC, Thompson PD, Barthel TK, Bartik L, Egan JB, Wu Y, Kubicek JL, Lowmiller CL, Moffet EW, Forster RE, Jurutka PW. 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(1–2):88–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pike JW, Meyer MB. The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D3. Rheum Dis Clin N Am. 2012;38(1):13–27.

    Article  Google Scholar 

  20. Wan LY, Zhang YQ, Chen MD, Liu CB, Wu JF. Relationship of structure and function of DNA-binding domain in vitamin D receptor. Molecules. 2015;20(7):12389–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Barminko J, Reinholt BM, Emmanuelli A, Lejeune AN, Baron MH. Activation of the vitamin D receptor transcription factor stimulates the growth of definitive erythroid progenitors. Blood Adv. 2018;2(11):1207–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rochel N. Vitamin D and its receptor from a structural perspective. Nutrients. 2022;14(14):2847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Haussler MR, Livingston S, Sabir ZL, Haussler CA, Jurutka PW. 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(1):e10432.

    Article  CAS  PubMed  Google Scholar 

  24. Pike JW, Christakos S. Biology and mechanisms of action of the vitamin D hormone. Endocrinol Metab Clin N Am. 2017;46(4):815–43.

    Article  Google Scholar 

  25. Pike JW, Meyer MB, Lee SM, Onal M, Benkusky NA. The vitamin D receptor: contemporary genomic approaches reveal new basic and translational insights. J Clin Invest. 2017;127(4):1146–54.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Costa PLF, Franca MM, Katayama ML, Carneiro ET, Martin RM, Folgueira MAK, Latronico AC, Ferraz-de-Souza B. Transcriptomic response to 1,25-dihydroxy vitamin D in human fibroblasts with or without a functional vitamin D receptor (VDR): novel target genes and insights into VDR basal transcriptional activity. Cells. 2019;8(4):318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Donati S, Palmini G, Aurilia C, Falsetti I, Miglietta F, Iantomasi T, Brandi ML. Rapid nontranscriptional effects of calcifediol and calcitriol. Nutrients. 2022;14(6):1291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zmijewski MA. Nongenomic activities of vitamin D. Nutrients. 2022;14(23):5104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Irazoqui AP, Boland RL, Buitrago CG. Actions of 1,25(OH)2-vitamin D3 on the cellular cycle depend on VDR and p38 MAPK in skeletal muscle cells. J Mol Endocrinol. 2014;53(3):331–43.

    Article  CAS  PubMed  Google Scholar 

  30. Zenata O, Vrzal R. Fine tuning of vitamin D receptor (VDR) activity by post-transcriptional and post-translational modifications. Oncotarget. 2017;8(21):35390–402.

    Article  PubMed  PubMed Central  Google Scholar 

  31. van Driel M, Pols HA, van Leeuwen JP. Osteoblast differentiation and control by vitamin D and vitamin D metabolites. Curr Pharm Des. 2004;10(21):2535–55.

    Article  PubMed  Google Scholar 

  32. Xu D, Gao HJ, Lu CY, Tian HM, Yu XJ. Vitamin D inhibits bone loss in mice with thyrotoxicosis by activating the OPG/RANKL and Wnt/beta-catenin signaling pathways. Front Endocrinol (Lausanne). 2022;13:1066089.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Haussler MR, Jurutka PW, Mizwicki M, Norman AW. Vitamin D receptor (VDR)-mediated actions of 1alpha,25(OH)(2)vitamin D(3): genomic and non-genomic mechanisms. Best Pract Res Clin Endocrinol Metab. 2011;25(4):543–59.

    Article  CAS  PubMed  Google Scholar 

  34. Chen Y, Zhang J, Ge X, Du J, Deb DK, Li YC. Vitamin D receptor inhibits nuclear factor kappaB activation by interacting with IkappaB kinase beta protein. J Biol Chem. 2013;288(27):19450–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chen Z, Huang D, Yongyut P, Li G, Esteban MA, Jintasataporn O, Deng J, Zhang W, Ai Q, Mai K, Zhang Y. Vitamin D(3) deficiency induced intestinal inflammatory response of turbot through nuclear factor-kappaB/inflammasome pathway, accompanied by the mutually exclusive apoptosis and autophagy. Front Immunol. 2022;13:986593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. He L, Liu T, Shi Y, Tian F, Hu H, Deb DK, Chen Y, Bissonnette M, Li YC. Gut epithelial vitamin D receptor regulates microbiota-dependent mucosal inflammation by suppressing intestinal epithelial cell apoptosis. Endocrinology. 2018;159(2):967–79.

    Article  CAS  PubMed  Google Scholar 

  37. Szeto FL, Sun J, Kong J, Duan Y, Liao A, Madara JL, Li YC. Involvement of the vitamin D receptor in the regulation of NF-kappaB activity in fibroblasts. J Steroid Biochem Mol Biol. 2007;103(3–5):563–6.

    Article  CAS  PubMed  Google Scholar 

  38. Ge X, Wang L, Li M, Xu N, Yu F, Yang F, Li R, Zhang F, Zhao B, Du J. Vitamin D/VDR signaling inhibits LPS-induced IFNgamma and IL-1beta in Oral epithelia by regulating hypoxia-inducible factor-1alpha signaling pathway. Cell Commun Signal. 2019;17(1):18.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kongsbak M, Levring TB, Geisler C, von Essen MR. The vitamin d receptor and T cell function. Front Immunol. 2013;4:148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fernandez Lahore G, Raposo B, Lagerquist M, Ohlsson C, Sabatier P, Xu B, Aoun M, James J, Cai X, Zubarev RA, Nandakumar KS, Holmdahl R. Vitamin D3 receptor polymorphisms regulate T cells and T cell-dependent inflammatory diseases. Proc Natl Acad Sci U S A. 2020;117(40):24986–97.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Saccone D, Asani F, Bornman L. Regulation of the vitamin D receptor gene by environment, genetics and epigenetics. Gene. 2015;561(2):171–80.

    Article  CAS  PubMed  Google Scholar 

  42. Kim JM, Lin C, Stavre Z, Greenblatt MB, Shim JH. Osteoblast–osteoclast communication and bone homeostasis. Cells. 2020;9(9):2073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bikle DD. Vitamin D and bone. Curr Osteoporos Rep. 2012;10(2):151–9.

    Article  PubMed  PubMed Central  Google Scholar 

  44. van de Peppel J, van Leeuwen JP. Vitamin D and gene networks in human osteoblasts. Front Physiol. 2014;5:137.

    PubMed  PubMed Central  Google Scholar 

  45. Atkins GJ, Anderson PH, Findlay DM, Welldon KJ, Vincent C, Zannettino AC, O’Loughlin PD, Morris HA. Metabolism of vitamin D3 in human osteoblasts: evidence for autocrine and paracrine activities of 1 alpha,25-dihydroxyvitamin D3. Bone. 2007;40(6):1517–28.

    Article  CAS  PubMed  Google Scholar 

  46. Kogawa M, Findlay DM, Anderson PH, Ormsby R, Vincent C, Morris HA, Atkins GJ. Osteoclastic metabolism of 25(OH)-vitamin D3: a potential mechanism for optimization of bone resorption. Endocrinology. 2010;151(10):4613–25.

    Article  CAS  PubMed  Google Scholar 

  47. 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(4):771–7.

    Article  CAS  PubMed  Google Scholar 

  48. van Driel M, Koedam M, Buurman CJ, Roelse M, Weyts F, Chiba H, Uitterlinden AG, Pols HA, van Leeuwen JP. Evidence that both 1alpha,25-dihydroxyvitamin D3 and 24-hydroxylated D3 enhance human osteoblast differentiation and mineralization. J Cell Biochem. 2006;99(3):922–35.

    Article  PubMed  Google Scholar 

  49. van Leeuwen JP, van Driel M, van den Bemd GJ, Pols HA. Vitamin D control of osteoblast function and bone extracellular matrix mineralization. Crit Rev Eukaryot Gene Expr. 2001;11(1–3):199–226.

    PubMed  Google Scholar 

  50. 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(6):1297–308.

    Article  CAS  PubMed  Google Scholar 

  51. Shymanskyi I, Lisakovska O, Mazanova A, Labudzynskyi D, Veliky M. Vitamin D(3) modulates impaired crosstalk between RANK and glucocorticoid receptor signaling in bone marrow cells after chronic prednisolone administration. Front Endocrinol (Lausanne). 2018;9:303.

    Article  PubMed  Google Scholar 

  52. Yamamoto Y, Yoshizawa T, Fukuda T, Shirode-Fukuda Y, Yu T, Sekine K, Sato T, Kawano H, Aihara K, Nakamichi Y, Watanabe T, Shindo M, Inoue K, Inoue E, Tsuji N, Hoshino M, Karsenty G, Metzger D, Chambon P, Kato S, Imai Y. Vitamin D receptor in osteoblasts is a negative regulator of bone mass control. Endocrinology. 2013;154(3):1008–20.

    Article  CAS  PubMed  Google Scholar 

  53. Florencio-Silva R, Sasso GR, Sasso-Cerri E, Simoes MJ, Cerri PS. Biology of bone tissue: structure, function, and factors that influence bone cells. Biomed Res Int. 2015;2015:421746.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Zarei A, Morovat A, Javaid K, Brown CP. Vitamin D receptor expression in human bone tissue and dose-dependent activation in resorbing osteoclasts. Bone Res. 2016;4:16030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Menaa C, Barsony J, Reddy SV, Cornish J, Cundy T, Roodman GD. 1,25-Dihydroxyvitamin D3 hypersensitivity of osteoclast precursors from patients with Paget’s disease. J Bone Miner Res. 2000;15(2):228–36.

    Article  CAS  PubMed  Google Scholar 

  56. Deng J, Yang Y, He J, Xie Z, Luo F, Xu J, et al. Vitamin D receptor activated by vitamin D administration alleviates Mycobacterium tuberculosis-induced bone destruction by inhibiting NFkappaB-mediated aberrant osteoclastogenesis. FASEB J. 2021;35(6):e21543.

    Article  CAS  PubMed  Google Scholar 

  57. Taymans SE, Pack S, Pak E, Orban Z, Barsony J, Zhuang Z, et al. The human vitamin D receptor gene (VDR) is localized to region 12cen-q12 by fluorescent in situ hybridization and radiation hybrid mapping: genetic and physical VDR map. J Bone Miner Res. 1999;14(7):1163–6.

    Article  CAS  PubMed  Google Scholar 

  58. Penna-Martinez M, Badenhoop K. Inherited variation in vitamin D genes and type 1 diabetes predisposition. Genes (Basel). 2017;8(4):125.

    Article  PubMed  Google Scholar 

  59. Kostner K, Denzer N, Koreng M, Reichrath S, Graber S, Klein R, Tilgen W, Vogt T, Reichrath J. Association of genetic variants of the vitamin D receptor (VDR) with cutaneous squamous cell carcinomas (SCC) and basal cell carcinomas (BCC): a pilot study in a German population. Anticancer Res. 2012;32(1):327–33.

    PubMed  Google Scholar 

  60. Santoro D, Lucisano S, Gagliostro G, Alibrandi A, Benvenga S, Ientile R, Bellinghieri G, Buemi M, Caccamo D. Vitamin D receptor polymorphism in chronic kidney disease patients with complicated cardiovascular disease. J Ren Nutr. 2015;25(2):187–93.

    Article  CAS  PubMed  Google Scholar 

  61. Ansari MGA, Mohammed AK, Wani KA, Hussain SD, Alnaami AM, Abdi S, Aljohani NJ, Al-Daghri NM. Vitamin D receptor gene variants susceptible to osteoporosis in arab post-menopausal women. Curr Issues Mol Biol. 2021;43(3):1325–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Banjabi AA, Al-Ghafari AB, Kumosani TA, Kannan K, Fallatah SM. Genetic influence of vitamin D receptor gene polymorphisms on osteoporosis risk. Int J Health Sci (Qassim). 2020;14(4):22–8.

    PubMed  PubMed Central  Google Scholar 

  63. Becherini L, Gennari L, Masi L, Mansani R, Massart F, Morelli A, Falchetti A, Gonnelli S, Fiorelli G, Tanini A, Brandi ML. Evidence of a linkage disequilibrium between polymorphisms in the human estrogen receptor alpha gene and their relationship to bone mass variation in postmenopausal Italian women. Hum Mol Genet. 2000;9(13):2043–50.

    Article  CAS  PubMed  Google Scholar 

  64. Boschitsch E, Suk EK, Mayr WR, Lang T, Schwartz WM, Panzer S. Genotypes of the vitamin-D-receptor gene and bone mineral density in Caucasoid postmenopausal females. Maturitas. 1996;24(1–2):91–6.

    CAS  PubMed  Google Scholar 

  65. Jiang LL, Zhang C, Zhang Y, Ma F, Guan Y. Associations between polymorphisms in VDR gene and the risk of osteoporosis: a meta-analysis. Arch Physiol Biochem. 2022;128(6):1637–44.

    Article  CAS  PubMed  Google Scholar 

  66. Kim SW, Lee JM, Ha JH, Kang HH, Rhee CK, Kim JW, Moon HS, Baek KH, Lee SH. Association between vitamin D receptor polymorphisms and osteoporosis in patients with COPD. Int J Chron Obstr Pulm Dis. 2015;10:1809–17.

    Article  CAS  Google Scholar 

  67. Kow M, Akam E, Singh P, Singh M, Cox N, Bhatti JS, Tuck SP, Francis RM, Datta H, Mastana S. Vitamin D receptor (VDR) gene polymorphism and osteoporosis risk in White British men. Ann Hum Biol. 2019;46(5):430–3.

    Article  PubMed  Google Scholar 

  68. Lau EM, Lam V, Li M, Ho K, Woo J. Vitamin D receptor start codon polymorphism (Fok I) and bone mineral density in Chinese men and women. Osteoporos Int. 2002;13(3):218–21.

    Article  CAS  PubMed  Google Scholar 

  69. Li Y, Xi B, Li K, Wang C. Association between vitamin D receptor gene polymorphisms and bone mineral density in Chinese women. Mol Biol Rep. 2012;39(5):5709–17.

    Article  CAS  PubMed  Google Scholar 

  70. Liao JL, Qin Q, Zhou YS, Ma RP, Zhou HC, Gu MR, Feng YP, Wang BY, Yang L. Vitamin D receptor Bsm I polymorphism and osteoporosis risk in postmenopausal women: a meta-analysis from 42 studies. Genes Nutr. 2020;15(1):20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Pedrera-Canal M, Moran JM, Vera V, Roncero-Martin R, Lavado-Garcia JM, Aliaga I, Pedrera-Zamorano JDI. Lack of influence of vitamin D receptor BsmI (rs1544410) polymorphism on the rate of bone loss in a cohort of postmenopausal Spanish women affected by osteoporosis and followed for five years. PLoS ONE. 2015;10(9):e0138606.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Techapatiphandee M, Tammachote N, Tammachote R, Wongkularb A, Yanatatsaneejit P. VDR and TNFSF11 polymorphisms are associated with osteoporosis in Thai patients. Biomed Rep. 2018;9(4):350–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Wang S, Ai Z, Song M, Yan P, Li J, Wang S. The association between vitamin D receptor FokI gene polymorphism and osteoporosis in postmenopausal women: a meta-analysis. Climacteric. 2021;24(1):74–9.

    Article  CAS  PubMed  Google Scholar 

  74. Yu M, Chen GQ, Yu F. Lack of association between vitamin D receptor polymorphisms ApaI (rs7975232) and BsmI (rs1544410) and osteoporosis among the Han Chinese population: A meta-analysis. Kaohsiung J Med Sci. 2016;32(12):599–606.

    Article  PubMed  Google Scholar 

  75. Zintzaras E, Rodopoulou P, Koukoulis GN. BsmI, TaqI, ApaI and FokI polymorphisms in the vitamin D receptor (VDR) gene and the risk of osteoporosis: a meta-analysis. Dis Markers. 2006;22(5–6):317–26.

    Article  CAS  PubMed  Google Scholar 

  76. Casado-Diaz A, Cuenca-Acevedo R, Navarro-Valverde C, Diaz-Molina C, Caballero-Villarraso J, Santiago-Mora R, Dorado G, Quesada-Gomez JM. Vitamin D status and the Cdx-2 polymorphism of the vitamin D receptor gene are determining factors of bone mineral density in young healthy postmenopausal women. J Steroid Biochem Mol Biol. 2013;136:187–9.

    Article  CAS  PubMed  Google Scholar 

  77. He W, Liu M, Huang X, Qing Z, Gao W. The influence of vitamin D receptor genetic variants on bone mineral density and osteoporosis in Chinese postmenopausal women. Dis Markers. 2015;2015:760313.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Ferrari S, Rizzoli R, Manen D, Slosman D, Bonjour JP. Vitamin D receptor gene start codon polymorphisms (FokI) and bone mineral density: interaction with age, dietary calcium, and 3′-end region polymorphisms. J Bone Min Res. 1998;13(6):925–30.

    Article  CAS  Google Scholar 

  79. Uitterlinden AG, Fang Y, Van Meurs JB, Pols HA, Van Leeuwen JP. Genetics and biology of vitamin D receptor polymorphisms. Gene. 2004;338(2):143–56.

    Article  CAS  PubMed  Google Scholar 

  80. Al Anouti F, Taha Z, Shamim S, Khalaf K, Al Kaabi L, Alsafar H. An insight into the paradigms of osteoporosis: from genetics to biomechanics. Bone Rep. 2019;11:100216.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Morrison NA, Qi JC, Tokita A, Kelly PJ, Crofts L, Nguyen TV, Sambrook PN, Eisman JA. Prediction of bone density from vitamin D receptor alleles. Nature. 1994;367(6460):284–7.

    Article  CAS  PubMed  Google Scholar 

  82. Veldurthy V, Wei R, Oz L, Dhawan P, Jeon YH, Christakos S. Vitamin D, calcium homeostasis and aging. Bone Res. 2016;4:16041.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Latic N, Erben RG. Interaction of vitamin D with peptide hormones with emphasis on parathyroid hormone, FGF23, and the renin-angiotensin-aldosterone system. Nutrients. 2022;14(23):5186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Iamartino L, Brandi ML. The calcium-sensing receptor in inflammation: Recent updates. Front Physiol. 2022;13:1059369.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Goodman WG. Calcium-sensing receptors. Semin Nephrol. 2004;24(1):17–24.

    Article  CAS  PubMed  Google Scholar 

  86. Emmanuelle NE, Marie-Cecile V, Florence T, Jean-Francois A, Francoise L, Coralie F, Alexia V. Critical role of estrogens on bone homeostasis in both male and female: from physiology to medical implications. Int J Mol Sci. 2021;22(4):1568.

    Article  PubMed  Google Scholar 

  87. Brown MA, Haughton MA, Grant SF, Gunnell AS, Henderson NK, Eisman JA. Genetic control of bone density and turnover: role of the collagen 1alpha1, estrogen receptor, and vitamin D receptor genes. J Bone Min Res. 2001;16(4):758–64.

    Article  CAS  Google Scholar 

  88. Long J, Liu P, Zhang Y, Shen H, Liu Y, Dvornyk V, Deng HW. Interaction effects between estrogen receptor alpha gene, vitamin D receptor gene, age, and sex on bone mineral density in Chinese. J Hum Genet. 2003;48(10):514–9.

    Article  CAS  PubMed  Google Scholar 

  89. Mills EG, Yang L, Nielsen MF, Kassem M, Dhillo WS, Comninos AN. The relationship between bone and reproductive hormones beyond estrogens and androgens. Endocr Rev. 2021;42:691–719.

    Article  PubMed  PubMed Central  Google Scholar 

  90. LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sunyecz JA. The use of calcium and vitamin D in the management of osteoporosis. Ther Clin Risk Manag. 2008;4(4):827–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Campbell MJ, Adorini L. The vitamin D receptor as a therapeutic target. Expert Opin Ther Targets. 2006;10(5):735–48.

    Article  CAS  PubMed  Google Scholar 

  93. Lason W, Jantas D, Leskiewicz M, Regulska M, Basta-Kaim A. The vitamin D receptor as a potential target for the treatment of age-related neurodegenerative diseases such as Alzheimer’s and Parkinson’s diseases: a narrative review. Cells. 2023;12(4):660.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Appelman-Dijkstra NM, Papapoulos SE. Modulating bone resorption and bone formation in opposite directions in the treatment of postmenopausal osteoporosis. Drugs. 2015;75(10):1049–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Plum LA, DeLuca HF. Vitamin D, disease and therapeutic opportunities. Nat Rev Drug Discov. 2010;9(12):941–55.

    Article  CAS  PubMed  Google Scholar 

  96. Noguchi Y, Kawate H, Nomura M, Takayanagi R. Eldecalcitol for the treatment of osteoporosis. Clin Interv Aging. 2013;8:1313–21.

    PubMed  PubMed Central  Google Scholar 

  97. Moore AE, Dulnoan D, Voong K, Ayis S, Mangelis A, Gorska R, Harrington DJ, Tang JCY, Fraser WD, Hampson G. The additive effect of vitamin K supplementation and bisphosphonate on fracture risk in post-menopausal osteoporosis: a randomised placebo controlled trial. Arch Osteoporos. 2023;18(1):83.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Golden NH, Abrams SA. Committee on N. Optimizing bone health in children and adolescents. Pediatrics. 2014;134:e122943.

    Article  Google Scholar 

  99. Bone HG, Bolognese MA, Yuen CK, Kendler DL, Miller PD, Yang YC, et al. Effects of denosumab treatment and discontinuation on bone mineral density and bone turnover markers in postmenopausal women with low bone mass. J Clin Endocrinol Metab. 2011;96(4):972–80.

    Article  CAS  PubMed  Google Scholar 

  100. Gomberg SJ, Wustrack RL, Napoli N, Arnaud CD, Black DM. Teriparatide, vitamin D, and calcium healed bilateral subtrochanteric stress fractures in a postmenopausal woman with a 13-year history of continuous alendronate therapy. J Clin Endocrinol Metab. 2011;96(6):1627–32.

    Article  CAS  PubMed  Google Scholar 

  101. Liu HJ, Kim SA, Shim DJ, Jung JM, Lee EJ. Influence of supplementary vitamin D on bone mineral density when used in combination with selective estrogen receptor modulators. J Menopausal Med. 2019;25(2):94–9.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Shapses SA, Kendler DL, Robson R, Hansen KE, Sherrell RM, Field MP, Woolf E, Berd Y, Mantz AM, Santora AC. Effect of alendronate and vitamin D(3) on fractional calcium absorption in a double-blind, randomized, placebo-controlled trial in postmenopausal osteoporotic women. J Bone Miner Res. 2011;26(8):1836–44.

    Article  CAS  PubMed  Google Scholar 

  103. Bischoff-Ferrari HA, Vellas B, Rizzoli R, Kressig RW, da Silva JAP, Blauth M, Felson DT, McCloskey EV, Watzl B, Hofbauer LC, Felsenberg D, Willett WC, Dawson-Hughes B, Manson JE, Siebert U, Theiler R, Staehelin HB, DO-HEALTH Research Group. Effect of vitamin D supplementation, omega-3 fatty acid supplementation, or a strength-training exercise program on clinical outcomes in older adults: the DO-HEALTH randomized clinical trial. JAMA. 2020;324(18):1855–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Sanders KM, Stuart AL, Williamson EJ, Simpson JA, Kotowicz MA, Young D, Nicholson GC. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA. 2010;303(18):1815–22.

    Article  CAS  PubMed  Google Scholar 

  105. Khaw KT, Stewart AW, Waayer D, Lawes CMM, Toop L, Camargo CA Jr, Scragg R. Effect of monthly high-dose vitamin D supplementation on falls and non-vertebral fractures: secondary and post-hoc outcomes from the randomised, double-blind, placebo-controlled ViDA trial. Lancet Diabetes Endocrinol. 2017;5(6):438–47.

    Article  CAS  PubMed  Google Scholar 

  106. Jackson RD, LaCroix AZ, Gass M, Wallace RB, Robbins J, Lewis CE, Bassford T, Beresford SA, Black HR, Blanchette P, Bonds DE, Brunner RL, Brzyski RG, Caan B, Cauley JA, Chlebowski RT, Cummings SR, Granek I, Hays J, Heiss G, Hendrix SL, Howard BV, Hsia J, Hubbell FA, Johnson KC, Judd H, Kotchen JM, Kuller LH, Langer RD, Lasser NL, Limacher MC, Ludlam S, Manson JE, Margolis KL, McGowan J, Ockene JK, O’Sullivan MJ, Phillips L, Prentice RL, Sarto GE, Stefanick ML, Van Horn L, Wactawski-Wende J, Whitlock E, Anderson GL, Assaf AR, Barad D, Women’s Health Initiative Investigators. Calcium plus vitamin D supplementation and the risk of fractures. N Engl J Med. 2006;354(7):669–83.

    Article  CAS  PubMed  Google Scholar 

  107. LeBoff MS, Chou SH, Ratliff KA, Cook NR, Khurana B, Kim E, Cawthon PM, Bauer DC, Black D, Gallagher JC, Lee IM, Buring JE, Manson JE. Supplemental vitamin D and incident fractures in midlife and older adults. N Engl J Med. 2022;387(4):299–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge the contributions of all the researchers, scientists, and clinicians whose work has significantly contributed to our understanding of VDR and its role in osteoporosis. Their dedication and commitment to advancing knowledge in this field are greatly appreciated.

Funding

This work is supported by the grant of the High-end Foreign Expert Program of the Ministry of Science and Technology of China (G2021003005L) and the Hebei Provincial Science and Technology Plan Project (223777118D).

Author information

Authors and Affiliations

Authors

Contributions

GL, KL, and YT conceived the structures of the article and revised the manuscript. YL searched the publications and wrote the manuscript. PZ, BJ, KL, LZ, and HW revised the manuscript.

Corresponding authors

Correspondence to Yansheng Tian, Kun Li or Guoqi Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Zhao, P., Jiang, B. et al. Modulation of the vitamin D/vitamin D receptor system in osteoporosis pathogenesis: insights and therapeutic approaches. J Orthop Surg Res 18, 860 (2023). https://doi.org/10.1186/s13018-023-04320-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13018-023-04320-4

Keywords