Next Article in Journal
Chronic Alcohol Exposure Promotes Cancer Stemness and Glycolysis in Oral/Oropharyngeal Squamous Cell Carcinoma Cell Lines by Activating NFAT Signaling
Next Article in Special Issue
The Role of Vitamin D and Vitamin D Binding Protein in Chronic Liver Diseases
Previous Article in Journal
Phospholipid Scramblase 4 (PLSCR4) Regulates Adipocyte Differentiation via PIP3-Mediated AKT Activation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases

1
2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
2
2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
3
Rheumatology Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania
4
Rheumatology Discipline, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(17), 9784; https://doi.org/10.3390/ijms23179784
Submission received: 23 July 2022 / Revised: 24 August 2022 / Accepted: 26 August 2022 / Published: 29 August 2022
(This article belongs to the Special Issue Vitamin D and Vitamin D Binding Protein in Health and Disease 2.0)

Abstract

:
Vitamin D intervenes in calcium and phosphate metabolism and bone homeostasis. Experimental studies have shown that 1,25-dihydroxyvitamin D (calcitriol) generates immunologic activities on the innate and adaptive immune system and endothelial membrane stability. Low levels of serum 25-hydroxyvitamin D (25(OH)D) are associated with an increased risk of developing immune-related diseases such as psoriasis, type 1 diabetes, multiple sclerosis, and autoimmune diseases. Various clinical trials describe the efficacy of supplementation of vitamin D and its metabolites for treating these diseases that result in variable outcomes. Different disease outcomes are observed in treatment with vitamin D as high inter-individual difference is present with complex gene expression in human peripheral blood mononuclear cells. However, it is still not fully known what level of serum 25(OH)D is needed. The current recommendation is to increase vitamin D intake and have enough sunlight exposure to have serum 25(OH)D at a level of 30 ng/mL (75 nmol/L) and better at 40–60 ng/mL (100–150 nmol/L) to obtain the optimal health benefits of vitamin D.

1. Introduction

The role of vitamin D in health was first defined by its deficiency that results in rickets in children and osteomalacia in adults [1]. Vitamin D was first described in the early 1600s [2], and despite its name, it is a prohormone because humans do not obtain it only from their diet. Vitamin D is produced after exposure to ultraviolet B radiation (wavelength 290–315 nm) and can also be obtained from diet and supplements [3].
Vitamin D (calciferol) has two forms D2 (ergocalciferol) and D3 (cholecalciferol) [4]. After exposure to ultraviolet B radiation, cholecalciferol is produced in the skin from the precursor protein 7-dehydrocholesterol. The main supply of vitamin D derives from the skin; the diet only contributes in a small amount. The proteins involved in the transport of vitamin D are albumin and lipoproteins, most of vitamin D being transported in an inactive form by D-binding protein (DBP) to the liver. DBP also has an immunomodulatory role and intervenes in bone development [1,3,4,5]. In the liver, vitamin D is converted to 25-hydroxyvitamin D (25(OH)D), an intermediate inactive form in the presence of 25-hydroxylase encoded by the CYP2R1 allele [6]. The 25(OH)D is then transported by DBP to the kidney and converted to the active form, also called calcitriol, in the presence of 1α-hydroxylase encoded by the CYP27B1 allele [6]. This enzyme can be found in various cell types (i.e., skin, bone cells, immune cells, placenta), but the highest concentration is expressed in the proximal tubule in the kidney [1,3,4,5]. In the kidney, hydroxylation is controlled by parathyroid hormone (PTH), calcium and phosphate levels [7]. After passing through the cell membrane, 1,25-hydroxyvitamin D forms the 1,25-hydroxyvitamin D—vitamin D receptor (VDR) complex that modulates gene expression in the nucleus [7]. The outcome of this interaction is calcium homeostasis which inflects intestinal calcium absorption. In the case of low 1,25-hydroxyvitamin D levels, calcium will be absorbed from the bone in favor of the intestine [8]. Breakdown of both 25(OH)D and 1,25-hydroxyvitamin D is performed by the same 24-hydroxylation enzyme (CYP24A1) [1,3,4,5].
There are alternative pathways of vitamin D activation different from 25(OH)D and 1,25-hydroxyvitamin D. This novel in vivo pathway of vitamin D metabolism initiated by P450sc includes: 20-hydroxyvitamin D3 [20(OH)D3], 22(OH)D3, 20,23(OH)2D3, 20,22(OH)2D3, 1,20(OH)2D3, 1,20,23(OH)3D3, and 17,20,23(OH)3D3. These novel metabolites are produced by the placenta, adrenal glands and, at low levels, by epidermal keratinocytes. The predominant human in vivo production of 20(OH)D3 is approximately 20 times more reduced than 25(OH)D. The role of cytochrome P450scc (CYP11A1) and CYP27B1 enzymes in vitamin D metabolism was demonstrated in studies using isolated mitochondria and purified enzymes [9,10]. In addition, CYP11A1-derived secosteroids produced in vivo in the skin, serum and adrenal gland manifest their biological activity as hormones [11]. In the placenta and adrenal glands, the main pathway comprised 20(OH)D3, 20,23(OH)2D3, 17,20,23(OH)3D3, and minor pathways included 25(OH)D and 1,25(OH)2D, and 22(OH)D3 and 20,22(OH)2D3. In epidermal keratinocytes, the predominant metabolites were 22(OH)D3 and 20,22(OH)2D3 [9].
Similarly, human placentas, rat and bovine adrenal glands, human epidermal keratinocytes, and colon cancer Caco-2 cells can produce alternative pathways of ergocalciferol. The novel hydroxy-derivatives include 20(OH)D2, 17,20(OH)2D2, 1,20(OH)2D2, 25(OH)D2 and 1,25(OH)2D2 with high CYP11A1 and CYP27B1 expression. These pathway products present tissue- and cell-type specificity with a higher production of 25(OH)D2 in placentas and Caco-2 cells, and 20(OH)D2 and 25(OH)D2 production in human keratinocytes [12].
CYP11A1 can convert vitamin D into the non-calcemic analog 20S-hydroxyvitamin D3 [20S(OH)D3]. This analog could be used as a potential treatment for rheumatoid arthritis (RA) and other autoimmune disorders by suppressing clinical signs of arthritis and repairing joint damage in a mouse model. 20S(OH)D3 can also decrease the number of CD4 and CD19 cells, reducing inflammatory cytokines. Arthritis can be attenuated by 20S(OH)D3 by reducing pro-inflammatory cytokines and antibodies against type II collagen [13].
Alternative nuclear receptors for CYP11A1-derived vitamin D-hydroxy-derivatives comprise the retinoid-related orphan receptors (ROR)α and γ [14], the arylhydrocarbon receptor (AhR) [15], and liver X receptors α and β [16,17]. Notably, CYP11A1 is expressed in the immune system, in human [18] and murine [19] T cells, in human CD4, CD8, B cells and monocytes [20]. CYP11A1 is involved in the local production of steroidogenesis in the skin and the specific vitamin D metabolism, lumisterol, and 7-dehydrocholesterol. CYP11A1 metabolites play a role in the protective barrier and skin immune functions. Malfunction of CYP11A1 can result in skin disorders [21]. CYP11A1 hydroxy-derivatives and lumisterol hydroxy-derivatives present photoprotective effects by stimulating intracellular free radical scavenging and DNA repair. As photoprotective agents in human keratinocytes, CYP11A1 hydroxy-derivatives are involved in p53-phosphorylation, in the antioxidant response regulated by NRF2, and induction of DNA repair [17].
Human epidermis and serum production of a photoproduct of pre-vitamin D, CYP11A1 and CYP27A1 hydroxylate tachysterol3 produce 20S-hydroxytachysterol3 [20S(OH)T3] and 25(OH)T3. These metabolites can inhibit the proliferation of epidermal keratinocytes and dermal fibroblasts similar to 1,25-dihydroxyvitamin D resulting in stimulation of the differentiation and anti-oxidative genes in keratinocytes. They attach to VDR, express CYP24A1, activate AhR, and bind to the ligan-binding domain (LBD) of LXR α and β, and the peroxisome proliferator-activated receptor γ (PPARγ). The biological function of tachysterol3 is demonstrated by endogenous production of 20S(OH)T3 and 25(OH)T3, which can interact with VDR, AhR, LXRs, and PPARγ [22].
VDR is associated with the nuclear receptor superfamily. Ligand-receptor dimerization is established with the retinoic X receptor (RXR). This complex attaches to the vitamin D responsive elements (VDREs), inducing gene expression by targeting promoter sequences of calcitriol, underlining its genomic function [23,24,25]. The non-genomic effect of vitamin D is highlighted by the connection of calcitriol to a membrane-bound to VDR and caveolin-1 [26].
After activation, vitamin D binds to calcium transporting proteins in the small intestine triggering calcium absorption [26,27]. Vitamin D stimulates osteoclasts leading to bone resorption and increased blood calcium [28]. In bone growth, vitamin D supports collagen matrices and osteoblasts mineralization [29]. Vitamin D induces the expression of osteocalcin, an important non-collagenous bone protein, and stimulates bone resorption RANKL (receptor activator of nuclear factor kappaB ligand), a TNF family member [3,30,31,32]. Vitamin D with parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) regulate calcium and phosphate homeostasis [33]. The negative feedback consists of vitamin D directly inhibiting PTH production, which decreases bone resorption and increases urinary calcium excretion. In this loop, osteocytes induce FGF23 production, increasing urinary phosphate excretion [30,33,34].
The clinically significant reserves of vitamin D are serum 25(OH)D levels (defined as the sum of ergocalciferol and 25(OH)D). In contrast, calcitriol has a short half-life (4–8 h) and depends on calcium homeostasis. An important debate is the proper standardization of tests [35]. Two different methodologies are used: competitive immunoassays (radio-immunoassays or binding-protein assays) and procedures that use high-performance liquid chromatography and liquid chromatography tandem-mass spectrometry. The latter methods are the gold standard procedures [28,36,37].
The Clinical Guidelines Subcommittee of The Endocrine Society defines the deficiency of 25(OH)D to be less than 20 ng/mL (50 nmol/L) [38], while the Institute of Medicine (US) Committee to Review Dietary Reference Intakes for Vitamin D and Calcium defined it to be less than 12 ng/mL (30 nmol/L) [39]. The first society defined insufficiency at levels of 21–29 ng/mL (52 to 72 nmol/L) [38], while the latter at levels of 12–20 ng/mL (30–50 nmol/L) [39]. Vitamin D intoxication is described at values higher than 150 ng/mL (374 nmol/L) [3].
Considering these values, a study based on 6275 American children and adolescents revealed that 61% presented 25(OH)D insufficiency and 9% deficiency [40]. A high prevalence (up to 40%) of vitamin D insufficiency and 6% deficiency is described in adults [41,42,43]. The first internationally standardized dataset of vitamin D status across a latitude gradient of 35° N to 69° N that enrolled almost 56,000 individuals reported that 13% of EU participants had serum 25(OH)D < 30 nmol/L and 40% presented levels < 50 nmol/L [44,45]. Although a trend is described toward lower 25(OH)D levels in the population [45], the current recommendation is not population-wide screening for vitamin D deficiency but testing the individuals that are at risk of developing vitamin D deficiency, such as patients suffering from hyper- and hypoparathyroidism, osteoporosis, and kidney disease [38,46,47,48].
There is a direct relationship between the dose of vitamin D and serum 25(OH)D with a linear curve at a daily dose of 1000–2000 IU (25–50 μg), but this curve flattens at higher doses [48]. A daily intake of 1040 IU (26 μg) in vitamin D deficiency and 400 IU (10 μg) in vitamin D insufficiency is needed to obtain a concentration above 20 ng/mL (50 nmol/L) in 97.5% of the population [49]. Several studies have tried finding the optimal vitamin D dose to correct the deficiency. These studies concluded that most substitutions resulted in adequate 25(OH)D levels regardless of the interval or dosing regimen. Higher loading regimen doses result in a faster insufficiency correction [50,51,52,53,54,55,56,57,58]. Once the values of vitamin D are corrected, the Endocrine Society recommends a daily dose of 600–2000 IU (correction depending on the exposure to sunlight), and IOM recommends a daily dose of 600 IU [38,39,59]. Daily exposure of 7–30 min to sunlight is estimated to be enough to accomplish vitamin D substitution doses considering the skin color, season, and latitude [59]. Vitamin D substitution in adults aged 70 years or older without major comorbidities did not lead to significant differences in improvement in blood pressure, physical performance, fractures, infection rates, or cognitive function [60,61]. One nationwide, randomized, controlled trial including 25,871 participants, proved that vitamin D supplementation for five years, with or without omega 3 fatty acids, can decrease the risk of autoimmune disease by 22%. Omega 3 fatty acid supplementation with or without vitamin D can decrease the risk of autoimmune disease by 15% [62]. Hypercalciuria can be more frequently encountered with higher doses. In a double-blind, randomized controlled trial with 373 participants, hypercalcemia was rare, mild, and transient at higher doses. The safety of vitamin D supplementation is comparable for doses of 400, 4000, and 10,000 IU/day [61,63].
Food-based solutions for optimal vitamin D nutrition and health through the life cycle (European Commission funded project ODIN) was a cross-disciplinary project that included 30 partners from 19 countries, which aimed to prevent vitamin D deficiency and develop a public health food-first approach [64,65]. To address the problem of dietary requirements for vitamin D, four Randomized Controlled Trials (RCTs) [66,67,68,69,70] were conducted in Northern Europe to obtain 25(OH)D targets of 25/30 to 50 nmol/L in almost all participants with absent or reduced sun exposure. One RCT revealed that vitamin D intake of 8 and 13 μg/day can prevent vitamin D deficiency in white children and adolescents [66,67]. One meta-analysis of RCTs evaluating the use of vitamin D-fortified food among healthy children aged 1–18 years old reported that vitamin D food fortification prevented vitamin D deficiency and improved IQ levels [71]. White women during pregnancy require 30 μg/day to maintain serum 25(OH)D levels ≥50 nmol/L during pregnancy to obtain 25(OH)D concentration of >25 nmol/L in 99% and ≥30 nmol/L in 95% of umbilical cord serum [70]. Among women of ethnic minorities in Northern Europe, white women in Finland required 8 μg/day to obtain the cut-off value of 30 nmol/L, while women of East African descent needed 18 μg/day during wintertime [69]. ODIN research demonstrated that diverse fortification strategies could prevent vitamin D deficiency in population subgroups [64,65]. Finland has a mandatory fortification program of milk products with a positive impact on vitamin D nutrition [68].
Genetic and environmental factors have been known to influence the appearance of autoimmune diseases [72]. Among the well-known function of vitamin D in supporting bone mineralization through calcium homeostasis [73], vitamin D has been demonstrated to intervene in innate [74] and adaptive [75] immunity by controlling the growth and differentiation of different immune cells such as T [76] and B lymphocytes [77], macrophages [78] and dendritic cells [79]. Vitamin D is associated with systemic [80] or organ-specific [81] autoimmune diseases.
Beyond its classical role in calcium homeostasis, evidence arose from studies demonstrating antioxidant [82,83,84] and anti-fibrotic [85,86,87] functions, preventing inflammatory response [88] and intervening in immune-mediated injuries [3,8,89,90,91,92,93,94]. Treatment opportunities influence the prognosis and quality of life in patients with autoimmune diseases, and the immunomodulatory effects of vitamin D represent a potential supplementation therapy as vitamin D is deficient in patients suffering from autoimmune diseases [29].
Vitamin D has pleiotropic effects suggested by the expression of VDR in lymphocytes and dendritic cells. Many studies are based on vitamin D’s role in various diseases such as autoimmune disorders, cardiovascular diseases and tumors. In autoimmune and inflammatory diseases, vitamin D intervenes in the innate and adaptive immune systems [3,8,83,85,88,91,92,93,94]. Animal studies have demonstrated that administering vitamin D or its analogs can influence the occurrence and progression of many immune-related disorders [5,29]. This underlines that vitamin D can lead to changes in the incidence and severity of various diseases such as infectious diseases, psoriasis, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis [95]. Therefore, this narrative review discusses the effects of vitamin D on the immune system and the role of vitamin D in the pathogenesis of various immune-mediated and autoimmune diseases.

2. Vitamin D Effects on the Innate Immune System

Calcitriol is a pluripotent regulator of the innate immune system (Figure 1). The bacterial infection triggers the activation of toll-like receptors (TLRs) that regulates VDRs expression and 25(OH)D-1α-hydroxylase activity. TLRs are a class of non-catalytic transmembrane pathogen-recognition receptors (PRRs) that interact with specific pathogen-associated molecular patterns (PAMPs) [95]. Calcitriol stimulates antimicrobial activities of macrophages and monocytes through VDR-RXR signaling, which triggers the production of cathelicidins that attach to microbial membranes to eliminate the bacteria and fungi [3,5,27,29,92,96,97,98]. Cathelicidin directly influences various respiratory viruses by disrupting viral envelopes [97,99,100,101]. This is also important in granulomatous inflammation such as TB, lymphomas, and sarcoidosis [3,79,102,103].
Many studies reported that low vitamin D levels are associated with an increased risk of infections and autoimmune diseases due to molecular mimicry [104,105]. Vitamin D can influence dendritic cell activity, inhibiting monocyte differentiation into dendritic cells and reducing IL-12 production [7,106,107,108]. 1,25-dihydroxivitamin D regulates NK cell activity, degranulation process, cytokine secretion, and TLR4 expression. This could demonstrate the benefit of vitamin D supplementation in patients with cancer [109]. Vitamin D regulates the intracellular TLRs differently, down-regulating TLR9, whereas TLR3 is unaffected. The decreased expression of TLR9 results in less IL-6 secreted. This highlights the association between vitamin D deficiency and the risk of developing autoimmune diseases [110].
Calcitriol intervenes in antigen-presenting cells (APC) differentiation and function by promoting APC to become more tolerogenic and decreasing the expression of major histocompatibility complex (MHC) class II and other similar molecules on the cell surface [111,112,113]. The data regarding whether calcitriol induces or inhibits NK cell function is uncertain [109,114,115].
In 1986 Rook and colleagues demonstrated that calcitriol inhibits the growth of Mycobacterium tuberculosis [116]. Liu P.T. and colleagues reported that TLR activation of human macrophages intervenes in killing intracellular Mycobacterium tuberculosis by stimulating the expression of VDR and vitamin D-1–hydroxylase genes. The authors also highlighted the role of TLRs and vitamin D in modulating innate immune responses. This link contributes to the differences found among human populations in synthesizing vitamin D and responding to microbial infection [96]. Other studies showed that calcitriol generates the expression of antimicrobial genes and the interaction between VDREs and containing promoter sequences of the cathelicidin antimicrobial peptide (camp) and defensin β2 (defB2) genes. Ligand binding results in VDR heterodimerization with DNA binding and retinoid X receptors to related VDREs composed of direct duplicates of consensus PuG(G/T)TCA motifs. This underlies the importance of new therapeutic strategies using calcitriol analogs in treating opportunistic infections [95,117,118]. Calcitriol and its analogs have the potential to induce the expression of camp gene in bronchial epithelial cells [119], keratinocytes [120], and myeloid cell lines [121]. 25(OH)D can also induce promoter sequences of the cathelicidin, underlining the antimicrobial function in cells that express 1α-hydroxylase encoded by the CYP27B1 allele [120]. CYP27B1 allele is expressed on various types of cells, and its function depends on the stimulation of each specific cell. Enhanced sensitivity to calcitriol is achieved through PAMPs, activating innate immune responses mediated via TLRs [95,122].
Activated human macrophages after being treated with mycobacterial 19 kDa lipoprotein, a TLR2 that interacts with specific PAMPs, demonstrated increased gene expression of both CYP27B1 and VDR, inducing promoter sequences of the cathelicidin through 25(OH)D and further bacterial killing [88,105,123,124]. A similar result regarding the innate immune response to infection was revealed by inducing cathelicidin expression in monocytes exposed to M. tuberculosis mediated through calcitriol. Calcitriol was able to induce bacterial killing through other factors such as nitric oxide synthase [74]. The expression of cathelicidin-small interfering RNA can enhance the pathway by which calcitriol interacts with the innate immune system. This underlies that cathelicidin is needed for calcitriol antimicrobial activity against M. tuberculosis [96].
Macrophages stimulated with bacterial lipopolysaccharide converted calcitriol to a more polar calcitriol-like metabolite [125]. Vitamin-D-mediated pathway regarding bacterial killing consists of the expression of cathelicidin that is suppressed by specific pathogens. In this matter, macrophages infected with Shigella suppress the expression of cathelicidin and defensin β2 to bypass innate antibacterial immunity [126]. Local induction of calcitriol synthesis can increase cathelicidin expression in promoting the antibacterial function. This mechanism is controlled by feedback pathways limiting a potential exaggerated inflammatory response that could arise from the activation of the immune system. Both down-regulation of TLR2 and TLR4 on monocytes and up-regulation of CD14 were inhibited by VDR antagonist ZK 159222, underlining that calcitriol requires VDR transcription factor activation on immunity receptors [127]. The reduced expression of TLR might inhibit inflammatory T lymphocyte responses that could generate T-helper 1 (Th1) lymphocyte autoimmunity. Feedback regulation of vitamin D is controlled by CYP24A1, the enzyme that catalyzes the synthesis of less active vitamin D metabolites [128].
Vitamin D and its metabolites intervene in vascular permeability and endothelial function through various genomic and non-genomic pathways. 25(OH)D and calcitriol stabilized vascular endothelium via a non-genomic pathway [129,130]. Calcitriol up-regulates endothelial nitric oxide synthase (eNOS), causing increased endothelial production of nitric oxide [131,132]. The increase in eNOS activity via calcitriol activation of VDR through intracellular adenylyl cyclase/cyclic adenosine monophosphate (AC/cAMP) and inositol trisphosphate/diacylglycerol (IP3/DAG) pathways leads to increased intracellular calcium level. VDR also triggers eNOS activation through the phosphoinositide 3-kinase/protein kinase b (PI3K/Akt) pathway, resulting in phosphorylation of serine-1779 on eNOS [133]. In an animal model, calcitriol stimulates endothelial-cadherin-based cellular junctions, inhibits stress fiber formation, inhibits the endothelial intracellular gaps organization, and limits endothelial damage in chronic kidney disease. Vitamin D and its metabolites prevent vascular dysfunction and local and systemic inflammation with tissue injury [134].
Calcitriol stimulates the expression of CYP24A1 in macrophages with no concomitant increase in CYP24A1 activity as calcitriol stimulates the expression of a splice variant form (CYP24A1-SV) that encodes a truncated amino-terminal protein [135]. The cytoplasm inactive state of CYP24A1-SV can limit vitamin D metabolism by confining the conversion to metabolites. The autocrine mechanism for bacterial killing is enhanced through the metabolism of vitamin D with PRR responses that interact with specific PAMPs. This mechanism is involved in macrophage-mediated immune responses, but also keratinocytes can induce cathelicidin through TLR2-mediated recognition of PAMPs in relation to the autocrine synthesis of calcitriol [136]. Keratinocytes can produce calcitriol in the presence of the transforming growth factor β1 and stimulate the production of cathelicidin mediated through TLR2 with the expression of CYP27B1 [136]. Interaction between transforming growth factor β1 and vitamin-D-mediated cathelicidin production involves wound repair through innate immune activation [136].
Vitamin D intervenes in gut integrity and maintaining a balanced relationship between host and gut microbiota. Vitamin D signaling maintains the integrity of intestinal epithelial cells and limits the bacterial lipopolysaccharide damage to intestinal epithelium [137,138]. Multiple studies have shown that vitamin D stimulates the expression of proteins that recognize intracellular pathogens [139,140] and promotes the production of antimicrobial proteins by the intestinal epithelial cells, intraepithelial lymphocytes, and Paneth cells [141,142]. These inhibit intestinal bacterial translocation and maintain intestinal homeostasis, which is thought to contribute to developing many auto-inflammatory and metabolic disorders.

3. Vitamin D Effects on the Adaptive Immune System

Vitamin D and VDR can influence B and T lymphocytes [6,27] (Figure 1). Several studies have reported that Vitamin D may intervene in B-cell differentiation and proliferation, decreasing antibodies and auto-antibodies synthesis with B-cell apoptosis. Vitamin D may also reduce T helper (Th) cell differentiation and proliferation and induce a more tolerogenic immune response than a pro-inflammatory status [143,144]. Vitamin D inhibits the synthesis of various pro-inflammatory Th1, Th9 and Th22 cytokines and stimulates the synthesis of anti-inflammatory Th2 cytokines. These findings could demonstrate the beneficial effect of vitamin D in minimizing the risk of developing autoimmune diseases [144].
Studies demonstrated that VDR expression in lymphocytes enhances vitamin D  as a pluripotent regulator of the innate immune system [75,145,146]. VDR expression is described in activated proliferating T lymphocytes [75] and B lymphocytes [147], implicating vitamin D as a modulator of the adaptive immune system. Inactive B lymphocytes do not present VDR; they upregulate VDR expression only when they are activated for proliferation through mitogens [77].
Initially, calcitriol suppression of immunoglobulin production was thought to be an indirect effect mediated via Th lymphocytes [148]. Further studies showed that calcitriol could directly influence B lymphocyte homeostasis [77]. In this concern, calcitriol can inhibit B cell differentiation into antibody-secreting plasma cells, inducing apoptosis, a process linked to DNA hypo-methylation [149,150]. This suggests the implication of vitamin D in B cell-related diseases such as systemic lupus erythematosus (SLE). Patients with SLE have significantly lower serum levels of calcitriol and 25(OH)D compared to controls [25,77].
As part of the adaptive immune system, Vitamin D intervenes in T lymphocyte proliferation and function [75]. Calcitriol targets Th lymphocytes by suppressing their proliferation and cytokine production, such as interleukin 2 (IL-2) [151]. The antigen-activated pluripotent Th0 lymphocytes generate various cytokines, including IL-2, IL-4, IL-10 and interferon γ (IFN-γ) [152]. Calcitriol directly inhibits [153] the expression of Th1 cytokines (IL-2, IFN-γ, tumor necrosis factor) [154] and stimulates Th2 (IL-3, IL-4, IL-5, IL-10) [153,154,155,156] or indirectly through APCs [113]. It also suppresses mature B cells to form plasma cells and class-switched memory B cells [77,157,158].
Calcitriol stimulates the expression of CTLA-4 and FoxP3, requiring the presence of IL-2 [159]. Calcitriol promotes Th1 cellular immunity more than Th2 humoral immunity. This is an essential key factor regarding the beneficial effects of vitamin D in autoimmune diseases [160]. Reports have shown that calcitriol stimulates the production of CD4+CD25+ Treg. Treg cells generate self-tolerance and contribute to preventing autoimmune disorders and graft-versus-host disease in transplantation [161].
Cytotoxic T lymphocytes (CTL), similar to Th cells, express both VDR and CYP27B1. Expression of VDR can appear in the presence of infection and mitogen stimulation, underlining a connection between the VDR signaling pathway and CTL activity [162,163]. Studies have reported that decreased CD4/CD8 ratio is associated with reduced levels of 25(OH)D [164]. Administration of 5000–10,000 IU of 25(OH)D increased CD4/CD8 ratio, suggesting immune suppression [165,166], but there is limited information about the direct effect of vitamin D on CTL. Induction and functions of CTL are influenced via direct activation of VDR and alteration in the expression of inflammatory cytokines through Th cells and APCs [143,163].
Calcitriol or its synthetic analogs can cause induction of Treg cells offering protection against autoimmune disorders and immune tolerance in organ transplantation [167,168]. Vitamin D targets for the adaptive immune system are the dendritic cells (DCs), especially a certain class of DCs known as myeloid DCs [169]. Calcitriol suppresses the maturation of DCs and the development of Th1 and promotes the induction of tolerogenic DCs and Treg [106,170]. A similar effect on DC could be obtained through CC-chemokine ligand (CCL) 22, which is a chemoattractant factor secreted by DCs that stimulates the synthesis of Treg. In myeloid DCs, calcitriol inhibits intracellular signaling nuclear factor κB [169].

4. Vitamin D Target Genes with Functions in the Immune System

Vitamin D target genes present a key role in the action of vitamin D in innate and adaptive immunity, and each function is demonstrated based on particular gene regulatory scenarios. A study described these genes by performing transcriptome-wide datasets based on peripheral blood mononuclear cells (PBMCs) and human monocytic cell line (THP-1), which were treated in vitro by calcitriol. These genes were described based on their VDR stimulation and their mRNA production. The following genes were identified in this study: ACVRL1, CAMP, CD14, CD93, CEBPB, FN1, MAPK13, NINJ1, LILRB4, LRRC25, SEMA6B, SRGN, THBD, THEMIS2 and TREM1. Vitamin D target genes were categorized based on their role in acute infection, infection in general and autoimmunity (Figure 2). These immune-related genes encoded proteins placed in the plasma membrane (ACVRL1, CD14, CD93, LILRB4, LRRC25, NINJ1, SEMA6B, THBD, TREM1) or are secreted (CAMP, FN1 and SRGN). In addition, the kinase MAPK13 is located in the cytoplasm, and the transcription factor CEBPB and the TLR scaffold protein THEMIS2 are placed in the nucleus [171].
CD14, TREM1, FN1, and CAMP are proteins involved in acute response to an infection via LPS/TLR4 signaling pathway [171]. The CD14 glycoprotein is mainly expressed in monocytes and macrophages and is located on the surface of the plasma membrane via glycosylphosphatidylinositol. CD 14 is a co-receptor for TLR 1-4, 6, 7 and 9 [172] and presents bacteria produced by LPS to TLR4 in inflammation [173]. Another vitamin D target gene located on monocytes and macrophages [174] is TREM1, associated with TLR4 signaling that generates inflammation in bacterial infection [175]. The extracellular matrix protein FN1 encoded by vitamin D, secreted by macrophages, epithelial cells and fibroblasts, is involved in inflammatory responses in LPS/TLR4 signaling, cell adhesion, and wound healing [176].
THBD, LILRB4, SEMA6B, LRRC25, MAPK13, and THEMIS2 genes encode proteins with a general function in infection [171]. THBD is secreted by monocytes, macrophages and endothelial cells [177] and reduces blood clots by turning thrombin pro-coagulative action to anti-coagulative. It also prevents pro-inflammatory responses of NF-kB signaling, binds to LPS and promotes LPS binding to CD14 and TLR4 [178]. The THBD gene is a vitamin D target in monocytes [179] and PBMCs [180]. LILRB4 is a receptor involved in the inhibition of APCs such as macrophages, monocytes, DCs and microglia, resulting in the production of TNF, bactericidal activity [181], and stimulation of Treg differentiation [182]. The LILRB4 gene is a vitamin D target gene in PBMCs [183]. One vitamin D target gene in bone is SEMA6B [184], belonging to the semaphorin protein family, which presents immune activity by controlling cell movements and cell-to-cell communication [185]. The LRRC25 protein acts on monocytes, granulocytes T cells, and DCs, inhibiting NF-kB signaling [186] and interferon [187]. It intervenes in response to viral infections by decreasing the production of inflammatory cytokines [188,189]. The kinase MAPK13 intervenes in LPS/TLR4 signaling and inflammatory responses along with MAPK11 [190]. It is a vitamin D target in skeletal muscle [191]. THEMIS2 is a TLR signal transduction modulatory protein that acts on macrophages, DCs, and B cells in TNF production induced by LPS [192] and T cell development [193].
CD93, NINJ1, CEBPB, ACVRL1, and SRGN genes encode proteins related to autoimmunity. CD93 glycoprotein acts on monocytes, endothelial cells, granulocytes, platelets and stem cells [194] and affects innate immunity in adhesion, phagocytosis and inflammation [195].    It functions as a DNA receptor for presenting to TLR9 and is involved in inflammatory responses in LPS/TLR4 signaling [196]. NINJ1 protein is expressed in myeloid and endothelial cells [197] and is described in the immune pathogenesis of multiple sclerosis [198] by promoting cell adhesion and leukocyte migration in inflammation of the endothelium. CEBPB gene is a vitamin D target in myeloid leukemia cells [199]. It proved to be involved in inflammation via Th17 in models of multiple sclerosis [200]. ACVRL1 is known to be a protein of the transforming growth factor-beta superfamily, which is associated with monocyte to macrophage differentiation [201], and with the signaling of the bone morphogenetic protein (BMP)9 and BMP10 [202]. SRGN is expressed by monocytes, macrophages, lymphocytes, mast cells, and endothelial cells [203]. It is involved in the secretion of inflammatory mediators via LPS [204].   
These 15 genes are considered the most important vitamin D targets in connection to immunity and biomarkers in diagnosing vitamin D deficiency related to immune diseases [171].

5. Vitamin D and Immune-Related and Autoimmune Diseases

The role of vitamin D supplementation in autoimmune diseases was studied based on the immunomodulatory effect of vitamin D. Animal experiments showed an essential role of calcitriol supplementation in autoimmune encephalomyelitis and collagen-induced arthritis, where vitamin D prevented the initiation and reduced disease progression [143,205,206]. Mouse models of enterocolitis presented a more severe disease course during vitamin D deficiency and decreased inflammation after vitamin D supplementation [207]. Despite various experimental studies, human clinical trials presented mixed results. Here we summarize the current findings in the literature regarding the role of vitamin D in immune-mediated and autoimmune diseases.

5.1. Psoriasis

Psoriasis is a chronic inflammatory disease, a hyper-proliferative disorder of the skin that affects 2–3% of the global population [208]. As 25(OH)D is mainly produced in the skin [209], and keratinocytes express VDR, calcitriol could be used to treat this disorder [210]. The inflammation generated in psoriasis after exposure to self-antigen is associated with Th1, Th17, and Th22 [211]. Calcitriol inhibits the differentiation of dendritic cells, antigen presentation, and the inflammatory activity of IL-1β, IL-6, IL-8, and TNF-α [212]. One study that analyzed if VDR gene polymorphisms are associated with psoriasis concluded that the rs7975232 allele was over-expressed in psoriasis patients compared to controls [213,214].
Even if it was initially described that cultured psoriatic fibroblasts were resistant to the anti-proliferative activity of calcitriol [215], other studies revealed that only approximately 20% of psoriatic patients presented this partial resistance [216]. A topical analog of calcitriol, calcipotriene (50 mcg/g), was developed to avoid a potentially hypercalcemic effect of calcitriol. Calcipotriene modulates the expression of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-2, and IL-8) and is effective for treating psoriasis, and it does not generate significant hypercalcemia [217]. It was reported that skin irritation appeared mainly when applied to the face [218]. After it was described that calcitriol was effective at 15 mcg/g and did not cause skin irritation, topical treatment was developed as a 3 mcg/g formula for treating psoriasis that affects less than 10% of the body surface [210].
Studies described the association between vitamin D deficiency and patients with psoriasis [212,219], possibly because these patients avoid more exposure to sunlight. In this regard, various clinical trials have tried to find the proper vitamin D supplementation. The 25(OH)D level was inversely associated with all-cause mortality in patients with psoriasis [214,220]. A beneficial result in the PASI (psoriasis area and severity index) score and the serum level of 25(OH)D in patients who followed a six-month treatment with 60,000 IU of vitamin D2 administered every 2 weeks [221]. A similar result was described in a study that comprised psoriatic patients treated with daily 35,000 IUs of 25(OH)D for six months [222]. One study that included hospitalized patients during seven years of experience reported three patients with psoriasis that presented clinical skin improvement after using 20,000 to 50,000 IUs/day [223]. Another clinical trial reported no improvement in disease activity in patients with psoriasis after one year of monthly 100,000 IU of 25(OH)D [224]. The Endocrine Society clinical guideline recommends that patients with psoriasis should be tested and treated for vitamin D deficiency so that the 25(OH)D level would be 40–60 ng/mL (100–150 nmol/L) [38]. However, more extensive clinical trials are needed to adequately describe the efficacy of oral calcitriol psoriasis treatment.

5.2. Type 1 Diabetes

Type 1 diabetes (T1D) is encountered more frequently in countries located at higher latitudes with reduced sunlight exposure [225,226,227]. Areas such as Finland and Norway that are not exposed to sunlight for more than half a year have a high incidence of vitamin D deficiency [225,228]. The world’s highest incidence of T1D is described in Finland [229]. One meta-analysis involving 16 studies and more than 10,000 participants proved a significant inverse association between 25(OH)D concentration and the risk of developing T1D [214,230]. Other risk factors are proposed for the development of autoimmunity, such as exposure to toxic substances [231] and coxsackievirus B infections [232].
On the other hand, one mendelian randomization study including single nucleotide polymorphisms (SNPs) that are associated with 25(OH)D levels included in a large vitamin D genome-wide association study (GWAS) on more than 400,000 Europeans demonstrated that 25(OH)D levels were unlikely to present an impact on the risk of developing T1D [214,233]. Another case-control study did not register a significant association of each SNP (DHCR7, GC, CYP2R1 and CYP24A1 genes) with T1D but demonstrated a combined effect of multiple risk alleles [214,234].
The mechanisms involved in T1D comprise the generation of autoantibody and autoreactive Th1 and CTL, resulting in immune-mediated destruction of insulin-producing pancreatic β cells [235,236]. In animal experiments, calcitriol administration led to Treg stimulation and Th1 inhibition, reducing the incidence of T1D [168]. Vitamin D can be protective in developing T1D by stimulating insulin secretion in the pancreatic β cells via direct binding with VDR [225]. Some studies report a reduced risk of T1D in children with a higher vitamin D intake [237]. A clinical trial developed in Finland described a reduction of 88% in the risk of developing T1D in children who were given daily 2000 IU of 25(OH)D during their first year of life [238]. The EURODIAB multicenter case-control study reported a reduced risk for T1D if vitamin D was supplemented in any dose [237]. Other trials revealed that vitamin D supplementation managed to promote the reduction of the daily insulin dose and increase C-peptide levels [239,240,241]. Vitamin D intake with a normal 25(OH)D level is important in early childhood as it offers protection in the development of T1D. Vitamin D supplementation can improve the disease activity but does not influence morbidities and mortalities.

5.3. Multiple Sclerosis

Multiple sclerosis (MS) is encountered more frequently in countries located at a higher latitude with reduced exposure to sunlight, similar to type 1 diabetes [200,227]. Studies showed that a decrease of 41% in the risk of MS was obtained with every 20 ng/mL (50 nmol/L) increase in serum 25(OH)D levels above 24 ng/mL (60 nmol/L) [242]. The same study reported that women who had an intake of daily vitamin D higher than 400 IU presented a 41% decreased risk of developing MS [242].
A Mendelian Randomization (MR) study using information regarding MS and 25(OH)D levels from large European populations described that a decrease in genetically lowered 25(OH)D level by one standard deviation (SD) resulted in a 2-fold increase in developing MS [243]. Another MR analysis using genome-wide 25(OH)D-associated SNPs from a meta-analysis conducted in 401,460 British UK Biobank participants and 42,274 samples of European ancestry. Increased 25(OH)D concentration is associated with a decreased risk of developing MS. The authors found limited evidence supporting the protective effect of 25(OH)D on obesity, breast and prostate cancer, and autoimmune, metabolic, cardiovascular, and neuro-psychiatric diseases [244]. The largest GWAS for serum 25(OH)D includes 401,460 participants and 14,498 MS cases and 24,091 controls of European ancestry. A causal association was established with the increase of 25(OH)D based on genetic changes, resulting in a decreased MS risk [245]. Another 2-sample MR determined the genetic causal effect of vitamin 25(OH)D and concluded that each increase in the natural-log-transformed vitamin D concentration resulted in a 43% decrease in the odds ratio of MS (OR 0.57, 95% CI 0.41–0.81, p = 0.001) [61,246].
Vitamin D deficiency in MS stimulates auto-inflammation of the central nervous system through the dysregulation of Th cells, CTL, B cells, and NK cells, damaging neurons and oligodendrocytes [247,248]. There is an individual predisposition for developing MS, such as HLA-DRB1*1501 [203]. Vitamin D response elements are also situated in the promoter region of the HLA-DRB1 gene [249,250,251]. Among gene variants that are associated with developing MS are CYP27B1 and CYP24A1, alleles that are involved in the vitamin D pathway [214,252]. Various immune actions of calcitriol are similar to those of interferon-beta, an MS immunomodulatory treatment. There are conflicting results based on the therapeutic role of vitamin D in MS. Some studies revealed improvement in the relapse rate and MRI findings on an increased dose of vitamin D (14,000 IU/day) [253].
The treatment dosages of vitamin D vary significantly among various studies. They can comprise high doses of vitamin D of 50,000 IU/day or 1000 IUs/kg/day to obtain an increase in serum 25(OH)D up to 200–300 ng/mL (500–750 nmol/L). This high vitamin D supplementation improved symptoms and MRI findings in MS patients who did not respond to or refused conventional MS treatment [224]. One study described the treatment applied to 319 patients with a broad spectrum of autoimmune diseases (MS and non-MS such as vitiligo, psoriasis, inflammatory bowel disease, RA) that were treated for more than 3.5 years with high doses of orally 25(OH)D up to 1000 IU/kg. The study demonstrated that these doses were safe regarding calcium metabolism and renal function when patients followed a strict diet and fluid recommendations [254]. There are no RCTs to support the safety of the high dose of vitamin D (1000 IU/kg/day). Still, supplementation with lower doses of vitamin D (up to 14,000 IUs/day) proved to control the disease activity.

5.4. Inflammatory Bowel Diseases

The risk of inflammatory bowel disease (IBD) is more common in countries located at a higher latitude with reduced exposure to sunlight, similar to T1D and MS [227,255]. IBD presents a defective innate and adaptive immunity, with increased intestinal permeability, and imbalanced gut microbiota, resulting in chronic inflammation of the intestine [256,257,258].   
Vitamin D deficiency in IBD is caused by micelles and chylomicron that cannot form and absorb intestinal vitamin D [259,260], resulting in an increased risk of osteomalacia and osteoporosis [259,260,261]. Therefore, these patients should be tested and treated for vitamin D deficiency with an increased dose of vitamin D so that the 25(OH)D level would be at least 30 ng/mL (75 nmol/L) [38]. The decreased 25(OH)D level is a biomarker for IBD activity and a predictor of poor prognosis in these patients [214,262].
The inflammatory response in CD comprises Th1 stimulation, while in UC, Th2 is promoted [256,263], and Th17 is involved in both diseases [256,264,265]. Studies reported that calcitriol stimulates Treg and inhibits Th1 and Th17 activity. It also intervenes on intestinal mucosal cells by stimulating proteins involved in membrane junction integrity and intracellular pathogen recognition. Vitamin D is essential in stimulating the production of antibacterial proteins such as cathelicidin, angiogenin, and defensin [139,140,262,265,266].
A prospective study from the Nurses’ Health Study revealed that a high 25(OH)D level was connected with a decrease of 46% in developing Crohn’s disease (CD) and 35% in developing ulcerative colitis (UC) [267]. Various clinical trials reported that vitamin D supplementation was beneficial in patients with IBD by decreasing relapse rate [268] and improving bacterial intestinal homeostasis in patients with CD [269]. One study based on healthy individuals with vitamin D deficiency who received daily doses of either 600, 4000, or 10,000 IU for 6 months presented improved immune response with increased expression of Treg and improvement of gut microbiota [270]. Unable to absorb vitamin D, patients with IBD need higher doses of vitamin D to achieve normal serum 25(OH)D levels and help prevent the development of bone fragility, osteoporosis, and osteomalacia.

5.5. Rheumatoid Arthritis

The risk of rheumatoid arthritis (RA) is associated with low levels of 25(OH)D [271]. Studies reported that women with higher doses of vitamin D presented a 33% reduced risk for developing RA [271], and lower serum 25(OH)D levels were associated with disease activity [271,272]. Although these patients have reduced exposure to sunlight because of their disease, treatment with vitamin D is considered beneficial due to Th1 and Th17 suppression and Treg activity stimulation [273]. These immunologic responses promote chronic synovial inflammation [274,275].
There are mixed results reported from studies using adjunctive treatment with vitamin D for RA. Clinical trials described important pain relief in RA patients that received 500 IU daily of 25(OH)D with disease-modifying anti-rheumatic drugs (DMARDs) and calcium compared to those that did not receive vitamin D supplementation [276].
Another study presented RA patients receiving either 22-oxa-1,25-dihydroxyvitamin D or calcitriol or placebo, which led to decreased swollen joints and improved disease activity scores in the group of patients that received 22-oxa-1,25-dihydroxyvitamin D and those with calcitriol compared with the placebo group [277]. Other clinical trials that used vitamin D2 [278], 25(OH)D [279,280], or 1α-hydroxyvitamin D3 [281] presented less convincing results in the treatment of RA patients. One meta-analysis identified 9 RCTs of vitamin D treatment for at least 3 months in autoimmune diseases, which included five studies of RA patients, but with no statistical significance. These patients presented a decrease in the number of flares, patient global visual analog scale score (VAS) and disease activity score 28 (DAS-28) with vitamin D supplementation [214,266,282]. Vitamin D intake associated with serum 25(OH)D levels of 40–60 ng/mL (100–150 nmol/L) may result in a lower risk of RA, but there is still scarce evidence on this subject.

5.6. Systemic Lupus Erythematosus

Systemic lupus erythematosus (SLE) is a chronic, multisystem, autoimmune disease that mostly appears in young women. Studies described that SLE is associated with low vitamin D levels in disease expression, relapses, and pathogenesis [214,283,284,285,286,287,288,289,290,291,292]. In SLE, there is no association between vitamin D and pro-inflammatory cytokines. Most studies describe that low vitamin D correlates with disease activity, but two studies reported that vitamin D did not influence disease activity [293]. Vitamin D deficiency was more prevalent in unsupplemented SLE participants living at a latitude beyond the 37˚ parallel north [294]. Some additional risk factors for vitamin D insufficiency are described in SLE patients, such as reduced sun exposure, renal disease, VDR genetic polymorphisms with decreased cell responsiveness to the hormone and the genetic variants that encode important enzyme regulators of endogenous production [295,296].
Moreover, vitamin D has a protective role in lupus nephritis induced by autoantibodies [297]. Reduced vitamin D concentration is associated with worse disease activity, fatigue [298], cardiovascular disease [299], and cognitive impairment [300,301]. One study demonstrated that vitamin D deficiency is associated with more active disease at first admission and over time, and more severe lupus flares [285,302].
There is also a high prevalence of vitamin D deficiency in newly diagnosed SLE patients and a significant association between vitamin D level and the decreased C3 level [214,303]. Low 25(OH)D concentration is associated with increased levels of memory B cells in well-controlled SLE patients [214,304] and an increased number of Treg cells [305].
Vitamin D status and CYP24A1 might contribute to the predisposition of SLE in individuals with genetic risk for SLE [306]. Pregnant women with SLE and low vitamin D presented higher morbidity [288]. Vitamin D deficiency often correlates with low bone mineral density (BMD) [287]. The presence of FokI and TaqI variants was demonstrated in an eastern Indian cohort of patients with SLE [307]. Even if vitamin D is associated with disease activity in SLE, further recommendations are needed regarding vitamin D target dose and target levels [7].
Vitamin D supplementation should be part of SLE treatment [294] to reduce disease SLE activity as recommended by two randomized controlled trials [308,309], one open clinical trial [310] and a cohort study [311]. On the other hand, two cohort studies [251,312] and an RCT [313] failed to describe any significant variation.
Vitamin D supplementation can also be used to prevent glucocorticoid-induced osteoporosis [314]. Current vitamin D supplementation strategies do not focus on individual recommendations and can be insufficient in raising vitamin D concentrations in every patient. In this regard, a treat-to-target approach could be more suitable. For a better disease approach, an initial measurement of vitamin D level should be performed for each patient.

5.7. Systemic Sclerosis

Systemic sclerosis (SSc) is an autoimmune multi-organ disease characterized by vasculopathy and fibrosis involving the skin and/or lungs, heart and kidneys [315,316,317]. Multiple studies described lower vitamin D levels in SSc patients [318,319]. Still, clinical manifestations were not connected to the degree of vitamin D deficiency [320]. Neither was age, gender, disease activity with digital ulceration or systemic involvement, nor the type of auto-antibodies. Still, the degree of vitamin D deficiency had an inverse correlation with skin sclerosis [321].   
Low vitamin D levels in SSc can be explained by reduced sunlight exposure, inadequate epidermal production of vitamin D in the epidermis due to skin fibrosis, and diminished gastrointestinal malabsorption [322]. Another reason for low levels of vitamin D could be the presence of antivitamin D antibodies, as shown in a study that demonstrated that the antivitamin D antibodies were found in 87% of SSc patients and 42% of controls [323].
Regarding the SSc pathogenesis, the transition from fibroblast to activated myofibroblast is an important event in SSc involving TGF-β signaling, epigenetic mechanisms and various other cellular pathways [324]. Vitamin D effect on fibrosis in SSc concerns impaired VDR signaling with reduced expression resulting in hyperactive TGF-β signaling and abnormal fibroblast activation [325]. When analyzing vitamin D and two major regulators, SSc patients presented a reduced FGF23/α-Klotho index and its log10 related to disease activity score [326].
The studies regarding the impact of vitamin D deficiency and clinical features and disease activity of SSc are controversial because of the small sample size, SSc being a rare disease [327]. A study that included 327 SSc patients described an inverse relationship between vitamin D concentration and skin involvement [322]. A cohort study reported that pulmonary involvement was more often encountered in patients with vitamin D deficiency than controls, but with no statistical significance [328]. Some studies suggested an association between vitamin D level and organ involvement, particularly pulmonary and cardiac [329], and pulmonary artery hypertension [330,331]. Further research is required to understand the role of vitamin D in SSc and whether its supplementation could impact disease activity or influence disease onset.

5.8. Miscellanea

Antiphospholipid syndrome is a multisystem autoimmune disease characterized by hypercoagulability, which may be associated with recurrent fetal loss and thromboembolic events in the presence of elevated titers of antiphospholipid antibodies (aPL) [332,333]. Studies described that low vitamin D levels are associated with antiphospholipid syndrome [334,335,336,337] and vitamin D deficiency with clinically-defined thrombotic events [338].
Immunoglobulin A nephropathy (IgAN) is the most frequent primary form of glomerulopathy encountered in the western world. It is a disease that involves genetic and autoimmune mechanisms and environmental factors. It is a significant cause of renal impairment, which requires proper treatment to avoid irreversible damage [339]. Studies have shown an association between vitamin D and IgAN. Vitamin D deficiency resulted in poorer clinical outcomes and more severe renal disease [340].
In autoimmune hepatitis (AIH), some of the VDR and CTLA4 variants are involved in triggering the immune process, these genotype variants being linked with fatty acid synthase (FAS) promoter variants or pro-inflammatory cytokines that are part of the TNF superfamily. This mechanism holds great promise for discovering disease-specific liver fibrosis [341,342,343,344].
Low vitamin D levels were correlated in patients with Sjögren syndrome (SS). Vitamin D may intervene in SS pathogenesis, influencing the presence of extra-glandular symptoms such as lymphoma or neuropathy [345]. One meta-analysis, including 18 studies with SS patients, concluded that patients with vitamin D deficiency associated higher severity scores of dry eye symptoms, such as lower Schirmer’s test scores and higher ocular surface disease index (OSDI) scores [214,346].
Moreover, vitamin D was also correlated with immune thrombocytopenic purpura, chronic idiopathic neutropenia, autoimmune hemolytic anemia, and Evans’ syndrome. Vitamin D deficiency can influence disease severity in autoimmune cytopenias, underlining the protective role in the development of autoimmunity [319].

6. Conclusions

In summary, vitamin D is essential in calcium, phosphate, and bone homeostasis and acts as an immunomodulator, influencing the innate and adaptive immune systems to fight against pathogens. Clinical trials underline the significance of maintaining vitamin D status within the normal range because a low serum level of 25(OH)D is correlated with various immune-related disorders, including autoimmune diseases. There is increased evidence that vitamin D supplementation could improve the treatment of autoimmune disorders. Another question is whether vitamin D is beneficial as an immunomodulatory agent in most diseases based on mixed findings from clinical trials. Healthcare institutions should develop programs to educate the public about the role of vitamin D. Food should be fortified with vitamin D to reduce the risk of vitamin D deficiency at risk ages for developing autoimmune disorders in childhood, in young and middle-aged adults, and during pregnancy. For vitamin D to have immunomodulatory functions, it is probably required the production of calcitriol in monocytes and macrophages. Further questions still need to be answered to fully understand vitamin D’s complete effect on the immune system.

Author Contributions

Conceptualization, C.S.; methodology, C.S., A.G., S.R. and T.L.P.; validation, S.R. and T.L.P.; formal analysis, C.S.; data curation, C.S., A.G. and S.R.; writing—original draft preparation, C.S.; writing—review and editing, C.S., A.G. and T.L.P.; visualization, S.R. and T.L.P.; supervision, T.L.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bouillon, R.; Carmeliet, G.; Verlinden, L.; van Etten, E.; Verstuyf, A.; Luderer, H.F.; Lieben, L.; Mathieu, C.; Demay, M. Vitamin D and human health: Lessons from vitamin D receptor null mice. Endocr. Rev. 2008, 29, 726–776. [Google Scholar] [CrossRef] [PubMed]
  2. Jones, G. 100 YEARS OF VITAMIN D: Historical aspects of vitamin D. Endocr. Connect. 2022, 11, e210594. [Google Scholar] [CrossRef]
  3. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef] [PubMed]
  4. Colotta, F.; Jansson, B.; Bonelli, F. Modulation of inflammatory and immune responses by vitamin D. J. Autoimmun. 2017, 85, 78–97. [Google Scholar] [CrossRef] [PubMed]
  5. Prietl, B.; Treiber, G.; Pieber, T.R.; Amrein, K. Vitamin D and immune function. Nutrients 2013, 5, 2502–2521. [Google Scholar] [CrossRef]
  6. Czaja, A.J.; Montano-Loza, A.J. Evolving Role of Vitamin D in Immune-Mediated Disease and Its Implications in Autoimmune Hepatitis. Dig. Dis. Sci. 2019, 64, 324–344. [Google Scholar] [CrossRef]
  7. Rosen, Y.; Daich, J.; Soliman, I.; Brathwaite, E.; Shoenfeld, Y. Vitamin D and autoimmunity. Scand. J. Rheumato. 2016, 45, 439–447. [Google Scholar] [CrossRef]
  8. Dankers, W.; Colin, E.M.; van Hamburg, J.P.; Lubberts, E. Vitamin D in Autoimmunity: Molecular Mechanisms and Therapeutic Potential. Front. Immunol. 2016, 7, 697. [Google Scholar] [CrossRef]
  9. Slominski, A.T.; Kim, T.K.; Shehabi, H.Z.; Semak, I.; Tang, E.K.; Nguyen, M.N.; Benson, H.A.; Korik, E.; Janjetovic, Z.; Chen, J.; et al. In vivo evidence for a novel pathway of vitamin D₃ metabolism initiated by P450scc and modified by CYP27B1. FASEB J. 2012, 26, 3901–3915. [Google Scholar] [CrossRef]
  10. Slominski, A.T.; Li, W.; Kim, T.K.; Semak, I.; Wang, J.; Zjawiony, J.K.; Tuckey, R.C. Novel activities of CYP11A1 and their potential physiological significance. J. Steroid Biochem. Mol. Biol. 2015, 151, 25–37. [Google Scholar] [CrossRef] [Green Version]
  11. Slominski, A.T.; Kim, T.K.; Li, W.; Postlethwaite, A.; Tieu, E.W.; Tang, E.K.Y.; Tuckey, R.C. Detection of novel CYP11A1-derived secosteroids in the human epidermis and serum and pig adrenal gland. Sci. Rep. 2015, 5, 14875. [Google Scholar] [CrossRef] [PubMed]
  12. Slominski, A.T.; Kim, T.K.; Shehabi, H.Z.; Tang, E.K.; Benson, H.A.; Semak, I.; Lin, Z.; Yates, C.R.; Wang, J.; Li, W.; et al. In vivo production of novel vitamin D2 hydroxy-derivatives by human placentas, epidermal keratinocytes, Caco-2 colon cells and the adrenal gland. Mol. Cell Endocrinol. 2014, 383, 181–192. [Google Scholar] [CrossRef] [PubMed]
  13. Postlethwaite, A.E.; Tuckey, R.C.; Kim, T.K.; Li, W.; Bhattacharya, S.K.; Myers, L.K.; Brand, D.D.; Slominski, A.T. 20S-Hydroxyvitamin D3, a Secosteroid Produced in Humans, Is Anti-Inflammatory and Inhibits Murine Autoimmune Arthritis. Front Immunol. 2021, 12, 678487. [Google Scholar] [CrossRef]
  14. Slominski, A.T.; Kim, T.K.; Takeda, Y.; Janjetovic, Z.; Brozyna, A.A.; Skobowiat, C.; Wang, J.; Postlethwaite, A.; Li, W.; Tuckey, R.C.; et al. RORα and ROR γ are expressed in human skin and serve as receptors for endogenously produced noncalcemic 20-hydroxy- and 20,23-dihydroxyvitamin D. FASEB J. 2014, 28, 2775–2789. [Google Scholar] [CrossRef] [PubMed]
  15. Slominski, A.T.; Kim, T.K.; Janjetovic, Z.; Brożyna, A.A.; Żmijewski, M.A.; Xu, H.; Sutter, T.R.; Tuckey, R.C.; Jetten, A.M.; Crossman, D.K. Differential and Overlapping Effects of 20,23(OH)2D3 and 1,25(OH)2D3 on Gene Expression in Human Epidermal Keratinocytes: Identification of AhR as an Alternative Receptor for 20,23(OH)2D3. Int. J. Mol. Sci. 2018, 19, 3072. [Google Scholar] [CrossRef]
  16. Slominski, A.T.; Kim, T.K.; Qayyum, S.; Song, Y.; Janjetovic, Z.; Oak, A.S.W.; Slominski, R.M.; Raman, C.; Stefan, J.; Mier-Aguilar, C.A.; et al. Vitamin D and lumisterol derivatives can act on liver X receptors (LXRs). Sci. Rep. 2021, 11, 8002. [Google Scholar] [CrossRef]
  17. Slominski, A.T.; Chaiprasongsuk, A.; Janjetovic, Z.; Kim, T.K.; Stefan, J.; Slominski, R.M.; Hanumanthu, V.S.; Raman, C.; Qayyum, S.; Song, Y.; et al. Photoprotective Properties of Vitamin D and Lumisterol Hydroxyderivatives. Cell Biochem. Biophys. 2020, 78, 165–180. [Google Scholar] [CrossRef] [PubMed]
  18. Costa, B.; Pini, S.; Gabelloni, P.; Da Pozzo, E.; Abelli, M.; Lari, L.; Preve, M.; Lucacchini, A.; Cassano, G.B.; Martini, C. The spontaneous Ala147Thr amino acid substitution within the translocator protein influences pregnenolone production in lymphomonocytes of healthy individuals. Endocrinology 2009, 150, 5438–5445. [Google Scholar] [CrossRef]
  19. Talaber, G.; Jondal, M.; Okret, S. Local glucocorticoid production in the thymus. Steroids 2015, 103, 58–63. [Google Scholar] [CrossRef]
  20. Slominski, R.M.; Tuckey, R.C.; Manna, P.R.; Jetten, A.M.; Postlethwaite, A.; Raman, C.; Slominski, A.T. Extra-adrenal glucocorticoid biosynthesis: Implications for autoimmune and inflammatory disorders. Genes Immun. 2020, 21, 150–168. [Google Scholar] [CrossRef] [PubMed]
  21. Slominski, R.M.; Raman, C.; Elmets, C.; Jetten, A.M.; Slominski, A.T.; Tuckey, R.C. The significance of CYP11A1 expression in skin physiology and pathology. Mol. Cell Endocrinol. 2021, 530, 111238. [Google Scholar] [CrossRef] [PubMed]
  22. Slominski, A.T.; Kim, T.K.; Slominski, R.M.; Song, Y.; Janjetovic, Z.; Podgorska, E.; Reddy, S.B.; Song, Y.; Raman, C.; Tang, E.K.Y.; et al. Metabolic activation of tachysterol3 to biologically active hydroxyderivatives that act on VDR, AhR, LXRs, and PPARγ receptors. FASEB J. 2022, 36, e22451. [Google Scholar] [CrossRef]
  23. Dusso, A.S.; Brown, A.J.; Slatopolsky, E. Vitamin D. Am. J. Physiol. Renal. Physiol. 2005, 289, F8–F28. [Google Scholar] [CrossRef] [PubMed]
  24. Nagata, A.; Akagi, Y.; Asano, L.; Kotake, K.; Kawagoe, F.; Mendoza, A.; Masoud, S.S.; Usuda, K.; Yasui, K.; Takemoto, Y.; et al. Synthetic Chemical Probes That Dissect Vitamin D Activities. ACS Chem. Biol. 2019, 14, 2851–2858. [Google Scholar] [CrossRef] [PubMed]
  25. Yamamoto, E.; Jørgensen, T.N. Immunological effects of vitamin D and their relations to autoimmunity. J. Autoimmun. 2019, 100, 7–16. [Google Scholar] [CrossRef] [PubMed]
  26. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin, D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef]
  27. Kamen, D.L.; Tangpricha, V. Vitamin D and molecular actions on the immune system: Modulation of innate and autoimmunity. J. Mol. Med. 2010, 88, 441–450. [Google Scholar] [CrossRef]
  28. Hollis, B.W. Assessment of vitamin D nutritional and hormonal status: What to measure and how to do it. Calcif. Tissue Int. 1996, 58, 4–5. [Google Scholar] [CrossRef]
  29. Aranow, C. Vitamin D and the Immune System. J. Investig. Med. 2011, 59, 881–886. [Google Scholar] [CrossRef]
  30. Charoenngam, N.; Shirvani, A.; Holick, M.F. Vitamin D for skeletal and non-skeletal health: What we should know. J. Clin. Orthop. Trauma 2019, 10, 1082–1093. [Google Scholar] [CrossRef]
  31. Charoenngam, N.; Holick, M.F. Immunologic Effects of Vitamin D on Human Health and Disease. Nutrients 2020, 12, 2097. [Google Scholar] [CrossRef]
  32. Saponaro, F.; Saba, A.; Zucchi, R. An Update on Vitamin D Metabolism. Int. J. Mol. Sci. 2020, 21, 6573. [Google Scholar] [CrossRef] [PubMed]
  33. Bergwitz, C.; Jüppner, H. Regulation of phosphate homeostasis by PTH, vitamin D, and FGF23. Annu. Rev. Med. 2010, 61, 91–104. [Google Scholar] [CrossRef] [PubMed]
  34. Charoenngam, N.; Rujirachun, P.; Holick, M.F.; Ungprasert, P. Oral vitamin D3 supplementation increases serum fibroblast growth factor 23 concentration in vitamin D-deficient patients: A systematic review and meta-analysis. Osteoporos. Int. 2019, 30, 2183–2193. [Google Scholar] [CrossRef] [PubMed]
  35. Binkley, N.; Carter, G.D. Toward Clarity in Clinical Vitamin D Status Assessment: 25(OH)D Assay Standardization. Endocrinol. Metab. Clin. N. Am. 2017, 46, 885–899. [Google Scholar] [CrossRef]
  36. Jiménez-Sousa, M.Á.; Martínez, I.; Medrano, L.M.; Fernández-Rodríguez, A.; Resino, S. Vitamin D in Human Immunodeficiency Virus Infection: Influence on Immunity and Disease. Front. Immunol. 2018, 9, 458. [Google Scholar] [CrossRef]
  37. Carlberg, C.; Muñoz, A. An update on vitamin D signaling and cancer. Semin. Cancer Biol. 2022, 79, 217–230. [Google Scholar] [CrossRef]
  38. Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef]
  39. Ross, A.C.; Manson, J.E.; Abrams, S.A.; Aloia, J.F.; Brannon, P.M.; Clinton, S.K.; Durazo-Arvizu, R.A.; Gallagher, J.C.; Gallo, R.L.; Jones, G.; et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: What clinicians need to know. J. Clin. Endocrinol. Metab. 2011, 96, 53–58. [Google Scholar] [CrossRef]
  40. Kumar, J.; Muntner, P.; Kaskel, F.J.; Hailpern, S.M.; Melamed, M.L. Prevalence and associations of 25-hydroxyvitamin D deficiency in US children: NHANES 2001-2004. Pediatrics 2009, 124, e362–e370. [Google Scholar] [CrossRef] [Green Version]
  41. Parva, N.R.; Tadepalli, S.; Singh, P.; Qian, A.; Joshi, R.; Kandala, H.; Nookala, V.K.; Cheriyath, P. Prevalence of Vitamin D Deficiency and Associated Risk Factors in the US Population (2011–2012). Cureus 2018, 10, e2741. [Google Scholar] [CrossRef] [PubMed]
  42. Palacios, C.; Gonzalez, L. Is vitamin D deficiency a major global public health problem? J. Steroid Biochem. Mol. Biol. 2014, 144, 138–145. [Google Scholar] [CrossRef] [PubMed]
  43. Martens, P.-J.; Gysemans, C.; Verstuyf, A.; Mathieu, C. Vitamin D’s Effect on Immune Function. Nutrients 2020, 12, 1248. [Google Scholar] [CrossRef] [PubMed]
  44. Cashman, K.D.; Dowling, K.G.; Škrabáková, Z.; Gonzalez-Gross, M.; Valtueña, J.; De Henauw, S.; Moreno, L.; Damsgaard, C.T.; Michaelsen, K.F.; Mølgaard, C.; et al. Vitamin D deficiency in Europe: Pandemic? Am. J. Clin. Nutr. 2016, 103, 1033–1044. [Google Scholar] [CrossRef]
  45. Ganji, V.; Zhang, X.; Tangpricha, V. Serum 25-hydroxyvitamin D concentrations and prevalence estimates of hypovitaminosis D in the U.S. population based on assay-adjusted data. J. Nutr. 2012, 142, 498–507. [Google Scholar] [CrossRef]
  46. LeFevre, M.L. U.S. Preventive Services Task Force. Screening for vitamin D deficiency in adults: U.S. Preventive Services Task Force recommendation statement. Ann. Intern. Med. 2015, 162, 133–140. [Google Scholar] [CrossRef]
  47. Rockwell, M.; Kraak, V.; Hulver, M.; Epling, J. Clinical Management of Low Vitamin D: A Scoping Review of Physicians’ Practices. Nutrients 2018, 10, 493. [Google Scholar] [CrossRef]
  48. Pilz, S.; Zittermann, A.; Trummer, C.; Theiler-Schwetz, V.; Lerchbaum, E.; Keppel, M.H.; Grübler, M.R.; März, W.; Pandis, M. Vitamin D testing and treatment: A narrative review of current evidence. Endocr. Connect. 2019, 8, R27–R43. [Google Scholar] [CrossRef]
  49. Cashman, K.D.; Ritz, C.; Kiely, M.; Collaborators, O. Improved Dietary Guidelines for Vitamin D: Application of Individual Participant Data (IPD)-Level Meta-Regression Analyses. Nutrients 2017, 9, 469. [Google Scholar] [CrossRef]
  50. De Niet, S.; Coffiner, M.; Da Silva, S.; Jandrain, B.; Souberbielle, J.C.; Cavalier, E. A Randomized Study to Compare a Monthly to a Daily Administration of Vitamin D3 Supplementation. Nutrients 2018, 10, 659. [Google Scholar] [CrossRef] [Green Version]
  51. Giusti, A.; Barone, A.; Pioli, G.; Girasole, G.; Razzano, M.; Pizzonia, M.; Pedrazzoni, M.; Palummeri, E.; Bianchi, G. Heterogeneity in serum 25-hydroxy-vitamin D response to cholecalciferol in elderly women with secondary hyperparathyroidism and vitamin D deficiency. J. Am. Geriatr. Soc. 2010, 58, 1489–1495. [Google Scholar] [CrossRef] [PubMed]
  52. Mittal, M.; Yadav, V.; Khadgawat, R.; Kumar, M.; Sherwani, P. Efficacy and Safety of 90,000 IU versus 300,000 IU Single Dose Oral Vitamin D in Nutritional Rickets: A Randomized Controlled Trial. Indian J. Endocrinol. Metab. 2018, 22, 760–765. [Google Scholar] [CrossRef] [PubMed]
  53. Mittal, H.; Rai, S.; Shah, D.; Mehrotra, G.; Malhotra, R.K. 300,000 IU or 600,000 IU of oral vitamin D3 for treatment of nutritional rickets: A randomized controlled trial. Indian Pediatr. 2014, 51, 265–272. [Google Scholar] [CrossRef] [PubMed]
  54. Ish-Shalom, S.; Segal, E.; Salganik, T.; Raz, B.; Bromberg, I.L.; Vieth, R. Comparison of Daily, Weekly, and Monthly Vitamin D3 in Ethanol Dosing Protocols for Two Months in Elderly Hip Fracture Patients. J. Clin. Endocrinol. Metab. 2008, 93, 3430–3435. [Google Scholar] [CrossRef]
  55. Chel, V.; Wijnhoven, H.A.H.; Smit, J.H.; Ooms, M.; Lips, P. Efficacy of different doses and time intervals of oral vitamin D supplementation with or without calcium in elderly nursing home residents. Osteoporos. Int. 2008, 19, 663–671. [Google Scholar] [CrossRef]
  56. Bacon, C.J.; Gamble, G.D.; Horne, A.M.; Scott, M.A.; Reid, I.R. High-dose oral vitamin D3 supplementation in the elderly. Osteoporos. Int. 2009, 20, 1407–1415. [Google Scholar] [CrossRef]
  57. Schleck, M.L.; Souberbielle, J.C.; Jandrain, B.; Da Silva, S.; De Niet, S.; Vanderbist, F.; Scheen, A.; Scheen, A.; Cavalier, E. A Randomized, Double-Blind, Parallel Study to Evaluate the Dose-Response of Three Different Vitamin D Treatment Schemes on the 25-Hydroxyvitamin D Serum Concentration in Patients with Vitamin D Deficiency. Nutrients 2015, 7, 5413–5422. [Google Scholar] [CrossRef]
  58. Sanders, K.M.; Stuart, A.L.; Williamson, E.J.; Simpson, A.J.; Kotowicz, M.A.; Young, D. Annual high-dose oral vitamin D and falls and fractures in older women: A randomized controlled trial. JAMA 2010, 303, 1815–1822. [Google Scholar] [CrossRef]
  59. Bouillon, R. Comparative analysis of nutritional guidelines for vitamin D. Nat. Rev. Endocrinol. 2017, 13, 466–479. [Google Scholar] [CrossRef]
  60. Bischoff-Ferrari, H.A.; Vellas, B.; Rizzoli, R.; Kressig, R.W.; da Silva, J.A.P.; Blauth, M.; Felson, D.T.; McCloskey, E.V.; Watzl, B.; Hofbauer, L.C.; et al. 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, 1855–1868. [Google Scholar] [CrossRef]
  61. Bouillon, R.; Manousaki, D.; Rosen, C.; Trajanoska, K.; Rivadeneira, F.; Richards, J.B. The health effects of vitamin D supplementation: Evidence from human studies. Nat. Rev. Endocrinol. 2022, 18, 96–110. [Google Scholar] [CrossRef] [PubMed]
  62. Hahn, J.; Cook, N.R.; Alexander, E.K.; Friedman, S.; Walter, J.; Bubes, V.; Kotler, G.; Lee, I.M.; Manson, J.E.; Costenbader, K.H. Vitamin D and marine omega 3 fatty acid supplementation and incident autoimmune disease: VITAL randomized controlled trial. BMJ 2022, 376, e066452. [Google Scholar] [CrossRef] [PubMed]
  63. Billington, E.O.; Burt, L.A.; Rose, M.S.; Davison, E.M.; Gaudet, S.; Kan, M.; Boyd, S.K.; Hanley, D.A. Safety of High-Dose Vitamin D Supplementation: Secondary Analysis of a Randomized Controlled Trial. J. Clin. Endocrinol. Metab. 2020, 105, dgz212. [Google Scholar] [CrossRef] [PubMed]
  64. Kiely, M.; Cashman, K.D. The ODIN project: Development of food-based approaches for prevention ofvitamin D deficiency throughout life. Nutr. Bull. 2015, 40, 235–246. [Google Scholar] [CrossRef]
  65. Kiely, M.; Cashman, K.D. Summary Outcomes of the ODIN Project on Food Fortification for Vitamin D Deficiency Prevention. Int. J. Environ. Res. Public Health 2018, 15, 2342. [Google Scholar] [CrossRef]
  66. Mortensen, C.; Damsgaard, C.T.; Hauger, H.; Ritz, C.; Lanham-New, S.A.; Smith, T.J.; Hennessy, Á.; Dowling, K.; Cashman, K.D.; Kiely, M.; et al. Estimation of the dietary requirement for vitamin D in white children aged 4–8 y: A randomized, controlled, dose-response trial. Am. J. Clin. Nutr. 2016, 104, 1310–1317. [Google Scholar] [CrossRef]
  67. Smith, T.J.; Tripkovic, L.; Damsgaard, C.T.; Molgaard, C.; Ritz, C.; Wilson-Barnes, S.L.; Dowling, K.G.; Hennessy, Á.; Cashman, K.D.; Kiely, M.; et al. Estimation of the dietary requirement for vitamin D in adolescents aged 14–18 y: A dose-response, double-blind, randomized placebo-controlled trial. Am. J. Clin. Nutr. 2016, 104, 1301–1309. [Google Scholar] [CrossRef]
  68. Adebayo, F.A.; Itkonen, S.T.; Ohman, T.; Skaffari, E.; Saarnio, E.M.; Erkkola, M.; Cashman, K.D.; Lamberg-Allardt, C. Vitamin D intake, serum 25-hydroxyvitamin D status and response to moderate vitamin D3 supplementation: A randomised controlled trial in East African and Finnish women. Br. J. Nutr. 2018, 119, 431–441. [Google Scholar] [CrossRef]
  69. Cashman, K.D.; Ritz, C.; Adebayo, F.A.; Dowling, K.G.; Itkonen, S.T.; Öhman, T.; Skaffari, E.; Saarnio, E.M.; Kiely, M.; Lamberg-Allardt, C. Differences in the dietary requirement for vitamin D among Caucasian and East African women at Northern latitude. Eur. J. Nutr. 2018, 58, 2281–2291. [Google Scholar] [CrossRef]
  70. O’Callaghan, K.M.; Hennessy, Á.; Hull, G.L.; Healy, K.; Ritz, C.; Kenny, L.C.; Cashman, K.D.; Kiely, M.E. Estimation of the maternal vitamin D intake that maintains circulating 25-hydroxyvitamin D in late gestation at a concentration sufficient to keep umbilical cord sera ≥25–30 nmol/L: A dose-response, double-blind, randomized placebo-controlled trial in pregnant women at northern latitude. Am. J. Clin. Nutr. 2018, 108, 77–91. [Google Scholar] [CrossRef] [Green Version]
  71. Al Khalifah, R.; Alsheikh, R.; Alnasser, Y.; Alsheikh, R.; Alhelali, N.; Naji, A.; Al Backer, N. The impact of vitamin D food fortification and health outcomes in children: A systematic review and meta-regression. Syst. Rev. 2020, 9, 144. [Google Scholar] [CrossRef] [PubMed]
  72. Ermann, J.; Fathman, C.G. Autoimmune diseases: Genes, bugs and failed regulation. Nat. Immunol. 2001, 2, 759–761. [Google Scholar] [CrossRef] [PubMed]
  73. Bouillon, R.; Suda, T. Vitamin D: Calcium and bone homeostasis during evolution. Bonekey Rep. 2014, 3, 480. [Google Scholar] [CrossRef] [PubMed]
  74. Martineau, A.R.; Wilkinson, K.A.; Newton, S.M.; Floto, R.A.; Norman, A.W.; Skolimowska, K.; Davidson, R.N.; Sørensen, O.E.; Kampmann, B.; Griffiths, C.J.; et al. IFN-gamma- and TNF-independent vitamin D-inducible human suppression of mycobacteria: The role of cathelicidin LL-37. J. Immunol. 2007, 178, 7190–7198. [Google Scholar] [CrossRef] [PubMed]
  75. Bhalla, A.K.; Amento, E.P.; Clemens, T.L.; Holick, M.F.; Krane, S.M. Specific high-affinity receptors for 1,25-dihydroxyvitamin D3 in human peripheral blood mononuclear cells: Presence in monocytes and induction in T lymphocytes following activation. J. Clin. Endocrinol. Metab. 1983, 57, 1308–1310. [Google Scholar] [CrossRef]
  76. Cadranel, J.; Garabedian, M.; Milleron, B.; Guillozo, H.; Akoun, G.; Hance, A.J. 1,25(OH)2D2 production by T lymphocytes and alveolar macrophages recovered by lavage from normocalcemic patients with tuberculosis. J. Clin. Invest 1990, 85, 1588–1593. [Google Scholar] [CrossRef]
  77. Chen, S.; Sims, G.P.; Chen, X.X.; Gu, Y.Y.; Chen, S.; Lipsky, P.E. Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation. J. Immunol. 2007, 179, 1634–1647. [Google Scholar] [CrossRef]
  78. Overbergh, L.; Decallonne, B.; Valckx, D.; Verstuyf, A.; Depovere, J.; Laureys, J.; Rutgeerts, O.; Saint-Arnaud, R.; Bouillon, R.; Mathieu, C.    Identification and immune regulation of 25-hydroxyvitamin D-1-alpha-hydroxylase in murine macrophages. Clin. Exp. Immunol. 2000, 120, 139–146. [Google Scholar] [CrossRef]
  79. Hewison, M.; Freeman, L.; Hughes, S.V.; Evans, K.N.; Bland, R.; Eliopoulos, A.G.; Kilby, M.D.; Moss, P.A.H.; Chakraverty, R. Differential regulation of vitamin D receptor and its ligand in human monocyte-derived dendritic cells. J. Immunol. 2003, 170, 5382–5390. [Google Scholar] [CrossRef]
  80. Borba, V.Z.C.; Vieira, J.G.H.; Kasamatsu, T.; Radominski, S.C.; Sato, E.I.; Lazaretti-Castro, M. Vitamin D deficiency in patients with active systemic lupus erythematosus. Osteoporos. Int. 2009, 20, 427–433. [Google Scholar] [CrossRef]
  81. Guardiola, J.; Xiol, X.; Nolla, J.M. Influence of vitamin D receptor gene polymorphism on bone mineral density in primary biliary cirrhosis. Gastroenterology 2000, 119, 599. [Google Scholar] [CrossRef] [PubMed]
  82. Seydel, S.; Beilfuss, A.; Kahraman, A.; Aksoy, K.; Gerken, G.; Akkiz, H.; Canbay, A. Vitamin D ameliorates stress ligand expression elicited by free fatty acids in the hepatic stellate cell line LX-2. Turk J. Gastroenterol. 2011, 22, 400–407. [Google Scholar] [CrossRef] [PubMed]
  83. Jain, S.K.; Micinski, D. Vitamin D upregulates glutamate cysteine ligase and glutathione reductase, and GSH formation, and decreases ROS and MCP-1 and IL-8 secretion in high-glucose exposed U937 monocytes. Biochem. Biophys. Res. Commun. 2013, 437, 7–11. [Google Scholar] [CrossRef] [PubMed]
  84. Alvarez, J.A.; Chowdhury, R.; Jones, D.P.; Martin, G.S.; Brigham, K.L.; Binongo, J.N.; Ziegler, T.R.; Tangpricha, V. Vitamin D status is independently associated with plasma glutathione and cysteine thiol/disulphide redox status in adults. Clin. Endocrinol. 2014, 81, 458–466. [Google Scholar] [CrossRef] [PubMed]
  85. Abramovitch, S.; Dahan-Bachar, L.; Sharvit, E.; Weisman, Y.; Tov, A.B.; Brazowski, E.; Reif, S. Vitamin D inhibits proliferation and profibrotic marker expression in hepatic stellate cells and decreases thioacetamide-induced liver fibrosis in rats. Gut 2011, 60, 1728–1737. [Google Scholar] [CrossRef]
  86. Abramovitch, S.; Sharvit, E.; Weisman, Y.; Bentov, A.; Brazowski, E.; Cohen, G.; Volovelsky, O.; Reif, S. Vitamin D inhibits development of liver fibrosis in an animal model but cannot ameliorate established cirrhosis. Am. J. Physiol. Gastrointest. Liver. Physiol. 2015, 308, G112–G120. [Google Scholar] [CrossRef]
  87. Reiter, F.P.; Hohenester, S.; Nagel, J.M.; Wimmer, R.; Wottke, L.; Makeschin, M.-C.; Mayr, D.; Rust, C.; Trauner, M.; Denk, G.U. 1,25-(OH)2-vitamin D3 prevents activation of hepatic stellate cells in vitro and ameliorates inflammatory liver damage but not fibrosis in the Abcb4(-/-) model. Biochem. Biophys. Res. Commun. 2015, 459, 227–233. [Google Scholar] [CrossRef]
  88. Zhang, Y.; Leung, D.Y.M.; Richers, B.N.; Liu, Y.; Remigio, L.K.; Riches, D.W.; Goleva, E. Vitamin D Inhibits Monocyte/macrophage Pro-inflammatory Cytokine Production by Targeting Mitogen-Activated Protein Kinase Phosphatase 1. J. Immunol. 2012, 188, 2127–2135. [Google Scholar] [CrossRef]
  89. Walker, V.P.; Modlin, R.L. The vitamin D connection to pediatric infections and immune function. Pediatr. Res. 2009, 65, 106R–113R. [Google Scholar] [CrossRef]
  90. Lemire, J.M. Immunomodulatory role of 1,25-dihydroxyvitamin D3. J. Cell. Biochem. 1992, 49, 26–31. [Google Scholar] [CrossRef]
  91. Arnson, Y.; Amital, H.; Shoenfeld, Y. Vitamin D and autoimmunity: New aetiological and therapeutic considerations. Ann. Rheum. Dis. 2007, 66, 1137–1142. [Google Scholar] [CrossRef] [PubMed]
  92. Adams, J.S.; Hewison, M. Unexpected actions of vitamin D: New perspectives on the regulation of innate and adaptive immunity. Nat. Clin. Pract. Endocrinol. Metab. 2008, 4, 80–90. [Google Scholar] [CrossRef] [PubMed]
  93. Grama, A.; Burac, L.; Aldea, C.O.; Bulata, B.; Delean, D.; Samasca, G.; Abrudan, C.; Sirbe, C.; Pop, T.L. Vitamin D-Binding Protein (Gc-Globulin) in Acute Liver Failure in Children. Diagnostics 2020, 10, 278. [Google Scholar] [CrossRef] [PubMed]
  94. Szabo, R.; Petri, C.; Tranc, S. Vitamin D and Iron Levels Correlate Weakly with Hepcidin Levels in Postoperative Patients with Digestive Neoplasms Undergoing Open Abdominal Surgery. Eur. Rev. Med. Pharmacol. Col. 2021, 25, 3530–3535. [Google Scholar] [CrossRef]
  95. Trinchieri, G.; Sher, A. Cooperation of Toll-like receptor signals in innate immune defence. Nat. Rev. Immunol. 2007, 7, 179–190. [Google Scholar] [CrossRef]
  96. Liu, P.T.; Stenger, S.; Tang, D.H.; Modlin, R.L. Cutting Edge: Vitamin D-Mediated Human Antimicrobial Activity against Mycobacterium tuberculosis Is Dependent on the Induction of Cathelicidin. J. Immunol. 2007, 179, 2060–2063. [Google Scholar] [CrossRef]
  97. Barlow, P.G.; Svoboda, P.; Mackellar, A.; Nash, A.A.; York, I.A.; Pohl, J.; Davidson, D.J.; Donis, R.O. Antiviral activity and increased host defense against influenza infection elicited by the human cathelicidin LL-37. PLoS ONE 2011, 6, e25333. [Google Scholar] [CrossRef]
  98. Shahmiri, M.; Enciso, M.; Adda, C.G.; Smith, B.J.; Perugini, M.A.; Mechler, A. Membrane Core-Specific Antimicrobial Action of Cathelicidin LL-37 Peptide Switches Between Pore and Nanofibre Formation. Sci. Rep. 2016, 6, 38184. [Google Scholar] [CrossRef]
  99. Sousa, F.H.; Casanova, V.; Findlay, F.; Stevens, C.; Svoboda, P.; Pohl, J.; Proudfoot, L.; Barlow, P.G. Cathelicidins display conserved direct antiviral activity towards rhinovirus. Peptides 2017, 95, 76–83. [Google Scholar] [CrossRef]
  100. Tripathi, S.; Tecle, T.; Verma, A.; Crouch, E.; White, M.; Hartshorn, K.L. The human cathelicidin LL-37 inhibits influenza A viruses through a mechanism distinct from that of surfactant protein D or defensins. J. Gen. Virol. 2013, 94, 40–49. [Google Scholar] [CrossRef]
  101. Siddiqui, M.; Manansala, J.S.; Abdulrahman, H.A.; Nasrallah, G.K.; Smatti, M.K.; Younes, N.; Althani, A.A.; Yassine, H.M. Immune Modulatory Effects of Vitamin D on Viral Infections. Nutrients 2020, 12, 2879. [Google Scholar] [CrossRef] [PubMed]
  102. Sharma, O.P. Hypercalcemia in granulomatous disorders: A clinical review. Curr. Opin. Pulm. Med. 2000, 6, 442–447. [Google Scholar] [CrossRef] [PubMed]
  103. Papapoulos, S.E.; Fraher, L.J.; Sandler, L.M.; Clemens, T.L.; Lewin, I.G.; O’Riordan, J.L.H. 1, 25-Dihydroxycholecalciferol In The Pathogenesis Of The Hypercalcæmia Of Sarcoidosis. Lancet 1979, 313, 627–630. [Google Scholar] [CrossRef]
  104. Wang, T.T.; Nestel, F.P.; Bourdeau, V.; Nagai, Y.; Wang, Q.; Liao, J.; Tavera-Mendoza, L.; Lin, R.; Hanrahan, J.W.; Mader, S.; et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J. Immunol. 2004, 173, 2909–2912. [Google Scholar] [CrossRef]
  105. Gombart, A.F. The vitamin D–antimicrobial peptide pathway and its role in protection against infection. Future Microbiol. 2009, 4, 1151. [Google Scholar] [CrossRef] [PubMed]
  106. Penna, G.; Adorini, L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J. Immunol. 2000, 164, 2405–2411. [Google Scholar] [CrossRef]
  107. Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef]
  108. Ao, T.; Kikuta, J.; Ishii, M. The Effects of Vitamin D on Immune System and Inflammatory Diseases. Biomolecules 2021, 11, 1624. [Google Scholar] [CrossRef]
  109. Ota, K.; Dambaeva, S.; Kim, M.W.I.; Han, A.-R.; Fukui, A.; Gilman-Sachs, A.; Beaman, K.; Kwak-Kim, J. 1,25-Dihydroxy-vitamin D3 regulates NK-cell cytotoxicity, cytokine secretion, and degranulation in women with recurrent pregnancy losses. Eur. J. Immunol. 2015, 45, 3188–3199. [Google Scholar] [CrossRef]
  110. Dickie, L.J.; Church, L.D.; Coulthard, L.R.; Mathews, R.J.; Emery, P.; McDermott, M.F. Vitamin D3 down-regulates intracellular Toll-like receptor 9 expression and Toll-like receptor 9-induced IL-6 production in human monocytes. Rheumatology 2010, 49, 1466–1471. [Google Scholar] [CrossRef] [Green Version]
  111. Adorini, L.; Penna, G. Induction of Tolerogenic Dendritic Cells by Vitamin D Receptor Agonists. In Dendritic Cells. Handbook of Experimental Pharmacology; Lombardi, G., Riffo-Vasquez, Y., Eds.; Springer: Berlin/Heidelberg, Germany, 2009; pp. 251–273. [Google Scholar] [CrossRef]
  112. Széles, L.; Keresztes, G.; Töröcsik, D.; Balajthy, Z.; Krenács, L.; Póliska, S.; Steinmeyer, A.; Zuegel, U.; Pruenster, M.; Rot, A.; et al. 1,25-Dihydroxyvitamin D3 Is an Autonomous Regulator of the Transcriptional Changes Leading to a Tolerogenic Dendritic Cell Phenotype. J. Immunol. 2009, 182, 2074–2083. [Google Scholar] [CrossRef] [PubMed]
  113. Piemonti, L.; Monti, P.; Sironi, M.; Fraticelli, P.; Leone, B.E.; Cin, E.D.; Allavena, P.; Di Carlo, V. Vitamin D3 affects differentiation, maturation, and function of human monocyte-derived dendritic cells. J. Immunol. 2000, 164, 4443–4451. [Google Scholar] [CrossRef]
  114. Cantorna, M.T.; Zhao, J.; Yang, L. Vitamin D, invariant natural killer T-cells and experimental autoimmune disease. Proc. Nutr. Soc. 2012, 71, 62–66. [Google Scholar] [CrossRef]
  115. Weeres, M.A.; Robien, K.; Ahn, Y.O.; Neulen, M.-L.; Bergerson, R.; Miller, J.S.; Verneris, M.R. The Effects of 1,25-Dihydroxyvitamin D3 on In Vitro Human NK Cell Development from Hematopoietic Stem Cells. J. Immunol. 2014, 193, 3456–3462. [Google Scholar] [CrossRef]
  116. Rook, G.A.; Steele, J.; Fraher, L.; Barker, S.; Karmali, R.; O’Riordan, J.; Stanford, J. Vitamin D3, gamma interferon, and control of proliferation of Mycobacterium tuberculosis by human monocytes. Immunology 1986, 57, 159–163. [Google Scholar] [PubMed]
  117. Carlberg, C. Nutrigenomics of Vitamin D. Nutrients 2019, 11, 676. [Google Scholar] [CrossRef] [PubMed]
  118. Zmijewski, M.A. Vitamin D and Human Health. Int. J. Mol. Sci. 2019, 20, 145. [Google Scholar] [CrossRef]
  119. Yim, S.; Dhawan, P.; Ragunath, C.; Christakos, S.; Diamond, G. Induction of cathelicidin in normal and CF bronchial epithelial cells by 1,25-dihydroxyvitamin D3. J. Cyst. Fibros. 2007, 6, 403–410. [Google Scholar] [CrossRef]
  120. Weber, G.; Heilborn, J.D.; Chamorro Jimenez, C.I.; Hammarsjo, A.; Törmä, H.; Stahle, M. Vitamin D induces the antimicrobial protein hCAP18 in human skin. J. Invest Dermatol. 2005, 124, 1080–1082. [Google Scholar] [CrossRef] [Green Version]
  121. Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef]
  122. Bishop, E.L.; Ismailova, A.; Dimeloe, S.; Hewison, M.; White, J.H. Vitamin D and Immune Regulation: Antibacterial, Antiviral, Anti-Inflammatory. JBMR Plus 2021, 5, e10405. [Google Scholar] [CrossRef]
  123. Kongsbak, M.; von Essen, M.R.; Levring, T.B.; Schjerling, P.; Woetmann, A.; Ødum, N.; Bonefeld, C.M.; Geisler, C. Vitamin D-binding protein controls T cell responses to vitamin D. BMC Immunol. 2014, 15, 35. [Google Scholar] [CrossRef] [PubMed]
  124. Hewison, M. Vitamin D and the intracrinology of innate immunity. Mol. Cell. Endocrinol. 2010, 321, 103–111. [Google Scholar] [CrossRef]
  125. Reichel, H.; Koeffler, H.P.; Bishop, J.E.; Norman, A.W. 25-Hydroxyvitamin D3 Metabolism by Lipopolysaccharide-Stimulated Normal Human Macrophages. J. Clin. Endocrinol. Metab. 1987, 64, 1–9. [Google Scholar] [CrossRef] [PubMed]
  126. Islam, D.; Bandholtz, L.; Nilsson, J.; Christensson, B.; Agerberth, B.; Gudmundsson, G. Downregulation of bactericidal peptides in enteric infections: A novel immune escape mechanism with bacterial DNA as a potential regulator. Nat. Med. 2001, 7, 180–185. [Google Scholar] [CrossRef] [PubMed]
  127. Sadeghi, K.; Wessner, B.; Laggner, U.; Ploder, M.; Tamandl, D.; Friedl, J.; Zügel, U.; Steinmeyer, A.; Pollak, A.; Roth, E.; et al. Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur. J. Immunol. 2006, 36, 361–370. [Google Scholar] [CrossRef]
  128. Sakaki, T.; Kagawa, N.; Yamamoto, K.; Inouye, K. Metabolism of vitamin D3 by cytochromes P450. Front. Biosci. 2005, 10, 119–134. [Google Scholar] [CrossRef] [PubMed]
  129. Gibson, C.C.; Davis, C.T.; Zhu, W.; Bowman-Kirigin, J.A.; Walker, A.E.; Tai, Z.; Thomas, K.R.; Donato, A.J.; Lesniewski, L.A.; Li, D.Y. Dietary Vitamin D and Its Metabolites Non-Genomically Stabilize the Endothelium. PLoS ONE 2015, 10, e0140370. [Google Scholar] [CrossRef] [PubMed]
  130. Molinari, C.; Uberti, F.; Grossini, E.; Vacca, G.; Carda, S.; Invernizzi, M.; Cisari, C. 1α,25-Dihydroxycholecalciferol Induces Nitric Oxide Production in Cultured Endothelial Cells. Cell. Physiol. Biochem. 2011, 27, 661–668. [Google Scholar] [CrossRef]
  131. Andrukhova, O.; Slavic, S.; Zeitz, U.; Riesen, S.C.; Heppelmann, M.S.; Ambrisko, T.D.; Markovic, M.; Kuebler, W.M.; Erben, R.G. Vitamin D Is a Regulator of Endothelial Nitric Oxide Synthase and Arterial Stiffness in Mice. Mol. Endocrinol. 2014, 28, 53–64. [Google Scholar] [CrossRef]
  132. Ma, R.; Deng, X.L.; Du, G.L.; Li, C.; Xiao, S.; Aibibai, Y.; Zhu, J. Active vitamin D3, 1,25-(OH)2D3, protects against macrovasculopathy in a rat model of type 2 diabetes mellitus. Genet. Mol. Res. 2016, 15, gmr.15028113. [Google Scholar] [CrossRef]
  133. Kim, D.H.; Meza, C.A.; Clarke, H.; Kim, J.S.; Hickner, R.C. Vitamin D and Endothelial Function. Nutrients 2020, 12, 575. [Google Scholar] [CrossRef]
  134. Vila Cuenca, M.; Ferrantelli, E.; Meinster, E.; Pouw, S.M.; Kovačević, I.; de Menezes, R.X.; Niessen, H.W.; Beelen, R.H.G.; Hordijk, P.L.; Vervloet, M.G. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J. Am. Heart Assoc. 2018, 7, e008776. [Google Scholar] [CrossRef]
  135. Ren, S.; Nguyen, L.; Wu, S.; Encinas, C.; Adams, J.S.; Hewison, M. Alternative splicing of vitamin D-24-hydroxylase: A novel mechanism for the regulation of extrarenal 1,25-dihydroxyvitamin D synthesis. J. Biol. Chem. 2005, 280, 20604–20611. [Google Scholar] [CrossRef]
  136. Schauber, J.; Dorschner, R.A.; Coda, A.B.; Büchau, A.S.; Liu, P.T.; Kiken, D.; Helfrich, Y.R.; Kang, S.; Elalieh, H.Z.; Steinmeyer, A.; et al. Injury enhances TLR2 function and antimicrobial peptide expression through a vitamin D-dependent mechanism. J. Clin. Invest 2007, 117, 803–811. [Google Scholar] [CrossRef]
  137. Lee, C.; Lau, E.; Chusilp, S.; Li, B.; Zhu, H.; Yamoto, H.; Pierro, A. Protective effects of vitamin D against injury in intestinal epithelium. Pediatr. Surg. Int. 2019, 35, 1395–1401. [Google Scholar] [CrossRef]
  138. He, Y.; Wu, W.; Wu, S.; Zheng, H.-M.; Li, P.; Sheng, H.-F.; Chen, M.-X.; Chen, Z.-H.; Ji, G.-Y.; Zheng, Z.-D.-X.; et al. Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome 2018, 6, 172. [Google Scholar] [CrossRef]
  139. Wang, T.T.; Dabbas, B.; Laperriere, D.; Bitton, A.J.; Soualhine, H.; Tavera-Mendoza, L.E.; Dionne, S.; Servant, M.J.; Bitton, A.; Seidman, E.G.; et al. Direct and Indirect Induction by 1,25-Dihydroxyvitamin D3 of the NOD2/CARD15-Defensin β2 Innate Immune Pathway Defective in Crohn Disease. J. Biol. Chem. 2010, 285, 2227–2231. [Google Scholar] [CrossRef]
  140. Zhang, Y.-G.; Wu, S.; Sun, J. Vitamin D, vitamin D receptor and tissue barriers. Tissue Barriers 2013, 1, e23118. [Google Scholar] [CrossRef]
  141. Su, D.; Nie, Y.; Zhu, A.; Zishuo, C.; Wu, P.; Zhang, L.; Luo, M.; Sun, Q.; Cai, L.; Lai, Y.; et al. Vitamin D Signaling through Induction of Paneth Cell Defensins Maintains Gut Microbiota and Improves Metabolic Disorders and Hepatic Steatosis in Animal Models. Front. Physiol. 2016, 7, 498. [Google Scholar] [CrossRef]
  142. Fakhoury, H.M.A.; Kvietys, P.R.; AlKattan, W.; Anouti, F.A.; Elahi, M.A.; Karras, S.N.; Grant, W.B. Vitamin D and intestinal homeostasis: Barrier, microbiota, and immune modulation. J. Steroid Biochem. Mol. Biol. 2020, 200, 105663. [Google Scholar] [CrossRef] [PubMed]
  143. Cantorna, M.T.; Snyder, L.; Lin, Y.D.; Yang, L. Vitamin D and 1,25(OH)2D Regulation of T cells. Nutrients 2015, 7, 3011–3021. [Google Scholar] [CrossRef] [PubMed]
  144. Hewison, M. An update on vitamin D and human immunity. Clin. Endocrinol. 2012, 76, 315–325. [Google Scholar] [CrossRef]
  145. Provvedini, D.M.; Tsoukas, C.D.; Deftos, L.J.; Manolagas, S.C. 1,25-dihydroxyvitamin D3 receptors in human leukocytes. Science 1983, 221, 1181–1183. [Google Scholar] [CrossRef] [PubMed]
  146. Amento, E.P.; Bhalla, A.K.; Kurnick, J.T.; Kradin, R.L.; Clemens, T.L.; Holick, S.A.; Holick, M.F.; Krane, S.M. 1 alpha,25-dihydroxyvitamin D3 induces maturation of the human monocyte cell line U937, and, in association with a factor from human T lymphocytes, augments production of the monokine, mononuclear cell factor. J. Clin. Investig. 1984, 73, 731–739. [Google Scholar] [CrossRef] [PubMed]
  147. Provvedini, D.M.; Tsoukas, C.D.; Deftos, L.J.; Manolagas, S.C. 1 alpha,25-Dihydroxyvitamin D3-binding macromolecules in human B lymphocytes: Effects on immunoglobulin production. J. Immunol. 1986, 136, 2734–2740. [Google Scholar]
  148. Lemire, J.M.; Adams, J.S.; Sakai, R.; Jordan, S.C. 1 alpha,25-dihydroxyvitamin D3 suppresses proliferation and immunoglobulin production by normal human peripheral blood mononuclear cells. J. Clin. Investig. 1984, 74, 657–661. [Google Scholar] [CrossRef]
  149. Barwick, B.G.; Scharer, C.D.; Martinez, R.J.; Price, M.J.; Wein, A.N.; Haines, R.R.; Bally, A.P.R.; Kohlmeier, J.E.; Boss, J.M. B cell activation and plasma cell differentiation are inhibited by de novo DNA methylation. Nat. Commun. 2018, 9, 1900. [Google Scholar] [CrossRef]
  150. Geldmeyer-Hilt, K.; Heine, G.; Hartmann, B.; Baumgrass, R.; Radbruch, A.; Worm, M. 1,25-dihydroxyvitamin D3 impairs NF-κB activation in human naïve B cells. Biochem. Biophys. Res. Commun. 2011, 407, 699–702. [Google Scholar] [CrossRef]
  151. Lemire, J.M.; Adams, J.S.; Kermani-Arab, V.; Bakke, A.C.; Sakai, R.; Jordan, S.C. 1,25-Dihydroxyvitamin D3 suppresses human T helper/inducer lymphocyte activity in vitro. J. Immunol. 1985, 134, 3032–3035. [Google Scholar]
  152. Abbas, A.K.; Murphy, K.M.; Sher, A. Functional diversity of helper T lymphocytes. Nature 1996, 383, 787–793. [Google Scholar] [CrossRef] [PubMed]
  153. Boonstra, A.; Barrat, F.J.; Crain, C.; Heath, V.L.; Savelkoul, H.F.; O’Garra, A. 1alpha,25-Dihydroxyvitamin d3 has a direct effect on naive CD4+ T cells to enhance the development of Th2 cells. J. Immunol. 2001, 167, 4974–4980. [Google Scholar] [CrossRef] [PubMed]
  154. Lemire, J.M.; Archer, D.C.; Beck, L.; Spiegelberg, H.L. Immunosuppressive actions of 1,25-dihydroxyvitamin D3: Preferential inhibition of Th1 functions. J. Nutr. 1995, 125, 1704S–1708S. [Google Scholar] [CrossRef] [PubMed]
  155. Urry, Z.; Xystrakis, E.; Richards, D.F.; McDonald, J.; Sattar, Z.; Cousins, D.J.; Corrigan, C.J.; Hickman, E.; Brown, Z.; Hawrylowicz, C.M. Ligation of TLR9 induced on human IL-10–secreting Tregs by 1α,25-dihydroxyvitamin D3 abrogates regulatory function. J. Clin. Investig. 2009, 119, 387–398. [Google Scholar] [CrossRef]
  156. Tang, J.; Zhou, R.; Luger, D.; Zhu, W.; Silver, P.B.; Grajewski, R.S.; Su, S.-B.; Chan, C.-C.; Adorini, L.; Caspi, R.R. Calcitriol Suppresses Antiretinal Autoimmunity through Inhibitory Effects on the Th17 Effector Response. J. Immunol. 2009, 182, 4624–4632. [Google Scholar] [CrossRef]
  157. Shirakawa, A.K.; Nagakubo, D.; Hieshima, K.; Nakayama, T.; Jin, Z.; Yoshie, O. 1,25-Dihydroxyvitamin D3 Induces CCR10 Expression in Terminally Differentiating Human B Cells. J. Immunol. 2008, 180, 2786–2795. [Google Scholar] [CrossRef]
  158. Heine, G.; Niesner, U.; Chang, H.D.; Steinmeyer, A.; Zügel, U.; Zuberbier, T.; Radbruch, A.; Worm, M. 1,25-dihydroxyvitamin D3 promotes IL-10 production in human B cells. Eur. J. Immunol. 2008, 38, 2210–2218. [Google Scholar] [CrossRef]
  159. Jeffery, L.E.; Burke, F.; Mura, M.; Jeffery, L.; Burke, F.; Mura, M.; Zheng, Y.; Qureshi, O.; Hewison, M.; Walker, L.; et al. 1,25-Dihydroxyvitamin D3 and IL-2 Combine to Inhibit T Cell Production of Inflammatory Cytokines and Promote Development of Regulatory T Cells Expressing CTLA-4 and FoxP3. J. Immunol. 2009, 183, 5458–5467. [Google Scholar] [CrossRef]
  160. Overbergh, L.; Decallonne, B.; Waer, M.; Rutgeerts, O.; Valckx, D.; Casteels, K.M.; Laureys, J.; Bouillon, R.; Mathieu, C. 1alpha,25-dihydroxyvitamin D3 induces an autoantigen-specific T-helper 1/T-helper 2 immune shift in NOD mice immunized with GAD65 (p524-543). Diabetes 2000, 49, 1301–1307. [Google Scholar] [CrossRef]
  161. Barrat, F.J.; Cua, D.J.; Boonstra, A.; Richards, D.F.; Crain, C.; Savelkoul, H.F.; de Waal-Malefyt, R.; Coffman, R.L.; Hawrylowicz, C.M.; O’Garra, A. In vitro generation of interleukin 10-producing regulatory CD4+ T cells is induced by immunosuppressive drugs and inhibited by T helper type 1 (Th1)- and Th2-inducing cytokines. J. Exp. Med. 2002, 195, 603–616. [Google Scholar] [CrossRef]
  162. Kongsbak, M.; Levring, T.; Geisler, C.; von Essen, M. The Vitamin D Receptor and T Cell Function. Front. Immunol. 2013, 4, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Sarkar, S.; Hewison, M.; Studzinski, G.P.; Li, Y.C.; Kalia, V. Role of vitamin D in cytotoxic T lymphocyte immunity to pathogens and cancer. Crit. Rev. Clin. Lab. Sci. 2016, 53, 132–145. [Google Scholar] [CrossRef] [PubMed]
  164. Mao, X.; Hu, B.; Zhou, Z.; Xing, X.; Wu, Y.; Gao, J.; He, Y.; Hu, Y.; Cheng, Q.; Gong, Q. Vitamin D levels correlate with lymphocyte subsets in elderly patients with age-related diseases. Sci. Rep. 2018, 8, 7708. [Google Scholar] [CrossRef] [PubMed]
  165. Eckard, A.R.; O’Riordan, M.A.; Rosebush, J.C.; Lee, S.T.; Habib, J.G.; Ruff, J.H.; Labbato, D.; Daniels, J.E.; Uribe-Leitz, M.; Tangpricha, V.; et al. Vitamin D Supplementation Decreases Immune Activation and Exhaustion in HIV-1-Infected Youth. Antivir. Ther. 2018, 23, 315–324. [Google Scholar] [CrossRef] [PubMed]
  166. Stallings, V.A.; Schall, J.I.; Hediger, M.L.; Zemel, B.S.; Tuluc, F.; Dougherty, K.A.; Samuel, J.L.; Rutstein, R.M. High-dose Vitamin D3 Supplementation in Children and Young Adults with HIV: A Randomized, Placebo-controlled Trial. Pediatr. Infect. Dis. J. 2015, 34, e32–e40. [Google Scholar] [CrossRef] [PubMed]
  167. Gregori, S.; Casorati, M.; Amuchastegui, S.; Smiroldo, S.; Davalli, A.M.; Adorini, L. Regulatory T cells induced by 1 alpha,25-dihydroxyvitamin D3 and mycophenolate mofetil treatment mediate transplantation tolerance. J. Immunol. 2001, 167, 1945–1953. [Google Scholar] [CrossRef]
  168. Gregori, S.; Giarratana, N.; Smiroldo, S.; Uskokovic, M.; Adorini, L. A 1α,25-Dihydroxyvitamin D3 Analog Enhances Regulatory T-Cells and Arrests Autoimmune Diabetes in NOD Mice. Diabetes 2002, 51, 1367–1374. [Google Scholar] [CrossRef]
  169. Penna, G.; Amuchastegui, S.; Giarratana, N.; Daniel, K.C.; Vulcano, M.; Sozzani, S.; Adorini, L. 1,25-Dihydroxyvitamin D3 selectively modulates tolerogenic properties in myeloid but not plasmacytoid dendritic cells. J. Immunol. 2007, 178, 145–153. [Google Scholar] [CrossRef]
  170. Fisher, S.A.; Rahimzadeh, M.; Brierley, C.; Gration, B.; Doree, C.; Kimber, C.E.; Plaza Cajide, A.; Lamikanra, A.A.; Roberts, D.J. The role of vitamin D in increasing circulating T regulatory cell numbers and modulating T regulatory cell phenotypes in patients with inflammatory disease or in healthy volunteers: A systematic review. PLoS ONE 2019, 14, e0222313. [Google Scholar] [CrossRef]
  171. Koivisto, O.; Hanel, A.; Carlberg, C. Key Vitamin D Target Genes with Functions in the Immune System. Nutrients 2020, 12, 1140. [Google Scholar] [CrossRef]
  172. Granucci, F.; Zanoni, I. Role of CD14 in host protection against infections and in metabolism regulation. Front. Cell. Infect. Microbiol. 2013, 3, 32. [Google Scholar] [CrossRef] [Green Version]
  173. Zanoni, I.; Tan, Y.; Gioia, M.D.; Springstead, J.R.; Kagan, J.C. By Capturing Inflammatory Lipids Released from Dying Cells, the Receptor CD14 Induces Inflammasome-Dependent Phagocyte Hyperactivation. Immunity 2017, 47, 697–709.e3. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, T.H.; Lee, B.; Kwon, E.; Choi, S.J.; Lee, Y.H.; Song, G.G.; Sohn, J.; Ji, J.D. Regulation of TREM-1 expression by 1,25-dihydroxyvitamin D3 in human monocytes/macrophages. Immunol. Lett. 2013, 154, 80–85. [Google Scholar] [CrossRef] [PubMed]
  175. Dower, K.; Ellis, D.K.; Saraf, K.; Jelinsky, S.A.; Lin, L.L. Innate Immune Responses to TREM-1 Activation: Overlap, Divergence, and Positive and Negative Cross-Talk with Bacterial Lipopolysaccharide. J. Immunol. 2008, 180, 3520–3534. [Google Scholar] [CrossRef]
  176. Okamura, Y.; Watari, M.; Jerud, E.S.; Young, D.W.; Ishizaka, S.T.; Rose, J.; Chow, J.C.; Strauss, J.F. The Extra Domain A of Fibronectin Activates Toll-like Receptor 4. J. Biol. Chem. 2001, 276, 10229–10233. [Google Scholar] [CrossRef]
  177. Loghmani, H.; Conway, E.M. Exploring traditional and nontraditional roles for thrombomodulin. Blood 2018, 132, 148–158. [Google Scholar] [CrossRef]
  178. Ma, C.Y.; Shi, G.Y.; Shi, C.S.; Kao, Y.C.; Lin, S.W.; Wu, H.L. Monocytic Thrombomodulin Triggers LPS- and Gram-Negative Bacteria-Induced Inflammatory Response. J. Immunol. 2012, 188, 6328–6337. [Google Scholar] [CrossRef]
  179. Heikkinen, S.; Väisänen, S.; Pehkonen, P.; Seuter, S.; Benes, V.; Carlberg, C. Nuclear hormone 1α,25-dihydroxyvitamin D3 elicits a genome-wide shift in the locations of VDR chromatin occupancy. Nucleic Acids Res. 2011, 39, 9181–9193. [Google Scholar] [CrossRef]
  180. Carlberg, C.; Seuter, S.; de Mello, V.D.F.; Schwab, U.; Voutilainen, S.; Pulkki, K.; Nurmi, T.; Virtanen, J.; Tuomainen, T.P.; Uusitupa, M. Primary Vitamin D Target Genes Allow a Categorization of Possible Benefits of Vitamin D3 Supplementation. PLoS ONE 2013, 8, e71042. [Google Scholar] [CrossRef]
  181. Park, M.; Liu, R.W.; An, H.; Geczy, C.L.; Thomas, P.S.; Tedla, N. A dual positive and negative regulation of monocyte activation by leukocyte Ig-like receptor B4 depends on the position of the tyrosine residues in its ITIMs. Innate Immun. 2017, 23, 381–391. [Google Scholar] [CrossRef]
  182. Chang, C.C.; Zhang, Q.Y.; Liu, Z.; Clynes, R.A.; Suciu-Foca, N.; Vlad, G. Downregulation of Inflammatory MicroRNAs by Ig-like Transcript 3 Is Essential for the Differentiation of Human CD8+ T Suppressor Cells. J. Immunol. 2012, 188, 3042–3052. [Google Scholar] [CrossRef] [Green Version]
  183. Waschbisch, A.; Sanderson, N.; Krumbholz, M.; Vlad, G.; Theil, D.; Schwab, S.; Mäurer, M.; Derfuss, T. Interferon Beta and Vitamin D Synergize to Induce Immunoregulatory Receptors on Peripheral Blood Monocytes of Multiple Sclerosis Patients. PLoS ONE 2014, 9, e115488. [Google Scholar] [CrossRef]
  184. Sutton, A.L.M.; Zhang, X.; Dowd, D.R.; Kharode, Y.P.; Komm, B.S.; MacDonald, P.N. Semaphorin 3B Is a 1,25-Dihydroxyvitamin D3-Induced Gene in Osteoblasts that Promotes Osteoclastogenesis and Induces Osteopenia in Mice. Mol. Endocrinol. 2008, 22, 1370–1381. [Google Scholar] [CrossRef]
  185. Alto, L.T.; Terman, J.R. Semaphorins and their Signaling Mechanisms. In Semaphorin Signaling: Methods and Protocols. Methods in Molecular Biology; Terman, J.R., Ed.; Springer: Berlin/Heidelberg, Germany, 2017; Volume 1493, pp. 1–25. [Google Scholar] [CrossRef]
  186. Feng, Y.; Duan, T.; Du, Y.; Jin, S.; Wang, M.; Cui, J.; Wang, R.F. LRRC25 Functions as an Inhibitor of NF-κB Signaling Pathway by Promoting p65/RelA for Autophagic Degradation. Sci. Rep. 2017, 7, 13448. [Google Scholar] [CrossRef]
  187. Du, Y.; Duan, T.; Feng, Y.; Liu, Q.; Lin, M.; Cui, J.; Wang, R.F. LRRC25 inhibits type I IFN signaling by targeting ISG15-associated RIG-I for autophagic degradation. EMBO J. 2018, 37, 351–366. [Google Scholar] [CrossRef]
  188. Teymoori-Rad, M.; Shokri, F.; Salimi, V.; Marashi, S.M. The interplay between vitamin D and viral infections. Rev. Med. Virol. 2019, 29, e2032. [Google Scholar] [CrossRef]
  189. Lee, C. Controversial Effects of Vitamin D and Related Genes on Viral Infections, Pathogenesis, and Treatment Outcomes. Nutrients 2020, 12, 962. [Google Scholar] [CrossRef]
  190. Risco, A.; del Fresno, C.; Mambol, A.; Alsina-Beauchamp, D.; MacKenzie, K.F.; Yang, H.T.; Barber, D.F.; Morcelle, C.; Arthur, J.S.C.; Ley, S.C.; et al. p38γ and p38δ kinases regulate the Toll-like receptor 4 (TLR4)-induced cytokine production by controlling ERK1/2 protein kinase pathway activation. Proc. Natl. Acad. Sci. USA 2012, 109, 11200–11205. [Google Scholar] [CrossRef]
  191. Ryan, Z.C.; Craig, T.A.; Folmes, C.D.; Wang, X.; Lanza, I.R.; Schaible, N.S.; Salisbury, J.L.; Nair, K.S.; Terzic, A.; Sieck, G.C.; et al. 1α,25-Dihydroxyvitamin D3 Regulates Mitochondrial Oxygen Consumption and Dynamics in Human Skeletal Muscle Cells. J. Biol. Chem. 2016, 291, 1514–1528. [Google Scholar] [CrossRef]
  192. Lesourne, R.; Zvezdova, E.; Song, K.D.; El-Khoury, D.; Uehara, S.; Barr, V.A.; Samelson, L.E.; Love, P.E. Interchangeability of Themis1 and Themis2 in Thymocyte Development Reveals Two Related Proteins with Conserved Molecular Function. J. Immunol. 2012, 189, 1154–1161. [Google Scholar] [CrossRef] [Green Version]
  193. Treeck, O.; Odani, T.; Itoh, N.; Imai, H.; Fujita, S.; Kohroki, J.; Nakanishi, T.; Diedrich, K.; Ortmann, O.; Tanaka, K.; et al. Detection of increased icb-1 transcript levels in maturing HL-60 cells: A novel marker for granulocytic and monocytic in vitro differentiation. Leuk. Res. 2002, 26, 765–769. [Google Scholar] [CrossRef]
  194. Duvetorp, A.; Slind Olsen, R.; Skarstedt, M.; Söderman, J.; Seifert, O. Psoriasis and Pro-angiogenetic Factor CD93: Gene6Expression and Association with Gene Polymorphism Suggests a Role in Disease Pathogenesis. Acta Derm. Venereol. 2017, 97, 916–921. [Google Scholar] [CrossRef] [PubMed]
  195. Nativel, B.; Ramin-Mangata, S.; Mevizou, R.; Figuester, A.; Andries, J.; Iwema, T.; Ikewaki, N.; Gasque, P.; Viranaïcken, W. CD93 is a cell surface lectin receptor involved in the control of the inflammatory response stimulated by exogenous DNA. Immunology 2019, 158, 85–93. [Google Scholar] [CrossRef] [PubMed]
  196. Griffiths, M.R.; Botto, M.; Morgan, B.P.; Neal, J.W.; Gasque, P. CD93 regulates central nervous system inflammation in two mouse models of autoimmune encephalomyelitis. Immunology 2018, 155, 346–355. [Google Scholar] [CrossRef]
  197. Jennewein, C.; Sowa, R.; Faber, A.C.; Figuester, A.; Andries, J.; Iwema, T.; Ikewaki, N.; Gasque, P.; Viranaïcken, W. Contribution of Ninjurin1 to Toll-Like Receptor 4 Signaling and Systemic Inflammation. Am. J. Respir. Cell. Mol. Biol. 2015, 53, 656–663. [Google Scholar] [CrossRef]
  198. Ahn, B.J.; Le, H.; Shin, M.W.; Bae, S.J.; Lee, E.J.; Lee, S.Y.; Yang, J.H.; Wee, H.J.; Cha, J.H.; Seo, J.H.; et al. Ninjurin1 Enhances the Basal Motility and Transendothelial Migration of Immune Cells by Inducing Protrusive Membrane Dynamics. J. Biol. Chem. 2014, 289, 21926–21936. [Google Scholar] [CrossRef]
  199. Marchwicka, A.; Marcinkowska, E. Regulation of Expression of CEBP Genes by Variably Expressed Vitamin D Receptor and Retinoic Acid Receptor α in Human Acute Myeloid Leukemia Cell Lines. Int. J. Mol. Sci. 2018, 19, 1918. [Google Scholar] [CrossRef]
  200. Simpson, S.; Blizzard, L.; Otahal, P.; der Mei, I.V.; Taylor, B. Latitude is significantly associated with the prevalence of multiple sclerosis: A meta-analysis. J. Neurol. Neurosurg. Psychiatry 2011, 82, 1132–1141. [Google Scholar] [CrossRef]
  201. Ojeda-Fernández, L.; Recio-Poveda, L.; Aristorena, M.; Lastres, P.; Blanco, F.J.; Sanz-Rodríguez, F.; Gallardo-Vara, E.; de las Casas-Engel, M.; Corbí, N.; Arthur, H.M.; et al. Mice Lacking Endoglin in Macrophages Show an Impaired Immune Response. PLoS Genet. 2016, 12, e1005935. [Google Scholar] [CrossRef]
  202. Bocci, M.; Sjölund, J.; Kurzejamska, E.; Lindgren, D.; Marzouka, N.A.D.; Bartoschek, M.; Höglund, M.; Pietras, K. Activin receptor-like kinase 1 is associated with immune cell infiltration and regulates CLEC14A transcription in cancer. Angiogenesis 2019, 22, 117–131. [Google Scholar] [CrossRef] [Green Version]
  203. Korpetinou, A.; Skandalis, S.; Labropoulou, V.; Smirlaki, G.; Noulas, A.; Karamanos, N.K.; Theocharis, A.D. Serglycin: At the Crossroad of Inflammation and Malignancy. Front. Oncol. 2014, 3, 327. [Google Scholar] [CrossRef]
  204. Kolseth, I.B.M.; Reine, T.M.; Vuong, T.T.; Meen, A.J.; Fan, Q.; Jenssen, T.G.; Grønning-Wang, L.M.; Kolset, S.O. Serglycin is part of the secretory repertoire of LPS-activated monocytes. Immun. Inflamm. Dis. 2015, 3, 23–31. [Google Scholar] [CrossRef] [PubMed]
  205. Lemire, J.M.; Archer, D.C. 1,25-dihydroxyvitamin D3 prevents the in vivo induction of murine experimental autoimmune encephalomyelitis. J. Clin. Investig. 1991, 87, 1103–1107. [Google Scholar] [CrossRef] [PubMed]
  206. Cantorna, M.T.; Hayes, C.E.; DeLuca, H.F. 1,25-Dihydroxycholecalciferol Inhibits the Progression of Arthritis in Murine Models of Human Arthritis. J. Nut. 1998, 128, 68–72. [Google Scholar] [CrossRef]
  207. Cantorna, M.T.; Hayes, C.E.; DeLuca, H.F. 1,25-Dihydroxyvitamin D3 reversibly blocks the progression of relapsing encephalomyelitis, a model of multiple sclerosis. Proc. Natl. Acad. Sci. USA 1996, 93, 7861–7864. [Google Scholar] [CrossRef] [PubMed]
  208. Alhassan Mohammed, H.; Mirshafiey, A.; Vahedi, H.; Hemmasi, G.; Moussavi Nasl Khameneh, A.; Parastouei, K.; Saboor-Yaraghi, A.A. Immunoregulation of Inflammatory and Inhibitory Cytokines by Vitamin D3 in Patients with Inflammatory Bowel Diseases. Scand. J. Immunol. 2017, 85, 386–394. [Google Scholar] [CrossRef]
  209. Parisi, R.; Symmons, D.P.M.; Griffiths, C.E.M.; Ashcroft, D.M. Global Epidemiology of Psoriasis: A Systematic Review of Incidence and Prevalence. J. Investig. Dermatol. 2013, 133, 377–385. [Google Scholar] [CrossRef]
  210. Holick, M.F. The Photobiology of Vitamin D and Its Consequences for Humansa. Ann. N. Y. Acad. Sci. 1985, 453, 1–13. [Google Scholar] [CrossRef]
  211. Holick, M.F. Active Vitamin D Compounds and Analogues: A New Therapeutic Era for Dermatology in the 21st Century. Mayo Clin. Proc. 1993, 68, 925–927. [Google Scholar] [CrossRef]
  212. Kagami, S.; Rizzo, H.L.; Lee, J.J.; Koguchi, Y.; Blauvelt, A. Circulating Th17, Th22, and Th1 Cells Are Increased in Psoriasis. J. Investig. Dermatol. 2010, 130, 1373–1383. [Google Scholar] [CrossRef] [Green Version]
  213. Liu, J.; Wang, W.; Liu, K.; Wan, D.; Wu, Z.; Cao, Z.; Luo, Y.; Xiao, C.; Yin, M. Vitamin D receptor gene polymorphisms are associated with psoriasis susceptibility and the clinical response to calcipotriol in psoriatic patients. Exp. Dermatol. 2020, 29, 1186–1190. [Google Scholar] [CrossRef] [PubMed]
  214. Fletcher, J.; Bishop, E.M.; Harrison, S.R.; Swift, A.; Cooper, S.C.; Dimeloe, S.K.; Raza, K.; Hewison, M. Autoimmune disease and interconnections with vitamin D. Endocr. Connect. 2022, 11, e210554. [Google Scholar] [CrossRef] [PubMed]
  215. Barrea, L.; Savanelli, M.C.; Di Somma, C.; Napolitano, M.; Megna, M.; Colao, A.; Savastano, S. Vitamin D and its role in psoriasis: An overview of the dermatologist and nutritionist. Rev. Endocr. Metab. Disord. 2017, 18, 195–205. [Google Scholar] [CrossRef] [PubMed]
  216. MacLaughlin, J.A.; Gange, W.; Taylor, D.; Smith, E.; Holick, M.F. Cultured psoriatic fibroblasts from involved and uninvolved sites have a partial but not absolute resistance to the proliferation-inhibition activity of 1,25-dihydroxyvitamin D3. Proc. Natl. Acad. Sci. USA 1985, 82, 5409–5412. [Google Scholar] [CrossRef]
  217. Illescas-Montes, R.; Melguizo-Rodríguez, L.; Ruiz, C.; Costela-Ruiz, V.J. Vitamin D and autoimmune diseases. Life Sci. 2019, 233, 116744. [Google Scholar] [CrossRef]
  218. Smith, E.L.; Pincus, S.H.; Donovan, L.; Holick, M.F. A novel approach for the evaluation and treatment of psoriasis: Oral or topical use of 1,25-dihydroxyvitamin D3 can be a safe and effective therapy for psoriasis. J. Am. Acad. Dermatol. 1988, 19, 516–528. [Google Scholar] [CrossRef]
  219. Dubertret, L.; Wallach, D.; Souteyrand, P.; Perussel, M.; Kalis, B.; Meynadier, J.; Chevrant-Breton, J.; Beylot, C.; Bazex, J.A.; Jurgensen, H.J. Efficacy and safety of calcipotriol (MC 903) ointment in psoriasis vulgaris: A randomized, double-blind, right/left comparative, vehicle-controlled study. J. Am. Acad. Dermatol. 1992, 27, 983–988. [Google Scholar] [CrossRef]
  220. Fu, H.; Tang, Z.; Wang, Y.; Ding, X.; Rinaldi, G.; Rahmani, J.; Xing, F. Relationship Between Vitamin D Level and Mortality in Adults With Psoriasis: A Retrospective Cohort Study of NHANES Data. Clin. Ther. 2021, 43, e33–e38. [Google Scholar] [CrossRef]
  221. Orgaz-Molina, J.; Buendía-Eisman, A.; Arrabal-Polo, M.A.; Ruiz, J.C.; Arias-Santiago, S. Deficiency of serum concentration of 25-hydroxyvitamin D in psoriatic patients: A case-control study. J. Am. Acad. Dermatol. 2012, 67, 931–938. [Google Scholar] [CrossRef]
  222. Disphanurat, W.; Viarasilpa, W.; Chakkavittumrong, P.; Pongcharoen, P. The Clinical Effect of Oral Vitamin D2 Supplementation on Psoriasis: A Double-Blind, Randomized, Placebo-Controlled Study. Derm. Res. Pract. 2019, 2019, e5237642. [Google Scholar] [CrossRef] [Green Version]
  223. McCullough, P.J.; Lehrer, D.S.; Amend, J. Daily oral dosing of vitamin D3 using 5000 TO 50,000 international units a day in long-term hospitalized patients: Insights from a seven year experience. J. Steroid Biochem. Mol. Biol. 2019, 189, 228–239. [Google Scholar] [CrossRef] [PubMed]
  224. Finamor, D.C.; Sinigaglia-Coimbra, R.; Neves, L.C.M.; Gutierrez, M.; Silva, J.J.; Torres, L.D.; Surano, F.; Neto, D.J.; Novo, N.F.; Juliano, Y.; et al. A pilot study assessing the effect of prolonged administration of high daily doses of vitamin D on the clinical course of vitiligo and psoriasis. Dermatoendocrinol 2013, 5, 222–234. [Google Scholar] [CrossRef] [PubMed]
  225. Chen, Y.L.; Huang, Y.C.; Qiao, Y.C.; Ling, W.; Pan, Y.H.; Geng, L.J.; Xiao, J.L.; Zhang, X.X.; Zhao, H.L. Climates on incidence of childhood type 1 diabetes mellitus in 72 countries. Sci. Rep. 2017, 7, 12810. [Google Scholar] [CrossRef] [PubMed]
  226. Mohr, S.B.; Garland, C.F.; Gorham, E.D.; Garland, F.C. The association between ultraviolet B irradiance, vitamin D status and incidence rates of type 1 diabetes in 51 regions worldwide. Diabetologia 2008, 51, 1391–1398. [Google Scholar] [CrossRef]
  227. Wessels, I.; Rink, L. Micronutrients in autoimmune diseases: Possible therapeutic benefits of zinc and vitamin D. J. Nut. Biochem. 2020, 77, 108240. [Google Scholar] [CrossRef]
  228. Holick, M.F. Vitamin D: A millenium perspective. J. Cell. Biochem. 2003, 88, 296–307. [Google Scholar] [CrossRef]
  229. Webb, A.R.; Kline, L.; Holick, M.F. Influence of Season and Latitude on the Cutaneous Synthesis of Vitamin D3: Exposure to Winter Sunlight in Boston and Edmonton Will Not Promote Vitamin D3 Synthesis in Human Skin. J. Clin. Endocrinol. Metab. 1988, 67, 373–378. [Google Scholar] [CrossRef]
  230. Hou, Y.; Song, A.; Jin, Y.; Xia, Q.; Song, G.; Xing, X. A dose–response meta-analysis between serum concentration of 25-hydroxy vitamin D and risk of type 1 diabetes mellitus. Eur. J. Clin. Nutr. 2021, 75, 1010–1023. [Google Scholar] [CrossRef]
  231. Harjutsalo, V.; Sund, R.; Knip, M.; Groop, P.H. Incidence of Type 1 Diabetes in Finland. JAMA 2013, 310, 427–428. [Google Scholar] [CrossRef]
  232. Junnila, S.K. Type 1 diabetes epidemic in Finland is triggered by zinc-containing amorphous silica nanoparticles. Med. Hypotheses 2015, 84, 336–340. [Google Scholar] [CrossRef]
  233. Manousaki, D.; Harroud, A.; Mitchell, R.E.; Ross, S.; Forgetta, V.; Timpson, N.J.; Smith, G.D.; Polychronakos, C.; Richards, J.B. Vitamin D levels and risk of type 1 diabetes: A Mendelian randomization study. PLoS Med. 2021, 18, e1003536. [Google Scholar] [CrossRef]
  234. Almeida, J.T.; Rodrigues, D.; Guimarães, J.; Lemos, M.C. Vitamin D Pathway Genetic Variation and Type 1 Diabetes: A Case–Control Association Study. Genes 2020, 11, 897. [Google Scholar] [CrossRef] [PubMed]
  235. Hyöty, H.; Leon, F.; Knip, M. Developing a vaccine for type 1 diabetes by targeting coxsackievirus B. Expert Rev. Vaccines 2018, 17, 1071–1083. [Google Scholar] [CrossRef] [PubMed]
  236. Li, M.; Song, L.J.; Qin, X.Y. Advances in the cellular immunological pathogenesis of type 1 diabetes. J. Cell. Mol. Med. 2014, 18, 749–758. [Google Scholar] [CrossRef]
  237. The EURODIAB Substudy 2 Study Group. Vitamin D supplement in early childhood and risk for Type I (insulin-dependent) diabetes mellitus. Diabetologia 1999, 42, 51–54. [Google Scholar] [CrossRef]
  238. Yoon, J.W.; Jun, H.S. Autoimmune Destruction of Pancreatic β Cells. Am. J. Ther. 2005, 12, 580–591. [Google Scholar] [CrossRef] [PubMed]
  239. Hyppönen, E.; Läärä, E.; Reunanen, A.; Järvelin, M.R.; Virtanen, S.M. Intake of vitamin D and risk of type 1 diabetes: A birth-cohort study. Lancet 2001, 358, 1500–1503. [Google Scholar] [CrossRef]
  240. Ataie-Jafari, A.; Loke, S.C.; Rahmat, A.B.; Larijani, B.; Abbasi, F.; Leow, M.K.; Yassin, Z. A randomized placebo-controlled trial of alphacalcidol on the preservation of beta cell function in children with recent onset type 1 diabetes. Clin. Nut. 2013, 32, 911–917. [Google Scholar] [CrossRef]
  241. Gabbay, M.A.L.; Sato, M.N.; Finazzo, C.; Duarte, A.J.S.; Dib, S.A. Effect of Cholecalciferol as Adjunctive Therapy With Insulin on Protective Immunologic Profile and Decline of Residual β-Cell Function in New-Onset Type 1 Diabetes Mellitus. Arch. Pediatric. Adolesc. Med. 2012, 166, 601–607. [Google Scholar] [CrossRef] [PubMed]
  242. Treiber, G.; Prietl, B.; Fröhlich-Reiterer, E.; Lechner, E.; Ribitsch, A.; Fritsch, M.; Rami-Merhar, B.; Steigleder-Schweiger, C.; Graninger, W.; Borkenstein, M.; et al. Cholecalciferol supplementation improves suppressive capacity of regulatory T-cells in young patients with new-onset type 1 diabetes mellitus—A randomized clinical trial. Clin. Immunol. 2015, 161, 217–224. [Google Scholar] [CrossRef]
  243. Mokry, L.E.; Ross, S.; Ahmad, O.S.; Forgetta, V.; Smith, G.D.; Leong, A.; Greenwood, C.M.T.; Thanassoulis, G.; Richards, J.B. Vitamin D and Risk of Multiple Sclerosis: A Mendelian Randomization Study. PLoS Med. 2015, 12, e1001866. [Google Scholar] [CrossRef] [PubMed]
  244. Jiang, X.; Ge, T.; Chen, C.-Y. The causal role of circulating vitamin D concentrations in human complex traits and diseases: A large-scale Mendelian randomization study. Sci. Rep. 2021, 11, 184. [Google Scholar] [CrossRef] [PubMed]
  245. Wang, R. Mendelian randomization study updates the effect of 25-hydroxyvitamin D levels on the risk of multiple sclerosis. J. Transl. Med. 2022, 20, 3. [Google Scholar] [CrossRef] [PubMed]
  246. Jacobs, B.M.; Noyce, A.J.; Giovannoni, G.; Dobson, R. BMI and low vitamin D are causal factors for multiple sclerosis: A Mendelian Randomization study. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e662. [Google Scholar] [CrossRef] [PubMed]
  247. Munger, K.L.; Zhang, S.M.; O’Reilly, E.; Hernan, M.A.; Olek, M.J.; Willett, W.C.; Ascherio, A. Vitamin D intake and incidence of multiple sclerosis. Neurology 2004, 62, 60–65. [Google Scholar] [CrossRef] [PubMed]
  248. Haines, J.D.; Inglese, M.; Casaccia, P. Axonal Damage in Multiple Sclerosis. Mt. Sinai. J. Med. J. Transl. Personal. Med. 2011, 78, 231–243. [Google Scholar] [CrossRef]
  249. Ghasemi, N.; Razavi, S.; Nikzad, E. Multiple Sclerosis: Pathogenesis, Symptoms, Diagnoses and Cell-Based Therapy. Cell J. 2017, 19, 1–10. [Google Scholar] [CrossRef]
  250. Cocco, E.; Meloni, A.; Murru, M.R.; Corongiu, D.; Tranquilli, S.; Fadda, E.; Murru, R.; Schirru, L.; Secci, M.A.; Costa, G.; et al. Vitamin D Responsive Elements within the HLA-DRB1 Promoter Region in Sardinian Multiple Sclerosis Associated Alleles. PLoS ONE 2012, 7, e41678. [Google Scholar] [CrossRef]
  251. Ruiz-Ballesteros, A.I.; Meza-Meza, M.R.; Vizmanos-Lamotte, B.; Parra-Rojas, I.; de la Cruz-Mosso, U. Association of Vitamin D Metabolism Gene Polymorphisms with Autoimmunity: Evidence in Population Genetic Studies. Int. J. Mol. Sci. 2020, 21, 9626. [Google Scholar] [CrossRef]
  252. Scazzone, C.; Agnello, L.; Bivona, G.; Lo Sasso, B.; Ciaccio, M. Vitamin D and Genetic Susceptibility to Multiple Sclerosis. Biochem. Genet. 2021, 59, 1–30. [Google Scholar] [CrossRef]
  253. Ramagopalan, S.V.; Maugeri, N.J.; Handunnetthi, L.; Lincoln, M.R.; Orton, S.M.; Dyment, D.A.; DeLuca, G.C.; Herrera, B.M.; Chao, M.J.; Sadovnick, A.D.; et al. Expression of the Multiple Sclerosis-Associated MHC Class II Allele HLA-DRB1*1501 Is Regulated by Vitamin D. PLoS Genet. 2009, 5, e1000369. [Google Scholar] [CrossRef] [PubMed]
  254. Amon, U.; Yaguboglu, R.; Ennis, M.; Holick, M.F.; Amon, J. Safety Data in Patients with Autoimmune Diseases during Treatment with High Doses of Vitamin D3 According to the ‘Coimbra Protocol’. Nutrients 2022, 14, 1575. [Google Scholar] [CrossRef] [PubMed]
  255. McLaughlin, L.; Clarke, L.; Khalilidehkordi, E.; Butzkueven, H.; Taylor, B.; Broadley, S.A. Vitamin D for the treatment of multiple sclerosis: A meta-analysis. J. Neurol. 2018, 265, 2893–2905. [Google Scholar] [CrossRef] [PubMed]
  256. Schultz, M.; Butt, A.G. Is the north to south gradient in inflammatory bowel disease a global phenomenon? Expert Rev. Gastroenterol. Hepatol. 2012, 6, 445–447. [Google Scholar] [CrossRef] [PubMed]
  257. Matricon, J.; Barnich, N.; Ardid, D. Immunopathogenesis of inflammatory bowel disease. Self Nonself 2010, 1, 299–309. [Google Scholar] [CrossRef]
  258. Wu, Z.; Liu, D.; Deng, F. The Role of Vitamin D in Immune System and Inflammatory Bowel Disease. J. Inflamm. Res. 2022, 15, 3167–3185. [Google Scholar] [CrossRef]
  259. Yue, B.; Luo, X.; Yu, Z.; Mani, S.; Wang, Z.; Dou, W. Inflammatory Bowel Disease: A Potential Result from the Collusion between Gut Microbiota and Mucosal Immune System. Microorganisms 2019, 7, 440. [Google Scholar] [CrossRef]
  260. Farraye, F.A.; Nimitphong, H.; Stucchi, A.; Dendrinos, K.; Boulanger, A.B.; Vijjeswarapu, A.; Tanennbaum, A.; Biancuzzo, R.; Chen, T.C.; Holick, M.F. Use of a novel vitamin D bioavailability test demonstrates that vitamin D absorption is decreased in patients with quiescent crohn’s disease1,2,3. Inflamm. Bowel Dis. 2011, 17, 2116–2121. [Google Scholar] [CrossRef]
  261. Lo, C.W.; Paris, P.W.; Clemens, T.L.; Nolan, J.; Holick, M.F. Vitamin D absorption in healthy subjects and in patients with intestinal malabsorption syndromes. Am. J. Clin. Nut. 1985, 42, 644–649. [Google Scholar] [CrossRef]
  262. Gubatan, J.; Moss, A.C. Vitamin D in inflammatory bowel disease: More than just a supplement. Curr. Opin. Gastroenterol. 2018, 34, 217–225. [Google Scholar] [CrossRef]
  263. Ludvigsson, J.F.; Mahl, M.; Sachs, M.C.; Björk, J.; Michaelsson, K.; Ekbom, A.; Askling, J.; Backman, A.S.; Olén, O. Fracture Risk in Patients With Inflammatory Bowel Disease: A Nationwide Population-Based Cohort Study From 1964 to 2014. Am. J. Gastroenterol. 2019, 114, 291–304. [Google Scholar] [CrossRef] [PubMed]
  264. Nemeth, Z.H.; Bogdanovski, D.A.; Barratt-Stopper, P.; Paglinco, S.R.; Antonioli, L.; Rolandelli, R.H. Crohn’s Disease and Ulcerative Colitis Show Unique Cytokine Profiles. Cureus 2017, 9, e1177. [Google Scholar] [CrossRef] [PubMed]
  265. Gálvez, J. Role of Th17 Cells in the Pathogenesis of Human IBD. ISRN Inflamm. 2014, 2014, e928461. [Google Scholar] [CrossRef]
  266. Fletcher, J.; Cooper, S.C.; Ghosh, S.; Hewison, M. The Role of Vitamin D in Inflammatory Bowel Disease: Mechanism to Management. Nutrients 2019, 11, 1019. [Google Scholar] [CrossRef] [PubMed]
  267. Xu, Y.; Baylink, D.J.; Cao, H.; Xiao, J.; Abdalla, M.I.; Wasnik, S.; Tang, X. Inflammation- and Gut-Homing Macrophages, Engineered to De Novo Overexpress Active Vitamin D, Promoted the Regenerative Function of Intestinal Stem Cells. Int. J. Mol. Sci. 2021, 22, 9516. [Google Scholar] [CrossRef]
  268. Ananthakrishnan, A.N.; Khalili, H.; Higuchi, L.M.; Bao, Y.; Korzenik, J.R.; Giovannucci, E.L.; Richter, J.M.; Fuchs, C.S.; Chan, A.T. Higher Predicted Vitamin D Status Is Associated With Reduced Risk of Crohn’s Disease. Gastroenterology 2012, 142, 482–489. [Google Scholar] [CrossRef]
  269. Li, J.; Chen, N.; Wang, D.; Zhang, J.; Gong, X. Efficacy of vitamin D in treatment of inflammatory bowel disease: A meta-analysis. Medicine 2018, 97, e12662. [Google Scholar] [CrossRef]
  270. Schäffler, H.; Herlemann, D.P.; Klinitzke, P.; Berlin, P.; Kreikemeyer, B.; Jaster, R.; Lamprecht, G. Vitamin D administration leads to a shift of the intestinal bacterial composition in Crohn’s disease patients, but not in healthy controls. J. Dig. Dis. 2018, 19, 225–234. [Google Scholar] [CrossRef]
  271. Charoenngam, N.; Shirvani, A.; Kalajian, T.A.; Song, A.; Holick, M.F. The Effect of Various Doses of Oral Vitamin D3 Supplementation on Gut Microbiota in Healthy Adults: A Randomized, Double-blinded, Dose-response Study. Anticancer Res. 2020, 40, 551–556. [Google Scholar] [CrossRef]
  272. Lee, Y.H.; Bae, S.C. Vitamin D level in rheumatoid arthritis and its correlation with the disease activity: A meta-analysis. Clin. Exp. Rheumatol. 2016, 34, 827–833. [Google Scholar]
  273. Meena, N.; Singh Chawla, S.P.; Garg, R.; Batta, A.; Kaur, S. Assessment of Vitamin D in Rheumatoid Arthritis and Its Correlation with Disease Activity. J. Nat. Sci. Biol. Med. 2018, 9, 54–58. [Google Scholar] [CrossRef] [PubMed]
  274. Aslam, M.M.; John, P.; Bhatti, A.; Jahangir, S.; Kamboh, M.I. Vitamin D as a Principal Factor in Mediating Rheumatoid Arthritis-Derived Immune Response. BioMed Res. Int. 2019, 2019, e3494937. [Google Scholar] [CrossRef] [PubMed]
  275. Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos, N.J.; Xu, J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018, 6, 1–14. [Google Scholar] [CrossRef]
  276. Li, S.; Yin, H.; Zhang, K.; Wang, T.; Yang, Y.; Liu, X.; Chang, X.; Zhang, M.; Yan, X.; Ren, Y.; et al. Effector T helper cell populations are elevated in the bone marrow of rheumatoid arthritis patients and correlate with disease severity. Sci. Rep. 2017, 7, 4776. [Google Scholar] [CrossRef]
  277. Gopinath, K.; Danda, D. Supplementation of 1,25 dihydroxy vitamin D3 in patients with treatment naive early rheumatoid arthritis: A randomised controlled trial—GOPINATH—2011. Int. J. Rheum. Dis. 2011, 14, 332–339. [Google Scholar] [CrossRef] [PubMed]
  278. Li, C.; Yin, S.; Yin, H.; Cao, L.; Zhang, T.; Wang, Y. Efficacy and Safety of 22-Oxa-Calcitriol in Patients with Rheumatoid Arthritis: A Phase II Trial. Med. Sci. Monit. 2018, 24, 9127–9135. [Google Scholar] [CrossRef] [PubMed]
  279. Hansen, K.E.; Bartels, C.M.; Gangnon, R.E.; Jones, A.N.; Gogineni, J. An Evaluation of High-Dose Vitamin D for Rheumatoid Arthritis. J.Clin. Rheumatol. 2014, 20, 112–114. [Google Scholar] [CrossRef]
  280. Dehghan, A.; Rahimpour, S.; Soleymani-Salehabadi, H.; Owlia, M.B. Role of vitamin D in flare ups of rheumatoid arthritis. Z Rheumatol 2014, 73, 461–464. [Google Scholar] [CrossRef]
  281. Salesi, M.; Farajzadegan, Z. Efficacy of Vitamin D in patients with active rheumatoid arthritis receiving methotrexate therapy. Rheumatol. Int. 2012, 32, 2129–2133. [Google Scholar] [CrossRef] [PubMed]
  282. Franco, A.S.; Freitas, T.Q.; Bernardo, W.M.; Pereira, R.M.R. Vitamin D supplementation and disease activity in patients with immune-mediated rheumatic diseases: A systematic review and meta-analysis. Medicine 2017, 96, e7024. [Google Scholar] [CrossRef] [PubMed]
  283. Amital, H.; Szekanecz, Z.; Szücs, G.; Dankó, K.; Nagy, E.; Csépány, T.; Kiss, E.; Rovensky, J.; Tuchynova, A.; Kozakova, D.; et al. Serum concentrations of 25-OH vitamin D in patients with systemic lupus erythematosus (SLE) are inversely related to disease activity: Is it time to routinely supplement patients with SLE with vitamin D? Ann. Rheum. Dis. 2010, 69, 1155–1157. [Google Scholar] [CrossRef]
  284. Birmingham, D.; Hebert, L.; Song, H.; Noonan, W.; Rovin, B.; Nagaraja, H.; Yu, C. Evidence that abnormally large seasonal declines in vitamin D status may trigger SLE flare in non-African Americans. Lupus 2012, 21, 855–864. [Google Scholar] [CrossRef] [PubMed]
  285. Mok, C.C.; Birmingham, D.J.; Leung, H.W.; Hebert, L.A.; Song, H.; Rovin, B.H. Vitamin D levels in Chinese patients with systemic lupus erythematosus: Relationship with disease activity, vascular risk factors and atherosclerosis. Rheumatology 2012, 51, 644–652. [Google Scholar] [CrossRef] [PubMed]
  286. AlSaleem, A.; AlE’ed, A.; AlSaghier, A.; Al-Mayouf, S.M. Vitamin D status in children with systemic lupus erythematosus and its association with clinical and laboratory parameters. Clin. Rheumatol. 2015, 34, 81–84. [Google Scholar] [CrossRef]
  287. Eloi, M.; Horvath, D.V.; Ortega, J.C.; Prado, M.S.; Andrade, L.E.C.; Szejnfeld, V.L.; de Moura Castro, C.H. 25-Hydroxivitamin D Serum Concentration, Not Free and Bioavailable Vitamin D, Is Associated with Disease Activity in Systemic Lupus Erythematosus Patients. PLoS ONE 2017, 12, e0170323. [Google Scholar] [CrossRef] [PubMed]
  288. Salman-Monte, T.C.; Torrente-Segarra, V.; Almirall, M.; Corzo, P.; Mojal, S.; Carbonell-Abelló, J. Prevalence and predictors of vitamin D insufficiency in supplemented and non-supplemented women with systemic lupus erythematosus in the Mediterranean region. Rheumatol. Int. 2016, 36, 975–985. [Google Scholar] [CrossRef]
  289. Murdaca, G.; Tonacci, A.; Negrini, S.; Greco, M.; Borro, M.; Puppo, F.; Gangemi, S. Emerging role of vitamin D in autoimmune diseases: An update on evidence and therapeutic implications. Autoimmun. Rev. 2019, 18, 102350. [Google Scholar] [CrossRef]
  290. Arshad, A.; Mahmood, S.B.Z.; Ayaz, A.; Al Karim Manji, A.; Ahuja, A.K. Association of vitamin D deficiency and disease activity in systemic lupus erythematosus patients: Two-year follow-up study. Arch. Rheumatol. 2021, 36, 101–106. [Google Scholar] [CrossRef]
  291. Stagi, S.; Rigante, D. Vitamin D and juvenile systemic lupus erythematosus: Lights, shadows and still unresolved issues. Autoimmun. Rev. 2018, 17, 290–300. [Google Scholar] [CrossRef]
  292. Pludowski, P.; Holick, M.F.; Pilz, S.; Wagner, C.L.; Hollis, B.W.; Grant, W.B.; Shoenfeld, Y.; Lerchbaum, E.; Llewellyn, D.J.; Kienreich, K.; et al. Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality-a review of recent evidence. Autoimmun. Rev. 2013, 12, 976–989. [Google Scholar] [CrossRef]
  293. Willis, R.; Smikle, M.; DeCeulaer, K.; Romay-Penabad, Z.; Papalardo, E.; Jajoria, P.; Harper, B.; Murthy, V.; Petri, M.; Gonzalez, E.B. Clinical associations of proinflammatory cytokines, oxidative biomarkers and vitamin D levels in systemic lupus erythematosus. Lupus 2017, 26, 1517–1527. [Google Scholar] [CrossRef]
  294. Islam, M.A.; Khandker, S.S.; Alam, S.S.; Kotyla, P.; Hassan, R. Vitamin D status in patients with systemic lupus erythematosus (SLE): A systematic review and meta-analysis. Autoimmun. Rev. 2019, 18, 102392. [Google Scholar] [CrossRef] [PubMed]
  295. Ritterhouse, L.L.; Crowe, S.R.; Niewold, T.B.; Kamen, D.L.; Macwana, S.R.; Roberts, V.C.; Dedeke, A.B.; Harley, J.B.; Scofield, R.H.; Guthridge, J.M.; et al. Vitamin D deficiency is associated with an increased autoimmune response in healthy individuals and in patients with systemic lupus erythematosus. Ann. Rheum. Dis. 2011, 70, 1569–1574. [Google Scholar] [CrossRef] [PubMed]
  296. Gittoes, N.J. Vitamin D – what is normal according to latest research and how should we deal with it? Clin. Med. 2015, 15, s54–s57. [Google Scholar] [CrossRef]
  297. Yu, Q.; Qiao, Y.; Liu, D.; Liu, F.; Gao, C.; Duan, J.; Liang, L.; Di, X.; Yuan, Y.; Gao, Y.; et al. Vitamin D protects podocytes from autoantibodies induced injury in lupus nephritis by reducing aberrant autophagy. Arthritis Res. Ther. 2019, 2, 19. [Google Scholar] [CrossRef] [PubMed]
  298. Nguyen, M.; Bryant, K.; O’Neill, S. Vitamin D in SLE: A role in pathogenesis and fatigue? A review of the literature. Lupus 2018, 27, 2003–2011. [Google Scholar] [CrossRef] [PubMed]
  299. Sabio, J.M.; Vargas-Hitos, J.A.; Martínez-Bordonado, J.; Mediavilla-García, J.D. Association between non-dipper hypertension and vitamin D deficiency in women with systemic lupus erythematosus. Clin. Exp. Rheumatol. 2019, 37, 286–292. [Google Scholar]
  300. Hussein, H.A.; Daker, L.I.; Fouad, N.A.; Elamir, A.; Mohamed, S.R. Does Vitamin D Deficiency Contribute to Cognitive Dysfunction in Patients with Systemic Lupus Erythematosus? Innov. Clin. Neurosci. 2018, 15, 25–29. [Google Scholar]
  301. Mak, A. The Impact of Vitamin D on the Immunopathophysiology, Disease Activity, and Extra-Musculoskeletal Manifestations of Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2018, 19, 2355. [Google Scholar] [CrossRef]
  302. Bellan, M.; Andreoli, L.; Mele, C.; Sainaghi, P.P.; Rigamonti, C.; Piantoni, S.; De Benedittis, C.; Aimaretti, G.; Pirisi, M.; Marzullo, P. Pathophysiological Role and Therapeutic Implications of Vitamin D in Autoimmunity: Focus on Chronic Autoimmune Diseases. Nutrients 2020, 12, 789. [Google Scholar] [CrossRef]
  303. Fakhfakh, R.; Feki, S.; Elleuch, A.; Neifar, M.; Marzouk, S.; Elloumi, N.; Hachicha, H.; Abida, O.; Bahloul, Z.; Ayadi, F.; et al. Vitamin D status and CYP27B1-1260 promoter polymorphism in Tunisian patients with systemic lupus erythematosus. Mol. Genet. Genomic. Med. 2021, 9, e1618. [Google Scholar] [CrossRef] [PubMed]
  304. Yamamoto, E.A.; Nguyen, J.K.; Liu, J.; Keller, E.; Campbell, N.; Zhang, C.J.; Smith, H.R.; Li, X.; Jørgensen, T.N. Low Levels of Vitamin D Promote Memory B Cells in Lupus. Nutrients 2020, 12, 291. [Google Scholar] [CrossRef] [PubMed]
  305. Berardi, S.; Giardullo, L.; Corrado, A.; Cantatore, F.P. Vitamin D and connective tissue diseases. Inflamm. Res. 2020, 69, 453–462. [Google Scholar] [CrossRef] [PubMed]
  306. Young, K.A.; Munroe, M.E.; Guthridge, J.M.; Kamen, D.L.; Niewold, T.B.; Gilkeson, G.S.; Weisman, M.H.; Ishimori, M.L.; Kelly, J.; Gaffney, P.M.; et al. Combined role of vitamin D status and CYP24A1 in the transition to systemic lupus erythematosus. Ann. Rheum. Dis. 2017, 76, 153–158. [Google Scholar] [CrossRef] [PubMed]
  307. Mahto, H.; Tripathy, R.; Das, B.K.; Panda, A.K. Association between vitamin D receptor polymorphisms and systemic lupus eryhematosus in an Indian cohort. Int. J. Rheum. Dis. 2017, 21, 468–476. [Google Scholar] [CrossRef]
  308. Aranow, C.; Kamen, D.L.; Dall’Era, M.; Massarotti, E.M.; Mackay, M.C.; Koumpouras, F.; Coca, A.; Chatham, W.W.; Clowse, M.E.; Criscione-Schreiber, L.G.; et al. Randomized, Double-Blind, Placebo-Controlled Trial of the Effect of Vitamin D3 on the Interferon Signature in Patients With Systemic Lupus Erythematosus. Arthr. Rheum. 2015, 67, 1848–1857. [Google Scholar] [CrossRef]
  309. Lima, G.L.; Paupitz, J.; Aikawa, N.E.; Takayama, L.; Bonfa, E.; Pereira, R.M. Vitamin D Supplementation in Adolescents and Young Adults With Juvenile Systemic Lupus Erythematosus for Improvement in Disease Activity and Fatigue Scores: A Randomized, Double-Blind, Placebo-Controlled Trial. Arthr. Care Res. 2016, 68, 91–98. [Google Scholar] [CrossRef]
  310. Rifaâ€TMi, A.; Kalim, H.; Kusworini, K.; Wahono, C.S. Effect of vitamin D supplementation on disease activity (SLEDAI) and fatigue in Systemic Lupus Erythematosus patients with hipovitamin D: An Open Clinical Trial. Indones. J. Rheumatol. 2016, 8, 2. [Google Scholar] [CrossRef]
  311. Petri, M.; Bello, K.J.; Fang, H.; Magder, L.S. Vitamin D in systemic lupus erythematosus: Modest association with disease activity and the urine protein-to-creatinine ratio. Arthr. Rheum. 2013, 65, 1865–1871. [Google Scholar] [CrossRef]
  312. Andreoli, L.; Dall’Ara, F.; Piantoni, S.; Zanola, A.; Piva, N.; Cutolo, M.; Tincani, A. A 24-month prospective study on the efficacy and safety of two different monthly regimens of vitamin D supplementation in pre-menopausal women with systemic lupus erythematosus. Lupus 2015, 24, 499–506. [Google Scholar] [CrossRef]
  313. Karimzadeh, H.; Shirzadi, M.; Karimifar, M. The effect of Vitamin D supplementation in disease activity of systemic lupus erythematosus patients with Vitamin D deficiency: A randomized clinical trial. J. Res. Med. Sci. 2017, 22, 4. [Google Scholar] [CrossRef] [PubMed]
  314. Dall’Ara, F.; Cutolo, M.; Andreoli, L.; Tincani, A.; Paolino, S. Vitamin D and systemic lupus erythematous: A review of immunological and clinical aspects. Clin. Exp. Rheumatol. 2018, 6, 153–162. [Google Scholar]
  315. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet. 2017, 390, 1685–1699. [Google Scholar] [CrossRef]
  316. Murdaca, G.; Contatore, M.; Gulli, R.; Mandich, P.; Puppo, F. Genetic factors and systemic sclerosis. Autoimmun. Rev. 2016, 15, 427–432. [Google Scholar] [CrossRef] [PubMed]
  317. Szabo, I.; Muntean, L.; Crisan, T.; Rednic, V.; Sirbe, C.; Rednic, S. Novel Concepts in Systemic Sclerosis Pathogenesis: Role for miRNAs. Biomedicines 2021, 9, 1471. [Google Scholar] [CrossRef] [PubMed]
  318. Vacca, A.; Cormier, C.; Piras, M.; Mathieu, A.; Kahan, A.; Allanore, Y. Vitamin D Deficiency and Insufficiency in 2 Independent Cohorts of Patients with Systemic Sclerosis. J. Rheumatol. 2009, 36, 1924–1929. [Google Scholar] [CrossRef]
  319. Fattizzo, B.; Zaninoni, A.; Giannotta, J.A.; Binda, F.; Cortelezzi, A.; Barcellini, W. Reduced 25-OH vitamin D in patients with autoimmune cytopenias, clinical correlations and literature review. Autoimmun. Rev. 2016, 15, 770–775. [Google Scholar] [CrossRef]
  320. An, L.; Sun, M.-H.; Chen, F.; Li, J.-R. Vitamin D levels in systemic sclerosis patients: A meta-analysis. Drug Des. Devel. Ther. 2017, 11, 3119–3125. [Google Scholar] [CrossRef]
  321. Gupta, S.; Mahajan, V.K.; Yadav, R.S.; Mehta, K.; Bhushan, S.; Chauhan, P.; Rawat, R.; Sharma, V. Evaluation of Serum Vitamin D Levels in Patients with Systemic Sclerosis and Healthy Controls: Results of a Pilot Study. Indian Dermatol. Online J. 2018, 9, 250–255. [Google Scholar] [CrossRef]
  322. Arnson, Y.; Amital, H.; Agmon-Levin, N.; Alon, D.; Sánchez-Castañón, M.; López-Hoyos, M.; Matucci-Cerinic, M.; Szücs, G.; Shapira, Y.; Szekanecz, Z.; et al. Serum 25-OH vitamin D concentrations are linked with various clinical aspects in patients with systemic sclerosis: A retrospective cohort study and review of the literature. Autoimmun. Rev. 2011, 10, 490–494. [Google Scholar] [CrossRef]
  323. Carmel, N.N.; Rotman-Pikielny, P.; Lavrov, A.; Levy, Y. Vitamin D Antibodies in Systemic Sclerosis Patients: Findings and Clinical Correlations. Isr. Med. Assoc. J. 2015, 17, 80–84. [Google Scholar] [PubMed]
  324. Korman, B. Evolving insights into the cellular and molecular pathogenesis of fibrosis in systemic sclerosis. Transl. Res. 2019, 209, 77–89. [Google Scholar] [CrossRef] [PubMed]
  325. Zerr, P.; Vollath, S.; Palumbo-Zerr, K.; Zerr, P.; Vollath, S.; Palumbo-Zerr, K.; Tomcik, M.; Huang, J.; Distler, A.; Beyer, C.; et al. Vitamin D receptor regulates TGF-β signalling in systemic sclerosis. Ann. Rheum. Dis. 2015, 74, e20. [Google Scholar] [CrossRef] [PubMed]
  326. Kotyla, P.J.; Kruszec-Zytniewska, A.; Owczarek, A.J.; Olszanecka-Glinianowicz, M.; Chudek, J. Fibroblast Growth Factor 23 to Alpha-Klotho Index Correlates with Systemic Sclerosis Activity: A Proposal for Novel Disease Activity Marker. J. Clin. Med. 2018, 7, 558. [Google Scholar] [CrossRef] [Green Version]
  327. Bivona, G.; Agnello, L.; Pivetti, A.; Milano, S.; Scazzone, C.; Sasso, B.L.; Ciaccio, M. Association between hypovitaminosis D and systemic sclerosis: True or fake? Clin. Chim. Acta. 2016, 458, 115–119. [Google Scholar] [CrossRef]
  328. Zhang, L.; Duan, Y.; Zhang, T.P.; Huang, X.L.; Li, B.Z.; Ye, D.Q.; Wang, J. Association between the serum level of vitamin D and systemic sclerosis in a Chinese population: A case control study. Int. J. Rheum. Dis. 2017, 20, 1002–1008. [Google Scholar] [CrossRef]
  329. Groseanu, L.; Bojinca, V.; Gudu, T.; Saulescu, I.; Predeteanu, D.; Balanescu, A.; Berghea, F.; Opris, D.; Borangiu, A.; Constantinescu, C.; et al. Low vitamin D status in systemic sclerosis and the impact on disease phenotype. Eur. J. Rheumatol. 2016, 3, 50–55. [Google Scholar] [CrossRef]
  330. Atteritano, M.; Santoro, D.; Corallo, G.; Visalli, E.; Buemi, M.; Catalano, A.; Lasco, A.; Bitto, A.; Squadrito, F. Skin Involvement and Pulmonary Hypertension Are Associated with Vitamin D Insufficiency in Scleroderma. Int. J. Mol. Sci. 2016, 17, 2103. [Google Scholar] [CrossRef]
  331. Demir, M.; Uyan, U.; Keçeoçlu, S.; Demir, C. The relationship between vitamin D deficiency and pulmonary hypertension. Prague Med. Rep. 2013, 114, 154–161. [Google Scholar] [CrossRef]
  332. Limper, M.; Scirè, C.A.; Talarico, R.; Amoura, Z.; Avcin, T.; Basile, M.; Burmester, G.; Carli, L.; Cervera, R.; Costedoat-Chalumeau, N.; et al. Antiphospholipid syndrome: State of the art on clinical practice guidelines. RMD Open 2018, 4, e000785. [Google Scholar] [CrossRef]
  333. Cervera, R. Antiphospholipid syndrome. Thromb. Res. 2017, 151, S43–S47. [Google Scholar] [CrossRef]
  334. Andreoli, L.; Piantoni, S.; Dall’Ara, F.; Allegri, F.; Meroni, P.; Tincani, A. Vitamin D and antiphospholipid syndrome. Lupus 2012, 21, 736–740. [Google Scholar] [CrossRef] [PubMed]
  335. Agmon-Levin, N.; Blank, M.; Zandman-Goddard, G.; Orbach, H.; Meroni, P.L.; Tincani, A.; Doria, A.; Cervera, R.; Miesbach, W.; Stojanovich, L.; et al. Vitamin D: An instrumental factor in the anti-phospholipid syndrome by inhibition of tissue factor expression. Ann. Rheum. Dis. 2011, 70, 145–150. [Google Scholar] [CrossRef] [PubMed]
  336. Klack, K.; de Carvalho, J.F. High frequency of vitamin D insufficiency in primary antiphospolipid syndrome. Jt. Bone Spine 2010, 77, 489–490. [Google Scholar] [CrossRef]
  337. Paupitz, J.; Freire de Carvalho, J.; Caparbo, V.; Klack, K.; Pereira, R. Primary antiphospholipid syndrome in premenopausal women: Low vitamin D, high fat mass and maintained bone mineral mass. Lupus 2010, 19, 1302–1306. [Google Scholar] [CrossRef]
  338. Riancho-Zarrabeitia, L.; Cubería, M.; Muñoz, P.; López-Hoyos, M.; García-Canale, S.; García-Unzueta, M.; Hernández, J.L.; Martínez-Taboada, V.M. Vitamin D and antiphospholipid syndrome: A retrospective cohort study and meta-analysis. Semin. Arthritis Rheum. 2018, 47, 877–882. [Google Scholar] [CrossRef]
  339. Al Hussain, T.; Hussein, M.H.; Al Mana, H.; Akhtar, M. Pathophysiology of IgA Nephropathy. Adv. Anat. Pathol. 2017, 24, 56–62. [Google Scholar] [CrossRef]
  340. Li, X.; Li, J.; Fang, X.; Bakzhan, K.; Wang, L.; Van der Bruggen, B. A synergetic analysis method for antifouling behavior investigation on PES ultrafiltration membrane with self-assembled TiO2 nanoparticles. J. Colloid Interface Sci. 2016, 469, 164–176. [Google Scholar] [CrossRef]
  341. Agarwal, K.; Czaja, A.J.; Donaldson, P.T. A functional Fas promoter polymorphism is associated with a severe phenotype in type 1 autoimmune hepatitis characterized by early development of cirrhosis. Tissue Antigens 2007, 69, 227–235. [Google Scholar] [CrossRef]
  342. Sirbe, C.; Simu, G.; Szabo, I.; Grama, A.; Pop, T.L. Pathogenesis of Autoimmune Hepatitis—Cellular and Molecular Mechanisms. Int. J. Mol. Sci. 2021, 22, 13578. [Google Scholar] [CrossRef]
  343. Agarwal, K.; Czaja, A.J.; Jones, D.E.; Donaldson, P.T. Cytotoxic T lymphocyte antigen-4 (CTLA-4) gene polymorphisms and susceptibility to type 1 autoimmune hepatitis. Hepatology 2000, 31, 49–53. [Google Scholar] [CrossRef] [PubMed]
  344. Vogel, A.; Strassburg, C.P.; Manns, M.P. Genetic association of vitamin D receptor polymorphisms with primary biliary cirrhosis and autoimmune hepatitis. Hepatology 2002, 35, 126–131. [Google Scholar] [CrossRef] [PubMed]
  345. Garcia-Carrasco, M.; Jiménez-Herrera, E.A.; Gálvez-Romero, J.L.; de Lara, L.V.; Mendoza-Pinto, C.; Etchegaray-Morales, I.; Munguía-Realpozo, P.; Ruíz-Argüelles, A.; Jose, R.; Vera-Recabarren, M.; et al. Vitamin D and Sjögren syndrome. Autoimmun. Rev. 2017, 16, 587–593. [Google Scholar] [CrossRef] [PubMed]
  346. Kuo, C.-Y.; Huang, Y.-C.; Lin, K.-J.; Tsai, T.-Y. Vitamin D Deficiency Is Associated with Severity of Dry Eye Symptoms and Primary Sjögren’s Syndrome: A Systematic Review and Meta-Analysis. J. Nutr. Sci. Vitaminol. 2020, 66, 386–388. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immunomodulatory actions of active vitamin D (1,25-dihydroxyvitamin D3; 1,25-(OH)2D3) include effects on the innate and adaptative immune systems. 1,25-(OH)2D3 exerts its effect via direct binding on both VDR on the dendritic cell (DC) and the T lymphocytes. Calcitriol intervenes in APC differentiation and function by promoting APC to become more tolerogenic and decreasing the expression of major histocompatibility complex (MHC) class II and other similar molecules on the cell surface. Vitamin D may also reduce T helper (Th) cell differentiation and proliferation and induce a more tolerogenic immune response than a pro-inflammatory status with induction of T helper-2 (Th 2)-lymphocytes and regulatory T lymphocytes (Tregs), with downregulation of the pro-inflammatory T helper-1 (Th 1) lymphocytes, T helper-17 (Th 17) lymphocytes, and T helper-9 (Th 9) lymphocytes. Other abbreviations: IL: interleukin; Ig M: immunoglobulin M; Ig G: immunoglobulin G; IFN-γ: interferon-γ; TNF-α: tumor necrosis factor-α; toll-like receptors (TLRs), GATA-3: GATA binding protein-3; FoxP3: forkhead box P3, CTLA-4: cytotoxic T lymphocyte-associated protein-4.
Figure 1. Immunomodulatory actions of active vitamin D (1,25-dihydroxyvitamin D3; 1,25-(OH)2D3) include effects on the innate and adaptative immune systems. 1,25-(OH)2D3 exerts its effect via direct binding on both VDR on the dendritic cell (DC) and the T lymphocytes. Calcitriol intervenes in APC differentiation and function by promoting APC to become more tolerogenic and decreasing the expression of major histocompatibility complex (MHC) class II and other similar molecules on the cell surface. Vitamin D may also reduce T helper (Th) cell differentiation and proliferation and induce a more tolerogenic immune response than a pro-inflammatory status with induction of T helper-2 (Th 2)-lymphocytes and regulatory T lymphocytes (Tregs), with downregulation of the pro-inflammatory T helper-1 (Th 1) lymphocytes, T helper-17 (Th 17) lymphocytes, and T helper-9 (Th 9) lymphocytes. Other abbreviations: IL: interleukin; Ig M: immunoglobulin M; Ig G: immunoglobulin G; IFN-γ: interferon-γ; TNF-α: tumor necrosis factor-α; toll-like receptors (TLRs), GATA-3: GATA binding protein-3; FoxP3: forkhead box P3, CTLA-4: cytotoxic T lymphocyte-associated protein-4.
Ijms 23 09784 g001
Figure 2. The proteins encoded by the 15 key genes are present at a certain location in the cell. The classification of the proteins is based on their transcriptome profile: orange—genes that encode proteins related to autoimmunity; purple—genes that encode proteins with a general function in infection; blue—proteins that are involved in acute response to infection.
Figure 2. The proteins encoded by the 15 key genes are present at a certain location in the cell. The classification of the proteins is based on their transcriptome profile: orange—genes that encode proteins related to autoimmunity; purple—genes that encode proteins with a general function in infection; blue—proteins that are involved in acute response to infection.
Ijms 23 09784 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sîrbe, C.; Rednic, S.; Grama, A.; Pop, T.L. An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases. Int. J. Mol. Sci. 2022, 23, 9784. https://doi.org/10.3390/ijms23179784

AMA Style

Sîrbe C, Rednic S, Grama A, Pop TL. An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases. International Journal of Molecular Sciences. 2022; 23(17):9784. https://doi.org/10.3390/ijms23179784

Chicago/Turabian Style

Sîrbe, Claudia, Simona Rednic, Alina Grama, and Tudor Lucian Pop. 2022. "An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases" International Journal of Molecular Sciences 23, no. 17: 9784. https://doi.org/10.3390/ijms23179784

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