Next Article in Journal
Suppression of Oral Sweet Sensations during Consumption of Sweet Food in Humans: Effects on Gastric Emptying Rate, Glycemic Response, Appetite, Food Satisfaction and Desire for Basic Tastes
Next Article in Special Issue
Impact of Epigenetics on Complications of Fanconi Anemia: The Role of Vitamin D-Modulated Immunity
Previous Article in Journal
Circulating Ionized Magnesium as a Measure of Supplement Bioavailability: Results from a Pilot Study for Randomized Clinical Trial
Previous Article in Special Issue
Vitamin D and Immunity in Infants and Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin D’s Effect on Immune Function

Clinical and Experimental Endocrinology (CEE), KU Leuven, Campus Gasthuisberg O&N1, Herestraat 49, box 902, 3000 Leuven, Belgium
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(5), 1248; https://doi.org/10.3390/nu12051248
Submission received: 5 April 2020 / Revised: 24 April 2020 / Accepted: 26 April 2020 / Published: 28 April 2020
(This article belongs to the Special Issue Vitamin D on Immune Function)

Abstract

:
Ever since its discovery by Windhaus, the importance of the active metabolite of vitamin D (1,25-dihydroxyvitamin D3; 1,25-(OH)2D3) has been ever expanding. In this review, the attention is shifted towards the importance of the extra-skeletal effects of vitamin D, with special emphasis on the immune system. The first hint of the significant role of vitamin D on the immune system was made by the discovery of the presence of the vitamin D receptor on almost all cells of the immune system. In vitro, the overwhelming effect of supra-physiological doses of vitamin D on the individual components of the immune system is very clear. Despite these promising pre-clinical results, the translation of the in vitro observations to solid clinical effects has mostly failed. Nevertheless, the evidence of a link between vitamin D deficiency and adverse outcomes is overwhelming and clearly points towards avoidance of vitamin D deficiency especially in early life.

1. Introduction

The significance of vitamin D for health was first demonstrated by the discovery that its deficiency causes rickets in children and osteomalacia in adults [1]. The identification of vitamin D was groundbreaking, and Windaus was awarded the Nobel Prize in 1938 for this discovery. Despite its name, it is not stricto sensu a vitamin, but in fact, a prohormone, as humans are not exclusively dependent on it by their diet. Vitamin D can be obtained from ultraviolet (UV)B-dependent (wavelength 290–315 nm) endogenous production, from the diet and from supplements [2]. The two major forms of vitamin D (calciferol) are vitamin D2 (ergocalciferol) and vitamin D3 (cholecalciferol) [3]. Vitamin D2 is formed by UV irradiation of ergosterol from vegetable origins, such as yeast and mushrooms, making them naturally rich sources of vitamin D. Vitamin D3 is formed in the skin by the UV irradiation of 7-dehydrocholesterol [2]. As only 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3 or calcitriol) is the active vitamin, both 25- and 1α-hydroxylation are required for activation. The 25-hydroxylation happens in the liver by at least five enzymes (i.e., CYP2DII, CYP2D25, CYP3A4, CYP2R1, and CYP27A1). The serum 25-hydroxyvitamin D3 (25-(OH)D3) reflects the nutritional vitamin D status. The next step to obtain active vitamin D is 1α-hydroxylation (CYP27B1). This enzyme is expressed by many cell types (i.e., skin, immune cells, bone cells, placenta), but is present at the highest concentration in the kidney proximal tubule cells. The activity of the 1α-hydroxylase enzyme in the kidney is highly regulated by calcium and phosphate. Regulation of the 1α-hydroxylase enzyme in the other tissues has only little feedback inhibition. Breakdown of both 25-(OH)D3 and 1,25-(OH)2D3 is done by the same 24-hydroxylation enzyme (CYP24A1). Vitamin D and its metabolites in the circulation are bound to the multifunctional vitamin D-binding protein (DBP), which, besides the transport of vitamin D, also functions as modulator of inflammatory and immune responses as well as a regulator of bone development [1,2,3,4].
The classic vitamin D receptor (VDR) belongs to the nuclear receptor superfamily. Ligand binding results in heterodimerization with the retinoic X receptor (RXR). This complex then classically binds to the vitamin D responsive elements (VDRE) in the promotor region of target genes to exert the genomic effects of 1,25-(OH)2D3 [5]. Non-genomic effects of calcitriol proceed via the binding of 1,25-(OH)2D3 to a membrane bound VDR complexed to caveolin-1 [6]. The pleiotropy of vitamin D effects is ever expanding, and vitamin D analogs are produced to help in this exploration [7].
To express clinically relevant reserves of vitamin D, serum 25-(OH)D3 levels are used (defined as the sum of 25-(OH)D2 and 25-(OH)D3), as 1,25-(OH)2D3 is homeostatic regulated and has a short half-life (4–8 h). Nonetheless, the major problem remains the standardization of tests [8]. There are mainly two different methodologies: competitive immunoassays (such as competitive binding-protein assays or radioimmunoassays) and methods based on high-performance liquid chromatography (HPLC) and direct detection with liquid chromatography tandem-mass spectrometry (LC-MS/MS), with the latter methods being the gold standard [9,10,11].
Besides there being issues with standardization of testing, there is also controversy about levels of adequacy. The Endocrine Society defines deficiency as 25-(OH)D3 levels less than 20 ng/mL (50 nmol/L) and insufficiency as levels 21–29 ng/mL (52 to 72 nmol/L) [12]. These cut-offs are determined based on parathyroid hormone (PTH) levels and intestinal calcium transporter activity that normalizes as 25-(OH)D3 levels reach the current cut-offs [13,14,15]. The Institute of Medicine (IOM) on the other hand, states there is no increased benefit of serum 25-(OH)D3 levels above 20 ng/mL (50 nmol/L) and defines deficiency as 25-(OH)D3 levels less than 12 ng/mL (30 nmol/L) and insufficiency as levels 12–20 ng/mL (30–50 nmol/L) [16]. Vitamin D intoxication is observed at concentrations higher than 150 ng/mL (374 nmol/L) [2].
Based on these cut-offs, there is a high prevalence (up to 40% in adults) of vitamin D insufficiency in both children and adults. A study in 6275 American children and adolescents aged 1–21 years showed that 61% were 25-(OH)D3 insufficient and 9% deficient [17]. In adults, up to 40% are 25-(OH)D3 insufficient and 6% deficient [18,19]. A more disturbing trend is the shift in the American population towards even lower 25-(OH)D3 levels without a logic explanation aside from a decrease in sun exposure as the population becomes more overweight and thus engages less in outdoor activities, next to a possible better sun protection [20]. In the face of this ominous trend, the current consensus remains that population-wide screening for vitamin D deficiency is not recommended, but to limit testing to those individuals at risk of developing deficiency (hyper- and hypoparathyroidism, kidney disease, osteoporosis, etc.) [12,21,22,23]. Despite these recommendations, the importance of vitamin D is rising and parallels an increasing trend in its testing (albeit with a significant healthcare cost) [22,23].
As the aforementioned stresses the controversy about cut-off values, there is even more discussion about the correct substitution regimen. It is demonstrated that with a daily dose of up to 1000–2000 IU (25–50 µg) per day, there is a linear dose-response curve between vitamin D intake and serum 25-(OH)D3, which flattens at higher intakes [23]. Based on randomized controlled trials it has been calculated that an intake of 1040 IU (26 µg) per day is required in vitamin D deficiency and 400 IU (10 µg) per day in vitamin D insufficiency in order to obtain a concentration >20 ng/mL (50 nmol/L) in 97.5% of the population [24].
Several studies have tried to determine the optimal dosing regimens to correct deficiency. Irrespective of the interval or the exact substitution dose, most dosing regimens result in adequate serum 25-(OH) D3 levels, although higher doses and especially loading regimens result in a faster accomplishment of sufficiency [25,26,27,28,29,30,31,32,33,34].
Once adequate vitamin D values are reached, to further preserve adequate vitamin D levels in adults, the IOM recommends a daily dose of 600 IU per day, while the Endocrine Society recommends a dose of 600–2000 IU per day (according to the amount of sunlight the individual is exposed to) [12,35,36]. To explain the role of sunlight, it is estimated that a daily exposure of 7–30 min (depending on skin color, latitude, and season) is required to meet vitamin D substitution doses [35].
One of the major caveats in the use of vitamin D substitution is the risk of excessive substitution resulting in renal failure and cardiac arrest because of hypercalcemia. Both the European Food Safety Authorization and the IOM determined that the upper tolerable limit of vitamin D intake in adults is 4000 IU/day (100 µg/day) [36,37], mostly because there seems to be no additional health benefit in doses higher than 4000 IU/day [38].

2. Vitamin D’s Role in Immune Function: In Vitro Data

The importance of vitamin D in the regulation of both the innate and adaptive immune system was demonstrated by the discovery of the presence of VDR expression in almost all cells of the immune system, as well as the presence of the metabolizing hormones in immune cells [39,40]. Also, gut epithelial VDR is important in protecting the mucosal barrier integrity and regulating the gut innate immunity (recently demonstrated by innate lymphoid cells) [41,42,43]. The effect of vitamin D on immune cells is complex, as illustrated by the fact that VDR expression in immune cells is differently controlled according to their corresponding activation status. For example, T-cells gain a higher concentration of VDR upon activation with an increase that is already significant after eight hours and reaches a maximum 48 h after activation [44]. Monocytes on the other hand lose VDR expression by differentiating into either macrophages or dendritic cells (DCs) [45]. In immune cells, the 1α-hydroxylase enzyme, although the same enzyme as in the renal tubules, is not regulated by negative feedback by 1,25-(OH)2D3 itself [46]. As immune cells also express 24-hydroxylase, this is only minimally regulated by 1,25-(OH)2D3 and depends on the activation status of the immune cells [45,47,48,49]. Essentially, vitamin D results in a shift in the immune status towards a more tolerogenic status [4].

2.1. Innate

2.1.1. Monocyte/Macrophage

Both monocytes as macrophages express the VDR, but as monocytes differentiate towards macrophages, there is a decrease in the expression levels of the VDR [45]. Additionally, the expression of the 1α-hydroxylase enzyme on monocytes and macrophages is upregulated by immune stimuli (i.e., signal transducer and activator of transcription-1α (STAT-1α), interferon-γ (IFN-γ), lipopolysaccharide (LPS), toll like receptors (TLR)) [50,51]. 1,25-(OH)2D3 results in an anti-inflammatory activity on macrophages as it increases interleukin (IL)-10 and decreases inflammatory stimuli (i.e., IL-1β, IL-6, tumor necrosis factor-α (TNF-α), receptor activator of nuclear factor kappa-Β ligand (RANKL), and cyclo-oxygenase-2 (COX-2)) [3]. The downregulation of inflammatory cytokines happens through the upregulation of mitogen-activated protein kinase (MAPK) phosphatase (MKP)-1 by 1,25-(OH)2D3 and subsequent inhibition of LPS-induced p38 activation [52].
Another pathway of inhibition of inflammatory cytokines is through inhibition of COX-2 expression by targeting thioesterase superfamily member 4 (an Akt modulator protein) [53]. In addition, activation of the TLR results in increased expression of VDR, however, TLR-mediated inflammation is controlled by 1,25-(OH)2D3 as it stimulates suppression of cytokine signaling 1 (SOCS-1) through miRNA-155 downregulation [54,55]. 1,25-(OH)2D3 has a direct antimicrobial role in monocytes and macrophages by induction of cathelicidin antimicrobial peptide (CAMP), with an increase of hCAP18 and LL-37, and by targeting defensing β2 (DEFB4) [56,57,58]. 1,25-(OH)2D3 also has an anti-oxidative effect on monocytes by upregulation of glutathione reductase (GR) and glutamate-cysteine ligase (GCL), which results in the reduced formation of oxygen radicals (such as reactive oxygen species (ROS) [59,60]. Some controversy exists in the involvement of 1,25-(OH)2D3 in the expression of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) formation, as some describe upregulation, but others report inhibition [61,62]. Recent studies suggest that 1,25-(OH)2D3 is able to modulate the epigenome of immune cells, especially monocytes during antigen encounter and differentiation of the innate immune system [11,63].

2.1.2. Dendritic Cells

1,25-(OH)2D3 modulates DCs towards a less mature and more tolerogenic phenotype with changes in both morphology (more adherent spindle-shaped cells), as in cytokine production and surface markers [64,65,66]. There is a decreased expression of major histocompatibility complex (MHC) II, cluster of differentiation (CD) 80, CD86 (co-stimulatory molecules), and CD54 (adhesion molecule), and increased expression of CCR5 (chemokine receptor), DEC205 (antigen-uptake receptor), F4/80 (macrophage marker), and CD40 [65,67,68]. The cytokines IL-6 and IL-12 decrease together with an increase in IL-10 [3,65,67,68]. 1,25-(OH)2D3 upregulates the expression of immunoglobulin-like transcript (ILT)-3 and programmed death-ligand 1 (PD-L1), the latter contributing to the induction of regulatory T-cells (Tregs) [69]. TNF secretion by DCs appears to be another essential regulator in the induction of antigen-specific suppressive T-cells by 1,25-(OH)2D3 (Figure 1) [70].

2.1.3. Others

Both natural-killer (NK) cells and neutrophils express the VDR [71]. 1,25-(OH)2D3 has an antithetical effect on neutrophils helping to minimize damage by pathogens. On the one hand, 1,25-(OH)2D3 increases the destructive power against pathogens by increased expression of cathelicidin, α and β-defensins [72,73]. Moreover, 1,25-(OH)2D3 helps to reduce bystander destruction of the pro-inflammatory response by reducing expression of Trappin-2/elafin/skin-derived anti-leucoproteinase (an inhibitor of elastase, associated with a pro-inflammatory response) and reducing migration of neutrophils [74,75,76]. Furthermore, vitamin D has been shown to reduce the formation of neutrophil extracellular traps (NETs), and thus reduces both the response against invading pathogens (NETs trap and kill pathogens) as well as the risk of autoimmunity (autoantigens are exposed and the complement system is activated in NETs) [77,78]. The effect of 1,25-(OH)2D3 on NK cells is also immunoregulatory as it results in a decreased expression of IFN-γ, CD107a (suggests decreased cytotoxic activity), and granzymes A and B [71,79,80].

2.2. Adaptive

2.2.1. T-lymphocytes

1,25-(OH)2D3 can both directly and indirectly influence T-lymphocytes. The indirect pathway involves the modulation of the T-lymphocyte stimulatory function of antigen-presenting cells (APC). In monocytes and macrophages, 1,25-(OH)2D3 downregulates surface expression of MHC class II and co-stimulatory molecules (such as CD40, CD80, and CD86), and thus decreases antigen presentation [81]. 1,25-(OH)2D3 has the same effect in DCs and also inhibits their production of IL-12 and IL-23, besides a stimulation of the release of IL-10 and macrophage inflammatory protein-3α (MIP-3α) [82]. Taken together the indirect effect of 1,25-(OH)2D3 (especially on DCs as they are believed to be its central target) is a modulation of T-lymphocyte response. There will be a decrease in autoreactive T-lymphocyte proliferation, induction of both early (annexin V+/PI-) and late (annexin V+/PI+) apoptosis of autoreactive T-lymphocytes, and even a rise of Tregs [49,83]. The DC derived cytokines will alter the Thelper (Th)–lymphocyte balance from a Th1 and Th17 predominance towards a Th2 phenotype (Figure 1) [82,84].
The direct effect of 1,25-(OH)2D3 is variable, as it is dependent on the activation state of the T-lymphocyte as they gain a higher VDR concentration upon activation [44,85]. 1,25-(OH)2D3 inhibits the production of Th1 cytokines (i.e., IL-2, IFN-γ), Th17 cytokines (i.e., IL-17, IL-21), and Th9 cytokines (i.e., IL-9) [84,86,87,88]. The direct effects on Th2 cytokines are more controversial with a proclaimed upregulation of GATA binding protein-3 (GATA-3), c-maf, and IL-4 [85,89]. Likewise, there is an induction of IL-10-producing Tregs by binding to the Forkhead box P3 (FoxP3) promotor region, and increased expression of FoxP3 alongside cytotoxic T-lymphocyte antigen- 4 (CTLA-4) [82,86,90,91]. Because CTLA-4 expression is a key mechanism for Treg suppression, the upregulation of CTLA-4 by 1,25-(OH)2D3 suggests its more tolerogenic character (Figure 1) [86].

2.2.2. B-lymphocytes

The presence of VDR in human B-lymphocytes with upregulation of both VDR as well as the 1α-hydroxylation enzyme, suggests a strong influence of vitamin D on B-lymphocytes [68]. It has been shown that 1,25-(OH)2D3 induces apoptosis of activated B-lymphocytes, and impedes the generation of plasma cells (by modulation of CD40 and thus NF-κB) and post-switch memory B-lymphocytes, without affecting B-lymphocyte differentiation [92,93,94]. It has been hypothesized that 1,25-(OH)2D3 may have a potential benefit in maintaining B-lymphocyte homeostasis in autoimmune diseases based on B-lymphocyte proliferation [71]. 1,25-(OH)2D3 also upregulates the production of IL-10 by B-lymphocytes, resulting in an additional regulatory effect [95]. Besides its direct role on B-lymphocyte function, 1,25-(OH)2D3 reduces the activation of T-lymphocytes by B-lymphocytes (by downregulation of CD86 expression and upregulation of CD74) [3,96].

3. The Impact of Vitamin D Deficiency on the Immune System: In Vitro and In Vivo Data

Epidemiological data link vitamin D deficiency to a defective functioning of the immune system with an increased risk of infections and a predisposition to autoimmune disease [97].
In particular, in the case of infections, associations have been described between 25-(OH)D3 deficiency and an increased risk for infections with mycobacterium tuberculosis and respiratory tract infections [98,99,100]. A large systematic review (of 10,933 subjects) showed that vitamin D supplementation (both D2 as D3) was protective against acute respiratory tract infections in a 25-(OH)D3 deficient population, especially in those receiving daily or weekly supplementation [101]. However, in children this protective effect could not be reproduced [102]. The mechanism by which vitamin D prevents respiratory tract infections is based on in vitro research that shows that 1,25-(OH)2D3 results in increased expression of cathelicidin, regulation of cytokine release, and suppression of the adaptive response by boosting the innate immune system [103]. In children and adults vitamin D3 as an adjunct to antibiotics did not have an additional beneficial effect in the treatment of acute bacterial pneumonia, although there was evidence that there was a trend towards faster resolution of radiographic manifestations in those with low baseline 25-(OH)D3 levels [104,105].
As vitamin D has an important effect on macrophages, and tremendous effort has been put into linking vitamin D to tuberculosis. It has been demonstrated that 25-(OH)D3 deficiency increases the risk of developing active tuberculosis [106]. Possible reasons are that 1,25-(OH)2D3 leads to activation and enhanced mycobactericidal activity of macrophages by induction of CAMP and DEFB4 [107,108]. Furthermore, adding vitamin D supplementation (both D2 as D3) to anti-tuberculosis treatment has been shown to have a beneficial effect [109]. Even in chronic obstructive pulmonary disease (COPD), it has been demonstrated that patients with COPD were more likely to suffer from 25-(OH)D3 deficiency than matched healthy smokers, with a deterioration of COPD-classification and exacerbation rate associated with a further decrease in serum 25-(OH)D3 [110,111]. Restoring 25-(OH)D3 deficiency reduces incidence of exacerbations, however only in cases of severe 25-(OH)D3 deficiency at baseline (at least <20 ng/mL or 50 nmol/L) [112,113].
In autoimmune diseases, there is a clear association between 25-(OH)D3 deficiency and the incidence of autoimmunity. In type 1 diabetes (T1D), multiple sclerosis (MS), rheumatoid arthritis (RA), systemic sclerosis (SSc), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD), circulating levels of 25-(OH)D3, independent of seasonal variation and latitude, are decreased at disease onset as well as during follow-up [114,115,116,117,118,119,120,121,122,123]. A systematic review (of 5942 subjects) demonstrated that in both MS as in T1D, as principal examples of immune-mediated diseases, there was a lower level of 25-(OH)D3 in affected patients than in the healthy control group [124]. In SLE, lower levels of 25-(OH)D3 were even associated with an increased frequency of lupus flares [125].
This was not completely confirmed in SSc, as the diffuse type has been shown to have significantly lower levels of 25-(OH)D3 than the limited type; although within both types, the level per se is not associated with disease severity [119]. In general, because only a relatively small percentage of the general population has a 25-(OH)D3 sufficiency, only a true deficiency (25-(OH)D3 levels less than 12 ng/mL or 30 nmol/L) seems to be correlated with an increased prevalence and aggressiveness of autoimmune diseases [124,126].
The link between vitamin D and autoimmune disease is furthermore supported by both seasonal variation (increased prevalence in children born in spring) and latitude (higher prevalence in northern countries with less UVB radiation) [127,128,129,130,131,132]. Additional evidence for the link between vitamin D and autoimmunity is found on the genome level as single nucleotide polymorphisms (SNPs) in three key vitamin D metabolism genes (i.e., DHCR7 and CYP2R1: determinants of circulating 25-(OH)D3, and CYP27B1: vitamin D signaling in T-lymphocytes) and VDR genes (i.e., TaqI and BsmI) have been linked to an increased risk of respectively T1D and MS [116,133,134]. Furthermore, SNPs in VDR genes (i.e., ApaI, BgII GT and TaqI) are associated with SSc and IBD, respectively [135,136]. Since these SNPs result in a decreased efficacy of vitamin D substitution, this supports the importance of vitamin D on autoimmunity. Moreover, there is an association between certain VDR polymorphisms and autoimmunity as VDR FokI and TaqI polymorphisms are associated with an increased risk in SLE and RA [137,138].
As the importance of restoring a 25-(OH)D3 deficiency was stressed by its role in infections as well as in immune-mediated diseases, this was extrapolated to critically ill patients, where a distinct association was demonstrated between low 25-(OH)D3 levels and adverse outcomes (both in morbidity as in mortality) [139,140,141,142,143,144,145]. It has been shown that in critically ill patients 25-(OH)D3 even continue to decrease if no substitution is started [140]. In contrast to these findings in adults, a recent large meta-analysis in critically ill children was not able to link a 25-(OH)D3 deficiency to a higher mortality [146]. The reason for a lower 25-(OH)D3 in critically ill patients (both at admission and the subsequent decrease) is still not completely understood. One explanation is a decrease in DBP due to reduced protein synthesis and increased clearance (although reduced levels of DBP do not affect the free 25-(OH)D3 concentration) [140]. Another possible explanation is that low 25-(OH)D3 is a marker of illness as inflammatory processes reduce 25-(OH)D3, so an important consideration in critically ill patients might be that serum 25-(OH)D3 is the consequence of illness and not the cause [147,148].
The exact mechanism of protection by vitamin D3 remains elusive. Based on current knowledge, the most important observation is that vitamin D3 results in a shift from an inflammatory Th1 response towards a pro-tolerogenic Th2 response with an arrest of cytotoxic T-lymphocyte infiltration and an increase in CD4+CD25+ Tregs [149,150,151]. In healthy individuals, vitamin D3 increases the absolute number of Tregs without altering their suppressor function [152]. On the other hand, a similar dose of vitamin D3, in vivo, was shown to improve the suppressor function of Tregs, in T1D and MS patients, without altering their absolute number [153,154,155,156,157]. Of note, the baseline frequency and absolute number of Tregs in peripheral blood before initiation of vitamin D3 supplementation was not different between healthy controls and patients [158]. It is clear that the exact effect on Tregs is still not completely known, as in SLE (in vivo), vitamin D3 resulted in an increase in Tregs, independent of the patients’ vitamin D status [159]. Besides its major impact on Tregs, vitamin D3 is able to directly reduce effector T-lymphocytes as has been demonstrated, in vivo, in both MS as SLE [160,161].
The change towards a more tolerogenic status is also reflected by a change in the cytokine profile. In vitro, 1,25-(OH)2D3 resulted in a reduction of IL-1, IL-6, and IL-17, together with a reduction in TNF-α, as demonstrated in RA, and an increase in IL-4, IL-5, and IL-10 and a reduction in IFN-γ, as demonstrated in IBD [162,163,164,165,166]. To extrapolate these in vitro findings to in vivo studies is more complicated, however results are also pointing towards a more tolerogenic state, as in MS there was a decrease in IL-17 and an increase in IL-10 with the latter also being observed in IBD [167,168,169,170]. Furthermore, in vivo, there was a decrease in inflammatory cytokines IL-23 and IL-17 in RA and even a direct antifibrotic effect by impairment of TGF-β in SSc [171,172].
Furthermore, 1,25-(OH)2D3 and even 25-(OH)D3 affect the maturation and migration of DCs, conferring an immunoregulatory role and tolerogenic phenotype, characterized by IL-10 production and thus again promoting tolerance [173,174]. This finding is supported by the comparison of transcriptomes of 1,25-(OH)2D3/dexamethasone-modulated tolerogenic with non-modulated mature inflammatory DCs, and it was shown that these modulated DCs had immunomodulating effects, including the induction of Tregs [175,176]. In addition, in SLE, both in vitro and in vivo experiments show that vitamin D counterbalances B-lymphocyte hyperactivity by inducing early apoptosis in B-lymphocytes with a possible favorable effect [94,161,171].

4. Vitamin D Metabolites as Immune Modulators in Autoimmune Diseases: Animal Models and Human Data

As vitamin D is an important regulator of the immune system with a preponderance towards tolerance induction, its therapeutic potential as an immune modulator is appealing in the treatment of immune-mediated diseases. Studies in animal models of autoimmune diseases show that restoring serum 25-(OH)D3 levels using high doses of vitamin D metabolites (i.e., 25-(OH)D3 and 1,25-(OH)2D3) or less calcemic vitamin D analogues, can alter the course of autoimmune diseases like T1D, MS, or RA [177]. One of the first studies to show a decrease in T1D incidence by vitamin D was performed in the non-obese diabetic (NOD) mouse model, which was able to demonstrate a significant decrease in T1D incidence due to long-term treatment with high dose 1,25-(OH)2D3 starting from a young age [151,178,179]. Later in this model it was demonstrated that T1D could be arrested by treatment with a 1,25-(OH)2D3 analog, possibly by increasing Tregs and inhibition of Th1-lymphocytes [150,180]. In the experimental autoimmune encephalitis (EAE) mouse model of MS, the effect on disease alterations has been even more intensely studied, with an inhibition of EAE by increasing IL-4 and transforming growth factor-β1 (TGF-β1) and by modulation of the JAK-STAT pathway in the IL-12/IFN-γ axis [181]. In the pristane-induced model of SLE, there was only a reduction in IFN-γ, but no effect on IL-4 [182]. In addition, a reduction in the pro-inflammatory cytokines IL-1β, IL-6, IL-8 and prostaglandin E2 was described in the type II collagen injection rat model of RA [183]. Recently in the Act1-/- mouse model of SLE and Sjögren syndrome, amongst other changes characterized by a peripheral B-lymphocyte expansion, it was demonstrated that lower levels of vitamin D3 are linked to an increase in memory B-lymphocytes [184]. Furthermore, in collagen-induced arthritis, the mouse model of RA 1,25-(OH)2D3 was able to decrease severity of arthritis by downregulation of Th17 cells and increasing Tregs [185].
Later these animal models were translated to humans as the European Diabetes Centers EURODIAB study showed a significant decrease in T1D onset in children receiving vitamin D supplementation [186]. The first major birth-cohort study was done in Finland and resulted in a staggering 80% reduction in T1D onset in 10,336 children receiving vitamin D supplementation before the age of 1 year, irrespective of the dose of supplementation [187]. Likewise, in MS, low neonatal 25-(OH)D3 concentrations are associated with an increased risk of disease incidence [126]. Based on cohort data of more than 7 million persons, it was proven in MS that vitamin D supplementation resulted in significant lower disease incidence [188,189].
As it was demonstrated in both animal and human studies that the age of intervention was critical in disease prevention, the rationale was that ensuring adequate 25-(OH)D3 levels as early as during pregnancy would be critical for disease prevention. In MS, a large systematic review showed that 25-(OH)D3 deficiency during pregnancy resulted in an increased prevalence of MS in the offspring [190]. Therefore, in theory, maternal vitamin D supplementation during pregnancy would seem protective. However, in T1D, studies indicated that maternal vitamin D supplementation during pregnancy did not reduce the risk on T1D in the offspring [191,192,193]. Even a recent large cohort study, The Environmental Determinants of Diabetes in the Young (TEDDY), conducted on 8676 European and American children with T1D-associated human leukocyte antigen (HLA) genotypes, and thus an increased risk for the development of islet autoimmunity and T1D, was not able to demonstrate an effect of maternal vitamin D supplementation on islet-autoimmunity in the offspring [194]. Unfortunately, to our knowledge there are also no studies investigating the effect of vitamin D supplementation during pregnancy on the incidence of MS or rheumatic diseases in offspring. An important note in a lot of these (smaller) studies, and one of the main reasons for continuing debate, is that most of these studies are performed in retrospect resulting in inconsistency in timing of the start of intervention, difference in dosing regimens and eventually conflicting results [193,195,196,197,198,199,200]. It is because of these conflicting results that the aim is currently changing into identifying subpopulations predicted to benefit most from vitamin D supplementation. The earlier mentioned TEDDY cohort study, conducted in children with T1D-associated HLA genotypes (and thus an increased risk for the development of islet autoimmunity and T1D), demonstrated that vitamin D supplementation seemed to be only beneficial in those with minor alleles at VDR ApaI [201]. Others found similar results in those homozygous for the VDR Cdx2 G/G [202]. These studies are examples of future tailored medicine as specific VDR genotypes seem to have distinct functional effects on immune cells, e.g., depending on the Fokl polymorphism, the effect of both 25-(OH)D3 and 1,25-(OH)2D3 results in different functional effects on lymphocytes and monocytes [203,204].

5. Discrepancy between Promising in Vitro Data, Animal Models and Human Intervention Trials

Whereas the hypothesis that vitamin D and its metabolites have a role in normal physiology as immune modulators is now well supported by in vitro studies showing that there is a dose-dependent effect of vitamin D or its metabolites and even synthetic analogues on many immune cell subsets, the translation of these observations into solid results in clinical trials has failed and the scientific community is starting to question the relevance of the in vitro observations and even interventions in animal models for human health. So, what is missing? Why are the in vitro observations not translated in success in clinical intervention trials?

5.1. Dose of Vitamin D Products

Studying effects of vitamin D3 (i.e., metabolites or analogues) on immune cells in vitro is an artificial situation, with continued exposure of these isolated immune cell subsets and too high doses of vitamin D products, mostly 1,25-(OH)D2D3, being induced. Often supra-physiological concentrations are employed that are not achievable in human peripheral blood. Still, these concentrations could be achieved in local sites of inflammation, as also many immune cells can produce vitamin D products themselves upon activation [68,82]. Thus, the physiological relevance of the in vitro observations seems valid, but the translation to interventions with supplements of vitamin D products is problematic.
In animal models, it has been shown that in order to see any effect on disease modulation, the dose and route of administration of the vitamin D products are crucial. As such, in the T1D studies in NOD mice, therapy was only successful when doses of vitamin D or its metabolites or analogues were used that were at the edge of toxicity [178,180,205]. In addition, continuous administration, leading to continued exposure to the high doses of the products was needed, often lifelong [180].
When looking at human intervention trials, many used safe doses of regular vitamin D, lifting levels of 25-(OH)D3 above sufficiency levels, but far from the very high levels observed in those animal studies that measured levels and correlated them to efficacy of treatment. In NOD mice, for example, we showed that a very high dose of regular vitamin D was able to prevent T1D, but this study was also done using a very high dosing regimen that for humans would require a lifelong daily dose of 12,500 IU [180].
Translating in vitro studies and animal studies to humans, would mean using much higher doses of vitamin D products and thus reaching levels at which also side effects of hypercalcemia would be seen [205,206,207]. In many studies in animals, investigators avoided hypercalcemia by lowering calcium intake of the animals, a detail often not noted by readers of the manuscripts [208].
In addition, in the in vitro experiments, continued exposure of immune cells to the vitamin D products was used, and in most animal models treatment was given continuously (via food supplementation or oral gavage). In human studies, often intermittent administration or bolus doses are used to improve compliance. These bolus doses have rarely been tested in animal models and have the potential to induce completely different immune effects than the continuous exposure used in vitro and in animal studies. Indeed, exposure to extremely high doses of vitamin D products of immune cells in vitro induces very different effects, such as apoptosis or even necrosis of specific cell types [209,210]. Also in animals, a bolus injection of vitamin D induces even the opposite effect for instance on macrophages, paralyzing them, rather than making them more efficacious [211]. In those clinical studies where continued administration of vitamin D products was studied, there is the major issue of compliance, which is not an issue in in vitro studies or animal studies.
Finally, the immune effects of vitamin D products in vitro are not a full immune suppression, but rather an immune modulation, shifting the adaptive immune system towards tolerance to antigens and the innate immune system to a better viral and bacterial clearance (as discussed above). These effects are subtle and may not be sufficient to achieve dramatic effects by themselves when administered in monotherapy in human disease. So, rather than studying the effect of vitamin D products alone, combinations may be needed.

5.2. Timing of Intervention and Duration of Exposure

In vitro studies typically start with naïve immune cells, freshly isolated from immune organs or blood of healthy subjects, which are then exposed for days to vitamin D3 products. In animal models, likewise, therapy is often started before disease onset (most autoimmune models) or early on in the disease. Few studies have looked at effects once disease is overt, and those that have indicate that at that stage vitamin D products by themselves, even at high doses, are not disease altering anymore [212]. In humans however, most intervention studies are late stage (disease is present) and study vitamin D monotherapies. In addition, in animal models, therapy is maintained for weeks and months, often during the whole life of the animal, whereas in humans, shorter duration studies happen.

5.3. Relevance of Animal Models for Human Disease

A major weakness in translation of our observations in vitro and confirmatory studies in animal models to humans, is the relevance of the animal models studied. In autoimmune diseases, animal models are criticized as they are, for instance, induced (most animal models of EAE and RA), using antigens that are sometimes not relevant for human disease, or have a much more dramatic course (e.g., NOD T1D mouse model) than what is observed in humans.
Thus, success in these animal models, using high doses of vitamin D products, starting therapy before induction or before disease is present, treating animals for long periods is not necessarily a guarantee for success in human disease.

6. Concluding Remarks

There is an indisputable relation between vitamin D and the immune system. With respect to in vitro, overwhelming evidence exists for a physiological role for the vitamin D system in immune regulation, and immune modulation can be observed by exposing immune cells to pharmacological doses of vitamin D metabolites. In animal models and humans, a correlation exists between adverse immune outcomes (infections and autoimmune diseases) and vitamin D deficiency, but translation of the in vitro observations of active vitamin D3 on the immune system to solid results of regular vitamin D supplementation in clinical trials have mostly failed. An important reason might be that the choice of the vitamin D metabolite, as well as its dose and frequency of administration are critical factors that need to be considered when designing clinical trials. Many in vitro effects on isolated immune cells are induced by supra-physiological concentrations of 1,25-(OH)D2D3, which are probably not achievable with regular vitamin D supplements in humans, as these concentrations risk hypercalcemia and soft tissue calcifications. Moreover, recurrent use of regular vitamin D, for instance daily or weekly (in comparable cumulative doses) instead of every 6–12 months, may enhance long-term compliance depending on the lifestyles of the target groups. In addition, the timing of vitamin D intervention will be crucial. In animal models, vitamin D metabolites work best in a preventive setting, a time window that is often missed in human trials. Therefore, future randomized and controlled trials will be needed to investigate whether supplementation with regular vitamin D can indeed prevent or modify the course of inflammatory or autoimmune diseases in at-risk subjects. For now, the bottom line on the effect of vitamin D in the immune system is that avoidance of severe vitamin D deficiency improves immune health and decreases susceptibility to autoimmune diseases.

Author Contributions

Writing—Original Draft Preparation: P.-J.M.; Writing—Review and Editing: P.-J.M., C.G., A.V., and C.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

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]
  2. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  3. 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]
  4. Prietl, B.; Treiber, G.; Pieber, T.R.; Amrein, K. Vitamin D and immune function. Nutrients 2013, 5, 2502–2521. [Google Scholar] [CrossRef] [PubMed]
  5. Dusso, A.S.; Brown, A.J.; Slatopolsky, E. Vitamin D. Am. J. Physiol. Renal Physiol. 2005, 289, F8–F28. [Google Scholar] [CrossRef] [PubMed]
  6. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. 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]
  8. 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]
  9. Hollis, B.W. Assessment and interpretation of circulating 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D in the clinical environment. Endocrinol. Metab. Clin. N. Am. 2010, 39, 271–286. [Google Scholar] [CrossRef]
  10. Jimenez-Sousa, M.A.; Martinez, I.; Medrano, L.M.; Fernandez-Rodriguez, A.; Resino, S. Vitamin D in Human Immunodeficiency Virus Infection: Influence on Immunity and Disease. Front. Immunol. 2018, 9, 458. [Google Scholar] [CrossRef]
  11. Carlberg, C. Vitamin D Signaling in the Context of Innate Immunity: Focus on Human Monocytes. Front. Immunol. 2019, 10, 2211. [Google Scholar] [CrossRef] [Green Version]
  12. 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.; Endocrine, S. 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] [PubMed] [Green Version]
  13. Heaney, R.P.; Dowell, M.S.; Hale, C.A.; Bendich, A. Calcium absorption varies within the reference range for serum 25-hydroxyvitamin D. J. Am. Coll. Nutr. 2003, 22, 142–146. [Google Scholar] [CrossRef] [PubMed]
  14. Serdar, M.A.; Batu Can, B.; Kilercik, M.; Durer, Z.A.; Aksungar, F.B.; Serteser, M.; Coskun, A.; Ozpinar, A.; Unsal, I. Analysis of Changes in Parathyroid Hormone and 25 (OH) Vitamin D Levels with Respect to Age, Gender and Season: A Data Mining Study. J. Med. Biochem. 2017, 36, 73–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Bischoff-Ferrari, H.A.; Giovannucci, E.; Willett, W.C.; Dietrich, T.; Dawson-Hughes, B. Estimation of optimal serum concentrations of 25-hydroxyvitamin D for multiple health outcomes. Am. J. Clin. Nutr. 2006, 84, 18–28. [Google Scholar] [CrossRef]
  16. 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 Dietary Reference Intakes for Calcium and Vitamin D: What dietetics practitioners need to know. J. Am. Diet. Assoc. 2011, 111, 524–527. [Google Scholar] [CrossRef]
  17. 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]
  18. 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] [Green Version]
  19. 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] [Green Version]
  20. 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] [Green Version]
  21. LeFevre, M.L.; Force, U.S.P.S.T. 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] [PubMed] [Green Version]
  22. 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] [PubMed] [Green Version]
  23. Pilz, S.; Zittermann, A.; Trummer, C.; Theiler-Schwetz, V.; Lerchbaum, E.; Keppel, M.H.; Grubler, M.R.; Marz, W.; Pandis, M. Vitamin D testing and treatment: A narrative review of current evidence. Endocr. Connect. 2019, 8, R27–R43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cashman, K.D.; Ritz, C.; Kiely, M.; Odin, C. Improved Dietary Guidelines for Vitamin D: Application of Individual Participant Data (IPD)-Level Meta-Regression Analyses. Nutrients 2017, 9, 469. [Google Scholar] [CrossRef] [Green Version]
  25. Takacs, I.; Toth, B.E.; Szekeres, L.; Szabo, B.; Bakos, B.; Lakatos, P. Randomized clinical trial to comparing efficacy of daily, weekly and monthly administration of vitamin D3. Endocrine 2017, 55, 60–65. [Google Scholar] [CrossRef]
  26. 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 D (3) Supplementation. Nutrients 2018, 10, 659. [Google Scholar] [CrossRef] [Green Version]
  27. 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]
  28. 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]
  29. 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] [Green Version]
  30. Chel, V.; Wijnhoven, H.A.; 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] [Green Version]
  31. Mittal, H.; Rai, S.; Shah, D.; Madhu, S.V.; Mehrotra, G.; Malhotra, R.K.; Gupta, P. 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]
  32. 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]
  33. Schleck, M.L.; Souberbielle, J.C.; Jandrain, B.; Da Silva, S.; De Niet, S.; Vanderbist, F.; 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] [Green Version]
  34. Sanders, K.M.; Stuart, A.L.; Williamson, E.J.; Simpson, J.A.; Kotowicz, M.A.; Young, D.; Nicholson, G.C. 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] [Green Version]
  35. Bouillon, R. Comparative analysis of nutritional guidelines for vitamin D. Nat. Rev. Endocrinol. 2017, 13, 466–479. [Google Scholar] [CrossRef]
  36. 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]
  37. Spiro, A.; Buttriss, J.L. Vitamin D: An overview of vitamin D status and intake in Europe. Nutr. Bull. 2014, 39, 322–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Bouillon, R. Safety of high dose vitamin D supplementation. J. Clin. Endocrinol. Metab. 2019. [Google Scholar] [CrossRef] [PubMed]
  39. 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]
  40. Veldman, C.M.; Cantorna, M.T.; DeLuca, H.F. Expression of 1,25-dihydroxyvitamin D(3) receptor in the immune system. Arch. Biochem. Biophys. 2000, 374, 334–338. [Google Scholar] [CrossRef] [PubMed]
  41. He, L.; Liu, T.; Shi, Y.; Tian, F.; Hu, H.; Deb, D.K.; Chen, Y.; Bissonnette, M.; Li, Y.C. Gut Epithelial Vitamin D Receptor Regulates Microbiota-Dependent Mucosal Inflammation by Suppressing Intestinal Epithelial Cell Apoptosis. Endocrinology 2018, 159, 967–979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. He, L.; Zhou, M.; Li, Y.C. Vitamin D/Vitamin D Receptor Signaling Is Required for Normal Development and Function of Group 3 Innate Lymphoid Cells in the Gut. iScience 2019, 17, 119–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Leyssens, C.; Verlinden, L.; De Hertogh, G.; Kato, S.; Gysemans, C.; Mathieu, C.; Carmeliet, G.; Verstuyf, A. Impact on Experimental Colitis of Vitamin D Receptor Deletion in Intestinal Epithelial or Myeloid Cells. Endocrinology 2017, 158, 2354–2366. [Google Scholar] [CrossRef] [PubMed]
  44. Baeke, F.; Korf, H.; Overbergh, L.; van Etten, E.; Verstuyf, A.; Gysemans, C.; Mathieu, C. Human T. lymphocytes are direct targets of 1,25-dihydroxyvitamin D3 in the immune system. J. Steroid Biochem. Mol. Biol. 2010, 121, 221–227. [Google Scholar] [CrossRef]
  45. Hewison, M.; Freeman, L.; Hughes, S.V.; Evans, K.N.; Bland, R.; Eliopoulos, A.G.; Kilby, M.D.; Moss, P.A.; 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] [Green Version]
  46. 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]
  47. Chen, K.S.; DeLuca, H.F. Cloning of the human 1 alpha,25-dihydroxyvitamin D-3 24-hydroxylase gene promoter and identification of two vitamin D-responsive elements. Biochim. Biophys. Acta 1995, 1263, 1–9. [Google Scholar] [CrossRef]
  48. Vidal, M.; Ramana, C.V.; Dusso, A.S. Stat1-vitamin D receptor interactions antagonize 1,25-dihydroxyvitamin D transcriptional activity and enhance stat1-mediated transcription. Mol. Cell. Biol. 2002, 22, 2777–2787. [Google Scholar] [CrossRef] [Green Version]
  49. Baeke, F.; Etten, E.V.; Overbergh, L.; Mathieu, C. Vitamin D3 and the immune system: Maintaining the balance in health and disease. Nutr. Res. Rev. 2007, 20, 106–118. [Google Scholar] [CrossRef] [Green Version]
  50. Stoffels, K.; Overbergh, L.; Giulietti, A.; Verlinden, L.; Bouillon, R.; Mathieu, C. Immune regulation of 25-hydroxyvitamin-D3-1alpha-hydroxylase in human monocytes. J. Bone Miner. Res. 2006, 21, 37–47. [Google Scholar] [CrossRef]
  51. Stoffels, K.; Overbergh, L.; Bouillon, R.; Mathieu, C. Immune regulation of 1alpha-hydroxylase in murine peritoneal macrophages: Unravelling the IFNgamma pathway. J. Steroid Biochem. Mol. Biol. 2007, 103, 567–571. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, Y.; Leung, D.Y.; Richers, B.N.; Liu, Y.; Remigio, L.K.; Riches, D.W.; Goleva, E. Vitamin D inhibits monocyte/macrophage proinflammatory cytokine production by targeting MAPK phosphatase-1. J. Immunol. 2012, 188, 2127–2135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Wang, Q.; He, Y.; Shen, Y.; Zhang, Q.; Chen, D.; Zuo, C.; Qin, J.; Wang, H.; Wang, J.; Yu, Y. Vitamin D inhibits COX-2 expression and inflammatory response by targeting thioesterase superfamily member 4. J. Biol. Chem. 2014, 289, 11681–11694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chen, Y.; Liu, W.; Sun, T.; Huang, Y.; Wang, Y.; Deb, D.K.; Yoon, D.; Kong, J.; Thadhani, R.; Li, Y.C. 1,25-Dihydroxyvitamin D promotes negative feedback regulation of TLR signaling via targeting microRNA-155-SOCS1 in macrophages. J. Immunol. 2013, 190, 3687–3695. [Google Scholar] [CrossRef] [Green Version]
  55. Liu, P.T.; Stenger, S.; Li, H.; Wenzel, L.; Tan, B.H.; Krutzik, S.R.; Ochoa, M.T.; Schauber, J.; Wu, K.; Meinken, C.; et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 2006, 311, 1770–1773. [Google Scholar] [CrossRef] [PubMed]
  56. 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] [Green Version]
  57. Liu, P.T.; Schenk, M.; Walker, V.P.; Dempsey, P.W.; Kanchanapoomi, M.; Wheelwright, M.; Vazirnia, A.; Zhang, X.; Steinmeyer, A.; Zugel, U.; et al. Convergence of IL-1beta and VDR activation pathways in human TLR2/1-induced antimicrobial responses. PLoS ONE 2009, 4, e5810. [Google Scholar]
  58. 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] [Green Version]
  59. Kanikarla-Marie, P.; Jain, S.K. 1,25(OH)2D3 inhibits oxidative stress and monocyte adhesion by mediating the upregulation of GCLC and GSH in endothelial cells treated with acetoacetate (ketosis). J. Steroid Biochem. Mol. Biol. 2016, 159, 94–101. [Google Scholar] [CrossRef] [Green Version]
  60. 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] [Green Version]
  61. Rockett, K.A.; Brookes, R.; Udalova, I.; Vidal, V.; Hill, A.V.; Kwiatkowski, D. 1,25-Dihydroxyvitamin D3 induces nitric oxide synthase and suppresses growth of Mycobacterium tuberculosis in a human macrophage-like cell line. Infect. Immun. 1998, 66, 5314–5321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Chang, J.M.; Kuo, M.C.; Kuo, H.T.; Hwang, S.J.; Tsai, J.C.; Chen, H.C.; Lai, YH. 1-alpha,25-Dihydroxyvitamin D3 regulates inducible nitric oxide synthase messenger RNA expression and nitric oxide release in macrophage-like RAW 264.7 cells. J. Lab. Clin. Med. 2004, 143, 14–22. [Google Scholar] [CrossRef] [PubMed]
  63. Carlberg, C. Molecular endocrinology of vitamin D on the epigenome level. Mol. Cell. Endocrinol. 2017, 453, 14–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Griffin, M.D.; Lutz, W.; Phan, V.A.; Bachman, L.A.; McKean, D.J.; Kumar, R. Dendritic cell modulation by 1alpha,25 dihydroxyvitamin D3 and its analogs: A vitamin D receptor-dependent pathway that promotes a persistent state of immaturity in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2001, 98, 6800–6805. [Google Scholar] [CrossRef] [Green Version]
  65. Ferreira, G.B.; van Etten, E.; Verstuyf, A.; Waer, M.; Overbergh, L.; Gysemans, C.; Mathieu, C. 1,25-Dihydroxyvitamin D3 alters murine dendritic cell behaviour in vitro and in vivo. Diabetes Metab. Res. Rev. 2011, 27, 933–941. [Google Scholar] [CrossRef]
  66. Ferreira, G.B.; Gysemans, C.A.; Demengeot, J.; da Cunha, J.P.; Vanherwegen, A.S.; Overbergh, L.; Van Belle, T.L.; Pauwels, F.; Verstuyf, A.; Korf, H.; et al. 1,25-Dihydroxyvitamin D3 promotes tolerogenic dendritic cells with functional migratory properties in NOD mice. J. Immunol. 2014, 192, 4210–4220. [Google Scholar] [CrossRef] [Green Version]
  67. Adorini, L.; Penna, G. Dendritic cell tolerogenicity: A key mechanism in immunomodulation by vitamin D receptor agonists. Hum. Immunol. 2009, 70, 345–352. [Google Scholar] [CrossRef]
  68. Vanherwegen, A.S.; Gysemans, C.; Mathieu, C. Vitamin D endocrinology on the cross-road between immunity and metabolism. Mol. Cell. Endocrinol. 2017, 453, 52–67. [Google Scholar] [CrossRef] [PubMed]
  69. Unger, W.W.; Laban, S.; Kleijwegt, F.S.; van der Slik, A.R.; Roep, B.O. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: Differential role for PD-L1. Eur. J. Immunol. 2009, 39, 3147–3159. [Google Scholar] [CrossRef]
  70. Kleijwegt, F.S.; Laban, S.; Duinkerken, G.; Joosten, A.M.; Zaldumbide, A.; Nikolic, T.; Roep, B.O. Critical role for TNF in the induction of human antigen-specific regulatory T cells by tolerogenic dendritic cells. J. Immunol. 2010, 185, 1412–1418. [Google Scholar] [CrossRef] [Green Version]
  71. Skrobot, A.; Demkow, U.; Wachowska, M. Immunomodulatory Role of Vitamin D: A Review. Adv. Exp. Med. Biol. 2018, 1108, 13–23. [Google Scholar]
  72. Lai, Y.; Gallo, R.L. AMPed up immunity: How antimicrobial peptides have multiple roles in immune defense. Trends Immunol. 2009, 30, 131–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Subramanian, K.; Bergman, P.; Henriques-Normark, B. Vitamin D Promotes Pneumococcal Killing and Modulates Inflammatory Responses in Primary Human Neutrophils. J Innate Immun. 2017, 9, 375–386. [Google Scholar] [CrossRef] [PubMed]
  74. Takahashi, K.; Nakayama, Y.; Horiuchi, H.; Ohta, T.; Komoriya, K.; Ohmori, H.; Kamimura, T. Human neutrophils express messenger RNA of vitamin D receptor and respond to 1alpha,25-dihydroxyvitamin D3. Immunopharmacol. Immunotoxicol. 2002, 24, 335–347. [Google Scholar] [CrossRef] [PubMed]
  75. Hoe, E.; Nathanielsz, J.; Toh, Z.Q.; Spry, L.; Marimla, R.; Balloch, A.; Mulholland, K.; Licciardi, P.V. Anti-Inflammatory Effects of Vitamin D on Human Immune Cells in the Context of Bacterial Infection. Nutrients 2016, 8, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chirumbolo, S.; Bjorklund, G.; Sboarina, A.; Vella, A. The Role of Vitamin D in the Immune System as a Pro-survival Molecule. Clin. Ther. 2017, 39, 894–916. [Google Scholar] [CrossRef] [Green Version]
  77. Barnado, A.; Crofford, L.J.; Oates, J.C. At the Bedside: Neutrophil extracellular traps (NETs) as targets for biomarkers and therapies in autoimmune diseases. J. Leukoc. Biol. 2016, 99, 265–278. [Google Scholar] [CrossRef] [PubMed]
  78. Agraz-Cibrian, J.M.; Giraldo, D.M.; Urcuqui-Inchima, S. 1,25-Dihydroxyvitamin D3 induces formation of neutrophil extracellular trap-like structures and modulates the transcription of genes whose products are neutrophil extracellular trap-associated proteins: A pilot study. Steroids 2019, 141, 14–22. [Google Scholar] [CrossRef]
  79. Alter, G.; Malenfant, J.M.; Altfeld, M. CD107a as a functional marker for the identification of natural killer cell activity. J. Immunol. Methods 2004, 294, 15–22. [Google Scholar] [CrossRef]
  80. Lee, G.Y.; Park, C.Y.; Cha, K.S.; Lee, S.E.; Pae, M.; Han, S.N. Differential effect of dietary vitamin D supplementation on natural killer cell activity in lean and obese mice. J. Nutr. Biochem. 2018, 55, 178–184. [Google Scholar] [CrossRef]
  81. Xu, H.; Soruri, A.; Gieseler, R.K.; Peters, J.H. 1,25-Dihydroxyvitamin D3 exerts opposing effects to IL-4 on MHC class-II antigen expression, accessory activity, and phagocytosis of human monocytes. Scand. J. Immunol. 1993, 38, 535–540. [Google Scholar] [CrossRef] [PubMed]
  82. Baeke, F.; Takiishi, T.; Korf, H.; Gysemans, C.; Mathieu, C. Vitamin D: Modulator of the immune system. Curr. Opin. Pharmacol. 2010, 10, 482–496. [Google Scholar] [CrossRef]
  83. van Halteren, A.G.; Tysma, O.M.; van Etten, E.; Mathieu, C.; Roep, B.O. 1alpha,25-dihydroxyvitamin D3 or analogue treated dendritic cells modulate human autoreactive T cells via the selective induction of apoptosis. J. Autoimmun. 2004, 23, 233–239. [Google Scholar] [CrossRef] [PubMed]
  84. Takiishi, T.; Van Belle, T.; Gysemans, C.; Mathieu, C. Effects of vitamin D on antigen-specific and non-antigen-specific immune modulation: Relevance for type 1 diabetes. Pediatr. Diabetes 2013, 14, 81–89. [Google Scholar] [CrossRef] [PubMed]
  85. Mahon, B.D.; Wittke, A.; Weaver, V.; Cantorna, M.T. The targets of vitamin D depend on the differentiation and activation status of CD4 positive T cells. J. Cell. Biochem. 2003, 89, 922–932. [Google Scholar] [CrossRef] [PubMed]
  86. Jeffery, L.E.; Burke, F.; Mura, M.; Zheng, Y.; Qureshi, O.S.; Hewison, M.; Ker, L.S.; Lammas, D.A.; Raza, K.; Sansom, D.M. 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] [PubMed] [Green Version]
  87. 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] [Green Version]
  88. Baeke, F.; Gysemans, C.; Korf, H.; Mathieu, C. Vitamin D insufficiency: Implications for the immune system. Pediatr. Nephrol. 2010, 25, 1597–1606. [Google Scholar] [CrossRef]
  89. Staeva-Vieira, T.P.; Freedman, L.P. 1,25-dihydroxyvitamin D3 inhibits IFN-gamma and IL-4 levels during in vitro polarization of primary murine CD4+ T cells. J. Immunol. 2002, 168, 1181–1189. [Google Scholar] [CrossRef] [Green Version]
  90. Kang, S.W.; Kim, S.H.; Lee, N.; Lee, W.W.; Hwang, K.A.; Shin, M.S.; Lee, S.H.; Kim, W.; Kang, I. 1,25-Dihyroxyvitamin D3 promotes FOXP3 expression via binding to vitamin D response elements in its conserved noncoding sequence region. J. Immunol. 2012, 188, 5276–5282. [Google Scholar] [CrossRef] [Green Version]
  91. Van Belle, T.L.; Vanherwegen, A.S.; Feyaerts, D.; De Clercq, P.; Verstuyf, A.; Korf, H.; Gysemans, C.; Mathieu, C. 1,25-Dihydroxyvitamin D3 and its analog TX527 promote a stable regulatory T cell phenotype in T cells from type 1 diabetes patients. PLoS ONE 2014, 9, e109194. [Google Scholar] [CrossRef] [PubMed]
  92. 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] [PubMed] [Green Version]
  93. Geldmeyer-Hilt, K.; Heine, G.; Hartmann, B.; Baumgrass, R.; Radbruch, A.; Worm, M. 1,25-dihydroxyvitamin D3 impairs NF-kappaB activation in human naive B cells. Biochem. Biophys. Res. Commun. 2011, 407, 699–702. [Google Scholar] [CrossRef]
  94. 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] [PubMed] [Green Version]
  95. Heine, G.; Niesner, U.; Chang, H.D.; Steinmeyer, A.; Zugel, U.; Zuberbier, T.; Radbruch, A.; Worm, M. 1,25-dihydroxyvitamin D(3) promotes IL-10 production in human B cells. Eur. J. Immunol. 2008, 38, 2210–2218. [Google Scholar] [CrossRef]
  96. Drozdenko, G.; Scheel, T.; Heine, G.; Baumgrass, R.; Worm, M. Impaired T cell activation and cytokine production by calcitriol-primed human B cells. Clin. Exp. Immunol. 2014, 178, 364–372. [Google Scholar] [CrossRef]
  97. Lang, P.O.; Aspinall, R. Vitamin D Status and the Host Resistance to Infections: What It Is Currently (Not) Understood. Clin. Ther. 2017, 39, 930–945. [Google Scholar] [CrossRef] [Green Version]
  98. Huang, S.J.; Wang, X.H.; Liu, Z.D.; Cao, W.L.; Han, Y.; Ma, A.G.; Xu, S.F. Vitamin D deficiency and the risk of tuberculosis: A meta-analysis. Drug Des. Dev. Ther. 2017, 11, 91–102. [Google Scholar] [CrossRef] [Green Version]
  99. Zdrenghea, M.T.; Makrinioti, H.; Bagacean, C.; Bush, A.; Johnston, S.L.; Stanciu, L.A. Vitamin D modulation of innate immune responses to respiratory viral infections. Rev. Med. Virol. 2017, 27, e1909. [Google Scholar] [CrossRef]
  100. Ginde, A.A.; Mansbach, J.M.; Camargo, C.A., Jr. Association between serum 25-hydroxyvitamin D level and upper respiratory tract infection in the Third National Health and Nutrition Examination Survey. Arch. Intern. Med. 2009, 169, 384–390. [Google Scholar] [CrossRef] [Green Version]
  101. Martineau, A.R.; Jolliffe, D.A.; Hooper, R.L.; Greenberg, L.; Aloia, J.F.; Bergman, P.; Dubnov-Raz, G.; Esposito, S.; Ganmaa, D.; Ginde, A.A.; et al. Vitamin D supplementation to prevent acute respiratory tract infections: Systematic review and meta-analysis of individual participant data. BMJ 2017, 356, i6583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yakoob, M.Y.; Salam, R.A.; Khan, F.R.; Bhutta, Z.A. Vitamin D supplementation for preventing infections in children under five years of age. Cochrane Database Syst. Rev. 2016, 11, CD008824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Mao, S.; Huang, S. Vitamin D supplementation and risk of respiratory tract infections: A meta-analysis of randomized controlled trials. Scand. J. Infect. Dis. 2013, 45, 696–702. [Google Scholar] [CrossRef] [PubMed]
  104. Das, R.R.; Singh, M.; Naik, S.S. Vitamin D as an adjunct to antibiotics for the treatment of acute childhood pneumonia. Cochrane Database Syst. Rev. 2018, 7, CD011597. [Google Scholar] [CrossRef] [PubMed]
  105. Slow, S.; Epton, M.; Storer, M.; Thiessen, R.; Lim, S.; Wong, J.; Chin, P.; Tovaranonte, P.; Pearson, J.; Chambers, S.T.; et al. Effect of adjunctive single high-dose vitamin D3 on outcome of community-acquired pneumonia in hospitalised adults: The VIDCAPS randomised controlled trial. Sci. Rep. 2018, 8, 13829. [Google Scholar] [CrossRef] [Green Version]
  106. Aibana, O.; Huang, C.C.; Aboud, S.; Arnedo-Pena, A.; Becerra, M.C.; Bellido-Blasco, J.B.; Bhosale, R.; Calderon, R.; Chiang, S.; Contreras, C.; et al. Vitamin D status and risk of incident tuberculosis disease: A nested case-control study, systematic review, and individual-participant data meta-analysis. PLoS Med. 2019, 16, e1002907. [Google Scholar] [CrossRef]
  107. Balcells, M.E.; Yokobori, N.; Hong, B.Y.; Corbett, J.; Cervantes, J. The lung microbiome, vitamin D, and the tuberculous granuloma: A balance triangle. Microb. Pathog. 2019, 131, 158–163. [Google Scholar] [CrossRef]
  108. Korf, H.; Decallonne, B.; Mathieu, C. Vitamin D for infections. Curr. Opin. Endocrinol. Diabetes Obes. 2014, 21, 431–436. [Google Scholar] [CrossRef]
  109. Wu, H.X.; Xiong, X.F.; Zhu, M.; Wei, J.; Zhuo, K.Q.; Cheng, D.Y. Effects of vitamin D supplementation on the outcomes of patients with pulmonary tuberculosis: A systematic review and meta-analysis. BMC Pulm. Med. 2018, 18, 108. [Google Scholar] [CrossRef]
  110. Janssens, W.; Bouillon, R.; Claes, B.; Carremans, C.; Lehouck, A.; Buysschaert, I.; Mathieu, C.; Decramer, M.; Lambrechts, D. Vitamin D deficiency is highly prevalent in COPD and correlates with variants in the vitamin D-binding gene. Thorax 2010, 65, 215–220. [Google Scholar] [CrossRef] [Green Version]
  111. Ferrari, R.; Caram, L.M.O.; Tanni, S.E.; Godoy, I.; Rupp de Paiva, S.A. The relationship between Vitamin D status and exacerbation in COPD patients—A literature review. Respir. Med. 2018, 139, 34–38. [Google Scholar] [CrossRef] [PubMed]
  112. Lehouck, A.; Mathieu, C.; Carremans, C.; Baeke, F.; Verhaegen, J.; Van Eldere, J.; Decallonne, B.; Bouillon, R.; Decramer, M.; Janssens, W. High doses of vitamin D to reduce exacerbations in chronic obstructive pulmonary disease: A randomized trial. Ann. Intern. Med. 2012, 156, 105–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Martineau, A.R.; James, W.Y.; Hooper, R.L.; Barnes, N.C.; Jolliffe, D.A.; Greiller, C.L.; Islam, K.; McLaughlin, D.; Bhowmik, A.; Timms, P.M.; et al. Vitamin D3 supplementation in patients with chronic obstructive pulmonary disease (ViDiCO): A multicentre, double-blind, randomised controlled trial. Lancet Respir. Med. 2015, 3, 120–130. [Google Scholar] [CrossRef]
  114. Pozzilli, P.; Manfrini, S.; Crino, A.; Picardi, A.; Leomanni, C.; Cherubini, V.; Valente, L.; Khazrai, M.; Visalli, N.; IMDIAB group. Low levels of 25-hydroxyvitamin D3 and 1,25-dihydroxyvitamin D3 in patients with newly diagnosed type 1 diabetes. Horm. Metab. Res. 2005, 37, 680–683. [Google Scholar] [CrossRef]
  115. Littorin, B.; Blom, P.; Scholin, A.; Arnqvist, H.J.; Blohme, G.; Bolinder, J.; Ekbom-Schnell, A.; Eriksson, J.W.; Gudbjornsdottir, S.; Nystrom, L.; et al. Lower levels of plasma 25-hydroxyvitamin D among young adults at diagnosis of autoimmune type 1 diabetes compared with control subjects: Results from the nationwide Diabetes Incidence Study in Sweden (DISS). Diabetologia 2006, 49, 2847–2852. [Google Scholar] [CrossRef] [Green Version]
  116. Cooper, J.D.; Smyth, D.J.; Walker, N.M.; Stevens, H.; Burren, O.S.; Wallace, C.; Greissl, C.; Ramos-Lopez, E.; Hypponen, E.; Dunger, D.B.; et al. Inherited variation in vitamin D genes is associated with predisposition to autoimmune disease type 1 diabetes. Diabetes 2011, 60, 1624–1631. [Google Scholar] [CrossRef] [Green Version]
  117. Duan, S.; Lv, Z.; Fan, X.; Wang, L.; Han, F.; Wang, H.; Bi, S. Vitamin D status and the risk of multiple sclerosis: A systematic review and meta-analysis. Neurosci. Lett. 2014, 570, 108–113. [Google Scholar] [CrossRef]
  118. Lin, J.; Liu, J.; Davies, M.L.; Chen, W. Serum Vitamin D Level and Rheumatoid Arthritis Disease Activity: Review and Meta-Analysis. PLoS ONE 2016, 11, e0146351. [Google Scholar] [CrossRef]
  119. An, L.; Sun, M.H.; Chen, F.; Li, J.R. Vitamin D levels in systemic sclerosis patients: A meta-analysis. Drug Des. Dev. Ther. 2017, 11, 3119–3125. [Google Scholar] [CrossRef] [Green Version]
  120. Bae, S.C.; Lee, Y.H. Association between Vitamin D level and/or deficiency, and systemic lupus erythematosus: A meta-analysis. Cell. Mol. Biol. 2018, 64, 7–13. [Google Scholar] [CrossRef]
  121. Del Pinto, R.; Pietropaoli, D.; Chandar, A.K.; Ferri, C.; Cominelli, F. Association Between Inflammatory Bowel Disease and Vitamin D Deficiency: A Systematic Review and Meta-analysis. Inflamm. Bowel Dis. 2015, 21, 2708–2717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Fabisiak, N.; Fabisiak, A.; Watala, C.; Fichna, J. Fat-soluble Vitamin Deficiencies and Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. J. Clin. Gastroenterol. 2017, 51, 878–889. [Google Scholar] [CrossRef] [PubMed]
  123. 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]
  124. Antico, A.; Tampoia, M.; Tozzoli, R.; Bizzaro, N. Can supplementation with vitamin D reduce the risk or modify the course of autoimmune diseases? A systematic review of the literature. Autoimmun. Rev. 2012, 12, 127–136. [Google Scholar] [CrossRef] [PubMed]
  125. Mok, C.C.; Bro, E.T.; Ho, L.Y.; Singh, R.J.; Jannetto, P.J. Serum 25-hydroxyvitamin D3 levels and flares of systemic lupus erythematosus: A longitudinal cohort analysis. Clin. Rheumatol. 2018, 37, 2685–2692. [Google Scholar] [CrossRef] [PubMed]
  126. Nielsen, N.M.; Munger, K.L.; Koch-Henriksen, N.; Hougaard, D.M.; Magyari, M.; Jorgensen, K.T.; Lundqvist, M.; Simonsen, J.; Jess, T.; Cohen, A.; et al. Neonatal vitamin D status and risk of multiple sclerosis: A population-based case-control study. Neurology 2017, 88, 44–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Simpson, S., Jr.; Blizzard, L.; Otahal, P.; Van der Mei, I.; 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]
  128. Staples, J.A.; Ponsonby, A.L.; Lim, L.L.; McMichael, A.J. Ecologic analysis of some immune-related disorders, including type 1 diabetes, in Australia: Latitude, regional ultraviolet radiation, and disease prevalence. Environ. Health Perspect. 2003, 111, 518–523. [Google Scholar] [CrossRef]
  129. Vieira, V.M.; Hart, J.E.; Webster, T.F.; Weinberg, J.; Puett, R.; Laden, F.; Costenbader, K.H.; Karlson, E.W. Association between residences in U.S. northern latitudes and rheumatoid arthritis: A spatial analysis of the Nurses’ Health Study. Environ. Health Perspect. 2010, 118, 957–961. [Google Scholar] [CrossRef] [Green Version]
  130. Kahn, H.S.; Morgan, T.M.; Case, L.D.; Dabelea, D.; Mayer-Davis, E.J.; Lawrence, J.M.; Marcovina, S.M.; Imperatore, G.; Group SfDiYS. Association of type 1 diabetes with month of birth among U.S. youth: The SEARCH for Diabetes in Youth Study. Diabetes Care 2009, 32, 2010–2015. [Google Scholar] [CrossRef] [Green Version]
  131. 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] [PubMed] [Green Version]
  132. Sloka, S.; Grant, M.; Newhook, L.A. The geospatial relation between UV solar radiation and type 1 diabetes in Newfoundland. Acta Diabetol. 2010, 47, 73–78. [Google Scholar] [CrossRef] [PubMed]
  133. Frederiksen, B.N.; Kroehl, M.; Fingerlin, T.E.; Wong, R.; Steck, A.K.; Rewers, M.; Norris, J.M. Association between vitamin D metabolism gene polymorphisms and risk of islet autoimmunity and progression to type 1 diabetes: The diabetes autoimmunity study in the young (DAISY). J. Clin. Endocrinol. Metab. 2013, 98, E1845–E1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Imani, D.; Razi, B.; Motallebnezhad, M.; Rezaei, R. Association between vitamin D receptor (VDR) polymorphisms and the risk of multiple sclerosis (MS): An updated meta-analysis. BMC Neurol. 2019, 19, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Li, J.; Chen, S.Y.; Liu, H.H.; Yin, X.D.; Cao, L.T.; Xu, J.H.; Li, X.M.; Ye, D.Q.; Wang, J. Associations of Vitamin D Receptor Single Nucleotide Polymorphisms with Susceptibility to Systemic Sclerosis. Arch. Med. Res. 2019, 50, 368–376. [Google Scholar] [CrossRef]
  136. Gisbert-Ferrandiz, L.; Salvador, P.; Ortiz-Masia, D.; Macias-Ceja, D.C.; Orden, S.; Esplugues, J.V.; Calatayud, S.; Hinojosa, J.; Barrachina, M.D.; Hernandez, C. A Single Nucleotide Polymorphism in the Vitamin D Receptor Gene Is Associated with Decreased Levels of the Protein and a Penetrating Pattern in Crohn’s Disease. Inflamm. Bowel. Dis. 2018, 24, 1462–1470. [Google Scholar] [CrossRef]
  137. Salimi, S.; Eskandari, F.; Rezaei, M.; Sandoughi, M. Vitamin D Receptor rs2228570 and rs731236 Polymorphisms are Susceptible Factors for Systemic Lupus Erythematosus. Adv. Biomed. Res. 2019, 8, 48. [Google Scholar] [CrossRef]
  138. Tizaoui, K.; Hamzaoui, K. Association between VDR polymorphisms and rheumatoid arthritis disease: Systematic review and updated meta-analysis of case-control studies. Immunobiology 2015, 220, 807–816. [Google Scholar] [CrossRef]
  139. Braun, A.B.; Gibbons, F.K.; Litonjua, A.A.; Giovannucci, E.; Christopher, K.B. Low serum 25-hydroxyvitamin D at critical care initiation is associated with increased mortality. Crit. Care Med. 2012, 40, 63–72. [Google Scholar] [CrossRef]
  140. Higgins, D.M.; Wischmeyer, P.E.; Queensland, K.M.; Sillau, S.H.; Sufit, A.J.; Heyland, D.K. Relationship of vitamin D deficiency to clinical outcomes in critically ill patients. JPEN J. Parenter. Enteral Nutr. 2012, 36, 713–720. [Google Scholar] [CrossRef]
  141. Quraishi, S.A.; Bittner, E.A.; Blum, L.; McCarthy, C.M.; Bhan, I.; Camargo, C.A., Jr. Prospective study of vitamin D status at initiation of care in critically ill surgical patients and risk of 90-day mortality. Crit. Care Med. 2014, 42, 1365–1371. [Google Scholar] [CrossRef] [Green Version]
  142. Amrein, K.; Schnedl, C.; Holl, A.; Riedl, R.; Christopher, K.B.; Pachler, C.; Urbanic Purkart, T.; Waltensdorfer, A.; Munch, A.; Warnkross, H.; et al. Effect of high-dose vitamin D3 on hospital length of stay in critically ill patients with vitamin D deficiency: The VITdAL-ICU randomized clinical trial. JAMA 2014, 312, 1520–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Han, J.E.; Jones, J.L.; Tangpricha, V.; Brown, M.A.; Brown, L.A.S.; Hao, L.; Hebbar, G.; Lee, M. J., Liu S., Ziegler T.R., et al. High Dose Vitamin D Administration in Ventilated Intensive Care Unit Patients: A Pilot Double Blind Randomized Controlled Trial. J. Clin. Transl. Endocrinol. 2016, 4, 59–65. [Google Scholar]
  144. de Haan, K.; Groeneveld, A.B.; de Geus, H.R.; Egal, M.; Struijs, A. Vitamin D deficiency as a risk factor for infection, sepsis and mortality in the critically ill: Systematic review and meta-analysis. Crit. Care 2014, 18, 660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhang, Y.P.; Wan, Y.D.; Sun, T.W.; Kan, Q.C.; Wang, L.X. Association between vitamin D deficiency and mortality in critically ill adult patients: A meta-analysis of cohort studies. Crit. Care 2014, 18, 684. [Google Scholar] [CrossRef] [Green Version]
  146. Razavi Khorasani, N.; Moazzami, B.; Zahedi Tajrishi, F.; Mohammadpour, Z.; Rouhi, F.; Alizadeh-Navaei, R.; Ghadimi, R. The Association Between Low Levels of Vitamin D and Clinical Outcomes in Critically-Ill Children: A Systematic Review and Meta-Analysis. Fetal Pediatr. Pathol. 2019, 2019, 1–15. [Google Scholar] [CrossRef] [PubMed]
  147. Autier, P.; Boniol, M.; Pizot, C.; Mullie, P. Vitamin D status and ill health: A systematic review. Lancet Diabetes Endocrinol. 2014, 2, 76–89. [Google Scholar] [CrossRef]
  148. Jagannath, V.A.; Filippini, G.; Di Pietrantonj, C.; Asokan, G.V.; Robak, E.W.; Whamond, L.; Robinson, S.A. Vitamin D for the management of multiple sclerosis. Cochrane Database Syst. Rev. 2018, 9, CD008422. [Google Scholar] [CrossRef]
  149. 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] [Green Version]
  150. Gregori, S.; Giarratana, N.; Smiroldo, S.; Uskokovic, M.; Adorini, L. A 1alpha,25-dihydroxyvitamin D(3) analog enhances regulatory T-cells and arrests autoimmune diabetes in NOD mice. Diabetes 2002, 51, 1367–1374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Gysemans, C.A.; Cardozo, A.K.; Callewaert, H.; Giulietti, A.; Hulshagen, L.; Bouillon, R.; Eizirik, D.L.; Mathieu, C. 1,25-Dihydroxyvitamin D3 modulates expression of chemokines and cytokines in pancreatic islets: Implications for prevention of diabetes in nonobese diabetic mice. Endocrinology 2005, 146, 1956–1964. [Google Scholar] [CrossRef] [PubMed]
  152. Prietl, B.; Treiber, G.; Mader, J.K.; Hoeller, E.; Wolf, M.; Pilz, S.; Graninger, W.B.; Obermayer-Pietsch, B.M.; Pieber, T.R. High-dose cholecalciferol supplementation significantly increases peripheral CD4(+) Tregs in healthy adults without negatively affecting the frequency of other immune cells. Eur J. Nutr. 2014, 53, 751–759. [Google Scholar] [CrossRef] [PubMed]
  153. Treiber, G.; Prietl, B.; Frohlich-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] [PubMed]
  154. Bogdanou, D.; Penna-Martinez, M.; Filmann, N.; Chung, T.L.; Moran-Auth, Y.; Wehrle, J.; Cappel, C.; Huenecke, S.; Herrmann, E.; Koehl, U.; et al. T-lymphocyte and glycemic status after vitamin D treatment in type 1 diabetes: A randomized controlled trial with sequential crossover. Diabetes Metab. Res. Rev. 2017, 33, e2865. [Google Scholar] [CrossRef]
  155. 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] [Green Version]
  156. Di Liberto, D.; Scazzone, C.; La Rocca, G.; Cipriani, P.; Lo Pizzo, M.; Ruscitti, P.; Agnello, L.; Ciaccio, M.; Dieli, F.; Giacomelli, R.; et al. Vitamin D increases the production of IL-10 by regulatory T cells in patients with systemic sclerosis. Clin. Exp. Rheumatol. 2019, 37 (Suppl. 119), 76–81. [Google Scholar]
  157. Muris, A.H.; Smolders, J.; Rolf, L.; Thewissen, M.; Hupperts, R.; Damoiseaux, J.; SOLARIUM study group. Immune regulatory effects of high dose vitamin D3 supplementation in a randomized controlled trial in relapsing remitting multiple sclerosis patients receiving IFNbeta; the SOLARIUM study. J. Neuroimmunol. 2016, 300, 47–56. [Google Scholar] [CrossRef]
  158. Brusko, T.; Wasserfall, C.; McGrail, K.; Schatz, R.; Viener, H.L.; Schatz, D.; Haller, M.; Rockell, J.; Gottlieb, P.; Clare-Salzler, M.; et al. No alterations in the frequency of FOXP3+ regulatory T-cells in type 1 diabetes. Diabetes 2007, 56, 604–612. [Google Scholar] [CrossRef] [Green Version]
  159. Marinho, A.; Carvalho, C.; Boleixa, D.; Bettencourt, A.; Leal, B.; Guimaraes, J.; Neves, E.; Oliveira, J.C.; Almeida, I.; Farinha, F.; et al. Vitamin D supplementation effects on FoxP3 expression in T cells and FoxP3(+)/IL-17A ratio and clinical course in systemic lupus erythematosus patients: A study in a Portuguese cohort. Immunol. Res. 2017, 65, 197–206. [Google Scholar] [CrossRef]
  160. Sotirchos, E.S.; Bhargava, P.; Eckstein, C.; Van Haren, K.; Baynes, M.; Ntranos, A.; Gocke, A.; Steinman, L.; Mowry, E.M.; Calabresi, P.A. Safety and immunologic effects of high- vs low-dose cholecalciferol in multiple sclerosis. Neurology 2016, 86, 382–390. [Google Scholar] [CrossRef]
  161. Terrier, B.; Derian, N.; Schoindre, Y.; Chaara, W.; Geri, G.; Zahr, N.; Mariampillai, K.; Rosenzwajg, M.; Carpentier, W.; Musset, L.; et al. Restoration of regulatory and effector T cell balance and B cell homeostasis in systemic lupus erythematosus patients through vitamin D supplementation. Arthritis Res. Ther. 2012, 14, R221. [Google Scholar] [CrossRef] [Green Version]
  162. 162. Avcioglu, G.; Ozbek Iptec, B.; Akcan, G.; Gorgun, B.; Fidan, K.; Carhan, A.; Yilmaz, G.; Kozaci, L.D. Effects of 1,25-Dihydroxy vitamin D3 on TNF-alpha induced inflammation in human chondrocytes and SW1353 cells: A possible role for toll-like receptors. Mol. Cell. Biochem. 2020, 464, 131–142. [Google Scholar] [CrossRef]
  163. Luo, J.; Wen, H.; Guo, H.; Cai, Q.; Li, S.; Li, X. 1,25-dihydroxyvitamin D3 inhibits the RANKL pathway and impacts on the production of pathway-associated cytokines in early rheumatoid arthritis. Biomed. Res. Int. 2013, 2013, 101805. [Google Scholar] [CrossRef] [Green Version]
  164. Neve, A.; Corrado, A.; Cantatore, F.P. Immunomodulatory effects of vitamin D in peripheral blood monocyte-derived macrophages from patients with rheumatoid arthritis. Clin. Exp. Med. 2014, 14, 275–283. [Google Scholar] [CrossRef]
  165. Wen, H.Y.; Luo, J.; Li, X.F.; Wei, D.D.; Liu, Y. 1,25-Dihydroxyvitamin D3 modulates T cell differentiation and impacts on the production of cytokines from Chinese Han patients with early rheumatoid arthritis. Immunol. Res. 2019, 67, 48–57. [Google Scholar] [CrossRef]
  166. 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] [Green Version]
  167. Ashtari, F.; Toghianifar, N.; Zarkesh-Esfahani, S.H.; Mansourian, M. Short-term effect of high-dose vitamin D on the level of interleukin 10 in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled clinical trial. Neuroimmunomodulation 2015, 22, 400–404. [Google Scholar] [CrossRef]
  168. da Costa, D.S.; Hygino, J.; Ferreira, T.B.; Kasahara, T.M.; Barros, P.O.; Monteiro, C.; Oliveira, A.; Tavares, F.; Vasconcelos, C.C.; Alvarenga, R.; et al. Vitamin D modulates different IL-17-secreting T cell subsets in multiple sclerosis patients. J. Neuroimmunol. 2016, 299, 8–18. [Google Scholar] [CrossRef]
  169. Bendix-Struve, M.; Bartels, L.E.; Agnholt, J.; Dige, A.; Jorgensen, S.P.; Dahlerup, J.F. Vitamin D3 treatment of Crohn’s disease patients increases stimulated T cell IL-6 production and proliferation. Aliment. Pharmacol. Ther. 2010, 32, 1364–1372. [Google Scholar] [CrossRef] [Green Version]
  170. Gubatan, J.; Mitsuhashi, S.; Longhi, M.S.; Zenlea, T.; Rosenberg, L.; Robson, S.; Moss, A.C. Higher serum vitamin D levels are associated with protective serum cytokine profiles in patients with ulcerative colitis. Cytokine 2018, 103, 38–45. [Google Scholar] [CrossRef]
  171. Shahin, D.; El-Farahaty, R.M.; Houssen, M.E.; Machaly, S.A.; Sallam, M.; ElSaid, T.O.; Neseem, N.O. Serum 25-OH vitamin D level in treatment-naive systemic lupus erythematosus patients: Relation to disease activity, IL-23 and IL-17. Lupus 2017, 26, 917–926. [Google Scholar] [CrossRef]
  172. Zerr, P.; Vollath, S.; Palumbo-Zerr, K.; Tomcik, M.; Huang, J.; Distler, A.; Beyer, C.; Dees, C.; Gela, K.; Distler, O.; et al. Vitamin D receptor regulates TGF-beta signalling in systemic sclerosis. Ann. Rheum. Dis. 2015, 74, e20. [Google Scholar] [CrossRef] [Green Version]
  173. Mauf, S.; Penna-Martinez, M.; Jentzsch, T.; Ackermann, H.; Henrich, D.; Radeke, H.H.; Bruck, P.; Badenhoop, K.; Ramos-Lopez, E. Immunomodulatory effects of 25-hydroxyvitamin D3 on monocytic cell differentiation and influence of vitamin D3 polymorphisms in type 1 diabetes. J. Steroid Biochem. Mol. Biol. 2015, 147, 17–23. [Google Scholar] [CrossRef]
  174. Gregori, S.; Tomasoni, D.; Pacciani, V.; Scirpoli, M.; Battaglia, M.; Magnani, C.F.; Hauben, E.; Roncarolo, M.G. Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway. Blood 2010, 116, 935–944. [Google Scholar] [CrossRef] [Green Version]
  175. Nikolic, T.; Woittiez, N.J.C.; van der Slik, A.; Laban, S.; Joosten, A.; Gysemans, C.; Mathieu, C.; Zwaginga, J.J.; Koeleman, B.; Roep, B.O. Differential transcriptome of tolerogenic versus inflammatory dendritic cells points to modulated T1D genetic risk and enriched immune regulation. Genes Immun. 2017, 18, 176–183. [Google Scholar] [CrossRef]
  176. Wu, H.J.; Lo, Y.; Luk, D.; Lau, C.S.; Lu, L.; Mok, M.Y. Alternatively activated dendritic cells derived from systemic lupus erythematosus patients have tolerogenic phenotype and function. Clin. Immunol. 2015, 156, 43–57. [Google Scholar] [CrossRef] [Green Version]
  177. Baeke, F.; van Etten, E.; Gysemans, C.; Overbergh, L.; Mathieu, C. Vitamin D signaling in immune-mediated disorders: Evolving insights and therapeutic opportunities. Mol. Aspects Med. 2008, 29, 376–387. [Google Scholar] [CrossRef]
  178. Mathieu, C.; Laureys, J.; Sobis, H.; Vandeputte, M.; Waer, M.; Bouillon, R. 1,25-Dihydroxyvitamin D3 prevents insulitis in NOD mice. Diabetes 1992, 41, 1491–1495. [Google Scholar] [CrossRef]
  179. Giulietti, A.; Gysemans, C.; Stoffels, K.; van Etten, E.; Decallonne, B.; Overbergh, L.; Bouillon, R.; Mathieu, C. Vitamin D deficiency in early life accelerates Type 1 diabetes in non-obese diabetic mice. Diabetologia 2004, 47, 451–462. [Google Scholar] [CrossRef] [Green Version]
  180. Takiishi, T.; Ding, L.; Baeke, F.; Spagnuolo, I.; Sebastiani, G.; Laureys, J.; Verstuyf, A.; Carmeliet, G.; Dotta, F.; Van Belle, T.L.; et al. Dietary supplementation with high doses of regular vitamin D3 safely reduces diabetes incidence in NOD mice when given early and long term. Diabetes 2014, 63, 2026–2036. [Google Scholar] [CrossRef] [Green Version]
  181. Niino, M.; Fukazawa, T.; Kikuchi, S.; Sasaki, H. Therapeutic potential of vitamin D for multiple sclerosis. Curr. Med. Chem. 2008, 15, 499–505. [Google Scholar] [CrossRef]
  182. Correa Freitas, E.; Evelyn Karnopp, T.; de Souza Silva, J.M.; Cavalheiro do Espirito Santo, R.; da Rosa, T.H.; de Oliveira, M.S.; da Costa Goncalves, F.; de Oliveira, F.H.; Guilherme Schaefer, P.; Andre Monticielo, O. Vitamin D supplementation ameliorates arthritis but does not alleviates renal injury in pristane-induced lupus model. Autoimmunity 2019, 52, 69–77. [Google Scholar] [CrossRef]
  183. Fan, P.; He, L.; Hu, N.; Luo, J.; Zhang, J.; Mo, L.F.; Wang, Y.H.; Pu, D.; Lv, X.H.; Hao, Z.M.; et al. Effect of 1,25-(OH)2D3 on Proliferation of Fibroblast-Like Synoviocytes and Expressions of Pro-Inflammatory Cytokines through Regulating MicroRNA-22 in a Rat Model of Rheumatoid Arthritis. Cell. Physiol. Biochem. 2017, 42, 145–155. [Google Scholar] [CrossRef] [Green Version]
  184. Yamamoto, E.A.; Nguyen, J.K.; Liu, J.; Keller, E.; Campbell, N.; Zhang, C.J.; Smith, H.R.; Li, X.; Jorgensen, T.N. Low Levels of Vitamin D Promote Memory B Cells in Lupus. Nutrients 2020, 12, 291. [Google Scholar] [CrossRef] [Green Version]
  185. Zhou, L.; Wang, J.; Li, J.; Li, T.; Chen, Y.; June, R.R.; Zheng, S.G. 1,25-Dihydroxyvitamin D3 Ameliorates Collagen-Induced Arthritis via Suppression of Th17 Cells Through miR-124 Mediated Inhibition of IL-6 Signaling. Front. Immunol. 2019, 10, 178. [Google Scholar] [CrossRef]
  186. 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] [Green Version]
  187. Hypponen, E.; Laara, E.; Reunanen, A.; Jarvelin, 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]
  188. 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]
  189. Munger, K.L.; Levin, L.I.; Hollis, B.W.; Howard, N.S.; Ascherio, A. Serum 25-hydroxyvitamin D levels and risk of multiple sclerosis. JAMA 2006, 296, 2832–2838. [Google Scholar] [CrossRef] [Green Version]
  190. Jasper, E.A.; Nidey, N.L.; Schweizer, M.L.; Ryckman, K.K. Gestational vitamin D and offspring risk of multiple sclerosis: A systematic review and meta-analysis. Ann. Epidemiol. 2020. [Google Scholar] [CrossRef]
  191. Thorsen, S.U.; Marild, K.; Olsen, S.F.; Holst, K.K.; Tapia, G.; Granstrom, C.; Halldorsson, T.I.; Cohen, A.S.; Haugen, M.; Lundqvist, M.; et al. Lack of Association Between Maternal or Neonatal Vitamin D Status and Risk of Childhood Type 1 Diabetes: A Scandinavian Case-Cohort Study. Am. J. Epidemiol. 2018, 187, 1174–1181. [Google Scholar] [CrossRef]
  192. Jacobsen, R.; Thorsen, S.U.; Cohen, A.S.; Lundqvist, M.; Frederiksen, P.; Pipper, C.B.; Pociot, F.; Thygesen, L.C.; Ascherio, A.; Svensson, J.; et al. Neonatal vitamin D status is not associated with later risk of type 1 diabetes: Results from two large Danish population-based studies. Diabetologia 2016, 59, 1871–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Dong, J.Y.; Zhang, W.G.; Chen, J.J.; Zhang, Z.L.; Han, S.F.; Qin, L.Q. Vitamin D intake and risk of type 1 diabetes: A meta-analysis of observational studies. Nutrients 2013, 5, 3551–3562. [Google Scholar] [CrossRef]
  194. Silvis, K.; Aronsson, C.A.; Liu, X.; Uusitalo, U.; Yang, J.; Tamura, R.; Lernmark, A.; Rewers, M.; Hagopian, W.; She, J.X.; et al. Maternal dietary supplement use and development of islet autoimmunity in the offspring: TEDDY study. Pediatr. Diabetes 2019, 20, 86–92. [Google Scholar] [CrossRef] [Green Version]
  195. Stene, L.C.; Joner, G.; Norwegian Childhood Diabetes Study Group. Use of cod liver oil during the first year of life is associated with lower risk of childhood-onset type 1 diabetes: A large, population-based, case-control study. Am. J. Clin Nutr. 2003, 78, 1128–1134. [Google Scholar] [CrossRef] [Green Version]
  196. Brekke, H.K.; Ludvigsson, J. Vitamin D supplementation and diabetes-related autoimmunity in the ABIS study. Pediatr. Diabetes 2007, 8, 11–14. [Google Scholar] [CrossRef]
  197. Zipitis, C.S.; Akobeng, A.K. Vitamin D supplementation in early childhood and risk of type 1 diabetes: A systematic review and meta-analysis. Arch. Dis. Child. 2008, 93, 512–517. [Google Scholar] [CrossRef] [Green Version]
  198. Simpson, M.; Brady, H.; Yin, X.; Seifert, J.; Barriga, K.; Hoffman, M.; Bugawan, T.; Baron, A.E.; Sokol, R.J.; Eisenbarth, G.; et al. No association of vitamin D intake or 25-hydroxyvitamin D levels in childhood with risk of islet autoimmunity and type 1 diabetes: The Diabetes Autoimmunity Study in the Young (DAISY). Diabetologia 2011, 54, 2779–2788. [Google Scholar] [CrossRef] [Green Version]
  199. Stene, L.C.; Ulriksen, J.; Magnus, P.; Joner, G. Use of cod liver oil during pregnancy associated with lower risk of Type I diabetes in the offspring. Diabetologia 2000, 43, 1093–1098. [Google Scholar] [CrossRef] [Green Version]
  200. Marjamaki, L.; Niinisto, S.; Kenward, M.G.; Uusitalo, L.; Uusitalo, U.; Ovaskainen, M.L.; Kronberg-Kippila, C.; Simell, O.; Veijola, R.; Ilonen, J.; et al. Maternal intake of vitamin D during pregnancy and risk of advanced beta cell autoimmunity and type 1 diabetes in offspring. Diabetologia 2010, 53, 1599–1607. [Google Scholar] [CrossRef]
  201. Norris, J.M.; Lee, H.S.; Frederiksen, B.; Erlund, I.; Uusitalo, U.; Yang, J.; Lernmark, A.; Simell, O.; Toppari, J.; Rewers, M.; et al. Plasma 25-Hydroxyvitamin D Concentration and Risk of Islet Autoimmunity. Diabetes 2018, 67, 146–154. [Google Scholar] [CrossRef] [Green Version]
  202. Tapia, G.; Marild, K.; Dahl, S.R.; Lund-Blix, N.A.; Viken, M.K.; Lie, B.A.; Njolstad, P.R.; Joner, G.; Skrivarhaug, T.; Cohen, A.S.; et al. Maternal and Newborn Vitamin D-Binding Protein, Vitamin D Levels, Vitamin D Receptor Genotype, and Childhood Type 1 Diabetes. Diabetes Care 2019, 42, 553–559. [Google Scholar] [CrossRef] [Green Version]
  203. van Etten, E.; Verlinden, L.; Giulietti, A.; Ramos-Lopez, E.; Branisteanu, D.D.; Ferreira, G.B.; Overbergh, L.; Verstuyf, A.; Bouillon, R.; Roep, B.O.; et al. The vitamin D receptor gene FokI polymorphism: Functional impact on the immune system. Eur. J. Immunol. 2007, 37, 395–405. [Google Scholar] [CrossRef]
  204. Moran-Auth, Y.; Penna-Martinez, M.; Badenhoop, K. VDR FokI polymorphism is associated with a reduced T-helper cell population under vitamin D stimulation in type 1 diabetes patients. J. Steroid Biochem. Mol. Biol. 2015, 148, 184–186. [Google Scholar] [CrossRef]
  205. Mathieu, C.; Waer, M.; Laureys, J.; Rutgeerts, O.; Bouillon, R. Prevention of autoimmune diabetes in NOD mice by 1,25 dihydroxyvitamin D3. Diabetologia 1994, 37, 552–558. [Google Scholar] [CrossRef]
  206. Becklund, B.R.; Hansen, D.W., Jr.; Deluca, H.F. Enhancement of 1,25-dihydroxyvitamin D3-mediated suppression of experimental autoimmune encephalomyelitis by calcitonin. Proc. Natl. Acad. Sci. USA 2009, 106, 5276–5281. [Google Scholar] [CrossRef] [Green Version]
  207. Malihi, Z.; Wu, Z.; Stewart, A.W.; Lawes, C.M.; Scragg, R. Hypercalcemia, hypercalciuria, and kidney stones in long-term studies of vitamin D supplementation: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2016, 104, 1039–1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Mallya, S.M.; Corrado, K.R.; Saria, E.A.; Yuan, F.F.; Tran, H.Q.; Saucier, K.; Atti, E.; Tetradis, S.; Arnold, A. Modeling vitamin D insufficiency and moderate deficiency in adult mice via dietary cholecalciferol restriction. Endocr. Res. 2016, 41, 290–299. [Google Scholar] [CrossRef] [Green Version]
  209. Kusunoki, Y.; Matsui, I.; Hamano, T.; Shimomura, A.; Mori, D.; Yonemoto, S.; Takabatake, Y.; Tsubakihara, Y.; St-Arnaud, R.; Isaka, Y.; et al. Excess 25-hydroxyvitamin D3 exacerbates tubulointerstitial injury in mice by modulating macrophage phenotype. Kidney Int. 2015, 88, 1013–1029. [Google Scholar] [CrossRef] [Green Version]
  210. Tabasi, N.; Rastin, M.; Mahmoudi, M.; Ghoryani, M.; Mirfeizi, Z.; Rabe, S.Z.; Reihani, H. Influence of vitamin D on cell cycle, apoptosis, and some apoptosis related molecules in systemic lupus erythematosus. Iran. J. Basic Med. Sci. 2015, 18, 1107–1111. [Google Scholar]
  211. Chen, L.; Eapen, M.S.; Zosky, G.R. Vitamin D both facilitates and attenuates the cellular response to lipopolysaccharide. Sci. Rep. 2017, 7, 45172. [Google Scholar] [CrossRef] [Green Version]
  212. Baeke, F.; Van Belle, T.L.; Takiishi, T.; Ding, L.; Korf, H.; Laureys, J.; Gysemans, C.; Mathieu, C. Low doses of anti-CD3, ciclosporin A and the vitamin D analogue, TX527, synergise to delay recurrence of autoimmune diabetes in an islet-transplanted NOD mouse model of diabetes. Diabetologia 2012, 55, 2723–2732. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Immunomodulatory actions of active vitamin D (1,25-dihydroxyvitamin D3; 1,25-(OH)2D3). Both the direct as the indirect effects on T-lymphocytes are shown as 1,25-(OH)2D3 exerts its effect through direct binding on both the vitamin D receptor of the antigen-presenting cell (APC), in this case the dendritic cell (DC), and the T-lymphocytes directly. The effect of 1,25-(OH)2D3 on the APC is both an upregulation of the direct inhibition of the APC, as well as a downregulation of its antigen presentation function. The direct effect of 1,25-(OH)2D3 on the T-lymphocytes is a change towards a more tolerogenic state with an induction of Thelper-2 (Th2)-lymphocytes and regulatory T-lymphocytes (Tregs; depicted in green text), together with a downregulation of the pro-inflammatory Thelper-1 (Th1)-lymphocytes, Thelper-17 (Th17)-lymphocytes, and Thelper-9 (Th9)-lymphocytes (depicted in red text). Other abbreviations: IL: interleukin; IFN-γ: interferon-γ; TNF-α: tumor necrosis factor-α; ILT-3: immunoglobulin-like transcript-3; 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). Both the direct as the indirect effects on T-lymphocytes are shown as 1,25-(OH)2D3 exerts its effect through direct binding on both the vitamin D receptor of the antigen-presenting cell (APC), in this case the dendritic cell (DC), and the T-lymphocytes directly. The effect of 1,25-(OH)2D3 on the APC is both an upregulation of the direct inhibition of the APC, as well as a downregulation of its antigen presentation function. The direct effect of 1,25-(OH)2D3 on the T-lymphocytes is a change towards a more tolerogenic state with an induction of Thelper-2 (Th2)-lymphocytes and regulatory T-lymphocytes (Tregs; depicted in green text), together with a downregulation of the pro-inflammatory Thelper-1 (Th1)-lymphocytes, Thelper-17 (Th17)-lymphocytes, and Thelper-9 (Th9)-lymphocytes (depicted in red text). Other abbreviations: IL: interleukin; IFN-γ: interferon-γ; TNF-α: tumor necrosis factor-α; ILT-3: immunoglobulin-like transcript-3; GATA-3: GATA binding protein-3; FoxP3: forkhead box P3, CTLA-4: cytotoxic T lymphocyte associated protein-4.
Nutrients 12 01248 g001

Share and Cite

MDPI and ACS Style

Martens, P.-J.; Gysemans, C.; Verstuyf, A.; Mathieu, C. Vitamin D’s Effect on Immune Function. Nutrients 2020, 12, 1248. https://doi.org/10.3390/nu12051248

AMA Style

Martens P-J, Gysemans C, Verstuyf A, Mathieu C. Vitamin D’s Effect on Immune Function. Nutrients. 2020; 12(5):1248. https://doi.org/10.3390/nu12051248

Chicago/Turabian Style

Martens, Pieter-Jan, Conny Gysemans, Annemieke Verstuyf, and Chantal Mathieu. 2020. "Vitamin D’s Effect on Immune Function" Nutrients 12, no. 5: 1248. https://doi.org/10.3390/nu12051248

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