Next Article in Journal
On the Potential Role of Phytate Against Neurodegeneration: It Protects Against Fe3+-Catalyzed Degradation of Dopamine and Ascorbate and Against Fe3+-Induced Protein Aggregation
Previous Article in Journal
Ageing Trajectories: Exposome-Driven Pathobiological Mechanisms and Implications for Prevention from Blue Zones and Italian Longevity Hotspots Such as Cilento and Sicilian Mountain Villages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Insight into the Role of Vitamin D in Immune-Mediated Inflammatory Diseases

1
Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy
2
Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90133 Palermo, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(10), 4798; https://doi.org/10.3390/ijms26104798
Submission received: 8 April 2025 / Revised: 4 May 2025 / Accepted: 6 May 2025 / Published: 16 May 2025
(This article belongs to the Section Molecular Biology)

Abstract

:
In the last decades, it has become increasingly evident that the role of vitamin D extends beyond the regulation of calcium homeostasis and the maintenance of bone health. A significant extraskeletal function of vitamin D is its role in modulating the immune system, particularly highlighted in the context of immune-mediated inflammatory diseases, where correlations between vitamin D status and genetic variations in the vitamin D receptor have been observed about the incidence and severity of these conditions. Additionally, different studies have reported the existence of immunomodulatory effects of vitamin D, particularly the effects of vitamin D on dendritic cell function, maturation, cytokine production, and antigen presentation, and that its deficiency may be associated with a sub-inflammatory state. In this sense, different clinical trials have been conducted to assess the therapeutic efficacy of vitamin D in different immune-mediated inflammatory disorders, including asthma, atopic dermatitis (AD), rheumatoid arthritis (RA), psoriasis, thyroid diseases, infectious diseases, and systemic lupus erythematosus (SLE). This review will provide a comprehensive overview of the current understanding of the molecular mechanisms underlying vitamin D’s immunomodulatory properties, its role, and innovative therapeutic applications in patients with immune-mediated inflammatory diseases.

1. Introduction

In recent years, attention has been paid to the role of vitamin D in different fields. vitamin D is a fat-soluble prohormone critical in various physiological processes, particularly in bone metabolism. In addition to its well-established role in promoting calcium absorption and bone health, vitamin D is implicated in many extraskeletal functions. Its functions encompass endocrine, autocrine, and paracrine mechanisms essential for overall health [1]. Different studies have increasingly highlighted the correlation between vitamin D deficiency and several pathological conditions. Some evidence shows its systemic effects, such as regulation of cell differentiation, proliferation, activation and death, xenobiotic detoxification, antioxidant and neuroprotective actions, antimicrobial defense, immunoregulation of both innate and adaptive immunity, and anti-inflammatory and anti-neoplastic effects in many cells and tissues [2,3,4,5,6]. In this sense, serum vitamin D levels are related to the incidence of chronic inflammatory diseases such as cardiovascular diseases, diabetes, insulin resistance, metabolic syndrome, autoimmune diseases, allergic disorders, infectious diseases, and tumors. Particularly, vitamin D may serve as a significant predisposing factor in diabetes pathogenesis, wherein dysregulation of its signaling pathways elevates susceptibility to autoimmune dysregulation and macrovascular and microvascular complications [7,8,9,10,11,12,13,14,15,16,17].
The recent COVID-19 pandemic has further emphasized the potential therapeutic role of vitamin D in managing infectious diseases. Emerging studies suggest that adequate levels of vitamin D favorably impact immune responses, influencing the severity of COVID-19 infections. Insufficient vitamin D levels have been linked to worse health outcomes in patients suffering from COVID-19, raising concerns about the implications of vitamin D deficiency during the pandemic [18,19,20]. This growing body of evidence underscores the importance of monitoring and potentially supplementing vitamin D to enhance health and mitigate the risk of various diseases. Some studies have reported the immunomodulatory effects of vitamin D, particularly the effects of vitamin D on dendritic cell (DC) function, maturation, cytokine production, and antigen presentation, and that its deficiency may be associated with a sub-inflammatory state [3,21,22]. Moreover, immune cells express the vitamin D receptor (VDR) and a vitamin D activating enzyme, indicating that these cells can produce and respond to activated vitamin D, suggesting that its deficiency may significantly impact inflammatory disorders [23,24,25,26] (Figure 1).
Given this background, an extensive search of SCOPUS, PubMed, and CENTRAL was performed using the following string: “((vitamin D) or (calciferol) or (cholecalciferol) or (calcifediol) or (calcitriol) or (ergocholecalciferol)) AND ((autoimmune diseases) OR (autoinflammatory disorders) OR (immune system disorders) OR (immunological disorders) OR (chronic inflammatory diseases)) AND (systematic review [pt] or meta-analysis [pt]) and 2015:2025 [dp])”. The search string retrieved 301 manuscripts. The hand-searching of principal generalist, human nutrition, and basic research journals was also carried out [27,28,29]. Two authors (V.O. and A.T.) independently reviewed the retrieved articles’ titles, abstracts, and full texts to determine their potential inclusion. Any disagreements were resolved via discussion with the third author (S.C.). Manuscripts regarding the role of vitamin D in immune-mediated inflammatory diseases were extracted for this review. This review explores the molecular basis, the role, and the innovative therapeutic applications of vitamin D in immune-mediated inflammatory diseases.

2. Vitamin D Metabolism

Originally classified as a vitamin, vitamin D is now recognized as a prohormone essential for multiple physiological functions. It is a liposoluble compound found in two inactive forms: cholecalciferol (vitamin D3), synthesized in the skin upon UVB exposure and found in animal-based foods (particularly fatty fish such as herring, mackerel, sardines, and salmon), and ergocalciferol (vitamin D2), derived from UV-exposed fungi and yeasts that, when exposed to UV light, produce vitamin D2 from a precursor called ergosterol, which is present in their cell membranes, and primarily ingested through fortified foods [3,14,30]. Despite structural differences in their side chains, D2 and D3 share the same metabolic pathway. Upon absorption in the small intestine, they are incorporated into chylomicrons and transported to the liver, where they bind to vitamin D Binding Protein (DBP) and are taken up by hepatocytes. Hepatic 25-hydroxylase (CYP2R1) converts them into 25-hydroxyvitamin D (25(OH)D or calcifediol), the main circulating form [4]. In the mitochondria of the proximal convoluted tubule cells in the kidneys, 25(OH)D undergoes further hydroxylation by mitochondrial 1α-hydroxylase (CYP27B1) to become the biologically active 1α,25-dihydroxyvitamin D (1α,25(OH)2D or calcitriol), or by 24-hydroxylase (CYP24A1) to form 24,25(OH)2D, a metabolite implicated in bone repair and autoregulation of the vitamin D pathway [4,14]. The expression of CYP24A1 is upregulated by high calcitriol levels and downregulated by low calcium/phosphate or elevated PTH, reflecting tight feedback control. A secondary metabolite, 24,25(OH)2D, is mainly produced when calcitriol levels are high, with a key role in regulating bone metabolism and vitamin D. High levels of calcitriol activate the expression of CYP24A1, which increases the synthesis of 24,25(OH)2D and promotes the degradation of calcitriol. Low levels of calcium and phosphate inhibit CYP24A1 and promote the production of calcitriol instead of 24,25(OH)2D. Parathyroid Hormone (PTH) inhibits CYP24A1 and stimulates the production of calcitriol. 24R,25(OH)2D plays a key role in modulating bone metabolism and promoting bone repair, particularly in fractures. It can degrade calcitriol, reducing levels of active vitamin D and protecting against hypercalcemia. It has also recently been suggested that it may play a role in regulating the immune response [26]. Calcitriol exerts its effects via the vitamin D receptor (VDR), a nuclear receptor that forms heterodimers with Retinoid X Receptor (RXR) to regulate gene expression through vitamin D Response Elements (VDREs). The VDR is widely expressed in various body tissues, including the intestine, pancreas, kidney, bone, bronchi, skin, osteoblasts, chondrocytes, endocrine glands, reproductive tissues, and immune cells. As a result, the VDR-RXR complex interacts with Vitamin D Response Elements (VDREs) located in the promoter regions of specific target genes, thereby positively or negatively regulating their expression by recruiting VDR-interacting nuclear proteins (coregulators) to the transcriptional preinitiation complex [31]. After ligand binding, the repositioning of helix 12 to the COOH terminus of the VDR ligand-binding domain, known as the ligand-dependent Activation Function 2 (AF2), confers a major conformational change in the three-dimensional structure of the VDR. This activation step appears necessary for the recruitment of motor proteins by the VDR [26,32], which are responsible for the rapid translocation of the cytoplasmic VDR along microtubules to the nucleus. Point mutations in the two nuclear localizing signals result in defective cytoplasmic-nuclear translocation and the type II vitamin D rickets phenotype [33,34]. Selective association between VDR and its protein partner, RXR, involves dimerization surfaces in three domains of the VDR molecule and induces a VDR conformation that is essential for the transactivating function of VDR. An interaction between the ligand binding and heterodimerization domains has been suggested by two naturally occurring mutations (I314S and R391C) in the LBD of VDR, which confer the vitamin D resistance phenotype by significantly impairing both VDR-RXR heterodimerization and ligand retention [35]. Recently discovered VDR interactions with nuclear coregulatory molecules provide new mechanistic insights into VDR-mediated positive and negative modulation of transcription [36]. Recent studies suggest a bifunctional role for the VDR co-modulator NCoA62/Skip: it can promote transcriptional activation or repression in a cell-specific manner, depending on the expression of coregulatory molecules. The coactivator p300 and the corepressors NCoR and SMRT interact with the same NH2-terminal region of the Skip molecule. The relative expression levels of the nuclear corepressor NCoR and the coactivator CBP/p300 in CV-1 and P19 cells determine whether Skip activates or represses VDR/RXR-dependent transcription [37]. More importantly, the 1,25(OH)2D-VDR complex selectively induces the expression of genes from two important families of coregulators, TIF2 among the p160 coactivators and SMRT among the corepressors [38]. Since SRC1/TIF2 ratios have been shown to influence important cellular functions, such as energy metabolism in mouse adipose tissue [39], the apparent cellular specificity of the 1,25(OH)2D-VDR complex in inducing gene transcription, and possibly TIF2 and SMRT protein abundance, suggests that 1,25(OH)2D itself may modulate the transcriptional competence of target cells. In addition to Skip, a novel ATP-dependent chromatin remodeling complex containing the Williams syndrome transcription factor potentiates ligand-induced VDR action in transactivation and gene repression. NCoA62/Skip also mediates a link between transcriptional regulation by the VDR (and other nuclear receptors) and RNA splicing by the spliceosome. Skip physically interacts with components of the splicing machinery and with nuclear matrix-associated proteins. Indeed, the expression of a dominant-negative Skip interfered with the proper splicing of 1,25(OH)2D-VDR transactivation-derived transcripts [40]. In conclusion, complex cell-specific and promoter-specific interactions of VDR nuclear coregulators are responsible for VDR regulation of vitamin D-responsive gene expression, including VDR coactivator and corepressor molecules. This intricate mechanism underlies the pleiotropic effects of vitamin D on numerous biological processes throughout the body [5,26,41] (Figure 2).
Further clarification is needed on the three forms of vitamin D, which are similar but not identical. The presence of vitamin D epimers in clinical trials is important because some methods of measuring 25(OH)D may include its epimers, potentially overestimating the levels of active vitamin D. The best-known epimers are:
1α,25(OH)2D (calcitriol), which is the biologically active form of vitamin D. It is essential for calcium metabolism and the regulation of many cellular processes. It is associated with osteoporosis, rickets, hypocalcemia, immune dysfunction, and some autoimmune diseases.
3-epi-1α,25(OH)2D (3-epi-calcitriol), which has a different configuration at carbon 3 of ring A. It has reduced affinity for the VDR and reduced biological effects. Its role is not fully understood, but it may be less calcium-affecting than calcitriol, with therapeutic implications in conditions requiring control of calcium metabolism. Do not alter the core message.
24R,25(OH)D, which is a less well-understood metabolite of vitamin D involved in bone repair and regulation of vitamin D metabolism. It has been linked to a protective role in certain bone diseases and the regulation of vitamin D excess [30].

3. Molecular Effects of Vitamin D on Immune Cells

Recent studies have shown that immune cells express VDR and produce enzymes when stimulated by vitamin D, so vitamin D deficiency may impact inflammatory diseases [26].
Monocytes and macrophages recognize bacteria, fungi, and viruses via Toll-Like Receptors (TLRs) on their surface. This activates VDR and CYP27B1 [26]. Vitamin D’s role in host defense is via an autocrine pathway in monocytes and macrophages, stimulated by TLRs2/1 and TLR4, Interferon-γ (IFN-γ), or CD40 [31,32]. Activation of these receptors initiates a signaling cascade responsible for the microsomal-mediated upregulation of cytoplasmic levels of VDR and CYP27B1, resulting in the conversion of 25(OH)D to 1,25(OH)2D. This results in calcitriol peaks in the cytosol, which drive signaling and calcitriol/VDR complexes, leading to the expression of multiple genes that modulate monocyte/macrophage function during infection. The circulating hormone calcitriol does not enter target cells at significant levels. This is important for balanced intracellular signaling, which is critical for immune cell functions [42]. Levels of vitamin D in the blood are tightly regulated to prevent too much VDR signaling. This is achieved through a metabolic feedback loop involving conversion by the inactivating enzyme 24-hydroxylase (CYP24A1) to inactive calcitroic acid, which is excreted in the bile. Immune cells express CYP27B1, which allows them to activate functional VDR signaling pathways by converting inactive vitamin D to an active form [33].
The heterodimer VDR-RXR binds to DNA and induces the assembly of cathelicidin antimicrobial peptide (cAMP), β-defensin 2 (HBD2), and other antimicrobial peptides (AMPs) with bactericidal and viricidal activity [26,34,35]. AMPs have dual roles in innate immune defense: antimicrobial activity and chemokine, cytokine, and ROS induction. CAMP is found in keratinocytes and cells of the adaptive immune system and helps with new blood vessel formation, wound healing, chemokine expression, and the movement of various immune cells, especially in response to mycobacterial organisms such as tuberculosis and leprosy, and other types of bacteria. The vitamin D pathway is the only one capable of regulating cAMP expression in keratinocytes by two mechanisms: direct regulation of cAMP and epigenetic action (histone acetylation) on VDRE, which is located in the promoter region of the cAMP gene [36,37]. Calcitriol activates the NF-kB transcription factor, which regulates NOD2 expression and muramyl dipeptide binding. The muramyl dipeptide binding is a peptidoglycan found in Gram-negative bacteria that promotes the transcription of DEFB4, the gene for β-defensin 2, via Nuclear Factor Kappa light chain (NF-κB) [26,37]. Some authors have discovered that transforming growth factor-1 (TGF-1) interferes with the signaling pathway activated by vitamin D, by reducing VDR expression and suppressing NF-kB transcription factor activity [38]. This reduces the expression of genes involved in host defense and impairs airway epithelial cells’ ability to fight respiratory pathogens. Vitamin D induces the transcription of Atg-5 and Beclin-1, which promote autophagy via the induction of cAMP and its downstream factors (p38, ERK, and C/EBPβ) [26,37]. Autophagy is important in the antimicrobial response in several immune-mediated inflammatory diseases, such as asthma, atopic dermatitis, rheumatoid arthritis and psoriasis, and in infectious diseases. In this context, vitamin D counteracts steroid resistance by inhibiting the gene for a steroid; therefore, lower serum vitamin D levels are associated with higher corticosteroid requirements, which has important implications, particularly for asthma [3,39,40,41,43].
Vitamin D, mediated by transforming growth factor-β (TGF-β), plays a role in the inflammatory response of cells of the monocyte/macrophage lineage. Macrophages contain 1α-hydroxylase and are therefore capable of autocrine or paracrine activation of vitamin D. In the context of local dermal inflammation, CD69 overexpression has been reported in systemic lupus erythematosus (SLE), Graves’ disease (GD), and Hashimoto’s thyroiditis (HT). The combination of TGF-β and 1,25(OH)2D has been shown to reinduce the differentiation antigen CD69 in monocytic cells [3] (Figure 3).
Calcitriol downregulates MHC class II and co-stimulates CD40, CD80, and CD86 in dendritic cells (DCs), activating T cells. It also prevents the maturation of DCs that express VDR, thereby inhibiting excessive inflammation in response to infectious diseases [44,45].
B and T cells express VDR and CYP27B1.VDR mRNA is expressed at low levels in B cells, but these cells may be capable of autocrine/intracrine signaling, and the receptors can be upregulated by stimulation with 1,25(OH)2D [46]. A recent study showed that calcitriol suppresses the differentiation of B cells and their maturation into memory B cells and plasma cells [25,26]. It may also inhibit B cell proliferation by upregulating mRNA at the level of p27, thereby stopping the cell cycle entry of previously cycling B cells.
It also has inhibitory effects on plasma cell differentiation and immunoglobulin (Ig) production, which has important implications for the role of vitamin D in some pathologies such as asthma, autoimmune thyroiditis, and SLE. Vitamin D suppresses IgE production by B cells and increases IL-10, promoting regulatory B cells. IL-10 increases IgG4, which is linked to positive outcomes after allergen desensitization immunotherapy [47]. The precursor 25(OH)D has similar effects to the active form on purified B cells, but at higher concentrations [47,48].
Naïve T cells express low levels of VDR, which increase upon activation. Calcitriol promotes anti-inflammatory cytokine secretion and suppresses pro-inflammatory cytokines, reducing Th1-type differentiation. Vitamin D suppresses the production of pro-inflammatory cytokines (IFNγ and TNF-α), specifically by inhibiting the p38 mitogen-activated protein (MAP) kinase pathway in monocytes [24,26,49,50,51]. Calcitriol inhibits Th17 secretion of IL-17, IFNγ, IL-21, IL-22, and granzyme B and downregulates Th17-type differentiation by suppressing IL-23 production and by acting on RAR-related orphan receptor C and aryl hydrocarbon receptor (Figure 4). Vitamin D probably acts by reducing the expression of RORγt/IL-17 by inhibiting the translocation of the p65 transcription factor to the nucleus, decreasing IL-17 mRNA levels [52,53,54,55]. Vitamin D may play a role in some inflammatory diseases, as Th17 cells are important in their pathogenesis. Conversely, calcitriol induces the FoxP3 transcription factor that promotes regulatory T cell (Treg) differentiation [26,56,57] (Figure 4). Finally, vitamin D induces the expression of CD200, an immunoglobulin-like anti-inflammatory protein, on T cells, regulating the immune response through its binding to cells expressing the CD200 receptor [3].
Regarding mast cells, vitamin D promotes anti-inflammatory IL-10 production by mast cells. It also suppresses mast cell activation, reducing histamine and TNF-α release, including IgE-dependent activation [47].
Vitamin D and its active metabolites play a significant role in endothelial function, influencing both genomic and non-genomic pathways. Specifically, 25(OH)D and calcitriol help stabilize the vascular endothelium through a non-genomic mechanism. Calcitriol interacts with the VDR, activating intracellular pathways, including adenylyl cyclase/cyclic adenosine monophosphate (AC/cAMP) and inositol trisphosphate/diacylglycerol (IP3/DAG), which leads to the upregulation of endothelial nitric oxide synthase (eNOS), which boosts the production of nitric oxide (NO) in the endothelium and enhances calcium signaling within endothelial cells. Additionally, VDR activation triggers eNOS activation via the PI3K/Akt pathway, facilitating the phosphorylation of eNOS at serine-1779 [58]. In animal models, calcitriol has been shown to stimulate endothelial cadherin-based cellular junctions, reduce the formation of stress fibers, and limit the organization of intracellular endothelial gaps, helping to prevent endothelial damage [59]. Furthermore, vitamin D metabolites play a crucial role in protecting against vascular dysfunction and systemic inflammation, both of which are key contributors to tissue injury. Vitamin D also regulates several genes involved in immunity, which are critical for understanding immune-related diseases and diagnosing vitamin D deficiency. For instance, CD93 glycoprotein plays a role in innate immunity, affecting cell adhesion, phagocytosis, and inflammation in monocytes, endothelial cells, granulocytes, platelets, and stem cells. CD93 functions as a receptor for TLR9, playing a key role in inflammatory responses through Lipopolysaccharide (LPS)/TLR4 signaling [60]. The Ninjurin1 protein, which is expressed in myeloid and endothelial cells, contributes to the immune pathogenesis of diseases such as multiple sclerosis, promoting cell adhesion and leukocyte migration, particularly during endothelial inflammation [61]. The CEBPB gene, a vitamin D target in myeloid leukemia cells, is involved in inflammation via the Th17 pathway, particularly in models of multiple sclerosis. Additionally, ACVRL1, a protein within the transforming growth factor-beta superfamily, is involved in monocyte differentiation to macrophages. Finally, SRGN, expressed by monocytes, macrophages, lymphocytes, mast cells, and endothelial cells, is involved in the secretion of inflammatory mediators following LPS exposure [62], and it is crucial in the inflammatory response and immune system modulation [63].

4. Allergic Diseases

Allergic diseases represent a pervasive public health concern. It has been estimated that one in four people in Western countries will become allergic [64].
An allergy is defined as an excessive immune response, categorized as a type I hypersensitivity reaction, directed towards various natural proteins, including pollen, house dust mites, food allergens, insect venom, or drugs. The allergic response is characterized by a sensitization phase, during which allergens are presented by Antigen-Presenting Cells (APCs) to naïve T cells through class II Major Histocompatibility Complex (MHC) molecules. This results in the differentiation of T cells into Th2 cells, which in turn produce IL-4 and IL-13, stimulating the production of IgE by B cells. Subsequently, in the second effector phase, following a new exposure to the allergen, IgE binds to the high-affinity IgE receptor (FcεRI) expressed by mast cells and basophils, resulting in degranulation, defined as a substantial discharge of inflammatory mediators stored in cytoplasmic secretory granules. These include, but are not limited to, histamine, leukotrienes, and heparin, which are responsible for allergic clinical manifestations. In addition, the release of IL-5 and IL-13 by T cells causes eosinophil activation and tissue infiltration, which is responsible for chronic inflammation [64,65].
Some studies have demonstrated a correlation between vitamin D and allergic diseases, including asthma, allergic rhinoconjunctivitis, atopic dermatitis, chronic urticaria, allergic contact dermatitis, and food allergies [66]. In the following paragraphs, we will analyze the possible therapeutic role of administering vitamin D in some allergic diseases, as recent clinical trials show.

4.1. Vitamin D, Asthma, and Rhinosinusitis

Asthma, the most common chronic respiratory disease in children, is characterized by chronic airway inflammation, remodeling, hyperresponsiveness, and airflow limitation, leading to symptoms such as wheezing, dyspnea, chest tightness, and cough, with its pathogenesis involving genetic, immunological, and environmental factors, including vitamin D status [67]. At the same time, rhinosinusitis, an inflammatory condition affecting the nasal mucosa and paranasal sinuses, presents with nasal obstruction, itching, rhinorrhea, sneezing, and ocular symptoms, affecting 10–20% of the global population and frequently associating with asthma [68]. Multiple studies demonstrate an inverse relationship between maternal and infant vitamin D levels and asthma prevalence and severity, with deficiency (<25 nmol/L) increasing exacerbation risk by 25% [69]. While definitions vary, most studies classify sufficient levels as ≥30 ng/mL (≥75 nmol/L), insufficient as 20–29.9 ng/mL (50–74.9 nmol/L), and deficient as <20 ng/mL (<50 nmol/L) [70,71,72]. Higher maternal vitamin D intake during pregnancy, particularly with levels > 30 ng/mL, reduces offspring asthma risk [73,74,75,76]. Genetic analyses suggest maternal 17q21 genotype influences this protection [77], with meta-analyses confirming lower vitamin D in asthmatic children [78] and a U-shaped dose-response relationship showing optimal protection at ~70 nmol/L but potential risk beyond ~130 nmol/L [79]. In contrast, asthmatics with low vitamin D levels exhibit reduced FEV1 and FEV1% [80]. In rhinosinusitis, deficiency (<10 ng/mL) correlates with severe allergic rhinitis and elevated IgE [81], and insufficiency (<30 ng/mL/<20 ng/mL) is a risk factor for chronic rhinosinusitis with nasal polyps [68,82,83], with in vitro studies showing vitamin D3 analogs reduce fibroblast proliferation via Regulated on Activation Normal T Expressed and Secreted (RANTES) and eotaxin suppression while enhancing cAMP expression [83]. Children with vitamin D > 75 nmol/L have lower aeroallergen sensitization. However, maternal intake or infant supplementation does not significantly alter allergic rhinitis risk, with observational data suggesting lower prevalence in those > 75 nmol/L versus < 50 nmol/L, particularly in males, though RCTs are needed [84]. Vitamin D modulates immunity by reducing Th17-driven inflammation [67,85] and urban particulate matter-induced Th17 responses [52], while supplementation may reduce rhinovirus replication, shift Th1/Th2 balance toward Th2 dominance (reducing IFN-γ, IL-5, IL-6, IL-8, and IgE while increasing TGF-β, IL-10, and IL-4) [77,79,86,87], enhance glucocorticoid responsiveness, in particular with a dose of 50,000 IU, via glucocorticoid receptor GRα upregulation [88], and inhibit remodeling by downregulating MMPs, fibronectin, and IL-6 [38,39]. Clinical trials show mixed results, with some reporting reduced exacerbations [89] but others such as the vitamin D Add On Therapy Enhances Corticosteroid Responsiveness in Asthma (VIDA) trial finding no significant benefit [90], potentially due to genetic variability, as VDR single-nucleotide polymorphisms (SNPs) such as ApaI (rs7975232), BsmI (rs1544410), FokI (rs2228570), TaqI (rs731236) and rs3782905a influence asthma susceptibility [91,92,93], while in allergic rhinitis, vitamin D with intranasal steroids improves symptom scores [like Total Nasal Symptom Score (TNSS) and Rhinitis Control Assessment Test (RACT)] and modulates CD4+/CD8+ ratios and cytokines (TNF-α↓, IFN-γ↑, IL-10↑) [94,95], and Sublingual ImmunoTherapy (SLIT) with vitamin D reduces symptoms in children, though larger trials are needed [96,97]. All these findings suggest that vitamin D supplementation may be a cost-effective, low-risk, and promising method for asthma control and symptom improvement in rhinosinusitis, both when added to intranasal steroid spray or antihistamine medication and when combined with SLIT, which in this context represents a new, effective, and well-tolerated immunotherapy modality. Further studies are needed to define this new therapeutic option.

4.2. Vitamin D and Atopic Dermatitis

Atopic dermatitis (AD), the most common chronic skin disease affecting 10–20% of children, is characterized by chronic pruritus and results from genetic predisposition, immune dysregulation, and impaired skin barrier function, increasing infection susceptibility [98,99,100,101]. The immune dysfunction in AD involves reduced innate immune cell activity at TLR sites, decreased IL-17 production, and impaired Treg cell function, all of which contribute to disease pathogenesis [100,101,102]. Vitamin D plays a crucial role in AD pathophysiology by enhancing cAMP expression in keratinocytes through Act1 and MEK/ERK activation, potentially explaining the therapeutic benefits of UVB therapy through vitamin D-mediated AMP production [36]. Importantly, vitamin D upregulates epidermal barrier proteins, including filaggrin, involucrin, loricrin, and transglutaminase [99], while its metabolism in skin and immune cells is regulated by Cyp27b1 and Cyp24a1 enzymes. Vitamin D also modulates immune responses by increasing Treg cell numbers and function through (DC)-mediated mechanisms involving PD-L1 expression and direct effects on CD4+ T cells to promote IL-10-secreting Tregs [102]. Clinical observations demonstrate that AD patients, particularly children, frequently exhibit vitamin D deficiency/insufficiency, with lower levels correlating with greater disease severity as measured by the Scoring of Atopic Dermatitis (SCORAD) index and elevated IL-4, IL-17A, and IL-22 levels [98,101]. Intervention studies report that daily supplementation with 1000–2000 IU vitamin D for 1–2 months significantly improves eczema scores [7,46,103,104,105,106,107], though these findings require cautious interpretation due to limitations including variable supplementation protocols, unmeasured confounders (sun exposure, diet, topical treatments), and lack of data in infants and pregnant women [108]. Neonatal vitamin D deficiency appears to increase later AD risk, while supplementation in children reduces symptoms and Th17 cell activity [101]. Genetic studies reveal associations between AD severity and polymorphisms in vitamin D metabolism genes, particularly elevated Cyp24a1 in severe cases and specific Cyp27b1 loss-of-function variants in adult subtypes [99]. While UVB therapy’s anti-inflammatory effects suggest potential for vitamin D as an oral or topical treatment [36], current evidence remains insufficient to establish standardized supplementation protocols or identify optimal candidate populations, necessitating further interdisciplinary research to translate biochemical improvements into clinically meaningful outcomes [98,107].

5. Vitamin D and Autoimmune Diseases

Several clinical studies have shown that vitamin D deficiency and receptor disorder/polymorphism may be associated with the development and progression of rheumatoid arthritis (RA), psoriatic arthritis (PA), SLE, multiple sclerosis, and autoimmune endocrine diseases such as Hashimoto’s thyroiditis (HT) and Graves’ disease (GD), type 1 diabetes, and Addison’s disease [2,5]. However, the role of vitamin D supplementation in treating these diseases is still unclear, as some studies have shown that vitamin D supplementation is beneficial, while others have shown no effect on key parameters of these diseases. These results may be due to differences in the type and dose of supplementation, as well as individual characteristics of the patients who participated in the studies. Therefore, new multicenter clinical trials are needed to elucidate potential underlying mechanisms [2].

5.1. Vitamin D and Rheumatoid Arthritis

Rheumatoid arthritis (RA) represents a complex autoimmune disorder characterized by chronic inflammation of synovial joints, leading to progressive cartilage and bone destruction. The pathophysiology of RA involves a multifaceted interplay between immune cells, inflammatory mediators, and genetic factors, with emerging evidence highlighting vitamin D’s significant immunomodulatory role.
At the molecular level, elevated IL-8 production by macrophages, neutrophils, and osteoclasts contributes substantially to the inflammatory milieu and bone resorption observed in RA [109]. This chemokine not only promotes leukocyte recruitment but also facilitates osteoclast activation, thereby accelerating joint destruction.
The immunopathogenesis of RA involves dysregulated interactions between DCs and T cells, influenced by metabolic factors such as glycolysis and reactive oxygen species (ROS), as well as vitamin D status [110].
Th17 cells in patients with early RA induce an inflammatory feedback loop for synovial fibroblast-mediated RA response interactions, including autocrine interleukin IL-17A production. The combination of 1,25(OH)2D and 1,25(OH)2D was found to regulate Th17 activity and synovial inflammation in patients with RA and potentially in patients with other Th17-mediated inflammatory diseases, suggesting that activation of VDR signaling may be more valuable than current TNF-neutralization strategies [104].
Clinically, vitamin D deficiency is prevalent in RA patients and correlates with increased disease activity (higher DAS28 scores), greater pain sensitivity by Visual Analog Scale (VAS), and reduced muscle strength [111,112,113,114].
Seasonal variations further support this relationship, as disease flares often coincide with lower vitamin D levels in winter and spring [115].
While high-dose 1,25(OH)2D therapy is limited by hypercalcemia, the non-calcemic analog 20S(OH)D3 has demonstrated efficacy in suppressing arthritis in preclinical models by activating a naturally suppressive receptor on leukemic cells called the Leukemic Immunoglobulin-1 Receptor (LAIR-1), highlighting its potential as a novel treatment. Vitamin D suppressed arthritis in LAIR-1-sufficient DR1 mice, while it was ineffective in LAIR-1-/- deficient mice [116].
In patients receiving abatacept, a frequently proposed alternative to rituximab that blocks T-cell co-stimulation, 1,25(OH)2D enhanced inhibition of T-cell activators and strongly suppressed T-cell activation induced by cross-linked anti-CD3, inhibiting T-cell activation, supporting its role as an adjunct therapy [117]. Considering the results of the studies presented in Table 1, oral alfacalcidiol (1α-hydroxyvitamin D3) at a dose of 2 micrograms/day has a positive effect on disease activity in the proportion of cases showing complete remission, symptom reduction, or no new symptoms. In addition, oral alfacalcidiol improved pain control, muscle function, and bone metabolism, though longer-term data are needed to assess its impact on fracture risk [118,119]. Interesting results from four meta-analyses show that TaqI and FokI VDR polymorphisms also have a significantly significant involvement in the development of RA. In particular VDR polymorphisms rs7975232, rs1544410, rs2228570, and rs731236 were significantly associated with RA susceptibility in some populations, particularly in Asians. The contribution of the VDR was modest, confirming that RA is a complex disease and therefore multiple genes may be involved, each of which makes a minor contribution to the risk of arthritis. Further characterization of VDR, in addition to traditional and associated risk factors, may contribute to the early identification of patients at high risk of developing RA and the development of new prevention and treatment strategies [111,112,113,120].
Furthermore, alfacalcidol has shown sustained benefits in preventing glucocorticoid-induced osteoporosis, and combination therapies with hormone replacement therapy (HRT) suggest additional immunomodulatory effects by reducing serum Ig levels and B- and T-cell-independent responses to recall antigens, though further research is needed to clarify these interactions [121,122].
The evolving understanding of vitamin D’s role in RA reflects the broader shift toward personalized medicine in rheumatology. Although it is not a panacea, the rational use of vitamin D supplementation is a safe and cost-effective adjunct to conventional therapies that has the potential to improve outcomes for many patients.
The challenge for the future is to better define which patients will benefit most and under which therapeutic conditions.
Current evidence supports routine vitamin D status assessment in RA patients, with supplementation recommended for those with deficiency. However, several questions remain regarding optimal dosing strategies, long-term effects on disease progression, and the potential benefits of vitamin D analogs with improved safety profiles. Future research should focus on personalized approaches that consider genetic factors, disease stage, and concurrent medications to maximize therapeutic outcomes while minimizing adverse effects.

5.2. Vitamin D and Psoriasis

Psoriasis is an autoimmune skin disease that causes plaques to appear on the skin, affecting about 2–3% of the world’s population. The most common type is psoriasis vulgaris, accounting for around 80–90% of cases, while other types include pustular, gutta-percha, inversion, and erythrodermic psoriasis, affecting children and adults, respectively. Psoriatic lesions appear on the scalp, skin folds, hands, feet, nails, and genitalia [123].
Psoriasis is a multifactorial skin disease. Although the inheritance pattern is unclear, a genetic predisposition plays a pivotal role in its susceptibility. The immunopathogenesis of psoriasis involves a complex inflammatory cascade. It is initially triggered by innate immune cells (keratinocytes, DCs, natural killer (NK) cells, macrophages, fibroblasts, and γδ T cells) activated by external or internal factors. Genetically predisposed individuals are susceptible to the disease. The cytokines produced by innate cells activate myeloid DCs. They increase the production of other cytokines. These are involved in the differentiation of lymphocytes into the primary adaptive immune cells Th1, Th22, and Th17. These cells play a central role in disease pathogenesis. The cytokines produced by these cells, including TNFα, IL-22, and IL-17A/F, lead to keratinocyte proliferation. They also lead to neoangiogenesis, chemokine production, neutrophil and CD8+ cell migration into the epidermis, and a chronic inflammatory process [124]. The activity of these cells is modulated by Treg, which can inhibit the immune response and prevent autoimmune responses against self-antigens [125]. Vitamin D [126] represents a potential therapeutic factor and protective agent in psoriasis.
The effects of vitamin D on the immune system in psoriasis are complex. Vitamin D inhibits IL-17-induced expression of IL-1Ra, IL-36α, IL-36β, and IL-36γ and TNF-α-induced expression of IL-1Ra, IL-36Ra, IL-36α, IL-36γ and HBD2 in human keratinocytes. In addition, vitamin D affects T-cell homing in the skin: the expression of the chemokine receptor CXCR10 on T cells is increased by vitamin D treatment, enabling these cells to respond to the skin-specific chemokine CCL27 secreted by keratinocytes. In contrast, vitamin D reduces the expression of cutaneous lymphocyte-associated antigen (CLA) on T cells. Epidermal keratinocytes synthesize 1,25(OH)2D and are targeted by this hormone, which exerts intracrine, autocrine, and paracrine effects on keratinocytes and neighboring cells. It regulates growth, differentiation, apoptosis, and other biological processes via genomic and non-genomic effects. The non-genomic effects of 1,25(OH)2D and analogs are partly related to effects on intracellular calcium [127], whereas the genomic effects of 1,25(OH)2D are mediated by binding to the VDR. This predominant nuclear receptor protein is present in target tissues and binds 1,25(OH)2D with high affinity and low capacity [128]. As vitamin D can promote terminal differentiation and inhibit proliferation of keratinocytes [129], impairment of VDR expression in epidermal skin cells may be involved in the pathogenesis of psoriasis, and not only changes in VDR expression but also VDR polymorphism are likely to be associated with psoriasis [126].
Clinically, psoriasis patients exhibit significantly lower serum 25(OH)D levels compared to healthy controls, with the most severe cases showing the greatest deficiency [118,119,126,130,131,132,133,134,135], though oral supplementation demonstrates limited efficacy in improving Psoriasis Area and Severity Index (PASI) scores, likely due to variable dosing and bioavailability [136].
Emerging evidence indicates that moderate to severe psoriasis is inversely correlated with serum vitamin D levels. However, these levels show no significant association with arthritis, family history, age at diagnosis, or disease duration [137]. This suggests that vitamin D supplementation may help mitigate systemic inflammation and oxidative stress, potentially delaying the development of psoriasis-associated comorbidities [138]. Vitamin D analogs, including maxacalcitol, tacalcitol, and calcipotriol, have been used as adjuvants in psoriasis treatment due to their ability to regulate epidermal cell differentiation, inhibit proliferation and angiogenesis, and modulate immune responses, all with a low risk of hypercalcemia [139,140,141,142]. In murine models, topical maxacalcitol significantly reduced inflammation and MHC class II APC infiltration in imiquimod (IMQ)-induced psoriasiform skin, though this was demonstrated in a prevention rather than treatment model [53]. Calcipotriol, introduced in the late 1980s, has shown consistent efficacy and safety, with studies reporting a 50–60% reduction in PASI scores within four weeks and no significant alterations in calcium metabolism [143,144]. Combined formulations, such as calcipotriene (0.005%) with betamethasone dipropionate (0.064%), have further improved treatment outcomes. PADTM technology cream containing this combination has demonstrated high efficacy and safety in two multicenter, randomized phase 3 trials [145,146], though limitations include missing data on scalp psoriasis, adolescent populations, and long-term intermittent use. Additional studies confirmed that calcipotriol/betamethasone foam, betamethasone foam, and clobetasol propionate ointment reduce epidermal thickness and improve tPASI scores. Notably, only the CAL/BDP foam combination significantly reduced CD8⁺ IFNγ⁺ T cells and neutrophils in lesional skin, though over a short treatment duration [147]. Long-term use of tacalcitol has also proven effective. A multicenter trial using tacalcitol (4 μg/g) daily for 18 months showed sustained efficacy and safety without affecting serum calcitriol, calcium, or PTH levels [148]. A separate study using a higher dose (20 μg/g) demonstrated PASI score reduction and minor local adverse effects, with stable serum calcium homeostasis despite reduced PTH and 1,25(OH)2D levels [149]. A recent meta-analysis found that topical corticosteroids and vitamin D analogs offered comparable outcomes in PASI, Investigator’s Global Assessment (IGA), and Patient Global Assessment (PGA) scores, although evidence certainty was low. Common limitations included a lack of scalp psoriasis data and withdrawal due to local irritation from vitamin D analogs [150]. Importantly, a systematic review found no benefit from oral vitamin D supplementation in patients with normal baseline levels and no clear superiority among different vitamin D formulations [151]. Taken together, current evidence supports the role of topical vitamin D analogs as an effective and safe component of psoriasis management, particularly when used in combination with other therapies.

5.3. Vitamin D and Systemic Lupus Erythematosus

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease marked by widespread immune dysregulation, including T-cell imbalance, B-cell hyperactivation, autoantibody production, and immune complex deposition, contributing to inflammation, vasculitis, and organ damage. While the precise role of vitamin D in SLE remains uncertain, whether causal or consequential, its immunomodulatory potential has drawn increasing interest. Vitamin D deficiency is prevalent in SLE patients, likely due to sun avoidance and sunscreen use, along with factors such as age, obesity, skin pigmentation, geographic location, and pharmacological treatments. Hydroxychloroquine impairs 1-α hydroxylation of 25(OH)D, while corticosteroids accelerate vitamin D catabolism, especially at doses >10–25 mg prednisone equivalent/day, reducing 1,25(OH)2D levels [111,152,153]. Vitamin D’s active form, 1,25(OH)2D, acts through the VDR, modulating immune responses by suppressing pro-inflammatory cytokines such as TNF-α, IL-17, Monocyte Chemoattractant Protein-1 (MCP-1), and supporting NK cell function [48,154,155]. Although vitamin D deficiency alone does not cause autoimmunity, it may contribute to the presence of genetic and environmental factors [115]. Some studies report lower IL-23 and higher pro-inflammatory cytokines in deficient patients, though findings vary. In addition, BsmI, FokI, and bb VDR polymorphisms have been associated with increased SLE risk, particularly in Asian populations [111,156,157]. Although anti-vitamin D antibodies have been detected in some patients, they do not consistently correlate with disease markers, except for anti-dsDNA [152,158]. In animal models (MRL/lpr and NZB/NZW), 1,25(OH)2D treatment reduced anti-dsDNA and anti-ssDNA antibodies, inflammatory cytokines, skin lesions, and renal pathology while improving survival effects comparable to high-dose corticosteroids [152,159]. Human studies support an association between low 25(OH)D levels and increased disease activity. The Nurses’ Health Study I and other case-control and cross-sectional studies showed lower vitamin D levels in SLE patients, particularly those with renal disease, photosensitivity, and reduced physical function [23,160,161,162]. While some inconsistencies exist, vitamin D deficiency correlates with fatigue, musculoskeletal symptoms, and lower quality of life. vitamin D supplementation has been shown to reduce disease activity measured by systemic lupus erythematosus disease activity index (SLEDAI), systemic lupus erythematosus disease activity index 2000 (SLEDAI-2K), European Consensus Lupus Activity Measurement Index (ECLAM), fatigue by fatigue severity scale (K-FSS), inflammatory markers (e.g., IL-6), flare frequency, and steroid/immunosuppressant use, although the results are not uniform [111,163,164,165,166,167,168]. Supplementation may also improve bone health and cognitive function, as well as reduce the risk of infection [152,169,170,171]. Novel strategies include combining vitamin D with tetanus toxoid to enhance immunogenicity [172] and using nonhypercalcemic analogues to avoid hypercalcemia while maintaining immunomodulatory effects [48]. Given the prevalence of hypovitaminosis D and its association with increased disease activity and comorbidities, routine monitoring and supplementation are recommended in SLE management. Optimizing vitamin D levels may help reduce disease burden, improve outcomes, and minimize long-term complications such as osteoporosis and cardiovascular disease [152,168,173].

5.4. Vitamin D and Autoimmune Thyroid Diseases

Hashimoto’s thyroiditis (HT) is a T cell-mediated autoimmune disorder where proinflammatory cytokines such as IFN-γ induce MHC class II HLA-DR antigen expression on thyrocytes, triggering immune-mediated thyroid damage. The active form of vitamin D, 1,25(OH)2D, exerts immunomodulatory effects by promoting B cell apoptosis, reducing thyroid autoantigens and antibodies, and modulating T cell and DC function, thereby protecting against thyroid autoimmunity.
A recent meta-analysis identified a significant association between the FokI VDR polymorphism and HT in Asians, but not Caucasians, while TaqI, ApaI, and BsmI showed no significant associations [174].
Several clinical studies report low vitamin D levels in HT patients. In an Indian study, weekly supplementation of 60,000 IU of vitamin D over eight weeks in deficient patients improved TSH levels and reduced antibody titers compared to placebo. However, patients with normal vitamin D levels were excluded, limiting generalizability [175].
A randomized trial also found that cholecalciferol supplementation reduced TPO-Ab and Tg-Ab levels, although the study’s small sample size, short duration, and limited demographic diversity constrained its conclusions [176]. These results are supported by three meta-analyses indicating that vitamin D3 supplementation, especially for periods >12 weeks, reduces thyroid autoantibodies and improves FT3 and FT4 levels in HT patients [116,177,178]. Despite promising outcomes, optimal dosing and treatment duration remain undefined, necessitating larger multicenter trials [175].
In addition to vitamin D, the vitamin D analog elocalcitol (BXL-628) has shown promise in modulating immune responses in thyroid cells. Compared to methimazole (MMI), elocalcitol reduced CXCL10 secretion and Th1-type cytokine activity while promoting a Th2 shift and lowering TRAb levels in Graves’ disease models. These findings highlight its potential as an immunomodulatory agent in autoimmune thyroid disorders and support exploring combination therapies to enhance efficacy and reduce side effects [179].

5.5. Vitamin D and Type 1 and Type 2 Diabetes Mellitus

Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disorder characterized by immune-mediated destruction of pancreatic beta-cells (β-cells), leading to impaired insulin production or function [180]. Current knowledge suggests that an important predisposing role to the onset of the disease could be played by vitamin D: impairment of its signaling, especially in the first years of life, which increases the risk of autoimmunity [181,182,183]. Given the role that vitamin D plays in the immune system, it is believed that it may have a protective role in the development of T1DM [184].
In the pancreas of affected patients, there is an inflammatory infiltrate composed of T-cells, B-cells, and macrophages; 1,25(OH)2D promotes macrophage differentiation, which is essential in animal models of T1DM for the activation of anti-inflammatory phenotype (M1→M2) via the VDR-PPARgamma signaling pathway; it also reduces effector T-cell numbers and promotes the induction of T-reg cells [185,186,187].
The Diabetes Autoimmunity Study in the Young (DAISY) reported that increased maternal intake of vitamin D in food reduced the risk of autoimmunity against pancreatic cells in their offspring, but there is no effect of 1,25(OH)2D on residual β-cell function and insulin requirements in adults [188].
Systematic reviews and meta-analyses indicate that maintaining adequate vitamin D status in the first years of life reduces the risk of diabetes [189,190,191,192]. Hypovitaminosis D is more common in people with T1DM, and the TEDDY study reported that a higher infant concentration of 25(OH)D is associated with lower islet autoimmunity [193]. Similarly, a Finnish birth cohort study suggested that sufficient vitamin D supplementation could mitigate T1DM risk [194].
A substudy of EURODIAB demonstrated that lower serum vitamin D levels were linked to an increased risk of developing T1DM before age 15 (OR 0.63) [195]. A substudy of EURODIAB demonstrated that lower serum vitamin D levels were linked to an increased risk of developing T1DM before age 15 (OR 0.63) [196]. Human studies have also identified associations between VDR polymorphisms and T1DM risk, as well as β-cell function. Although 25(OH)D is the primary circulating form, pancreatic β-cells can convert it to 1,25(OH)2D [135]. Elevated 25(OH)D levels may serve as a substrate for local 1,25(OH)2D synthesis in β-cells, while circulating 1,25(OH)2D could exert autocrine and paracrine effects. However, vitamin D is unlikely to be beneficial after clinical diagnosis due to established β-cell damage [197]. A meta-analysis revealed that vitamin D supplementation in T1DM patients reduced daily insulin requirements and improved fasting C-peptide (FCP), stimulated C-peptide (SCP), and HbA1c levels [198]. While some studies disagree [199], numerous observational studies support a strong association between vitamin D deficiency and T1DM [14,200].
Type 2 Diabetes Mellitus (T2DM) is a chronic metabolic disorder characterized by inadequate insulin production and, consequently, hyperglycemia.
Vitamin D deficiency or insufficiency is implicated in both macrovascular and microvascular complications of T2DM through multiple mechanisms. Macrovascular complications, such as endothelial dysfunction, arterial stiffness [201,202], peripheral arterial disease, and carotid plaque formation [203], are exacerbated by hypovitaminosis D. Microvascular complications, including diabetic retinopathy, neuropathy, and nephropathy, are also influenced by vitamin D status. For instance, vitamin D deficiency may worsen retinopathy severity by affecting angiogenesis and immune cell activity [204] and impair nociceptor function in neuropathy [205]. In addition, VDR is also expressed in highly insulin-sensitive tissues (pancreas, adipose tissue, and muscle) [206], and vitamin D works like an epigenetic factor mediating the transcription level and enhancing insulin sensitivity [180].
In line with recent studies, vitamin D supplementation is implicated in plasma HbA1c reduction, suggesting that vitamin D can contribute to reducing the development of diabetic complications [196]. Additionally, supplementation has been shown to improve β-cell function [207] and insulin sensitivity [206,208,209,210], particularly in high-risk individuals.
In pancreatic tissue, vitamin D protects β-cell function by the activation of the VDR expressed in β-cells, reducing local inflammation and increasing insulin secretion [211,212,213]. Vitamin D may play a role in the transcriptional activation of a gene contained in the human insulin receptor and in regulating the opening and closure of calcium channels, which regulates insulin secretion, mediating the calcium flux in β-cells, and interacting with receptors (VDR and 1,25D3-MARRS). Therefore, vitamin D deficiency, causing an alteration in calcium flux, could interfere with normal insulin secretion [214,215]. Increasing circulating vitamin D concentration could affect tissue energy and metabolism, improving systemic insulin sensitivity. The skeletal muscle is crucial in insulin sensitivity, involving 70–90% of total glucose disposal during the postprandial period [216,217,218]. Thus, vitamin D supplementation might improve skeletal muscle glucose handling and, as a consequence, insulin sensitivity [219]. Vitamin D also regulates the adipose tissue, and hypovitaminosis may play a role in obesity and fat mass due to the restoration of vitamin D, a fat-soluble vitamin, in the adipose tissue [220]. Hypovitaminosis also seems to increase microvascular complications such as diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, and diabetic foot ulcers [221], and a meta-analysis demonstrated that increased circulating vitamin D levels protect the kidney from injury and ameliorate proteinuria in T2DM patients [197].

6. Vitamin D and Infectious Diseases

The immune system’s defense against pathogens involves a coordinated response between innate and adaptive immunity [198]. The innate immune system is activated to neutralize the pathogen when it encounters an infectious agent. Subsequently, the adaptive immune response is initiated, typically taking 5–7 days to eliminate remaining pathogens. If the adaptive response fails, the innate system intensifies its activity, releasing pro-inflammatory cytokines [199,200]. Elderly and chronically ill individuals often exhibit an enhanced innate response, potentially due to immuno-inflammatory dysregulation associated with cellular aging, increasing their susceptibility to cytokine storms [201]. The NLRP3 inflammasome, a critical component of the innate immune system, promotes the production of IL-1β and IL-18, which stimulate downstream cytokines such as IL-6 [137,202]. Its activation is implicated in COVID-19 pathogenesis and cytokine storms, with associated markers such as CRP and LDH indicating inflammation [203]. Oxidative stress, characterized by excessive reactive oxygen species (ROS) and impaired detoxification, is a known trigger for NLRP3 activation, resulting in decreased immune recognition and ineffective pathogen clearance [204]. Vitamin D modulates inflammasome activation. While it inhibits NLRP3, it promotes NLRC4 activation, which is involved in bacterial intracellular immunity [205]. Studies have shown a link between adequate vitamin D levels and reduced inflammatory markers such as IL-6 and CRP [222,223,224]. Moreover, calcitriol has demonstrated protective effects against upper respiratory infections, including influenza [225,226]. Vitamin D also interacts with the Renin-Angiotensin-Aldosterone System (RAAS), which regulates immune responses, blood pressure, and metabolism. Angiotensin II (Ang II), a primary effector, can promote inflammation and oxidative stress via the AT1 receptor pathway. Excessive RAAS activation reduces ACE2 activity, contributing to acute lung injury and ARDS [227,228,229,230,231,232]. SARS-CoV-2 binds ACE2, impeding Ang II conversion to its anti-inflammatory counterpart Ang (1–7), exacerbating inflammatory damage [233,234,235]. Vitamin D, through its active form 1,25(OH)2D, downregulates RAAS by suppressing renin and enhancing ACE2 expression, mitigating Ang II-induced damage [236,237,238]. This action reduces oxidative stress and inflammation, potentially alleviating lung injury, as shown in LPS-induced models [237,239]. Chronic deficiency, however, may dysregulate RAAS, promoting lung fibrosis [240]. Adequate vitamin D increases ACE2 and Ang (1–7) levels, supporting vascular and immune stability [241,242,243]. By tightening epithelial junctions and reducing pro-inflammatory cytokines, vitamin D deficiency is a key risk factor for severe COVID-19 outcomes [244,245,246,247]. Vitamin D also supports antimicrobial defenses through the regulation of autophagy and T-cell differentiation. It stabilizes VDR proteins, enhancing immune signaling and reducing proteasomal degradation [248,249]. Deficiency correlates with increased susceptibility to respiratory pathogens and elevated inflammatory cytokines such as IL-6 and IL-8, with NF-κB inhibition observed upon supplementation [250,251,252]. While evidence suggests vitamin D reduces sepsis-related inflammation, clinical findings are mixed. Some trials indicate a reduction in RTI incidence and antibiotic use, especially with lower daily doses (≤1000 IU), though effects on asthma or COPD exacerbations remain inconclusive [253,254,255,256,257]. Concerning COVID-19, meta-analyses of randomized controlled trials (RCTs) do not show consistent results. While vitamin D supplementation did not significantly reduce mortality, it was associated with reduced ICU admissions and ventilator use. Observational studies link low 25(OH)D levels to increased COVID-19 risk, severity, and mortality [258,259]. In VAP patients, supplementation improved serum vitamin D and reduced procalcitonin, although no significant effect on SOFA scores was observed—likely due to limited sample size and duration [260]. Another RCT in healthcare workers found that high-dose vitamin D, an oral bolus of 100,000 IU with 10,000 IU/week of vitamin D, rapidly raised serum levels and may confer immunomodulatory benefits, although the trial ended prematurely [261].

Vitamin D and Acquired Immunodeficiency Syndrome (AIDS)

Recent studies have shown the role of vitamin D in regulating host-bacteria interactions, and its supplementation may reduce inflammation and HIV complications [41]. Vitamin D supplementation stimulating the expression of cAMP improves antibacterial immunity in monocyte cultures and HIV plasma [44] and is associated with reduced bacterial DNA translocation in HIV/HCV-coinfected patients. Low vitamin D levels are linked to high plasma HIV viral load, decreased CD4+ T-cells [262,263], faster-progressing AIDS, and lower survival in HIV patients [264,265]. High concentrations of vitamin D can boost autophagy activity, suppressing HIV replication in white cells [266].
Vitamin D deficiency may play a role in the development of HIV and could contribute to the progression of the infection by weakening the immune system. Low vitamin D levels can increase the risk of inflammation, immune system activation, and non-AIDS-related complications in people with HIV.
A potential benefit of vitamin D supplementation may be particularly relevant for children and adolescents with HIV. Low levels of vitamin D reduce the immune system’s ability to fight off infections such as Mycobacterium tuberculosis, the main cause of AIDS deaths in many regions worldwide. Some authors defined vitamin D supplementation in HIV-infected children and adolescents as safe and effective. They found that a dose of 100,000 IU every 2 months, together with 1 g/day of calcium, increased serum vitamin D concentrations significantly [267].
Vitamin D is linked to a higher risk of HIV-related health problems and death, so taking extra vitamin D might stop HIV from replicating, improve tuberculosis (TB) inflammation, and reduce the progression of HIV-TB co-infection. Some authors investigated the role of vitamin D in their meta-analysis, showing that HIV-TB patients had a higher prevalence of vitamin D deficiency than HIV patients, but supplementation had no effect on CD4 count or viral load [268]. Bone health is also compromised in people with HIV infection, and vitamin D supplementation may have beneficial effects on bone health. A recent meta-analysis found that vitamin D supplementation can improve serum 25(OH)D in HIV-infected patients; BMD and PTH need further investigation on a larger scale [269]. The current evidence highlights the relationship between vitamin D status and disease progression, demonstrating both potential promise and significant unanswered questions. While observational data consistently associate deficiency with worse outcomes, intervention studies reveal more nuanced results. These limitations suggest that while vitamin D assessment and repletion may be reasonable in HIV care, its role as a disease-modifying intervention requires substantially more evidence before widespread clinical implementation can be recommended.

7. Vitamin D and Multiple Sclerosis

Multiple sclerosis (MS) is a chronic autoimmune disorder affecting over 2 million people, predominantly young adults. It involves inflammation and demyelination in the central nervous system, impacting myelin, oligodendrocytes, axons, and neurons [270]. Clinically, MS manifests with paresthesias, ataxia, cognitive deficits, and impairments in vision and mobility [271]. While its etiology remains unclear, MS is associated with over 200 genetic loci [272] and environmental risk factors, notably Epstein-Barr virus infection, vitamin D insufficiency, and smoking [273]. Although the optimal serum 25(OH)D level is debated [269], levels above 40 ng/mL are considered protective in MS [274]. Sun exposure during childhood and its interaction with VDR gene variants may influence MS risk [275,276]. Polymorphisms in the VDR gene have been linked to MS, although findings remain inconsistent [277]. The FokI variant, which alters VDR protein structure, is associated with higher transcriptional activity [278,279], while other polymorphisms may affect RNA stability and VDR binding, with potential implications for immune regulation [280,281,282]. These genetic variations may increase MS susceptibility, particularly under conditions of vitamin D deficiency [283,284]. Geographical latitude and vitamin D levels also appear to correlate with MS prevalence [273]. Low serum 25(OH)D is associated with increased MS risk, disease activity, and progression [285,286,287]. Patients with MS tend to have lower vitamin D levels compared to the general population [288], and some studies link higher 25(OH)D levels with better disability outcomes [289]. Evidence on the efficacy of vitamin D supplementation in MS is mixed. A meta-analysis of 12 studies (950 patients) examined outcomes such as annualized relapse rate (ARR), Expanded Disability Status Scale (EDSS), and MRI lesion activity. Results showed a modest increase in ARR with high-dose vitamin D (mean difference 0.15; 95% CI 0.01–0.30) and a non-significant trend toward EDSS improvement (mean difference −0.12; 95% CI −0.78 to 0.01) [290]. However, high-dose supplementation did not outperform low-dose, and findings did not consistently support clinical benefit, highlighting the need for further placebo-controlled trials. Another meta-analysis of 12 RCTs (933 participants) found no significant effects of vitamin D (mostly D3) on ARR, EDSS, or gadolinium-enhancing lesions over 52 weeks [287]. Outcomes related to relapse timing, hospitalization, cognition, and psychological symptoms were not reported, limiting conclusions. An additional review of 10 RCTs on vitamin D (10 to 98,000 IU/day) found mixed results. While most trials showed no clinical benefit, some reported reduced IL-17 and increased IL-10 and TGF-β cytokines, along with improvement in active lesions and EDSS in patients with low baseline vitamin D [291,292]. Importantly, no adverse effects were reported. A study on pregnant women with MS and low 25(OH)D levels (<20 ng/mL) found that weekly supplementation with 50,000 IU of vitamin D3 improved serum levels (mean 33.7 ng/mL vs. 14.6 ng/mL, p < 0.05) and was associated with fewer relapses during pregnancy and up to six months postpartum. EDSS remained stable in the intervention group but worsened in the control group (p < 0.07) [293]. No significant adverse events were noted. Lastly, the D-Lay MS trial [294], a parallel, double-blind, randomized, placebo-controlled clinical trial including 316 individuals aged 18 to 55 years, provides compelling evidence for vitamin D’s therapeutic potential in early MS. High-dose cholecalciferol (100,000 IU every 2 weeks) significantly reduced disease activity in patients with clinically isolated syndrome (CIS), as measured by relapse rates and MRI lesions. These findings align with vitamin D’s known immunoregulatory properties, supporting its role as a potential adjunct therapy in neuroinflammatory disorders, significantly reducing disease activity in CIS and early relapsing-remitting MS.
In conclusion, while vitamin D is implicated in MS risk and may have immunomodulatory effects, current evidence from clinical trials does not conclusively support its use to improve clinical outcomes. Further large-scale, well-controlled studies are warranted to clarify its role in MS management.

8. Conclusions

The growing body of evidence highlights the multifaceted role of vitamin D in modulating immune responses and its potential impact on immune-mediated diseases. In Table 1, we provide a detailed summary of the findings investigating the effectiveness of vitamin D supplementation, including specific dosage recommendations for the diverse conditions addressed in this review. While preclinical and observational studies consistently demonstrate immunomodulatory properties of vitamin D, such as promoting regulatory T cell development, inhibiting pro-inflammatory cytokines, and enhancing innate immunity, translating these findings into clinical practice remains complex. Critically, the association between vitamin D deficiency and increased susceptibility to autoimmune and infectious diseases, including MS, SLE, T1DM, HIV, and COVID-19, is compelling but not conclusive. Much of the current evidence is correlative, and interventional studies show heterogeneous results. This suggests that while vitamin D supplementation may offer therapeutic or preventive benefits in deficient individuals, it is unlikely to serve as a universal remedy. Furthermore, variability in study design, dosing regimens, baseline vitamin D status, genetic polymorphisms affecting vitamin D metabolism, and disease heterogeneity complicate the interpretation of results. A one-size-fits-all approach is, therefore, inappropriate. Future research should prioritize large-scale randomized controlled trials with standardized protocols and stratified analyses to clarify the contexts in which vitamin D supplementation is most effective.
In conclusion, although maintaining adequate vitamin D status appears prudent, given its pleiotropic biological roles, current evidence does not support widespread therapeutic use in immune-mediated diseases. Further studies are necessary to provide more sophisticated, personalized approaches that go beyond blanket supplementation strategies to target specific pathological mechanisms in defined patient subgroups. Vitamin D remains an intriguing immunomodulator, but its clinical utilization requires much more rigorous investigation.
Table 1. Synthesis of different clinical studies on the effectiveness of various dosages of vitamin D administration in pathologic conditions analyzed.
Table 1. Synthesis of different clinical studies on the effectiveness of various dosages of vitamin D administration in pathologic conditions analyzed.
AuthorDesignDurationParticipants
(CA/CO)
Dose of Vitamin DResults
VITAMIN D AND ASTHMA
Majak et al.
[295]
RCT6 months 48 children (aged 5 to 18 years) with new diagnoses of asthma (24/24)Participants were randomly assigned to:
- Budesonide 800 mg/day administered as a dry powder and vitamin D placebo
or
- Budesonide 800 mg/day administered as a dry powder and 500 IU vitamin D cholecalciferol
The vitamin D group showed a significantly lower number of children who experienced asthma exacerbation and a lower number of children with a decrease of 25(OH)D than the steroid group (p = 0.029), so vitamin D supplementation prevented the reduction of serum concentrations of 25(OH)D and reduced the risk of asthma exacerbation triggered by acute RTIs.
Nanzer et al.
[296]
RCT4 weeks24 patients (aged 18 to 75 years) with glucocorticoid-resistant asthma (13/11)In the screening period, patients received a 2-week course of oral prednisolone; after a 4-week washout period, patients were randomly divided into two groups that received calcitriol 0.25 μg soft capsules
or placebo twice daily for 4 weeks with a second course of oral prednisolone identical to the first repeated during the final 2 weeks.
The calcitriol group showed a modest but significant improvement in absolute and predicted FEV1 compared to the placebo group (p = 0.03), so 1,25(OH)2D supplementation improved the clinical response to oral steroids.
Castro et al.
[90]
RCT28 weeks408 adults (aged 18 years or older) with asthma and a serum 25(OH)D level of less than 30 ng/mL (201/207)Participants were randomly assigned to either a placebo or a high-dose of vitamin D (100,000 IU once), followed by 4000 IU/day for 28 weeks added to inhaled ciclesonide (320 µg/d; 2 puffs twice/day) and levalbuterol; subsequently, inhaled corticosteroids were tapered by 50% if the participant’s asthma symptoms were controlled.Treatment with vitamin D did not alter the rate of first treatment failure or exacerbation compared with the placebo group; however, among secondary outcomes, the only statistically significant one was a small difference in the overall dose of ciclesonide required to maintain asthma control in the vitamin D group compared to the placebo group (111.3 µg/d [95% CI, 102.2–120.4 µg/d] in the vitamin D group vs. 126.2 µg/d [95% CI, 117.2–135.3 µg/d] in the placebo group; difference of 14.9 µg/d [95% CI, 2.1–27.7 µg/d]).
Mahboub et al.
[88]
RCT8 weeks54 adults (aged 18 to 65 years) with moderate-to-severe asthma and 25(OH)D levels less than 20 ng/mL (34/20)Participants were randomly assigned to:
- The experimental group received 50,000 IU of vitamin D orally
weekly
- The placebo group received the placebo
There was a significant increase in GR-α expression in the experimental group compared to the placebo control (p < 0.05), while no change in GR-β expression was observed; consequently, the GR-α/GR-β ratio increased significantly (p < 0.05).
Wang Q. et al.
[78]
MT (12 RCTs)From 6 weeks to 6 months1871 children with asthma (898/973)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 200 IU/day to 60,000 IU/month)
- The placebo group received the placebo
The experimental group had a significantly lower recurrence rate than the placebo group (18.4% versus 35.9%, RR = 0.35, 95% CI = 0.35–0.79, p = 0.002).
Wang M. et al.
[297]
MT (14 RCTs)From 3 to 12 months1421 patients with asthma (711/710)Four studies compared vitamin D (from 500 IU/day to 50,000 IU/week) to placebo as a treatment individually, while other studies received vitamin D as an adjunct treatment (corticosteroids/SABA/LABA/Montelukast/conventional therapy not described).Vitamin D supplementation was associated with:
- a significant improvement of FEV1% in patients with vitamin D insufficiency and airflow limitation (baseline FEV1% < 80%) (MD: 8.30, 95% Cl (5.95, 10.64)
- a reduction in the rate of exacerbation compared with placebo (RR: 0.7395% Cl (0.58,0.92)).
Wang Y. et al.
[77]
MT (19 RCTs)From 6 weeks to 48 weeks2063 patients with asthma (1039/1024)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 100 IU/day to 60,000 IU/week)
- The placebo group received placebo
In the vitamin D supplementation group:
-Number of exacerbations was less, while there was no statistical difference (OR = 0.73, p = 0.06, I2 = 59%)
- The FEV1/FVC was significantly improved (OR = 4.33, p = 0.02, I2 = 99%)
- Length of hospital stay and mortality were not changed
- Levels of IL-5 and IgE were significantly decreased (OR = −9.18, p = 0.0004, I2 = 99%; OR = −100.85, p < 0.00001, I2 = 0%)
- Levels of IL-6 and IL-10 and eosinophil counts were not significantly different, while in subgroup analysis based on serum vitamin D, the IL-10 level of the vitamin D deficiency group was significantly increased (OR = 2.51, p < 0.00001, I2 = 32%).
Fedora et al.
[86]
MT (10 RCTs)From 6 weeks to 12 months1243 asthmatic patients from 0 to 18 years old (631/612)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 150 IU/day up to 4000 IU/day)
- The placebo group received placebo
In the vitamin D supplementation group there was:
- A significant reduction of asthma exacerbations (RR 0.62; 95% CI: 0.44, 0.87; p = 0.006) (primary outcome)
- A significant improvement in predicted percentage of FEV1 levels
Pojsupap et al.
[298]
MT (5 RCTs)From 4 to 52 weeks625 asthmatic patients (from 5 to 18 years old)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 500 IU/day up to 2000 IU/day)
- The placebo group received placebo or budesonide ICS
In vitamin D supplementation group, there was a statistically significant reduction in asthma exacerbation (RR 0.41, CI 0.27–0.63).
Jolliffe et al.
[89]
MT (7 RCTs)From 15 weeks to 1 year955 asthmatic patientsParticipants were randomly assigned to:
- The experimental group received vitamin D supplements (from 500 IU/day up to 2000 IU/day)
- The placebo group received placebo
Vitamin D supplementation reduced the rate of asthma exacerbation requiring treatment with systemic corticosteroids among all participants (adjusted incidence rate ratio [aIRR] 0.74, 95% CI 0.56–0.97; p = 0.03). There were no significant differences between vitamin D and placebo in the proportion of participants with at least one exacerbation or time to first exacerbation.
Niu et al.
[299]
MT (12 RCTs)From 9 weeks to 9 months1295 asthmatic patients (649/646)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 400 IU up to 100,000 IU)
- The placebo group received placebo
Vitamin D supplementation significantly reduced the number of asthma exacerbations, including the rate of exacerbations requiring systemic corticosteroid therapy and the rate of acute exacerbations requiring emergency department or hospital visits or both.
Riverin et al.
[300]
MT (8 RCTs)From 1 to 12 months573 asthmatic children aged 3 to 18 yearsParticipants were randomly assigned to:
- The experimental group received vitamin D supplements (from 1000 IU/week up to 60,000 IU/month)
- The placebo group received placebo, budesonide, fluticasone, or prednisone
In vitamin D supplementation group there were:
- a reduced risk of asthma exacerbations (RR 0.41, 95% CI 0.27 to 0.63, 3 studies, n = 378).
- no significant effect for asthma symptom scores and lung function.
Chen et al.
[292]
MT (12 RCTs)From 2 to 12 months1543 asthmatic patients (769/774)Participants were randomly assigned to:
- The experimental group received vitamin D supplements (from 1000 IU/week up to 60,000 IU/week)
- The placebo group received placebo
Vitamin D supplementation significantly reduced the risk of asthma exacerbation (pooled risk ratio (RR) 0.70, 95% confidence interval (CI), 0.59, 0.83; p < 0.05), but did not improve ACT score or lung function among patients with asthma treated with corticosteroids.
VITAMIN D AND ALLERGIC RHINITIS
Bhardwaj et al.
[94]
RCT2 years and 5 months87 patients (aged 16 to 60 years) with allergic rhinitis and serum vitamin D levels less than 20 ng/mLParticipants were randomly divided into two groups:
- Group A received Fluticasone 50 mcg nasal spray, two puffs BID for 4 weeks
- Group B received Fluticasone 50 mcg nasal spray, two puffs BID, and Oral vitamin D cholecalciferol 60,000 IU once a week for 4 weeks
The pre-treatment TNSS score in Group A was 12.5 ± 2.68, while the post-treatment score was 8.98 ± 1.009, with the difference in both scores of Group A as 3.52 (the t score was 117.387 and p < 0.000). The pre-treatment TNSS score in Group B was 11.64 ± 3.09 while the post-treatment score was 6.3 ± 1.45, with the difference in both scores of Group B as 5.34 (the t score was 131.1403 and p < 0.0001). The post-treatment RCAT in Group A and Group B was 19.72 ± 2.84 and 28.2 ± 1.53, respectively, with the difference between the two groups as 8.48 (t score as 135.27 and p < 0.05).
Guo
[95]
RCT5 years140 patients (aged 16 to 60 years) with moderate-to-severe allergic rhinitis (70/70)Participants were randomly divided into two groups:
- The control group received 200 µg mometasone nasal spray and two puffs of BID for 4 weeks.
- The experimental group received 200 µg mometasone nasal spray, two puffs BID, and two oral vitamin D capsules (400 IU vitamin D per capsule) for 4 weeks.
Total TNSS scores were significantly reduced compared with those before treatment in both groups for all four main symptoms assessed (p < 0.05), and the improvements with vitamin D supplementation were significantly greater than those of the control group in the symptoms assessed, except for sneezing (p < 0.05). RQLQ scores in both groups were significantly lower after the therapy than before the therapy in all aspects (p < 0.05). The improvements with vitamin D supplementation were significantly higher than those in the control group, except for eye symptoms, which were improved. Still, the difference was not statistically significant compared to the control (p < 0.05).
Bakhshaee et al.
[96]
RCT1 year80 patients (aged 18 to 40 years) with allergic rhinitis and vitamin D level of 10–20 ng/mL (40/40)Participants were randomly divided into two groups:
- The control group received a placebo plus cetirizine for eight weeks
- Intervention group received
vitamin D (weekly 50,000 IU) plus cetirizine for eight weeks
There was no significant difference between the two groups at the onset of the study and 4 weeks later regarding the mean scores of symptom severity (p = 0.073), whereas a significant difference was obtained between the two groups in terms of symptom severity at baseline and 8 weeks post-treatment (p = 0.007).
Jerzynska et al.
[97]
RCT5 months50 children (aged 5 to 12 years) with grass-related moderate-to-severe rhinoconjunctivitis; eight patients had concomitant asthma (25/25)Participants were randomly divided into two groups:
- The control group received SLIT with a placebo
- The experimental group received SLIT with vitamin D 1000 IU daily supplementation
The experimental group shows a reduction of nasal symptoms (p = 0.04), asthma symptoms (p = 0.001), and combined symptom-medication score (p = 0.001); there was no significant difference between groups in medication and ocular scores.
VITAMIN D AND ATOPIC DERMATITIS (AD)
Kim et al.
[46]
MT (11 studies: 7 observational trials and 4 RCTs)In observational trials: N/A; in clinical trials: from 60 days to 2 monthsIn observation trials, 1643 children and adults with AD or healthy subjects (986/657); in clinical trials, 194 children and adults with AD (104/90)In observational trials: N/A; in clinical trials: from cholecalciferol or ergocalciferol 1000 IU daily to cholecalciferol 1600 IU daily vs. placeboIn observational trials, the AD group had lower serum 25(OH)D levels compared to the healthy group (statistically significant in the pediatric AD patients, not statistically significant in the adult AD group); in clinical trials, the SCORAD index or EASI score decreased significantly (mean difference: −5.85 points) in the vitamin D group.
Hattangdi-Haridas et al.
[103]
MT (16 studies: 11 observational studies and five interventional studies, including RCT, not RCT, clinical intervention, and audit trials)In observational trials: N/A; in interventional trials: from 60 days to 3 monthsIn observational trials: adults and children or adults only or children only with AD or healthy subjects (1221/982); in interventional trials: adults and children with AD (165/170)In observational trials: N/A; in interventional trials: cholecalciferol 1000 IU/daily or 1600 IU/daily or 2000 IU/daily In observational trials, the AD group had a statistically significantly lower vitamin D concentration compared to the healthy group (p = 0.02); in interventional trials, for the repeated measures interventions, the SCORAD index decreased significantly (mean difference: 21 points; p < 0.0001) in vitamin D group; for the randomized control trials, the SCORAD index reduced significantly (mean difference: 11 points; p < 0.0001) in the vitamin D group.
Huang et al.
[105]
SR (21 articles: 4 RCTs, 5 cohort studies, 6 case-control studies, 6cCross-sectional studies; vitamin D supplementation was investigated in 6 studies: 4 RCTs and 2 cohort studies)From 4 to 12.86 weeksA total of 354 patients with an average age of 6.79 years; participants involved with an RCT are 227 (114/113). Vitamin D doses ranged from 1000 to 2000 IU/daily; cholecalciferol was used in 2 articles, while 2 studies were supplemented with ergocalciferol, and the remaining 2 studies did not specify what type of vitamin D was used.Significant improvement of AD when supplemented with vitamin D was found in 67% (4/6) of studies.
Nielsen et al.
[107]
MT (12 RCTs)From 4 to 12 weeks686 patients with AD (347/339)From 1000 to 5000 IU/daily and 8000 to 60,000 IU/weekly; cholecalciferol was predominantly used, except for one study that utilized ergocalciferol.Significant improvement in AD symptoms following vitamin D supplementation was found in 27% (3/11) of studies in the intervention group compared with the control group.
VITAMIN D AND RHEUMATOID ARTHRITIS (RA)
Andjelkovic et al.
[301]
RCT3 months 19 patients treated with standard DMARD therapy for acute RA Oral alfacalcidol
(2 micrograms/day)
Positive effect on disease activity in 89% of the patients:
- 45% or nine patients with complete remission
- 44% or eight patients with a satisfactory effect
- 11% no improvement, but no new symptoms occurred
Scharla et al.
[121]
RCT4 weeks 71 inpatients with RA and osteopenia Patients were randomly assigned to:
- Group 1 treated with 1000 IU plain vitamin D + 500 mg calcium daily
- Group 2 treated with 1 µg of alfacalcidol + 500 mg calcium daily
In patients with RA, alfacalcidol, but not plain vitamin D, improved calcium and bone metabolism, modulated inflammation, improved muscular function, and decreased pain symptoms.
Forsblad et al.
[122]
RCT2 years 88 women with RA aged between 45 and 65 years All patients were treated with a daily dose of 400 IU vitamin D;
- 41 women were randomized to receive HRT (oestradiol and noretisterone acetate), vitamin D and calcium
- 47 to the control group receiving vitamin D and calcium alone
There are no differences between the HRT and control groups concerning
the concentration of Ig in serum and to B- and T-cell-dependent recall antigen reactions. Thus, HRT did not
stimulate nor suppress these immunological responses.
VITAMIN D AND PSORIASIS
Pinter et al.
[146]
MT (2 Phase 3 RCTs)8 weeks1271 patients: 783 male and 488 female subjects above the age of 18 years with mild-to-moderate psoriasis according to PGA and with a treatment area of 2–30% of the body (trunk and/or limbs), with a PASI as a baseline of at least of 2 or 3Subjects were randomly allocated to one of three arms:
- 551 for CAL/BDP PAD-cream;
- 178 for cream vehicle;
- 542 for active comparator [a marketed CAL/BDP gel/topical suspension.
The number of subjects who completed the trial was, 526 (for CAL/BDP PAD cream), 149 (for cream vehicle), and 513 (for active comparator) respectively.
CAL/BDP PAD cream demonstrated superiority after 8 weeks compared to CAL/BDP TS for all efficacy endpoints, including PGA treatment success, mPASI, and PASI75. Superiority was also demonstrated for patient-reported DLQI and PTCS.
Heim et al.
[147]
RCT4 weeks30 patients (18 male and 12 female subjects) with mild psoriasis defined by a PASI score below 10 and at least two symmetric lesions of 2 cm2 affecting the knees or the elbows. Subjects were randomly allocated into three groups of ten. All patients applied calcipotriol/betamethasone foam to their elbows on one side of the body. Three groups of 10 patients applied either placebo foam (emollient), betamethasone foam or clobetasol propionate ointment to elbows on the opposite side of their body. The side of the treatment allocation was randomly assigned using central randomization. All treatments, including placebo, provided a significant decrease in tPASI. The treatment with calcipotriol/betamethasone foam was the most effective in decreasing CD8+ cell infiltrate in both the dermis and epidermis after 4 weeks of treatment. They noticed that CD8+ IFNγ+ T cells decreased significantly after both betamethasone and combined calcipotriol/betamethasone treatment but also that calcipotriol/betamethasone
significantly reduced the number of MPO+ neutrophils which were predominantly IL-17+
Venegas-Iribarren et al.
[150]
MT (eight SRs including 26 studies overall, of which 22 were RCTs)N/A4238 adult patients (15 to 90 years) with plaque psoriasis in the trunk and limbs, not including the scalp and with low, moderate, or severe disease.All trials used topical corticosteroids as intervention (fluocinonide 0.05% twice a day, betamethasone dipropionate 0.05% once or twice a day, betamethasone 17-valerate 0.1% once or twice a day, desoxymethasone 0.25% twice a day, clobetasol propionate 0.05% twice a day, and diflorasone diacetate 0.05% twice a day). As a comparison, topical treatment with calcipotriol 50 mcg/g once or twice a day, calcitriol 3 mcg/g twice a day, and tacalcitol
4 mcg/g once a day.
There might be little or no difference in PASI, IAGI, and PAGI scores between topical corticosteroids and topical vitamin D analogues. The certainty of the evidence is low.
Topical corticosteroids lead to fewer local adverse events (skin irritation) and fewer withdrawals than topical vitamin D analogues. With a high certainty of the evidence.
No studies were found that evaluated the impact of topical corticosteroids and topical vitamin D analogues in cutaneous atrophy.
VITAMIN D AND SYSTEMIC LUPUS ERYTHEMATOSUS (SLE)
Andreoli et al.
[163]
RCT 24 months34 patients
(18/16)
Participants were randomly assigned to:
- Group 1:
Standard Regimen (SR) = 25,000 IU/month- Group 2:
Intensive Regimen (IR) = 300,000 IU bolus + 50,000 IU/month for one year and then switched to the other regimen in the second year.
After 12 months, values above 30 ng/mL were found in 75% of Intensive Regimen patients, while only 28% were in the Standard Regimen.
No significant differences in disease activity and SLE serology were found between SR and IR at any time.
No changes in the mineral metabolism were observed.
Lima et al.
[167]
RCT 24 weeks 40 patients with juvenile-onset SLE
(20/20)
Participants were randomly assigned to:
- Group 1: cholecalciferol 50,000 IU/week
- Group 2: placebo
SLEDAI score and ECLAM significantly improved (p = 0.011, p = 0.006)
Fatigue score and K-FSS significantly improved (p = 0.012, p = 0.008)
No significant improvements in cutaneous and articular manifestations and proteinuria (p = 0.66, p = 0.18, p = 0.32).
Karimzadeh et al.
[164]
RCT3 months90 patients (with levels of 25(OH)D < 30 ng/mL)
(45/45)
Participants were randomly assigned to:
- Interventional group patients: vitamin D3
soft gel capsules (50,000 IU/weekly for 12 weeks and then 50,000 IU/monthly for 3 months)
- Placebo group: placebo
The mean of vitamin D levels significantly increased in the interventional group (p < 0.001)
Not significantly different in SLEDAI score before and after vitamin D administration
(p = 0.39).
Singgih Wahono et al.
[165]
RCT14 months40 patientsParticipants were randomly assigned to:
- Group I: 3 × cholecalciferol 400 IU and 3 × 1 tablet placebo
- Group II: 3 × cholecalciferol 400 IU and curcumin (Curcuma xanthorrhiza) 3 × 20 mg
Supplementation of cholecalciferol 1200 IU either with placebo (p = 0.000)
or with curcumin (p = 0.003)
significantly increased serum 25(OH)D levels.
Serum vitamin D levels differed significantly, with higher levels in group I (p = 0.047).
The delta of vitamin D levels (the difference between vitamin D levels after supplementation and before supplementation) did not differ significantly between the two groups
(p = 0.166).
SLEDAI scores significantly decreased in both groups (Group I: p = 0.001); Group II: p = 0.003)
Serum IL-6 levels significantly decreased in both groups
(Group I: p = 0.001); Group II: p = 0.013).
Pakchotanon et al.
[166]
RCT24 weeks104 patients
(52/52)
Participants were randomly assigned to:
- Group A: ergocalciferol 100,000 IU weekly for 4 weeks, followed by ergocalciferol 40,000 IU weekly for 20 weeks,
- Group B: placebo
The mean standard deviation of serum levels of vitamin D in group A was significantly higher than in group B (p < 0.001).
There is no difference between groups A and B concerning SLEDAI-2K, flare events, ESR, CRP, and dosage of immunosuppressive drugs.
VITAMIN D AND AUTOIMMUNE THYROID DISEASES
Zhang et al.
[302]
MT (8 RCTs)From 4 to 24 weeks652 patients with HT (332/320)Participants were randomly assigned to:
- Vitamin D (in different formulations and dosages) ± LT4
- LT4/Sunshine and diet/placebo
Vitamin D supplementation:
- Significantly decrease TPO-Ab levels in the subgroups in whom the treatment duration > 3 months (p = 0.009)
and
in the subgroups treated with vitamin D3 (p = 0.006)
- Was effective in reducing the Tg-Ab levels (p = 0.009)
(due to limited data, no subgroup analysis and meta- regression analysis were performed).
Wang S. et al.
[177]
MT (6 RCTs)From 1 to 6 months344 patients with AIT (178/166)Participants were randomly assigned to:
- Vitamin D (in different formulations and dosages) ± LT4/elemental calcium 500 mg/day
- LT4/elemental calcium 500 mg/day/placebo/no treatment
Vitamin D supplementation significantly decrease TPO-Ab levels at six months
(3 RCT, p < 0.01)
and
significantly decrease Tg-Ab levels (4 RCT, p = 0.033; even if a significant heterogeneity was found)
No significant effect on changes in TSH, FT3, or FT4 after vitamin D supplementation.
Tang et al.
[178]
MT (12 RCTs)From 12 to 24 weeks862 patients with HT (429/423)Participants were randomly assigned to:
- Vitamin D (in different formulations and dosages) ± LT4
- LT4/placebo/no treatment
Vitamin D supplementation:
- Significantly decrease TPO-Ab levels
(p = 0.03)
- Significantly increase fT3 and fT4 levels (p < 0.001)
at 12 weeks
- Significantly decrease Tg-Ab levels
(p < 0.001; even if a significant heterogeneity was found)
-Significantly decreases TSH levels
VITAMIN D AND TYPE 1 AND TYPE 2 DIABETES MELLITUS
Gregoriou et al. [303]MT (7 RCTs)From 4 to 52 weeks287 patients with T1DMParticipants were randomly assigned to:
- Calcitriol 0.25 μg per day or on alternate days plus insulin
- Alphacalcidol 0.5 μg daily plus insulin cholecalciferol 2000 IU per day plus insulin for 18 mo
- Cholecalciferol 70 IU/kg body weight/day plus insulin
Vitamin D supplementation in the form of alphacalcidol and chole-calciferol appears to be beneficial in DID, FCP, SCP, and HbA1c.
Najjar et al.
[304]
MT (10 studies: 3 cohort;
5 case–control; 2 matched case–control)
N/A39,884 patients with T1DM (16,370/23,514)N/ANo large effect of a genetically determined reduction in 25(OH)D concentrations by selected polymorphisms on T1DM risk.
Nascimento et al. 2022
[305]
RS (10 studies)From 6 to 52 weeksChildren and adolescents (0–19 years) with T1DM- Cholecalciferol with dosages ranging from 1000 to 160,000 IU.
- Just one study used vitamin D in the form of alfacalcidol at a dosage of 0.25 to 0.5 μg/day
This study did not provide evidence to support the effect of vitamin D supplementation on glycemic control to aid in the treatment of T1DM.
Yu et al.
[306]
RS (13 studies: 9 RCTs;
2 open-label case–control, 1 open label, and 1 cohort
From 4 to 12 weeks527 patients with T1DMThe following therapeutic regimens were used:
1.25 D 0.25 μg twice daily; 25D 2000 IUdaily; 25D to achieve serum
25 D > 125 nmol/L; alfacalcidol 0.25 μg BD 25 D; 60,000 IU monthly;
Ergocalciferol (D2) 2 m of 50,000 IU/w; 25D 2000 IU/d; 25D 3000 IU/d;
Calciferol 2000 IU/d + etanercept + GAD-alum
The maintenance of optimal circulating 25 D levels may reduce the risk of T1DM, and that may have potential for benefits in delaying the development of absolute or near-absolute C-peptide deficiency.
Hu et al.
[196]
MT
(19 RCTs)
From 4 to 24 weeks1374 patients with T2DM (747/627)- Up to 50,000 UI/weekly vitamin D3
- 300,000 UI single injection of vitamin D3
Significant reduction in HbA1c, IR (marked by a decrease in HOMA-IR)
and insulin levels in the short-term vitamin D supplementation group.
Krul-Poel et al.
[219]
MT
(23 RCTs)
From 4 to 52 weeks1797 patients with T2DM: for the effect on HbA1c, 1475 patients (755/720); for the effect on FBG 1180 patients (608/572)- From 1000 IU/day vitamin D3 to 45,000 IU/week
- Vitamina D3 or 1200 IU/day
- Vitamin D3 for 2 weeks followed by 5600 IU/day for 10 weeks or from 100,000 to 300,000 IU
- Vitamin D3 single dose
Significant effect on FBG in a subgroup of studies (n = 4); no significant effect on change in HbA1c.
VITAMIN D AND INFECTIOUS DISEASES
Bergman et al.
[254]
RCT12 months140 patients with antibody deficiency disorder and
increased susceptibility to RTIs (70/70)
Participants were randomly assigned to:
- Group 1:
cholecalciferol 4000 UI daily
for 12 months
- Group 2: placebo
The overall infectious score was significantly
reduced for patients allocated to the vitamin D group compared with the placebo group.
Lehouck et al.
[256]
RCT12 months182 patients with moderate to very severe COPD and a history of recent exacerbations (91/91)Participants were randomly assigned to:
- Group 1:
100,000 IU of vitamin D supplementation every 4 weeks for 1 year
- Group 2: placebo
High-dose vitamin D supplementation in a sample of patients with COPD did not reduce the incidence of exacerbations.
In participants with severe vitamin D deficiency at baseline, supplementation may reduce exacerbations.
(rate ratio, 0.57 [CI, 0.33 to 0.98]; p = 0.042).
Slow et al.
[255]
RCT3 years135 patients were admitted to the hospital with CAP
(mean baseline plasma 25(OH)D concentration was 48.7  ±  21.6 (SD) nmol/L) (68/67)
Participants were randomly assigned to:
- Group 1:
An oral single dose of vitamin D 20,000 UI
- Group 2: placebo
Vitamin D supplementation had a significantly greater effect on those with low baseline vitamin D (<50 nmol/L) in improving the radiological resolution of pneumonia.
Miroliaee et al.
[260]
RCT10 months51 patients with VAP who were suffering from vitamin D deficiency (26/25)Participants were randomly assigned to:
- Group 1:
received 300,000 units of intramuscular vitamin D.
- Group 2: placebo
The levels of PCT were
significantly decreased (p = 0.001) in the treatment group (–0.02 ± 0.59 ng/mL) compared to that
of the placebo group (0.68 ± 1.03 ng/mL), no significant
changes in SOFA and CPIS scores
(p = 0.37 and p = 0.46,
respectively).
Arpadi et al.
[267]
RCT12 months56 children with HIV and
vitamin D levels insufficiency (29/27)
Participants were randomly assigned to:
- Group 1: HIV-infected children and adolescents who were aged 6 to 16 years receive vitamin D (100,000 IU bimonthly) and calcium
- Group 2: double placebo
No group differences were seen in the change in CD4 count or CD4% or viral load. The overall mean monthly serum 25(OH)D concentrations were higher in the group that received vitamin D.
Hosseini et al.
[261]
RCT16 weeks34 patients (healthcare workers as a primary prevention of SARS-CoV-2 infection) (19/15)Participants were randomly assigned to:
- Group 1: 100,000 IU vitamin D oral bolus at randomization followed by a weekly dose of 10,000 IU of vitamin D3
- Group 2: placebo
The groups which received vitamin D supplementation (95% CI) were in favor of supplementation; 77.8% of intervention, and 13.3% of control, patients were vitamin Dsufficient (OR:6.11, 95% CI:1.6, 22.9).
Anitua et al.
[257]
MT (65 RCTs)From 5 days to 3 years50,554 participantsAll trials used vitamin D as intervention as
dose ranging from 400 to 1000 IU/day
The incidence of RTIs in terms of count data (OR: 0.87; 95%CI [0.80–0.95]; p = 0.0028; I2 = 43%) and event rate (IRR: 0.81; 95%CI [0.70–0.95]; p = 0.010; I2 = 79%) was significantly reduced in the intervention group. However, no effect of vitamin D on duration or upper RTIs severity was observed.
Petrelli et al.
[259]
MT (43 observational trials)N/A612,601 patientsAll trials used an intervention group with vitamin D supplementation (from 1000 UI daily to
80,000/100,000 IU every 2–3 months or 80,000 IU within a few hours of the diagnosis of COVID-19).
Risk of COVID-19 infection was higher compared to those with replete values (OR = 1.26; 95% CI, 1.19–1.34; p < 0.01). Vitamin D deficiency was also associated with worse severity and higher mortality than in non-deficient patients (OR = 2.6; 95% CI, 1.84–3.67; p < 0.01 and OR = 1.22; 95% CI, 1.04–1.43; p < 0.01, respectively).
Meng et al.
[258]
MT (25 RCTs)From 7 days to 1 year8128 participantsPreventing effect group
- Group 1: 1949 participants received vitamin D supplementation, with daily dosages ranging from 800 IU to 5000 IU daily or weekly.
- Group 2: 3703 participants received a placebo.
Regarding the therapeutic effects
- Group 1: 1270 participants received vitamin D supplementation
- Group 2: 1206 participants did not receive vitamin D supplementation or received low dosage of vitamin D
The studies included in our analysis mainly administered a single-dose, high-dosage vitamin D supplementation (from 800 IU to 5000 IU) therapy for severe SARS-CoV-2 infection patients. Single-dose supplementation was found to reduce the rate of mechanical ventilation, while the multiple-dose therapy was found to reduce the rate of ICU admission and mortality.
VITAMIN D AND MULTIPLE SCLEROSIS (MS)
McLaughlin et al.
[290]
MT (12 interventional studies)6 to 22 months950 patientsStudies were divided into four groups because of heterogeneity in study design.
Three studies only included patients with serum 25(OH)D levels below 50, 75, or 100 nmol/L
The effective daily dosage of cholecalciferol (or equivalent ergocalciferol) used ranged from 2857–10,400 IU in the active or high-dose arms. The low-dose arms ranged from 800 to 1000 IU.
Non-significant trends in favor of vitamin D for all outcome measures: high-dose vitamin D has worse outomes in ARR (mean difference 0.15 [95% CI 0.01–0.30]) and non-significant trends of increased EDSS (mean difference −0.12 EDSS points [95% CI −0.78) to 0.01] and gadolinium-enhancing lesions for the higher-dose arms.
Jagannath et al.
[287]
MT (12 RCTs)From six months (24/26 weeks) to 12 months (48/52 weeks)933 participantsParticipants were enrolled in two groups: 464 were randomized to the vitamin D group, and 469 to the comparator group.Vitamin D has no effect on ARR: the mean in the intervention group was 0.05 lower (−0.17 lower to 0.07 higher); worsening of EDSS: the mean score in the intervention group was 0.25 lower (0.61 lower to 0.10 higher); or new MRI gadolinium-enhancing T1 lesions: in the intervention group was 0.02 higher (0.45 lower to 0.48 higher).
Berezowska et al.
[291]
MT (10 RCTs)From 2012 to 2018627 participantsAll studies evaluated the use of vitamin D supplementation (ranging from 10 to 98,000 IU), comparing it to a placebo or low-dose vitamin D. The duration of the intervention ranged from 12 to 96 weeks.One trial demonstrated that vitamin D supplementation resulted in fewer new T2 lesions (a mean of 0.5 compared to a mean of 1.1 in the placebo group).
Four of ten studies showed no significant differences between the vitamin D and control groups.
One trial found that the EDSS score increased significantly (p < 0.01) in a placebo group from a mean of 1.7 to 1.94, with no significant difference in scores at the end of the trial between intervention and control groups (p > 0.05).
Etemadifar et al. [293]RCT12 to 16 weeks of gestation till delivery15 participants Participants were randomly allocated into two groups:
- Group 1: 50,000 IU/week vitamin D
- Group 2: routine care from 12 to 16 weeks of gestation till delivery
Average serum 25(OH)D level at the end of the trial in vitamin D supplemented group was higher than routine care group (33.7 ng/mL vs. 14.6 ng/mL, p < 0.050). In the vitamin D group, the mean EDSS did not change 6 months after delivery (p > 0.050), whereas in the routine care group, the mean EDSS increased from 1.3 (0.4) to 1.7 (0.6) (p < 0.070).
Thouvenot et al. [294]RCTFrom July 2013 to December 2020 (final follow-up on January 2023)316 participantsPatients were randomized 1:1 to receive oral cholecalciferol 100,000 IU every 2 weeks for 24 months:
- Group 1:
163 participants
- Group 2 placebo: 153 participants
In group 1 vs. the placebo group:
MRI activity (89 patients [57.1%] vs. 96 patients [65.3%]; HR, 0.71 [95% CI, 0.53–0.95]; p = 0.02), new lesions (72 patients [46.2%] vs. 87 patients [59.2%]; HR, 0.61 [95% CI, 0.44–0.84]; p = 0.003), and contrast-enhancing lesions (29 patients [18.6%] vs. 50 patients [34.0%]; HR, 0.47 [95% CI, 0.30–0.75]; p = 0.001).
The vitamin D oral supplementation significantly reduces the disease activity in CIS and relapsing-remitting MS.
CA/CO: Case/Control; RCT: randomized controlled trial; IU: International Unit; p: p-value; RTIs: Respiratory Tract Infections; GR: glucocorticoid receptor; AD: atopic dermatitis; MT: meta-analysis; N/A: Not Applicable; FEV1: Forced Expiratory Volume in the first second; SABA: Short Acting Beta-adrenoceptor Agonists; LABA: Long Acting Beta-adrenoceptor Agonists; ICS: Inhaled CorticoSteroids; ACT: Asthma Control Test; BID: Bis In Die; TNSS: Total Nasal Symptom Score; RACT: Rhinitis Control Assessment Test; RQLQ: Rhinoconjunctivitis Quality of Life Questionnaire; SLIT: Sublingual Immunotherapy; HRT: hormone replacement therapy; mPASI: modified (excluding the head) Psoriasis Area and Severity Index; PGA: Physician’s global assessment; PASI75: at least 75% reduction in mPASI from Baseline; PTCS: Psoriasis Treatment Convenience Scale; SCORAD: Scoring of Atopic Dermatitis; EASI: Eczema Area and Severity; SR: Systematic Review; RA: rheumatoid arthritis; HRT: Hormon Replacement Therapy; CAL/BDP PAD: calcipotriol and betamethasone dipropionate cream based on PAD™ Technology; CAL/BDP TS: calcipotriol and betamethasone dipropionate topical suspension/gel; DLQI: Dermatology Life Quality Index; IAGI: Investigators Assessment of overall Global Improvement, PAGI: Patient Assessment of Global Improvement; SLE: systemic lupus erythematosus; SR: Standard Regimen; IR: Intensive Regimen SLEDAI-2K: SLE Disease Activity Index 2000; SLEDAI: SLE Disease Activity Index; ECLAM: European Consensus Lupus Activity Measurement; K-FSS: Krupp Fatigue Severity Scale; ESR: Erythrocyte Sedimentation Rate; CRP: C-Reactive Protein; HT: Hashimoto Thyroiditis; anti-TPO: anti-Thyroid PerOxidase; LT4: levothyroxine; AIT: autoImmune thyroiditis; DID: Daily Insulin Dose; FCP: Fasting C- Peptide; SCP: Stimulated C-Peptide; HbA1c: emoglobin A1c; T1DM: Type 1 Diabetes Mellitus; T2DM: Type 2 Diabetes Mellitus; IR: insulin resistance; HOMA-IR: Homeostatic Model Assessment of Insulin Resistance; FBG: Fasting Blood Glucose; RTIs: Respiratory Tract Infections; COPD: Chronic Obstructive Pulmonary Disease; CAP: Community-Acquired Pneumonia; VAP: Ventilator-associated Pneumonia; PCT: procalcitonin; SOFA: Sequential Organ Failure Assessment; CIPS: Critical Illness Polineuropathy; HIV: Human Immunodeficiency Virus; ICU: Intensive Care Unit; multiple sclerosis (MS); ARR: annualized relapse rate; EDSS: Expanded Disability Status Scale; MRI: Magnetic Resonance Imaging; CIS: clinically isolated syndrome.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dattola, A.; Silvestri, M.; Bennardo, L.; Passante, M.; Scali, E.; Patruno, C.; Nisticò, S.P. Role of Vitamins in Skin Health: A Systematic Review. Curr. Nutr. Rep. 2020, 9, 226–235. [Google Scholar] [CrossRef]
  2. Holick, M.F.; Mazzei, L.; García Menéndez, S.; Martín Giménez, V.M.; Al Anouti, F.; Manucha, W. Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases. Nutrients 2023, 15, 767. [Google Scholar] [CrossRef]
  3. Wöbke, T.K.; Sorg, B.L.; Steinhilber, D. Vitamin D in Inflammatory Diseases. Front. Physiol. 2014, 5. [Google Scholar] [CrossRef]
  4. Gil, Á.; Plaza-Diaz, J.; Mesa, M.D. Vitamin D: Classic and Novel Actions. Ann. Nutr. Metab. 2018, 72, 87–95. [Google Scholar] [CrossRef]
  5. Alswailmi, F.K.; Shah, S.I.A.; Nawaz, H.; Al-Mazaideh, G.M. Molecular Mechanisms of Vitamin D-Mediated Immunomodulation. Galen Med. J. 2021, 10, e2097. [Google Scholar] [CrossRef]
  6. Coussens, A.K. Immunomodulatory Actions of Vitamin D Metabolites and Their Potential Relevance to Human Lung Disease. Curr. Respir. Med. Rev. 2011, 7, 444–453. [Google Scholar] [CrossRef]
  7. Umar, M.; Sastry, K.S.; Chouchane, A.I. Role of Vitamin D Beyond the Skeletal Function: A Review of the Molecular and Clinical Studies. Int. J. Mol. Sci. 2018, 19, 1618. [Google Scholar] [CrossRef]
  8. Wimalawansa, S.J. Non-Musculoskeletal Benefits of Vitamin D. J. Steroid Biochem. Mol. Biol. 2018, 175, 60–81. [Google Scholar] [CrossRef]
  9. Maddaloni, E.; Cavallari, I.; Napoli, N.; Conte, C. Vitamin D and Diabetes Mellitus. In Frontiers of Hormone Research; Giustina, A., Bilezikian, J.P., Eds.; S. Karger AG: Basel, Switzerland, 2018; Volume 50, pp. 161–176. ISBN 978-3-318-06338-7. [Google Scholar]
  10. Mitri, J.; Muraru, M.D.; Pittas, A.G. Vitamin D and Type 2 Diabetes: A Systematic Review. Eur. J. Clin. Nutr. 2011, 65, 1005–1015. [Google Scholar] [CrossRef]
  11. Chen, X.; Zhou, M.; Yan, H.; Chen, J.; Wang, Y.; Mo, X. Association of Serum Total 25-Hydroxy-Vitamin D Concentration and Risk of All-Cause, Cardiovascular and Malignancies-Specific Mortality in Patients with Hyperlipidemia in the United States. Front. Nutr. 2022, 9, 971720. [Google Scholar] [CrossRef] [PubMed]
  12. Targher, G.; Bertolini, L.; Scala, L.; Cigolini, M.; Zenari, L.; Falezza, G.; Arcaro, G. Associations between Serum 25-Hydroxyvitamin D3 Concentrations and Liver Histology in Patients with Non-Alcoholic Fatty Liver Disease. Nutr. Metab. Cardiovasc. Dis. 2007, 17, 517–524. [Google Scholar] [CrossRef]
  13. Khademi, Z.; Hamedi-Shahraki, S.; Amirkhizi, F. Vitamin D Insufficiency Is Associated with Inflammation and Deregulation of Adipokines in Patients with Metabolic Syndrome. BMC Endocr. Disord. 2022, 22, 223. [Google Scholar] [CrossRef]
  14. Argano, C.; Mirarchi, L.; Amodeo, S.; Orlando, V.; Torres, A.; Corrao, S. The Role of Vitamin D and Its Molecular Bases in Insulin Resistance, Diabetes, Metabolic Syndrome, and Cardiovascular Disease: State of the Art. Int. J. Mol. Sci. 2023, 24, 15485. [Google Scholar] [CrossRef]
  15. Ismailova, A.; White, J.H. Vitamin D, Infections and Immunity. Rev. Endocr. Metab. Disord. 2022, 23, 265–277. [Google Scholar] [CrossRef]
  16. Teleni, L.; Baker, J.; Koczwara, B.; Kimlin, M.G.; Walpole, E.; Tsai, K.; Isenring, E.A. Clinical Outcomes of Vitamin D Deficiency and Supplementation in Cancer Patients. Nutr. Rev. 2013, 71, 611–621. [Google Scholar] [CrossRef]
  17. Liu, W.; Zhang, L.; Xu, H.-J.; Li, Y.; Hu, C.-M.; Yang, J.-Y.; Sun, M.-Y. The Anti-Inflammatory Effects of Vitamin D in Tumorigenesis. Int. J. Mol. Sci. 2018, 19, 2736. [Google Scholar] [CrossRef]
  18. Argano, C.; Mallaci Bocchio, R.; Lo Monaco, M.; Scibetta, S.; Natoli, G.; Cavezzi, A.; Troiani, E.; Corrao, S. An Overview of Systematic Reviews of the Role of Vitamin D on Inflammation in Patients with Diabetes and the Potentiality of Its Application on Diabetic Patients with COVID-19. Int. J. Mol. Sci. 2022, 23, 2873. [Google Scholar] [CrossRef]
  19. Corrao, S.; Mallaci Bocchio, R.; Lo Monaco, M.; Natoli, G.; Cavezzi, A.; Troiani, E.; Argano, C. Does Evidence Exist to Blunt Inflammatory Response by Nutraceutical Supplementation during COVID-19 Pandemic? An Overview of Systematic Reviews of Vitamin D, Vitamin C, Melatonin, and Zinc. Nutrients 2021, 13, 1261. [Google Scholar] [CrossRef] [PubMed]
  20. Argano, C.; Mallaci Bocchio, R.; Natoli, G.; Scibetta, S.; Lo Monaco, M.; Corrao, S. Protective Effect of Vitamin D Supplementation on COVID-19-Related Intensive Care Hospitalization and Mortality: Definitive Evidence from Meta-Analysis and Trial Sequential Analysis. Pharmaceuticals 2023, 16, 130. [Google Scholar] [CrossRef]
  21. Yu, X.P.; Bellido, T.; Manolagas, S.C. Down-Regulation of NF-Kappa B Protein Levels in Activated Human Lymphocytes by 1,25-Dihydroxyvitamin D3. Proc. Natl. Acad. Sci. USA 1995, 92, 10990–10994. [Google Scholar] [CrossRef] [PubMed]
  22. Barragan, M.; Good, M.; Kolls, J. Regulation of Dendritic Cell Function by Vitamin D. Nutrients 2015, 7, 8127–8151. [Google Scholar] [CrossRef]
  23. 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]
  24. Skrobot, A.; Demkow, U.; Wachowska, M. Immunomodulatory Role of Vitamin D: A Review. In Current Trends in Immunity and Respiratory Infections; Pokorski, M., Ed.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2018; Volume 1108, pp. 13–23. ISBN 978-3-030-01634-0. [Google Scholar]
  25. 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]
  26. Ao, T.; Kikuta, J.; Ishii, M. The Effects of Vitamin D on Immune System and Inflammatory Diseases. Biomolecules 2021, 11, 1624. [Google Scholar] [CrossRef]
  27. Corrao, S.; Colomba, D.; Arnone, S.; Argano, C.; Di Chiara, T.; Scaglione, R.; Licata, G. Improving Efficacy of PubMed Clinical Queries for Retrieving Scientifically Strong Studies on Treatment. J. Am. Med. Inform. Assoc. 2006, 13, 485–487. [Google Scholar] [CrossRef]
  28. Corrao, S.; Colomba, D.; Argano, C.; Calvo, L.; Scaglione, R.; Licata, G. Optimized Search Strategy for Detecting Scientifically Strong Studies on Treatment through PubMed. Intern. Emerg. Med. 2012, 7, 283–287. [Google Scholar] [CrossRef]
  29. Haynes, R.B.; McKibbon, K.A.; Wilczynski, N.L.; Walter, S.D.; Werre, S.R. Hedges Team Optimal Search Strategies for Retrieving Scientifically Strong Studies of Treatment from Medline: Analytical Survey. BMJ 2005, 330, 1179. [Google Scholar] [CrossRef]
  30. Al-Zohily, B.; Al-Menhali, A.; Gariballa, S.; Haq, A.; Shah, I. Epimers of Vitamin D: A Review. Int. J. Mol. Sci. 2020, 21, 470. [Google Scholar] [CrossRef] [PubMed]
  31. Adams, J.S.; Ren, S.; Liu, P.T.; Chun, R.F.; Lagishetty, V.; Gombart, A.F.; Borregaard, N.; Modlin, R.L.; Hewison, M. Vitamin D-Directed Rheostatic Regulation of Monocyte Antibacterial Responses. J. Immunol. 2009, 182, 4289–4295. [Google Scholar] [CrossRef]
  32. Kroner, J.; Sommer, A.; Fabri, M. Vitamin D Every Day to Keep the Infection Away? Nutrients 2015, 7, 4170–4188. [Google Scholar] [CrossRef]
  33. Yang, R.; Yang, E.; Shen, L.; Modlin, R.L.; Shen, H.; Chen, Z.W. IL-12+IL-18 Cosignaling in Human Macrophages and Lung Epithelial Cells Activates Cathelicidin and Autophagy, Inhibiting Intracellular Mycobacterial Growth. J. Immunol. 2018, 200, 2405–2417. [Google Scholar] [CrossRef]
  34. Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human Cathelicidin Antimicrobial Peptide (CAMP) Gene Is a Direct Target of the Vitamin D Receptor and Is Strongly up-Regulated in Myeloid Cells by 1,25-Dihydroxyvitamin D3. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef]
  35. Yuk, J.-M.; Shin, D.-M.; Lee, H.-M.; Yang, C.-S.; Jin, H.S.; Kim, K.-K.; Lee, Z.-W.; Lee, S.-H.; Kim, J.-M.; Jo, E.-K. Vitamin D3 Induces Autophagy in Human Monocytes/Macrophages via Cathelicidin. Cell Host Microbe 2009, 6, 231–243. [Google Scholar] [CrossRef]
  36. Dombrowski, Y.; Peric, M.; Koglin, S.; Ruzicka, T.; Schauber, J. Control of Cutaneous Antimicrobial Peptides by Vitamin D3. Arch. Dermatol. Res. 2010, 302, 401–408. [Google Scholar] [CrossRef]
  37. 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]
  38. Salameh, L.; Mahmood, W.; Hamoudi, R.; Almazrouei, K.; Lochanan, M.; Seyhoglu, S.; Mahboub, B. The Role of Vitamin D Supplementation on Airway Remodeling in Asthma: A Systematic Review. Nutrients 2023, 15, 2477. [Google Scholar] [CrossRef]
  39. Iqbal, S.F.; Freishtat, R.J. Mechanism of Action of Vitamin D in the Asthmatic Lung. J. Investig. Med. 2011, 59, 1200–1202. [Google Scholar] [CrossRef]
  40. Pfeffer, P.E.; Mann, E.H.; Hornsby, E.; Chambers, E.S.; Chen, Y.-H.; Rice, L.; Hawrylowicz, C.M. Vitamin D Influences Asthmatic Pathology through Its Action on Diverse Immunological Pathways. Ann. Am. Thorac. Soc. 2014, 11 (Suppl. S5), S314–S321. [Google Scholar] [CrossRef]
  41. Galowitz, S.; Chang, C. Immunobiology of Critical Pediatric Asthma. Clin. Rev. Allerg. Immunol. 2015, 48, 84–96. [Google Scholar] [CrossRef]
  42. Wimalawansa, S.J. Infections and Autoimmunity—The Immune System and Vitamin D: A Systematic Review. Nutrients 2023, 15, 3842. [Google Scholar] [CrossRef]
  43. Sundar, I.K.; Rahman, I. Vitamin D and Susceptibility of Chronic Lung Diseases: Role of Epigenetics. Front. Pharmacol. 2011, 2, 50. [Google Scholar] [CrossRef]
  44. Gois, P.; Ferreira, D.; Olenski, S.; Seguro, A. Vitamin D and Infectious Diseases: Simple Bystander or Contributing Factor? Nutrients 2017, 9, 651. [Google Scholar] [CrossRef]
  45. Watkins, R.R.; Lemonovich, T.L.; Salata, R.A. An Update on the Association of Vitamin D Deficiency with Common Infectious Diseases. Can. J. Physiol. Pharmacol. 2015, 93, 363–368. [Google Scholar] [CrossRef]
  46. Kim, M.; Kim, S.-N.; Lee, Y.; Choe, Y.; Ahn, K. Vitamin D Status and Efficacy of Vitamin D Supplementation in Atopic Dermatitis: A Systematic Review and Meta-Analysis. Nutrients 2016, 8, 789. [Google Scholar] [CrossRef]
  47. Pfeffer, P.E.; Hawrylowicz, C.M. Vitamin D in Asthma. Chest 2018, 153, 1229–1239. [Google Scholar] [CrossRef]
  48. Di Rosa, M.; Malaguarnera, M.; Nicoletti, F.; Malaguarnera, L. Vitamin D3: A Helpful Immuno-Modulator. Immunology 2011, 134, 123–139. [Google Scholar] [CrossRef]
  49. He, L.; Zhou, X.; Mo, H.; Li, X.; Guo, S. The Association between Vitamin D Receptor Gene Polymorphisms and Asthma: A Systematic Review and Meta-Analysis. Ann. Palliat. Med. 2022, 11, 574–587. [Google Scholar] [CrossRef]
  50. Penna, G.; Adorini, L. 1 Alpha,25-Dihydroxyvitamin D3 Inhibits Differentiation, Maturation, Activation, and Survival of Dendritic Cells Leading to Impaired Alloreactive T Cell Activation. J. Immunol. 2000, 164, 2405–2411. [Google Scholar] [CrossRef]
  51. Berer, A.; Stöckl, J.; Majdic, O.; Wagner, T.; Kollars, M.; Lechner, K.; Geissler, K.; Oehler, L. 1,25-Dihydroxyvitamin D(3) Inhibits Dendritic Cell Differentiation and Maturation In Vitro. Exp. Hematol. 2000, 28, 575–583. [Google Scholar] [CrossRef]
  52. Flayer, C.H.; Larson, E.D.; Haczku, A. Breaking Steroid Resistance: Effect of Vitamin D on IL-23. Am. J. Respir. Cell Mol. Biol. 2017, 57, 267–269. [Google Scholar] [CrossRef] [PubMed]
  53. Hau, C.S.; Shimizu, T.; Tada, Y.; Kamata, M.; Takeoka, S.; Shibata, S.; Mitsui, A.; Asano, Y.; Sugaya, M.; Kadono, T.; et al. The Vitamin D3 Analog, Maxacalcitol, Reduces Psoriasiform Skin Inflammation by Inducing Regulatory T Cells and Downregulating IL-23 and IL-17 Production. J. Dermatol. Sci. 2018, 92, 117–126. [Google Scholar] [CrossRef]
  54. Martens, P.-J.; Gysemans, C.; Verstuyf, A.; Mathieu, C. Vitamin D’s Effect on Immune Function. Nutrients 2020, 12, 1248. [Google Scholar] [CrossRef]
  55. Sun, D.; Luo, F.; Xing, J.; Zhang, F.; Xu, J.; Zhang, Z. 1,25(OH)2 D3 Inhibited Th17 Cells Differentiation via Regulating the NF-κB Activity and Expression of IL-17. Cell Prolif. 2018, 51, e12461. [Google Scholar] [CrossRef]
  56. Ikeda, U.; Wakita, D.; Ohkuri, T.; Chamoto, K.; Kitamura, H.; Iwakura, Y.; Nishimura, T. 1α,25-Dihydroxyvitamin D3 and All-Trans Retinoic Acid Synergistically Inhibit the Differentiation and Expansion of Th17 Cells. Immunol. Lett. 2010, 134, 7–16. [Google Scholar] [CrossRef]
  57. Joshi, S.; Pantalena, L.-C.; Liu, X.K.; Gaffen, S.L.; Liu, H.; Rohowsky-Kochan, C.; Ichiyama, K.; Yoshimura, A.; Steinman, L.; Christakos, S.; et al. 1,25-Dihydroxyvitamin D3 Ameliorates Th17 Autoimmunity via Transcriptional Modulation of Interleukin-17A. Mol. Cell. Biol. 2011, 31, 3653–3669. [Google Scholar] [CrossRef]
  58. Kim, D.-H.; Meza, C.A.; Clarke, H.; Kim, J.-S.; Hickner, R.C. Vitamin D and Endothelial Function. Nutrients 2020, 12, 575. [Google Scholar] [CrossRef]
  59. Vila Cuenca, M.; Ferrantelli, E.; Meinster, E.; Pouw, S.M.; Kovačević, I.; De Menezes, R.X.; Niessen, H.W.; Beelen, R.H.J.; Hordijk, P.L.; Vervloet, M.G. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J. Am. Heart Assoc. 2018, 7, e008776. [Google Scholar] [CrossRef]
  60. Nativel, B.; Ramin-Mangata, S.; Mevizou, R.; Figuester, A.; Andries, J.; Iwema, T.; Ikewaki, N.; Gasque, P.; Viranaïcken, W. CD 93 Is a Cell Surface Lectin Receptor Involved in the Control of the Inflammatory Response Stimulated by Exogenous DNA. Immunology 2019, 158, 85–93. [Google Scholar] [CrossRef]
  61. Jennewein, C.; Sowa, R.; Faber, A.C.; Dildey, M.; Von Knethen, A.; Meybohm, P.; Scheller, B.; Dröse, S.; Zacharowski, K. Contribution of Ninjurin1 to Toll-Like Receptor 4 Signaling and Systemic Inflammation. Am. J. Respir. Cell Mol. Biol. 2015, 53, 656–663. [Google Scholar] [CrossRef] [PubMed]
  62. Kolseth, I.B.M.; Reine, T.M.; Vuong, T.T.; Meen, A.J.; Fan, Q.; Jenssen, T.G.; Grønning-Wang, L.M.; Kolset, S.O. Serglycin Is Part of the Secretory Repertoire of LPS-activated Monocytes. Immunity Inflam. Dis. 2015, 3, 23–31. [Google Scholar] [CrossRef]
  63. Sîrbe, C.; Rednic, S.; Grama, A.; Pop, T.L. An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases. Int. J. Mol. Sci. 2022, 23, 9784. [Google Scholar] [CrossRef]
  64. Grijincu, M.; Buzan, M.-R.; Zbîrcea, L.-E.; Păunescu, V.; Panaitescu, C. Prenatal Factors in the Development of Allergic Diseases. Int. J. Mol. Sci. 2024, 25, 6359. [Google Scholar] [CrossRef]
  65. Anogeianaki, A.; Castellani, M.L.; Tripodi, D.; Toniato, E.; De Lutiis, M.A.; Conti, F.; Felaco, P.; Fulcheri, M.; Theoharides, T.C.; Galzio, R.; et al. Vitamins and Mast Cells. Int. J. Immunopathol. Pharmacol. 2010, 23, 991–996. [Google Scholar] [CrossRef]
  66. Murdaca, G.; Allegra, A.; Tonacci, A.; Musolino, C.; Ricciardi, L.; Gangemi, S. Mast Cells and Vitamin D Status: A Clinical and Biological Link in the Onset of Allergy and Bone Diseases. Biomedicines 2022, 10, 1877. [Google Scholar] [CrossRef]
  67. Ladeira, J.M.C.D.; Zacas, O.; Ferreira, A.M.; Gomes Stegun, P.C.; Grotta, M.B.; Toro, A.A.D.C. The Role of Vitamin D in the Severity and Control of Asthma in Children and Adolescents: A Protocol for Systematic Review and Meta-Analysis. Medicine 2022, 101, e31457. [Google Scholar] [CrossRef]
  68. Sansoni, E.R.; Sautter, N.B.; Mace, J.C.; Smith, T.L.; Yawn, J.R.; Lawrence, L.A.; Schlosser, R.J.; Soler, Z.M.; Mulligan, J.K. Vitamin D3 as a Novel Regulator of Basic Fibroblast Growth Factor in Chronic Rhinosinusitis with Nasal Polyposis. Int. Forum Allergy Rhinol. 2015, 5, 191–196. [Google Scholar] [CrossRef]
  69. Confino-Cohen, R.; Brufman, I.; Goldberg, A.; Feldman, B.S. Vitamin D, Asthma Prevalence and Asthma Exacerbations: A Large Adult Population-Based Study. Allergy 2014, 69, 1673–1680. [Google Scholar] [CrossRef]
  70. Cassim, R.; Russell, M.A.; Lodge, C.J.; Lowe, A.J.; Koplin, J.J.; Dharmage, S.C. The Role of Circulating 25 Hydroxyvitamin D in Asthma: A Systematic Review. Allergy 2015, 70, 339–354. [Google Scholar] [CrossRef] [PubMed]
  71. Aziz, D.A.; Abbas, A.; Viquar, W.; Munawar Hussain, A. Association of Vitamin D Levels and Asthma Exacerbations in Children and Adolescents: Experience from a Tertiary Care Center. Monaldi Arch. Chest Dis. 2022, 93. [Google Scholar] [CrossRef] [PubMed]
  72. Kaur, N.; Kumar, V.; Singh, J.; Jain, H.; Paras, P.; Kaur, N.; Sareen, A.K. Assessment of the Relation Between Asthma Severity and Serum Vitamin D Levels: A Cross-Sectional Study. Cureus 2023, 15, e46826. [Google Scholar] [CrossRef] [PubMed]
  73. Sobczak, M.; Pawliczak, R. Relationship between Vitamin D and Asthma from Gestational to Adulthood Period: A Meta-Analysis of Randomized Clinical Trials. BMC Pulm. Med. 2023, 23, 212. [Google Scholar] [CrossRef] [PubMed]
  74. Wolsk, H.M.; Chawes, B.L.; Litonjua, A.A.; Hollis, B.W.; Waage, J.; Stokholm, J.; Bønnelykke, K.; Bisgaard, H.; Weiss, S.T. Prenatal Vitamin D Supplementation Reduces Risk of Asthma/Recurrent Wheeze in Early Childhood: A Combined Analysis of Two Randomized Controlled Trials. PLoS ONE 2017, 12, e0186657. [Google Scholar] [CrossRef]
  75. Shi, D.; Wang, D.; Meng, Y.; Chen, J.; Mu, G.; Chen, W. Maternal Vitamin D Intake during Pregnancy and Risk of Asthma and Wheeze in Children: A Systematic Review and Meta-Analysis of Observational Studies. J. Matern.-Fetal Neonatal Med. 2021, 34, 653–659. [Google Scholar] [CrossRef]
  76. Feng, H.; Xun, P.; Pike, K.; Wills, A.K.; Chawes, B.L.; Bisgaard, H.; Cai, W.; Wan, Y.; He, K. In Utero Exposure to 25-Hydroxyvitamin D and Risk of Childhood Asthma, Wheeze, and Respiratory Tract Infections: A Meta-Analysis of Birth Cohort Studies. J. Allergy Clin. Immunol. 2017, 139, 1508–1517. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, Y.; Wang, J.; Chen, L.; Zhang, H.; Yu, L.; Chi, Y.; Chen, M.; Cai, Y. Efficacy of Vitamin D Supplementation on COPD and Asthma Control: A Systematic Review and Meta-Analysis. J. Glob. Health 2022, 12, 04100. [Google Scholar] [CrossRef]
  78. Wang, Q.; Ying, Q.; Zhu, W.; Chen, J. Vitamin D and Asthma Occurrence in Children: A Systematic Review and Meta-Analysis. J. Pediatr. Nurs. 2022, 62, e60–e68. [Google Scholar] [CrossRef]
  79. Song, H.; Yang, L.; Jia, C. Maternal Vitamin D Status during Pregnancy and Risk of Childhood Asthma: A Meta-analysis of Prospective Studies. Mol. Nutr. Food Res. 2017, 61, 1600657. [Google Scholar] [CrossRef]
  80. Liu, J.; Dong, Y.-Q.; Yin, J.; Yao, J.; Shen, J.; Sheng, G.-J.; Li, K.; Lv, H.-F.; Fang, X.; Wu, W.-F. Meta-Analysis of Vitamin D and Lung Function in Patients with Asthma. Respir. Res. 2019, 20, 161. [Google Scholar] [CrossRef]
  81. Alnori, H.; Alassaf, F.A.; Alfahad, M.; Qazzaz, M.E.; Jasim, M.; Abed, M.N. Vitamin D and Immunoglobulin E Status in Allergic Rhinitis Patients Compared to Healthy People. J. Med. Life 2020, 13, 463–468. [Google Scholar] [CrossRef]
  82. Frieri, M. Asthma Linked with Rhinosinusitis: An Extensive Review. Allergy Rhinol. 2014, 5, ar.2014.5.0083. [Google Scholar] [CrossRef]
  83. Stokes, P.J.; Rimmer, J. The Relationship between Serum Vitamin D and Chronic Rhinosinusitis: A Systematic Review. Am. J. Rhinol. Allergy 2016, 30, 23–28. [Google Scholar] [CrossRef]
  84. Aryan, Z.; Rezaei, N.; Camargo, C.A. Vitamin D Status, Aeroallergen Sensitization, and Allergic Rhinitis: A Systematic Review and Meta-Analysis. Int. Rev. Immunol. 2017, 36, 41–53. [Google Scholar] [CrossRef]
  85. Hamzaoui, A.; Berraïes, A.; Hamdi, B.; Kaabachi, W.; Ammar, J.; Hamzaoui, K. Vitamin D Reduces the Differentiation and Expansion of Th17 Cells in Young Asthmatic Children. Immunobiology 2014, 219, 873–879. [Google Scholar] [CrossRef]
  86. Fedora, K.; Setyoningrum, R.A.; Aina, Q.; Rosyidah, L.N.; Ni’mah, N.L.; Titiharja, F.F. Vitamin D Supplementation Decrease Asthma Exacerbations in Children: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Ann. Med. 2024, 56, 2400313. [Google Scholar] [CrossRef]
  87. El Abd, A.; Dasari, H.; Dodin, P.; Trottier, H.; Ducharme, F.M. The Effects of Vitamin D Supplementation on Inflammatory Biomarkers in Patients with Asthma: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Immunol. 2024, 15, 1335968. [Google Scholar] [CrossRef]
  88. Mahboub, B.; Al Heialy, S.; Hachim, M.Y.; Ramakrishnan, R.K.; Alzaabi, A.; Seliem, R.M.; Salameh, L.I.; Toor, S.M.; Shendi, F.S.; Al Ali, O.M.; et al. Vitamin D Regulates the Expression of Glucocorticoid Receptors in Blood of Severe Asthmatic Patients. J. Immunol. Res. 2021, 2021, 1–10. [Google Scholar] [CrossRef]
  89. Jolliffe, D.A.; Greenberg, L.; Hooper, R.L.; Griffiths, C.J.; Camargo, C.A.; Kerley, C.P.; Jensen, M.E.; Mauger, D.; Stelmach, I.; Urashima, M.; et al. Vitamin D Supplementation to Prevent Asthma Exacerbations: A Systematic Review and Meta-Analysis of Individual Participant Data. Lancet Respir. Med. 2017, 5, 881–890. [Google Scholar] [CrossRef]
  90. Castro, M.; King, T.S.; Kunselman, S.J.; Cabana, M.D.; Denlinger, L.; Holguin, F.; Kazani, S.D.; Moore, W.C.; Moy, J.; Sorkness, C.A.; et al. Effect of Vitamin D3 on Asthma Treatment Failures in Adults With Symptomatic Asthma and Lower Vitamin D Levels: The VIDA Randomized Clinical Trial. JAMA 2014, 311, 2083. [Google Scholar] [CrossRef]
  91. Zhao, D.; Yu, D.; Ren, Q.; Dong, B.; Zhao, F.; Sun, Y. Association of Vitamin D Receptor Gene Polymorphisms with Susceptibility to Childhood Asthma: A Meta-analysis. Pediatr. Pulmonol. 2017, 52, 423–429. [Google Scholar] [CrossRef]
  92. Han, J.-C.; Du, J.; Zhang, Y.-J.; Qi, G.-B.; Li, H.-B.; Zhang, Y.J.; Yu, X. Vitamin D Receptor Polymorphisms May Contribute to Asthma Risk. J. Asthma 2016, 53, 790–800. [Google Scholar] [CrossRef]
  93. Makoui, M.H.; Imani, D.; Motallebnezhad, M.; Azimi, M.; Razi, B. Vitamin D Receptor Gene Polymorphism and Susceptibility to Asthma. Ann. Allergy Asthma Immunol. 2020, 124, 57–69. [Google Scholar] [CrossRef] [PubMed]
  94. Bhardwaj, B.; Singh, J. Efficacy of Vitamin D Supplementation in Allergic Rhinitis. Indian. J. Otolaryngol. Head. Neck Surg. 2021, 73, 152–159. [Google Scholar] [CrossRef] [PubMed]
  95. Guo, M. Vitamin D Supplementation Improves the Therapeutic Effect of Mometasone on Allergic Rhinitis. Acta Biochim. Pol. 2023. [Google Scholar] [CrossRef]
  96. Bakhshaee, M.; Sharifian, M.; Esmatinia, F.; Rasoulian, B.; Mohebbi, M. Therapeutic Effect of Vitamin D Supplementation on Allergic Rhinitis. Eur. Arch. Otorhinolaryngol. 2019, 276, 2797–2801. [Google Scholar] [CrossRef] [PubMed]
  97. Jerzynska, J.; Stelmach, W.; Rychlik, B.; Lechańska, J.; Podlecka, D.; Stelmach, I. The Clinical Effect of Vitamin D Supplementation Combined with Grass-Specific Sublingual Immunotherapy in Children with Allergic Rhinitis. Allergy Asthma Proc. 2016, 37, 105–114. [Google Scholar] [CrossRef]
  98. Fenner, J.; Silverberg, N.B. Oral Supplements in Atopic Dermatitis. Clin. Dermatol. 2018, 36, 653–658. [Google Scholar] [CrossRef]
  99. Hallau, J.; Hamann, L.; Schumann, R.; Worm, M.; Heine, G. A Promoter Polymorphism of the Vitamin D Metabolism Gene Cyp24a1 Is Associated with Severe Atopic Dermatitis in Adults. Acta Derm. Venerol. 2016, 96, 169–172. [Google Scholar] [CrossRef]
  100. Massoumi, R. The Central Role of Bcl-3 in Atopic Dermatitis. J. Investig. Dermatol. 2009, 129, 2088–2090. [Google Scholar] [CrossRef]
  101. Barlianto, W.; Wulandari, D.; Sari, T.; Firdayanti, V.; Avandi, M. Vitamin D, Cytokine Profiles, and Disease Severity in Infants with Atopic Dermatitis: A Single Centre, Cross-Sectional Study. Postępy Dermatol. Alergol. 2022, 39, 793–799. [Google Scholar] [CrossRef]
  102. Nahm, D.-H. Regulatory T Cell-Targeted Immunomodulatory Therapy for Long-Term Clinical Improvement of Atopic Dermatitis: Hypotheses and Perspectives. Life 2023, 13, 1674. [Google Scholar] [CrossRef]
  103. Hattangdi-Haridas, S.R.; Lanham-New, S.A.; Wong, W.H.S.; Ho, M.H.K.; Darling, A.L. Vitamin D Deficiency and Effects of Vitamin D Supplementation on Disease Severity in Patients with Atopic Dermatitis: A Systematic Review and Meta-Analysis in Adults and Children. Nutrients 2019, 11, 1854. [Google Scholar] [CrossRef] [PubMed]
  104. Hidayati, A.N.; Sawitri, S.; Sari, D.W.; Prakoeswa, C.R.S.; Indramaya, D.M.; Damayanti, D.; Zulkarnain, I.; Citrashanty, I.; Widia, Y.; Anggraeni, S. Efficacy of Vitamin D Supplementation on the Severity of Atopic Dermatitis in Children: A Systematic Review and Meta-Analysis. F1000Research 2022, 11, 274. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, C.M.; Lara-Corrales, I.; Pope, E. Effects of Vitamin D Levels and Supplementation on Atopic Dermatitis: A Systematic Review. Pediatr. Dermatol. 2018, 35, 754–760. [Google Scholar] [CrossRef] [PubMed]
  106. Fu, H.; Li, Y.; Huang, H.; Wang, D. Serum Vitamin D Level and Efficacy of Vitamin D Supplementation in Children with Atopic Dermatitis: A Systematic Review and Meta-Analysis. Comput. Math. Methods Med. 2022, 2022, 9407888. [Google Scholar] [CrossRef]
  107. Nielsen, A.Y.; Høj, S.; Thomsen, S.F.; Meteran, H. Vitamin D Supplementation for Treating Atopic Dermatitis in Children and Adults: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 4128. [Google Scholar] [CrossRef]
  108. Lim, J.J.; Liu, M.H.; Chew, F.T. Dietary Interventions in Atopic Dermatitis: A Comprehensive Scoping Review and Analysis. Int. Arch. Allergy Immunol. 2024, 185, 545–589. [Google Scholar] [CrossRef]
  109. Ishikawa, L.L.W.; Colavite, P.M.; Fraga-Silva, T.F.D.C.; Mimura, L.A.N.; França, T.G.D.; Zorzella-Pezavento, S.F.G.; Chiuso-Minicucci, F.; Marcolino, L.D.; Penitenti, M.; Ikoma, M.R.V.; et al. Vitamin D Deficiency and Rheumatoid Arthritis. Clinic Rev. Allerg. Immunol. 2017, 52, 373–388. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, Z.; Zhang, J.; An, F.; Zhang, J.; Meng, X.; Liu, S.; Xia, R.; Wang, G.; Yan, C. The Mechanism of Dendritic Cell-T Cell Crosstalk in Rheumatoid Arthritis. Arthritis Res. Ther. 2023, 25, 193. [Google Scholar] [CrossRef]
  111. Hysa, E.; Gotelli, E.; Campitiello, R.; Paolino, S.; Pizzorni, C.; Casabella, A.; Sulli, A.; Smith, V.; Cutolo, M. Vitamin D and Muscle Status in Inflammatory and Autoimmune Rheumatic Diseases: An Update. Nutrients 2024, 16, 2329. [Google Scholar] [CrossRef]
  112. Miler, M.; Nikolac Gabaj, N.; Grazio, S.; Vahtarić, A.; Vrtarić, A.; Grubišić, F.; Skala Kavanagh, H.; Doko Vajdić, I.; Vrkić, N. Lower Concentration of Vitamin D Is Associated with Lower DAS28 and VAS-Pain Scores in Patients with Inflammatory Rheumatic Diseases Treated with Infliximab: A Pilot Study. Rheumatol. Int. 2020, 40, 1455–1461. [Google Scholar] [CrossRef]
  113. 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] [PubMed]
  114. Lee, Y.H.; Bae, S.-C. Vitamin D Level in Rheumatoid Arthritis and Its Correlation with the Disease Activity: A Meta-Analysis. Clin. Exp. Rheumatol. 2016, 34, 827–833. [Google Scholar]
  115. Cutolo, M.; Pizzorni, C.; Sulli, A. Vitamin D Endocrine System Involvement in Autoimmune Rheumatic Diseases. Autoimmun. Rev. 2011, 11, 84–87. [Google Scholar] [CrossRef] [PubMed]
  116. Myers, L.K.; Winstead, M.; Kee, J.D.; Park, J.J.; Zhang, S.; Li, W.; Yi, A.-K.; Stuart, J.M.; Rosloniec, E.F.; Brand, D.D.; et al. 1,25-Dihydroxyvitamin D3 and 20-Hydroxyvitamin D3 Upregulate LAIR-1 and Attenuate Collagen Induced Arthritis. Int. J. Mol. Sci. 2021, 22, 13342. [Google Scholar] [CrossRef] [PubMed]
  117. Gardner, D.H.; Jeffery, L.E.; Soskic, B.; Briggs, Z.; Hou, T.Z.; Raza, K.; Sansom, D.M. 1,25(OH)2D3 Promotes the Efficacy of CD28 Costimulation Blockade by Abatacept. J. Immunol. 2015, 195, 2657–2665. [Google Scholar] [CrossRef]
  118. Orgaz-Molina, J.; Buendía-Eisman, A.; Arrabal-Polo, M.A.; Ruiz, J.C.; Arias-Santiago, S. Deficiency of Serum Concentration of 25-Hydroxyvitamin D in Psoriatic Patients: A Case-Control Study. J. Am. Acad. Dermatol. 2012, 67, 931–938. [Google Scholar] [CrossRef]
  119. Bhat, G.H.; Guldin, S.; Khan, M.S.; Yasir, M.; Prasad, G. Vitamin D Status in Psoriasis: Impact and Clinical Correlations. BMC Nutr. 2022, 8, 115. [Google Scholar] [CrossRef]
  120. Van Hamburg, J.P.; Asmawidjaja, P.S.; Davelaar, N.; Mus, A.M.C.; Cornelissen, F.; Van Leeuwen, J.P.T.M.; Hazes, J.M.W.; Dolhain, R.J.E.M.; Bakx, P.A.G.M.; Colin, E.M.; et al. TNF Blockade Requires 1,25(OH)2D3 to Control Human Th17-Mediated Synovial Inflammation. Ann. Rheum. Dis. 2012, 71, 606–612. [Google Scholar] [CrossRef]
  121. Scharla, S.; Schmidt-Gayk, H.; Sattar, P. Exclusion of Autonomous 1,25(OH)2Vitamin D3 (Calcitriol) Formation as a Pathogenetic Principle of Inflammation-Mediated Osteopenia (IMO). Acta Endocrinol. 1984, 104, S7. [Google Scholar] [CrossRef]
  122. Forsblad d′Elia, H.; Carlsten, H. The Impact of Hormone Replacement Therapy on Humoral and Cell-mediated Immune Responses In Vivo in Post-menopausal Women with Rheumatoid Arthritis. Scand. J. Immunol. 2008, 68, 661–667. [Google Scholar] [CrossRef]
  123. Damiani, G.; Bragazzi, N.L.; Karimkhani Aksut, C.; Wu, D.; Alicandro, G.; McGonagle, D.; Guo, C.; Dellavalle, R.; Grada, A.; Wong, P.; et al. The Global, Regional, and National Burden of Psoriasis: Results and Insights From the Global Burden of Disease 2019 Study. Front. Med. 2021, 8, 743180. [Google Scholar] [CrossRef] [PubMed]
  124. Brożyna, A.A.; Slominski, R.M.; Nedoszytko, B.; Zmijewski, M.A.; Slominski, A.T. Vitamin D Signaling in Psoriasis: Pathogenesis and Therapy. Int. J. Mol. Sci. 2022, 23, 8575. [Google Scholar] [CrossRef]
  125. Murdaca, G.; Tonacci, A.; Negrini, S.; Greco, M.; Borro, M.; Puppo, F.; Gangemi, S. Emerging Role of Vitamin D in Autoimmune Diseases: An Update on Evidence and Therapeutic Implications. Autoimmun. Rev. 2019, 18, 102350. [Google Scholar] [CrossRef] [PubMed]
  126. Mattozzi, C.; Paolino, G.; Richetta, A.G.; Calvieri, S. Psoriasis, Vitamin D and the Importance of the Cutaneous Barrier’s Integrity: An Update. J. Dermatol. 2016, 43, 507–514. [Google Scholar] [CrossRef]
  127. Bittiner, B.; Bleehen, S.S.; MacNeil, S. 1 Alpha,25(OH)2 Vitamin D3 Increases Intracellular Calcium in Human Keratinocytes. Br. J. Dermatol. 1991, 124, 230–235. [Google Scholar] [CrossRef]
  128. Reichrath, J.; Reichrath, S.; Heyne, K.; Vogt, T.; Roemer, K. Tumor Suppression in Skin and Other Tissues via Cross-Talk between Vitamin D- and P53-Signaling. Front. Physiol. 2014, 5, 166. [Google Scholar] [CrossRef] [PubMed]
  129. Reichrath, J.; Saternus, R.; Vogt, T. Challenge and Perspective: The Relevance of Ultraviolet (UV) Radiation and the Vitamin D Endocrine System (VDES) for Psoriasis and Other Inflammatory Skin Diseases. Photochem. Photobiol. Sci. 2017, 16, 433–444. [Google Scholar] [CrossRef]
  130. Morimoto, S.; Yoshikawa, K.; Fukuo, K.; Shiraishi, T.; Koh, E.; Imanaka, S.; Kitano, S.; Ogihara, T. Inverse Relation between Severity of Psoriasis and Serum 1,25-Dihydroxyvitamin D Level. J. Dermatol. Sci. 1990, 1, 277–282. [Google Scholar] [CrossRef]
  131. Tajjour, R.; Baddour, R.; Redwan, F.; Hassan, F. The Relationship between Psoriasis and Serum Levels of Vitamin D. J. Adv. Med. Med. Res. 2018, 26, 1–12. [Google Scholar] [CrossRef]
  132. Bergler-Czop, B.; Brzezińska-Wcisło, L. Serum Vitamin D Level—The Effect on the Clinical Course of Psoriasis. Postępy Dermatol. Alergol. 2016, 6, 445–449. [Google Scholar] [CrossRef]
  133. Ricceri, F.; Pescitelli, L.; Tripo, L.; Prignano, F. Deficiency of Serum Concentration of 25-Hydroxyvitamin D Correlates with Severity of Disease in Chronic Plaque Psoriasis. J. Am. Acad. Dermatol. 2013, 68, 511–512. [Google Scholar] [CrossRef] [PubMed]
  134. Soleymani, T.; Hung, T.; Soung, J. The Role of Vitamin D in Psoriasis: A Review. Int. J. Dermatol. 2015, 54, 383–392. [Google Scholar] [CrossRef] [PubMed]
  135. Lee, Y.H.; Song, G.G. Association between Circulating 25-Hydroxyvitamin D Levels and Psoriasis, and Correlation with Disease Severity: A Meta-Analysis. Clin. Exp. Dermatol. 2018, 43, 529–535. [Google Scholar] [CrossRef]
  136. Formisano, E.; Proietti, E.; Borgarelli, C.; Pisciotta, L. Psoriasis and Vitamin D: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 3387. [Google Scholar] [CrossRef]
  137. Gamonal, S.B.L.; Gamonal, A.C.C.; Marques, N.C.V.; Brandão, M.A.F.; Raposo, N.R.B. Is Vitamin D Status Relevant to Psoriasis and Psoriatic Arthritis? A Retrospective Cross-Sectional Study. Sao Paulo Med. J. 2022, 141, e2022216. [Google Scholar] [CrossRef] [PubMed]
  138. Chandrashekar, L.; Kumari, G.R.K.; Rajappa, M.; Revathy, G.; Munisamy, M.; Thappa, D.M. 25-Hydroxy Vitamin D and Ischaemia-Modified Albumin Levels in Psoriasis and Their Association with Disease Severity. Br. J. Biomed. Sci. 2015, 72, 56–60. [Google Scholar] [CrossRef]
  139. Reichrath, J.; Zouboulis, C.C.; Vogt, T.; Holick, M.F. Targeting the Vitamin D Endocrine System (VDES) for the Management of Inflammatory and Malignant Skin Diseases: An Historical View and Outlook. Rev. Endocr. Metab. Disord. 2016, 17, 405–417. [Google Scholar] [CrossRef] [PubMed]
  140. Barrea, L.; Savanelli, M.C.; Di Somma, C.; Napolitano, M.; Megna, M.; Colao, A.; Savastano, S. Vitamin D and Its Role in Psoriasis: An Overview of the Dermatologist and Nutritionist. Rev. Endocr. Metab. Disord. 2017, 18, 195–205. [Google Scholar] [CrossRef]
  141. Disphanurat, W.; Viarasilpa, W.; Chakkavittumrong, P.; Pongcharoen, P. The Clinical Effect of Oral Vitamin D2 Supplementation on Psoriasis: A Double-Blind, Randomized, Placebo-Controlled Study. Dermatol. Res. Pract. 2019, 2019, 5237642. [Google Scholar] [CrossRef]
  142. van de Kerkhof, P.C. Biological Activity of Vitamin D Analogues in the Skin, with Special Reference to Antipsoriatic Mechanisms. Br. J. Dermatol. 1995, 132, 675–682. [Google Scholar] [CrossRef]
  143. Oxholm, A.; Oxholm, P.; Staberg, B.; Bendtzen, K. Expression of Interleukin-6-like Molecules and Tumour Necrosis Factor after Topical Treatment of Psoriasis with a New Vitamin D Analogue (MC 903). Acta Derm. Venereol. 1989, 69, 385–390. [Google Scholar] [PubMed]
  144. Van De Kerkhof, P.C.M.; Green, C.; Hamberg, K.J.; Hutchinson, P.E.; Jensen, J.K.; Kidson, P.; Kragballe, K.; Larsen, F.G.; Munro, C.S.; Tillman, D.M. Safety and Efficacy of Combined High-Dose Treatment with Calcipotriol Ointment and Solution in Patients with Psoriasis. Dermatology 2002, 204, 214–221. [Google Scholar] [CrossRef] [PubMed]
  145. Kircik, L.H. Topical Calcipotriene 0.005% and Betamethasone Dipropionate 0.064% Maintains Efficacy of Etanercept after Step-down Dose in Patients with Moderate-to-Severe Plaque Psoriasis: Results of an Open Label Trial. J. Drugs Dermatol. 2011, 10, 878–882. [Google Scholar]
  146. Pinter, A.; Green, L.J.; Selmer, J.; Praestegaard, M.; Gold, L.S.; Augustin, M.; trial investigator group. A Pooled Analysis of Randomized, Controlled, Phase 3 Trials Investigating the Efficacy and Safety of a Novel, Fixed Dose Calcipotriene and Betamethasone Dipropionate Cream for the Topical Treatment of Plaque Psoriasis. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 228–236. [Google Scholar] [CrossRef]
  147. Heim, M.; Irondelle, M.; Duteil, L.; Cardot-Leccia, N.; Rocchi, S.; Passeron, T.; Tulic, M.K. Impact of Topical Emollient, Steroids Alone or Combined with Calcipotriol, on the Immune Infiltrate and Clinical Outcome in Psoriasis. Exp. Dermatol. 2022, 31, 1764–1778. [Google Scholar] [CrossRef] [PubMed]
  148. van de Kerkhof, P.C.M.; Berth-Jones, J.; Griffiths, C.E.M.; Harrison, P.V.; Hönigsmann, H.; Marks, R.; Roelandts, R.; Schöpf, E.; Trompke, C. Long-Term Efficacy and Safety of Tacalcitol Ointment in Patients with Chronic Plaque Psoriasis. Br. J. Dermatol. 2002, 146, 414–422. [Google Scholar] [CrossRef]
  149. Miyachi, Y.; Ohkawara, A.; Ohkido, M.; Harada, S.; Tamaki, K.; Nakagawa, H.; Hori, Y.; Nishiyama, S. Long-Term Safety and Efficacy of High-Concentration (20 Microg/g) Tacalcitol Ointment in Psoriasis Vulgaris. Eur. J. Dermatol. 2002, 12, 463–468. [Google Scholar]
  150. Venegas-Iribarren, S.; Andino, R. Topical corticosteroids or vitamin D analogues for plaque psoriasis? Medwave 2017, 17, e6981. [Google Scholar] [CrossRef] [PubMed]
  151. Ford, A.R.; Siegel, M.; Bagel, J.; Cordoro, K.M.; Garg, A.; Gottlieb, A.; Green, L.J.; Gudjonsson, J.E.; Koo, J.; Lebwohl, M.; et al. Dietary Recommendations for Adults With Psoriasis or Psoriatic Arthritis From the Medical Board of the National Psoriasis Foundation: A Systematic Review. JAMA Dermatol. 2018, 154, 934–950. [Google Scholar] [CrossRef]
  152. Zold, E.; Barta, Z.; Bodolay, E. Vitamin D Deficiency and Connective Tissue Disease. In Vitamins & Hormones; Elsevier: Amsterdam, The Netherlands, 2011; Volume 86, pp. 261–286. ISBN 978-0-12-386960-9. [Google Scholar]
  153. Wright, T.B.; Shults, J.; Leonard, M.B.; Zemel, B.S.; Burnham, J.M. Hypovitaminosis D Is Associated with Greater Body Mass Index and Disease Activity in Pediatric Systemic Lupus Erythematosus. J. Pediatr. 2009, 155, 260–265. [Google Scholar] [CrossRef]
  154. Baxter, A.G.; Smyth, M.J. The Role of NK Cells in Autoimmune Disease. Autoimmunity 2002, 35, 1–14. [Google Scholar] [CrossRef] [PubMed]
  155. Grammer, A.C.; Lipsky, P.E. B Cell Abnormalities in Systemic Lupus Erythematosus. Arthritis Res. Ther. 2003, 5 (Suppl. S4), S22-27. [Google Scholar] [CrossRef]
  156. Zhou, T.-B.; Jiang, Z.-P.; Lin, Z.-J.; Su, N. Association of Vitamin D Receptor Gene Polymorphism with the Risk of Systemic Lupus Erythematosus. J. Recept. Signal Transduct. 2015, 35, 8–14. [Google Scholar] [CrossRef] [PubMed]
  157. Magro, R.; Saliba, C.; Camilleri, L.; Scerri, C.; Borg, A.A. Vitamin D Supplementation in Systemic Lupus Erythematosus: Relationship to Disease Activity, Fatigue and the Interferon Signature Gene Expression. BMC Rheumatol. 2021, 5, 53. [Google Scholar] [CrossRef]
  158. Carvalho, J.F.; Blank, M.; Kiss, E.; Tarr, T.; Amital, H.; Shoenfeld, Y. Anti-Vitamin D, Vitamin D in SLE: Preliminary Results. Ann. New York Acad. Sci. 2007, 1109, 550–557. [Google Scholar] [CrossRef]
  159. Koizumi, T.; Nakao, Y.; Matsui, T.; Nakagawa, T.; Matsuda, S.; Komoriya, K.; Kanai, Y.; Fujita, T. Effects of Corticosteroid and 1,24R-Dihydroxy-Vitamin D3 Administration on Lymphoproliferation and Autoimmune Disease in MRL/MP-Lpr/Lpr Mice. Int. Arch. Allergy Immunol. 1985, 77, 396–404. [Google Scholar] [CrossRef] [PubMed]
  160. O’Regan, S.; Chesney, R.W.; Hamstra, A.; Eisman, J.A.; O’Gorman, A.M.; Deluca, H.F. Reduced Serum 1,25-(OH)2 Vitamin D3 Levels in Prednisone-Treated Adolescents with Systemic Lupus Erythematosus. Acta Paediatr. Scand. 1979, 68, 109–111. [Google Scholar] [CrossRef]
  161. Müller, K.; Kriegbaum, N.J.; Baslund, B.; Sørensen, O.H.; Thymann, M.; Bentzen, K. Vitamin D3 Metabolism in Patients with Rheumatic Diseases: Low Serum Levels of 25-Hydroxyvitamin D3 in Patients with Systemic Lupus Erythematosus. Clin. Rheumatol. 1995, 14, 397–400. [Google Scholar] [CrossRef]
  162. Sarİ, F.; Saraç, D.; Bayram, S.; Baglan Yentur, S.; Tore, N.G.; Oskay, D.; Tufan, A. Pos1583-Hpr The Effect of Vitamin D Deficiency on Exercise Capacity, Respiratory Muscle Strength, and Peripheral Muscle Strength in Patients with Connective Tissue Diseases. Ann. Rheum. Dis. 2023, 82, 1168. [Google Scholar] [CrossRef]
  163. Andreoli, L.; Dall’Ara, F.; Piantoni, S.; Zanola, A.; Piva, N.; Cutolo, M.; Tincani, A. A 24-Month Prospective Study on the Efficacy and Safety of Two Different Monthly Regimens of Vitamin D Supplementation in Pre-Menopausal Women with Systemic Lupus Erythematosus. Lupus 2015, 24, 499–506. [Google Scholar] [CrossRef]
  164. Karimzadeh, H.; Shirzadi, M.; Karimifar, M. The Effect of Vitamin D Supplementation in Disease Activity of Systemic Lupus Erythematosus Patients with Vitamin D Deficiency: A Randomized Clinical Trial. J. Res. Med. Sci. 2017, 22, 4. [Google Scholar] [CrossRef] [PubMed]
  165. Singgih Wahono, C.; Diah Setyorini, C.; Kalim, H.; Nurdiana, N.; Handono, K. Effect of Curcuma xanthorrhiza Supplementation on Systemic Lupus Erythematosus Patients with Hypovitamin D Which Were Given Vitamin D3 towards Disease Activity (SLEDAI), IL-6, and TGF- β 1 Serum. Int. J. Rheumatol. 2017, 2017, 1–8. [Google Scholar] [CrossRef]
  166. Pakchotanon, R.; Lomarat, W.; Narongroeknawin, P.; Chaiamnuay, S.; Asavatanabodee, P. Randomized Double-Blind Controlled Trial to Evaluate Efficacy of Vitamin D Supplementation Among Patients with Systemic Lupus Erythematosus. J. Southeast. Asian Med. Res. 2020, 4, 24–32. [Google Scholar] [CrossRef]
  167. Lima, G.L.; Paupitz, J.; Aikawa, N.E.; Takayama, L.; Bonfa, E.; Pereira, R.M.R. Vitamin D Supplementation in Adolescents and Young Adults With Juvenile Systemic Lupus Erythematosus for Improvement in Disease Activity and Fatigue Scores: A Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Care Res. 2016, 68, 91–98. [Google Scholar] [CrossRef] [PubMed]
  168. Cutolo, M.; Smith, V.; Paolino, S.; Gotelli, E. Involvement of the Secosteroid Vitamin D in Autoimmune Rheumatic Diseases and COVID-19. Nat. Rev. Rheumatol. 2023, 19, 265–287. [Google Scholar] [CrossRef] [PubMed]
  169. Hyppönen, E.; Boucher, B.J.; Berry, D.J.; Power, C. 25-Hydroxyvitamin D, IGF-1, and Metabolic Syndrome at 45 Years of Age: A Cross-Sectional Study in the 1958 British Birth Cohort. Diabetes 2008, 57, 298–305. [Google Scholar] [CrossRef]
  170. Przybelski, R.J.; Binkley, N.C. Is Vitamin D Important for Preserving Cognition? A Positive Correlation of Serum 25-Hydroxyvitamin D Concentration with Cognitive Function. Arch. Biochem. Biophys. 2007, 460, 202–205. [Google Scholar] [CrossRef]
  171. Sousa, J.R.; Rosa, É.P.C.; Nunes, I.F.D.O.C.; Carvalho, C.M.R.G.D. Effect of Vitamin D Supplementation on Patients with Systemic Lupus Erythematosus: A Systematic Review. Rev. Bras. Reumatol. 2017, 57, 466–471. [Google Scholar] [CrossRef]
  172. Heine, G.; Drozdenko, G.; Lahl, A.; Unterwalder, N.; Mei, H.; Volk, H.-D.; Dörner, T.; Radbruch, A.; Worm, M. Efficient Tetanus Toxoid Immunization on Vitamin D Supplementation. Eur. J. Clin. Nutr. 2011, 65, 329–334. [Google Scholar] [CrossRef]
  173. Salman-Monte, T.C.; Torrente-Segarra, V.; Vega-Vidal, A.L.; Corzo, P.; Castro-Dominguez, F.; Ojeda, F.; Carbonell-Abelló, J. Bone Mineral Density and Vitamin D Status in Systemic Lupus Erythematosus (SLE): A Systematic Review. Autoimmun. Rev. 2017, 16, 1155–1159. [Google Scholar] [CrossRef]
  174. Wang, X.; Cheng, W.; Ma, Y.; Zhu, J. Vitamin D Receptor Gene FokI but Not TaqI, ApaI, BsmI Polymorphism Is Associated with Hashimoto’s Thyroiditis: A Meta-Analysis. Sci. Rep. 2017, 7, 41540. [Google Scholar] [CrossRef]
  175. Bhakat, B.; Pal, J.; Das, S.; Charaborty, S.K.; SircarMedical, N.R.; Kolkata; RGKar; Bengal, N.; Siliguri. A Prospective Study to Evaluate the Possible Role of Cholecalciferol Supplementation on Autoimmunity in Hashimoto’s Thyroiditis. J. Assoc. Physicians India 2023, 71, 1. [Google Scholar] [PubMed]
  176. Simsek, Y.; Cakir, I.; Yetmis, M.; Dizdar, O.; Baspinar, O.; Gokay, F. Effects of Vitamin D Treatment on Thyroid Autoimmunity. J. Res. Med. Sci. 2016, 21, 85. [Google Scholar] [CrossRef]
  177. Wang, S.; Wu, Y.; Zuo, Z.; Zhao, Y.; Wang, K. The Effect of Vitamin D Supplementation on Thyroid Autoantibody Levels in the Treatment of Autoimmune Thyroiditis: A Systematic Review and a Meta-Analysis. Endocrine 2018, 59, 499–505. [Google Scholar] [CrossRef]
  178. Tang, J.; Shan, S.; Li, F.; Yun, P. Effects of Vitamin D Supplementation on Autoantibodies and Thyroid Function in Patients with Hashimoto’s Thyroiditis: A Systematic Review and Meta-Analysis. Medicine 2023, 102, e36759. [Google Scholar] [CrossRef] [PubMed]
  179. Borgogni, E.; Sarchielli, E.; Sottili, M.; Santarlasci, V.; Cosmi, L.; Gelmini, S.; Lombardi, A.; Cantini, G.; Perigli, G.; Luconi, M.; et al. Elocalcitol Inhibits Inflammatory Responses in Human Thyroid Cells and T Cells. Endocrinology 2008, 149, 3626–3634. [Google Scholar] [CrossRef] [PubMed]
  180. Rafiq, S.; Jeppesen, P.B. Vitamin D Deficiency Is Inversely Associated with Homeostatic Model Assessment of Insulin Resistance. Nutrients 2021, 13, 4358. [Google Scholar] [CrossRef]
  181. Luong, K.V.Q.; Nguyen, L.T.H.; Pham Nguyen, D.N. The Role of Vitamin D in Protecting Type 1 Diabetes Mellitus. Diabetes Metabolism Res. 2005, 21, 338–346. [Google Scholar] [CrossRef]
  182. Danescu, L.G.; Levy, S.; Levy, J. Vitamin D and Diabetes Mellitus. Endocr 2009, 35, 11–17. [Google Scholar] [CrossRef]
  183. Yang, C.-Y.; Leung, P.S.C.; Adamopoulos, I.E.; Gershwin, M.E. The Implication of Vitamin D and Autoimmunity: A Comprehensive Review. Clinic Rev. Allerg. Immunol. 2013, 45, 217–226. [Google Scholar] [CrossRef]
  184. Norris, J.M.; Johnson, R.K.; Stene, L.C. Type 1 Diabetes—Early Life Origins and Changing Epidemiology. Lancet Diabetes Endocrinol. 2020, 8, 226–238. [Google Scholar] [CrossRef]
  185. Zella, J.B.; McCary, L.C.; DeLuca, H.F. Oral Administration of 1,25-Dihydroxyvitamin D3 Completely Protects NOD Mice from Insulin-Dependent Diabetes Mellitus. Arch. Biochem. Biophys. 2003, 417, 77–80. [Google Scholar] [CrossRef] [PubMed]
  186. 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] [PubMed]
  187. Casteels, K.M.; Mathieu, C.; Waer, M.; Valckx, D.; Overbergh, L.; Laureys, J.M.; Bouillon, R. Prevention of Type I Diabetes in Nonobese Diabetic Mice by Late Intervention with Nonhypercalcemic Analogs of 1,25-Dihydroxyvitamin D3 in Combination with a Short Induction Course of Cyclosporin A. Endocrinology 1998, 139, 95–102. [Google Scholar] [CrossRef]
  188. Fronczak, C.M.; Barón, A.E.; Chase, H.P.; Ross, C.; Brady, H.L.; Hoffman, M.; Eisenbarth, G.S.; Rewers, M.; Norris, J.M. In Utero Dietary Exposures and Risk of Islet Autoimmunity in Children. Diabetes Care 2003, 26, 3237–3242. [Google Scholar] [CrossRef]
  189. Hou, Y.; Song, A.; Jin, Y.; Xia, Q.; Song, G.; Xing, X. A Dose–Response Meta-Analysis between Serum Concentration of 25-Hydroxy Vitamin D and Risk of Type 1 Diabetes Mellitus. Eur. J. Clin. Nutr. 2021, 75, 1010–1023. [Google Scholar] [CrossRef] [PubMed]
  190. Storm, T.L.; Sørensen, O.H.; Lund, B.; Lund, B.; Christiansen, J.S.; Andersen, A.R.; Lumholtz, I.B.; Parving, H.-H. Vitamin D Metabolism in Insulin-Dependent Diabetes Mellitus. Metab. Bone Dis. Relat. Res. 1983, 5, 107–110. [Google Scholar] [CrossRef]
  191. Mathieu, C.; Badenhoop, K. Vitamin D and Type 1 Diabetes Mellitus: State of the Art. Trends Endocrinol. Metab. 2005, 16, 261–266. [Google Scholar] [CrossRef]
  192. Dong, J.-Y.; Zhang, W.; Chen, 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]
  193. Norris, J.M.; Lee, H.-S.; Frederiksen, B.; Erlund, I.; Uusitalo, U.; Yang, J.; Lernmark, Å.; 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]
  194. Hyppönen, E.; Läärä, E.; Reunanen, A.; Järvelin, M.-R.; Virtanen, S.M. Intake of Vitamin D and Risk of Type 1 Diabetes: A Birth-Cohort Study. Lancet 2001, 358, 1500–1503. [Google Scholar] [CrossRef] [PubMed]
  195. 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] [PubMed]
  196. Hu, Z.; Chen, J.; Sun, X.; Wang, L.; Wang, A. Efficacy of Vitamin D Supplementation on Glycemic Control in Type 2 Diabetes Patients: A Meta-Analysis of Interventional Studies. Medicine 2019, 98, e14970. [Google Scholar] [CrossRef]
  197. Zhao, J.; Dong, J.; Wang, H.; Shang, H.; Zhang, D.; Liao, L. Efficacy and Safety of Vitamin D3 in Patients with Diabetic Nephropathy: A Meta-Analysis of Randomized Controlled Trials. Chin. Med. J. 2014, 127, 2837–2843. [Google Scholar]
  198. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus–Infected Pneumonia in Wuhan, China. JAMA 2020, 323, 1061. [Google Scholar] [CrossRef]
  199. Corrao, S.; Gervasi, F.; Di Bernardo, F.; Natoli, G.; Raspanti, M.; Catalano, N.; Argano, C. Immunological Characteristics of Non-Intensive Care Hospitalized COVID-19 Patients: A Preliminary Report. J. Clin. Med. 2021, 10, 849. [Google Scholar] [CrossRef]
  200. Bergsbaken, T.; Fink, S.L.; Cookson, B.T. Pyroptosis: Host Cell Death and Inflammation. Nat. Rev. Microbiol. 2009, 7, 99–109. [Google Scholar] [CrossRef] [PubMed]
  201. Engelmann, B.; Massberg, S. Thrombosis as an Intravascular Effector of Innate Immunity. Nat. Rev. Immunol. 2013, 13, 34–45. [Google Scholar] [CrossRef] [PubMed]
  202. Zhao, N.; Di, B.; Xu, L. The NLRP3 Inflammasome and COVID-19: Activation, Pathogenesis and Therapeutic Strategies. Cytokine Growth Factor Rev. 2021, 61, 2–15. [Google Scholar] [CrossRef]
  203. Rodrigues, T.S.; De Sá, K.S.G.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Gonçalves, A.V.; Perucello, D.B.; Andrade, W.A.; Castro, R.; et al. Inflammasomes Are Activated in Response to SARS-CoV-2 Infection and Are Associated with COVID-19 Severity in Patients. J. Exp. Med. 2021, 218, e20201707. [Google Scholar] [CrossRef]
  204. Corrao, S.; Raspanti, M.; Agugliaro, F.; Gervasi, F.; Di Bernardo, F.; Natoli, G.; Argano, C. Safety of High-Dose Vitamin C in Non-Intensive Care Hospitalized Patients with COVID-19: An Open-Label Clinical Study. J. Clin. Med. 2024, 13, 3987. [Google Scholar] [CrossRef]
  205. Chen, X.; Zhang, Z.; Sun, N.; Li, J.; Ma, Z.; Rao, Z.; Sun, X.; Zeng, Q.; Wu, Y.; Li, J.; et al. Vitamin D Receptor Enhances NLRC4 Inflammasome Activation by Promoting NAIPs–NLRC4 Association. EMBO Rep. 2022, 23, e54611. [Google Scholar] [CrossRef] [PubMed]
  206. Gagnon, C.; Lu, Z.X.; Magliano, D.J.; Dunstan, D.W.; Shaw, J.E.; Zimmet, P.Z.; Sikaris, K.; Grantham, N.; Ebeling, P.R.; Daly, R.M. Serum 25-Hydroxyvitamin D, Calcium Intake, and Risk of Type 2 Diabetes After 5 Years. Diabetes Care 2011, 34, 1133–1138. [Google Scholar] [CrossRef] [PubMed]
  207. Mitri, J.; Dawson-Hughes, B.; Hu, F.B.; Pittas, A.G. Effects of Vitamin D and Calcium Supplementation on Pancreatic β Cell Function, Insulin Sensitivity, and Glycemia in Adults at High Risk of Diabetes: The Calcium and Vitamin D for Diabetes Mellitus (CaDDM) Randomized Controlled Trial. Am. J. Clin. Nutr. 2011, 94, 486–494. [Google Scholar] [CrossRef]
  208. Nazarian, S.; St Peter, J.V.; Boston, R.C.; Jones, S.A.; Mariash, C.N. Vitamin D3 Supplementation Improves Insulin Sensitivity in Subjects with Impaired Fasting Glucose. Transl. Res. 2011, 158, 276–281. [Google Scholar] [CrossRef] [PubMed]
  209. Pramono, A.; Jocken, J.W.E.; Blaak, E.E. Vitamin D Deficiency in the Aetiology of Obesity-related Insulin Resistance. Diabetes Metab. Res. 2019, 35, e3146. [Google Scholar] [CrossRef]
  210. Alvarez, J.A.; Ashraf, A. Role of Vitamin D in Insulin Secretion and Insulin Sensitivity for Glucose Homeostasis. Int. J. Endocrinol. 2010, 2010, 1–18. [Google Scholar] [CrossRef]
  211. Altieri, B.; Grant, W.B.; Della Casa, S.; Orio, F.; Pontecorvi, A.; Colao, A.; Sarno, G.; Muscogiuri, G. Vitamin D and Pancreas: The Role of Sunshine Vitamin in the Pathogenesis of Diabetes Mellitus and Pancreatic Cancer. Crit. Rev. Food Sci. Nutr. 2017, 57, 3472–3488. [Google Scholar] [CrossRef]
  212. Cade, C.; Norman, A.W. Vitamin D3 Improves Impaired Glucose Tolerance and Insulin Secretion in the Vitamin D-Deficient Rat in Vivo. Endocrinology 1986, 119, 84–90. [Google Scholar] [CrossRef]
  213. Zeitz, U.; Weber, K.; Soegiarto, D.W.; Wolf, E.; Balling, R.; Erben, R.G. Impaired Insulin Secretory Capacity in Mice Lacking a Functional Vitamin D Receptor. FASEB J. 2003, 17, 1–14. [Google Scholar] [CrossRef]
  214. Bland, R.; Markovic, D.; Hills, C.E.; Hughes, S.V.; Chan, S.L.F.; Squires, P.E.; Hewison, M. Expression of 25-Hydroxyvitamin D3-1α-Hydroxylase in Pancreatic Islets. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 121–125. [Google Scholar] [CrossRef]
  215. Reusch, J.E.-B.; Begum, N.; Sussman, K.E.; Draznin, B. Regulation of GLUT-4 Phosphorylation by Intracellular Calcium in Adipocytes. Endocrinology 1991, 129, 3269–3273. [Google Scholar] [CrossRef] [PubMed]
  216. Baron, A.D.; Brechtel, G.; Wallace, P.; Edelman, S.V. Rates and Tissue Sites of Non-Insulin- and Insulin-Mediated Glucose Uptake in Humans. Am. J. Physiol.-Endocrinol. Metab. 1988, 255, E769–E774. [Google Scholar] [CrossRef] [PubMed]
  217. DeFronzo, R.A.; Jacot, E.; Jequier, E.; Maeder, E.; Wahren, J.; Felber, J.P. The Effect of Insulin on the Disposal of Intravenous Glucose: Results from Indirect Calorimetry and Hepatic and Femoral Venous Catheterization. Diabetes 1981, 30, 1000–1007. [Google Scholar] [CrossRef]
  218. Jefferson, G.E.; Schnell, D.M.; Thomas, D.T.; Bollinger, L.M. Calcitriol Concomitantly Enhances Insulin Sensitivity and Alters Myocellular Lipid Partitioning in High Fat-Treated Skeletal Muscle Cells. J. Physiol. Biochem. 2017, 73, 613–621. [Google Scholar] [CrossRef] [PubMed]
  219. Krul-Poel, Y.H.M.; Ter Wee, M.M.; Lips, P.; Simsek, S. Management of Endocrine Disease: The Effect of Vitamin D Supplementation on Glycaemic Control in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Eur. J. Endocrinol. 2017, 176, R1–R14. [Google Scholar] [CrossRef]
  220. Earthman, C.P.; Beckman, L.M.; Masodkar, K.; Sibley, S.D. The Link between Obesity and Low Circulating 25-Hydroxyvitamin D Concentrations: Considerations and Implications. Int. J. Obes. 2012, 36, 387–396. [Google Scholar] [CrossRef]
  221. Bajaj, S.; Singh, R.; Dwivedi, N.; Singh, K.; Gupta, A.; Mathur, M. Vitamin D Levels and Microvascular Complications in Type 2 Diabetes. Indian. J. Endocr. Metab. 2014, 18, 537. [Google Scholar] [CrossRef]
  222. De Vita, F.; Lauretani, F.; Bauer, J.; Bautmans, I.; Shardell, M.; Cherubini, A.; Bondi, G.; Zuliani, G.; Bandinelli, S.; Pedrazzoni, M.; et al. Relationship between Vitamin D and Inflammatory Markers in Older Individuals. AGE 2014, 36, 9694. [Google Scholar] [CrossRef]
  223. Laird, E.; McNulty, H.; Ward, M.; Hoey, L.; McSorley, E.; Wallace, J.M.W.; Carson, E.; Molloy, A.M.; Healy, M.; Casey, M.C.; et al. Vitamin D Deficiency Is Associated With Inflammation in Older Irish Adults. J. Clin. Endocrinol. Metab. 2014, 99, 1807–1815. [Google Scholar] [CrossRef]
  224. Kruit, A.; Zanen, P. The Association between Vitamin D and C-Reactive Protein Levels in Patients with Inflammatory and Non-Inflammatory Diseases. Clin. Biochem. 2016, 49, 534–537. [Google Scholar] [CrossRef] [PubMed]
  225. Ilie, P.C.; Stefanescu, S.; Smith, L. The Role of Vitamin D in the Prevention of Coronavirus Disease 2019 Infection and Mortality. Aging Clin. Exp. Res. 2020, 32, 1195–1198. [Google Scholar] [CrossRef] [PubMed]
  226. Giannini, S.; Passeri, G.; Tripepi, G.; Sella, S.; Fusaro, M.; Arcidiacono, G.; Torres, M.O.; Michielin, A.; Prandini, T.; Baffa, V.; et al. Effectiveness of In-Hospital Cholecalciferol Use on Clinical Outcomes in Comorbid COVID-19 Patients: A Hypothesis-Generating Study. Nutrients 2021, 13, 219. [Google Scholar] [CrossRef]
  227. Vargas Vargas, R.A.; Varela Millán, J.M.; Fajardo Bonilla, E. Renin–Angiotensin System: Basic and Clinical Aspects—A General Perspective. Endocrinol. Diabetes Nutr. 2022, 69, 52–62. [Google Scholar] [CrossRef] [PubMed]
  228. Gaddam, R.; Chambers, S.; Bhatia, M. ACE and ACE2 in Inflammation: A Tale of Two Enzymes. Inflamm. Allergy-Drug Targets-Inflamm. Allergy 2014, 13, 224–234. [Google Scholar] [CrossRef]
  229. Lei, C.; Qian, K.; Li, T.; Zhang, S.; Fu, W.; Ding, M.; Hu, S. Neutralization of SARS-CoV-2 Spike Pseudotyped Virus by Recombinant ACE2-Ig. Nat. Commun. 2020, 11, 2070. [Google Scholar] [CrossRef]
  230. Fedson, D.S. Treating the Host Response to Emerging Virus Diseases: Lessons Learned from Sepsis, Pneumonia, Influenza and Ebola. Ann. Transl. Med. 2016, 4, 421. [Google Scholar] [CrossRef]
  231. Wee, C.L.; Azemi, A.K.; Mokhtar, S.S.; Yahaya, S.; Yaacob, N.S.; Rasool, A.H.G. Vitamin D Deficiency Enhances Vascular Oxidative Stress, Inflammation, and Angiotensin II Levels in the Microcirculation of Diabetic Patients. Microvasc. Res. 2023, 150, 104574. [Google Scholar] [CrossRef]
  232. Kong, J.; Zhu, X.; Shi, Y.; Liu, T.; Chen, Y.; Bhan, I.; Zhao, Q.; Thadhani, R.; Li, Y.C. VDR Attenuates Acute Lung Injury by Blocking Ang-2-Tie-2 Pathway and Renin-Angiotensin System. Mol. Endocrinol. 2013, 27, 2116–2125. [Google Scholar] [CrossRef]
  233. Marshall, R.P.; McANULTY, R.J.; Laurent, G.J. Angiotensin II Is Mitogenic for Human Lung Fibroblasts via Activation of the Type 1 Receptor. Am. J. Respir. Crit. Care Med. 2000, 161, 1999–2004. [Google Scholar] [CrossRef]
  234. Li, L.; Huang, T.; Wang, Y.; Wang, Z.; Liang, Y.; Huang, T.; Zhang, H.; Sun, W.; Wang, Y. COVID-19 Patients’ Clinical Characteristics, Discharge Rate, and Fatality Rate of Meta-analysis. J. Med. Virol. 2020, 92, 577–583. [Google Scholar] [CrossRef] [PubMed]
  235. Bertram, S.; Glowacka, I.; Müller, M.A.; Lavender, H.; Gnirss, K.; Nehlmeier, I.; Niemeyer, D.; He, Y.; Simmons, G.; Drosten, C.; et al. Cleavage and Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein by Human Airway Trypsin-Like Protease. J. Virol. 2011, 85, 13363–13372. [Google Scholar] [CrossRef]
  236. Ali, R.M.; Al-Shorbagy, M.Y.; Helmy, M.W.; El-Abhar, H.S. Role of Wnt4/β-Catenin, Ang II/TGFβ, ACE2, NF-κB, and IL-18 in Attenuating Renal Ischemia/Reperfusion-Induced Injury in Rats Treated with Vit D and Pioglitazone. Eur. J. Pharmacol. 2018, 831, 68–76. [Google Scholar] [CrossRef]
  237. Cui, C.; Xu, P.; Li, G.; Qiao, Y.; Han, W.; Geng, C.; Liao, D.; Yang, M.; Chen, D.; Jiang, P. Vitamin D Receptor Activation Regulates Microglia Polarization and Oxidative Stress in Spontaneously Hypertensive Rats and Angiotensin II-Exposed Microglial Cells: Role of Renin-Angiotensin System. Redox Biol. 2019, 26, 101295. [Google Scholar] [CrossRef]
  238. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
  239. Xu, J.; Yang, J.; Chen, J.; Luo, Q.; Zhang, Q.; Zhang, H. Vitamin D Alleviates Lipopolysaccharide-Induced Acute Lung Injury via Regulation of the Renin-Angiotensin System. Mol. Med. Rep. 2017, 16, 7432–7438. [Google Scholar] [CrossRef]
  240. Zhou, C.; Lu, F.; Cao, K.; Xu, D.; Goltzman, D.; Miao, D. Calcium-Independent and 1,25(OH)2D3-Dependent Regulation of the Renin-Angiotensin System in 1α-Hydroxylase Knockout Mice. Kidney Int. 2008, 74, 170–179. [Google Scholar] [CrossRef]
  241. Tiwari, V.; Singh, J.; Tiwari, P.; Chaturvedi, S.; Gupta, S.; Mishra, A.; Singh, S.; Wahajuddin, M.; Hanif, K.; Shukla, S. ACE2/ANG-(1-7)/Mas Receptor Axis Activation Prevents Inflammation and Improves Cognitive Functions in Streptozotocin Induced Rat Model of Alzheimer’s Disease-like Phenotypes. Eur. J. Pharmacol. 2023, 946, 175623. [Google Scholar] [CrossRef]
  242. Cui, D.; Liu, Y.; Jiang, X.; Ding, C.; Poon, L.C.; Wang, H.; Yang, H. Single-cell RNA Expression Profiling of SARS-CoV-2-related ACE2 and TMPRSS2 in Human Trophectoderm and Placenta. Ultrasound Obs. Gyne 2021, 57, 248–256. [Google Scholar] [CrossRef]
  243. Lin, S.; Pan, H.; Wu, H.; Ren, D.; Lu, J. Role of the ACE2-Ang-(1-7)-Mas Axis in Blood Pressure Regulation and Its Potential as an Antihypertensive in Functional Foods. Mol. Med. Rep. 2017, 16, 4403–4412. [Google Scholar] [CrossRef] [PubMed]
  244. Getachew, B.; Tizabi, Y. Vitamin D and COVID-19: Role of ACE2, Age, Gender, and Ethnicity. J. Med. Virol. 2021, 93, 5285–5294. [Google Scholar] [CrossRef]
  245. Beltrán-García, J.; Osca-Verdegal, R.; Pallardó, F.V.; Ferreres, J.; Rodríguez, M.; Mulet, S.; Sanchis-Gomar, F.; Carbonell, N.; García-Giménez, J.L. Oxidative Stress and Inflammation in COVID-19-Associated Sepsis: The Potential Role of Anti-Oxidant Therapy in Avoiding Disease Progression. Antioxidants 2020, 9, 936. [Google Scholar] [CrossRef] [PubMed]
  246. Ghosh, A.; Mukhyaprana Prabhu, M.; Prabhu, K.; Changani, M.; Patel, N.; Belle, V.S. Vitamin D as a Biomarker in Predicting Sepsis Outcome at a Tertiary Care Hospital. Asian J. Med. Sci. 2023, 14, 89–94. [Google Scholar] [CrossRef]
  247. Corrao, S.; Gervasi, F.; Di Bernardo, F.; Argano, C. Immune Response Failure in Paucisymptomatic Long-Standing SARS-CoV-2 Spreaders. Clin. Pract. 2021, 11, 151–161. [Google Scholar] [CrossRef] [PubMed]
  248. Kongsbak, M.; Von Essen, M.R.; Boding, L.; Levring, T.B.; Schjerling, P.; Lauritsen, J.P.H.; Woetmann, A.; Ødum, N.; Bonefeld, C.M.; Geisler, C. Vitamin D Up-Regulates the Vitamin D Receptor by Protecting It from Proteasomal Degradation in Human CD4+ T Cells. PLoS ONE 2014, 9, e96695. [Google Scholar] [CrossRef]
  249. Abdel-Mohsen, M.A.; El-Braky, A.A.-A.; Ghazal, A.A.E.-R.; Shamseya, M.M. Autophagy, Apoptosis, Vitamin D, and Vitamin D Receptor in Hepatocellular Carcinoma Associated with Hepatitis C Virus. Medicine 2018, 97, e0172. [Google Scholar] [CrossRef] [PubMed]
  250. Gunville, C.; Mourani, P.; Ginde, A. The Role of Vitamin D in Prevention and Treatment of Infection. Inflamm. Allergy-Drug Targets 2013, 12, 239–245. [Google Scholar] [CrossRef]
  251. Bouillon, R.; Marcocci, C.; Carmeliet, G.; Bikle, D.; White, J.H.; Dawson-Hughes, B.; Lips, P.; Munns, C.F.; Lazaretti-Castro, M.; Giustina, A.; et al. Skeletal and Extraskeletal Actions of Vitamin D: Current Evidence and Outstanding Questions. Endocr. Rev. 2019, 40, 1109–1151. [Google Scholar] [CrossRef]
  252. Equils, O.; Naiki, Y.; Shapiro, A.M.; Michelsen, K.; Lu, D.; Adams, J.; Jordan, S. 1,25-Dihydroxyvitamin D3 Inhibits Lipopolysaccharide-Induced Immune Activation in Human Endothelial Cells. Clin. Exp. Immunol. 2005, 143, 58–64. [Google Scholar] [CrossRef]
  253. Ginde, A.A.; Camargo, C.A.; Shapiro, N.I. Vitamin D Insufficiency and Sepsis Severity in Emergency Department Patients With Suspected Infection. Acad. Emerg. Med. 2011, 18, 551–554. [Google Scholar] [CrossRef]
  254. Bergman, P.; Norlin, A.-C.; Hansen, S.; Rekha, R.S.; Agerberth, B.; Björkhem-Bergman, L.; Ekström, L.; Lindh, J.D.; Andersson, J. Vitamin D3 Supplementation in Patients with Frequent Respiratory Tract Infections: A Randomised and Double-Blind Intervention Study. BMJ Open 2012, 2, e001663. [Google Scholar] [CrossRef]
  255. 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] [PubMed]
  256. 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. [Google Scholar] [CrossRef]
  257. Anitua, E.; Tierno, R.; Alkhraisat, M.H. Current Opinion on the Role of Vitamin D Supplementation in Respiratory Infections and Asthma/COPD Exacerbations: A Need to Establish Publication Guidelines for Overcoming the Unpublished Data. Clin. Nutr. 2022, 41, 755–777. [Google Scholar] [CrossRef] [PubMed]
  258. Meng, J.; Li, X.; Liu, W.; Xiao, Y.; Tang, H.; Wu, Y.; Xiong, Y.; Gao, S. The Role of Vitamin D in the Prevention and Treatment of SARS-CoV-2 Infection: A Meta-Analysis of Randomized Controlled Trials. Clin. Nutr. 2023, 42, 2198–2206. [Google Scholar] [CrossRef] [PubMed]
  259. Petrelli, F.; Luciani, A.; Perego, G.; Dognini, G.; Colombelli, P.L.; Ghidini, A. Therapeutic and Prognostic Role of Vitamin D for COVID-19 Infection: A Systematic Review and Meta-Analysis of 43 Observational Studies. J. Steroid Biochem. Mol. Biol. 2021, 211, 105883. [Google Scholar] [CrossRef]
  260. Miroliaee, A.E.; Salamzadeh, J.; Shokouhi, S.; Fatemi, A.; Ardehali, S.H.; Hajiesmaeili, M.R.; Sahraei, Z. Effect of Vitamin D Supplementation on Procalcitonin as Prognostic Biomarker in Patients with Ventilator Associated Pneumonia Complicated with Vitamin D Deficiency. Iran. J. Pharm. Res. 2017, 16, 1254–1263. [Google Scholar]
  261. Hosseini, B.; Tremblay, C.L.; Longo, C.; Glochi, S.; White, J.H.; Quach, C.; Ste-Marie, L.-G.; Platt, R.W.; Ducharme, F.M. Oral Vitamin D Supplemental Therapy to Attain a Desired Serum 25-Hydroxyvitamin D Concentration in Essential Healthcare Teams. Trials 2022, 23, 1019. [Google Scholar] [CrossRef]
  262. Theodorou, M.; Sersté, T.; Van Gossum, M.; Dewit, S. Factors Associated with Vitamin D Deficiency in a Population of 2044 HIV-Infected Patients. Clin. Nutr. 2014, 33, 274–279. [Google Scholar] [CrossRef]
  263. Coelho, L.; Cardoso, S.W.; Luz, P.M.; Hoffman, R.M.; Mendonça, L.; Veloso, V.G.; Currier, J.S.; Grinsztejn, B.; Lake, J.E. Vitamin D3 Supplementation in HIV Infection: Effectiveness and Associations with Antiretroviral Therapy. Nutr. J. 2015, 14, 81. [Google Scholar] [CrossRef]
  264. Shepherd, L.; Souberbielle, J.-C.; Bastard, J.-P.; Fellahi, S.; Capeau, J.; Reekie, J.; Reiss, P.; Blaxhult, A.; Bickel, M.; Leen, C.; et al. Prognostic Value of Vitamin D Level for All-Cause Mortality, and Association With Inflammatory Markers, in HIV-Infected Persons. J. Infect. Dis. 2014, 210, 234–243. [Google Scholar] [CrossRef] [PubMed]
  265. Viard, J.-P.; Souberbielle, J.-C.; Kirk, O.; Reekie, J.; Knysz, B.; Losso, M.; Gatell, J.; Pedersen, C.; Bogner, J.R.; Lundgren, J.D.; et al. Vitamin D and Clinical Disease Progression in HIV Infection: Results from the EuroSIDA Study. AIDS 2011, 25, 1305–1315. [Google Scholar] [CrossRef]
  266. Campbell, G.R.; Spector, S.A. Hormonally Active Vitamin D3 (1α,25-Dihydroxycholecalciferol) Triggers Autophagy in Human Macrophages That Inhibits HIV-1 Infection. J. Biol. Chem. 2011, 286, 18890–18902. [Google Scholar] [CrossRef]
  267. Arpadi, S.M.; McMahon, D.; Abrams, E.J.; Bamji, M.; Purswani, M.; Engelson, E.S.; Horlick, M.; Shane, E. Effect of Bimonthly Supplementation With Oral Cholecalciferol on Serum 25-Hydroxyvitamin D Concentrations in HIV-Infected Children and Adolescents. Pediatrics 2009, 123, e121–e126. [Google Scholar] [CrossRef] [PubMed]
  268. Xie, K.; Zhang, Y.; Zhang, M.; Wu, H.; Zheng, L.; Ji, J.; Li, Z.; Wang, W.; Zhang, T. Association of Vitamin D with HIV Infected Individuals, TB Infected Individuals, and HIV-TB Co-Infected Individuals: A Systematic Review and Meta-Analysis. Front. Public Health 2024, 12, 1344024. [Google Scholar] [CrossRef]
  269. Rezamand, G.; Estêvão, M.D.; Morvaridzadeh, M.; Akbari, A.; Tabaeian, S.P.; Pizarro, A.B.; Malekahmadi, M.; Hasani, M.; Roffey, D.M.; Mirzaei, A.; et al. Effects of Vitamin D Supplementation on Bone Health and Bone-Related Parameters in HIV-Infected Patients: A Systematic Review and Meta-Analysis. Clin. Ther. 2022, 44, e11-25.e8. [Google Scholar] [CrossRef]
  270. Pierrot-Deseilligny, C.; Souberbielle, J.-C. Vitamin D and Multiple Sclerosis: An Update. Mult. Scler. Relat. Disord. 2017, 14, 35–45. [Google Scholar] [CrossRef]
  271. Lassmann, H.; Bradl, M. Multiple Sclerosis: Experimental Models and Reality. Acta Neuropathol. 2017, 133, 223–244. [Google Scholar] [CrossRef] [PubMed]
  272. The Australia and New Zealand Multiple Sclerosis Genetics Consortium (ANZgene) Genome-Wide Association Study Identifies New Multiple Sclerosis Susceptibility Loci on Chromosomes 12 and 20. Nat. Genet. 2009, 41, 824–828. [CrossRef]
  273. Pierrot-Deseilligny, C.; Souberbielle, J.-C. Contribution of Vitamin D Insufficiency to the Pathogenesis of Multiple Sclerosis. Ther. Adv. Neurol. Disord. 2013, 6, 81–116. [Google Scholar] [CrossRef]
  274. Vieth, R.; Fraser, D. Vitamin D Insufficiency: No Recommended Dietary Allowance Exists for This Nutrient. CMAJ 2002, 166, 1541–1542. [Google Scholar]
  275. Mowry, E.M.; Waubant, E.; McCulloch, C.E.; Okuda, D.T.; Evangelista, A.A.; Lincoln, R.R.; Gourraud, P.; Brenneman, D.; Owen, M.C.; Qualley, P.; et al. Vitamin D Status Predicts New Brain Magnetic Resonance Imaging Activity in Multiple Sclerosis. Ann. Neurol. 2012, 72, 234–240. [Google Scholar] [CrossRef]
  276. Dickinson, J.; Perera, D.; Van Der Mei, A.; Ponsonby, A.-L.; Polanowski, A.; Thomson, R.; Taylor, B.; McKay, J.; Stankovich, J.; Dwyer, T. Past Environmental Sun Exposure and Risk of Multiple Sclerosis: A Role for the Cdx-2 Vitamin D Receptor Variant in This Interaction. Mult. Scler. 2009, 15, 563–570. [Google Scholar] [CrossRef]
  277. Lv, Y.; Yao, Q.; Ma, W.; Liu, H.; Ji, J.; Li, X. Associations of Vitamin D Deficiency and Vitamin D Receptor (Cdx-2, Fok I, Bsm I and Taq I) Polymorphisms with the Risk of Primary Open-Angle Glaucoma. BMC Ophthalmol. 2016, 16, 116. [Google Scholar] [CrossRef] [PubMed]
  278. Jurutka, P.W.; Remus, L.S.; Whitfield, G.K.; Thompson, P.D.; Hsieh, J.-C.; Zitzer, H.; Tavakkoli, P.; Galligan, M.A.; Dang, H.T.L.; Haussler, C.A.; et al. The Polymorphic N Terminus in Human Vitamin D Receptor Isoforms Influences Transcriptional Activity by Modulating Interaction with Transcription Factor IIB. Mol. Endocrinol. 2000, 14, 401–420. [Google Scholar] [CrossRef]
  279. Uitterlinden, A.G.; Fang, Y.; Van Meurs, J.B.J.; Pols, H.A.P.; Van Leeuwen, J.P.T.M. Genetics and Biology of Vitamin D Receptor Polymorphisms. Gene 2004, 338, 143–156. [Google Scholar] [CrossRef] [PubMed]
  280. O’Gorman, C.; Lucas, R.; Taylor, B. Environmental Risk Factors for Multiple Sclerosis: A Review with a Focus on Molecular Mechanisms. Int. J. Mol. Sci. 2012, 13, 11718–11752. [Google Scholar] [CrossRef] [PubMed]
  281. Handel, A.E.; Sandve, G.K.; Disanto, G.; Berlanga-Taylor, A.J.; Gallone, G.; Hanwell, H.; Drabløs, F.; Giovannoni, G.; Ebers, G.C.; Ramagopalan, S.V. Vitamin D Receptor ChIP-Seq in Primary CD4+ Cells: Relationship to Serum 25-Hydroxyvitamin D Levels and Autoimmune Disease. BMC Med. 2013, 11, 163. [Google Scholar] [CrossRef]
  282. 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]
  283. Smolders, J.; Peelen, E.; Thewissen, M.; Menheere, P.; Cohen Tervaert, J.W.; Hupperts, R.; Damoiseaux, J. The Relevance of Vitamin D Receptor Gene Polymorphisms for Vitamin D Research in Multiple Sclerosis. Autoimmun. Rev. 2009, 8, 621–626. [Google Scholar] [CrossRef]
  284. Simon, K.C.; Munger, K.L.; Yang, X.; Ascherio, A. Polymorphisms in Vitamin D Metabolism Related Genes and Risk of Multiple Sclerosis. Mult. Scler. 2010, 16, 133–138. [Google Scholar] [CrossRef] [PubMed]
  285. Boltjes, R.; Knippenberg, S.; Gerlach, O.; Hupperts, R.; Damoiseaux, J. Vitamin D Supplementation in Multiple Sclerosis: An Expert Opinion Based on the Review of Current Evidence. Expert Rev. Neurother. 2021, 21, 715–725. [Google Scholar] [CrossRef]
  286. Yuan, X.; Guo, L.; Jiang, C.; Yang, X.; Huang, J. The Effect of Different Administration Time and Dosage of Vitamin D Supplementation in Patients with Multiple Sclerosis: A Meta-Analysis of Randomized Controlled Trials. Neuroimmunomodulation 2021, 28, 118–128. [Google Scholar] [CrossRef]
  287. Jagannath, V.A.; Filippini, G.; Borges Do Nascimento, I.J.; Di Pietrantonj, C.; Robak, E.W.; Whamond, L. Vitamin D for the Management of Multiple Sclerosis. Cochrane Database Syst. Rev. 2018, 2023. [Google Scholar] [CrossRef]
  288. Nielsen, N.M.; Munger, K.L.; Koch-Henriksen, N.; Hougaard, D.M.; Magyari, M.; Jørgensen, 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]
  289. Mei, I.A.F.; Ponsonby, A.-L.; Dwyer, T.; Blizzard, L.; Taylor, B.V.; Kilpatrick, T.; Butzkueven, H.; McMichael, A.J. Vitamin D Levels in People with Multiple Sclerosis and Community Controls in Tasmania, Australia. J. Neurol. 2007, 254, 581–590. [Google Scholar] [CrossRef]
  290. McLaughlin, L.; Clarke, L.; Khalilidehkordi, E.; Butzkueven, H.; Taylor, B.; Broadley, S.A. Vitamin D for the Treatment of Multiple Sclerosis: A Meta-Analysis. J. Neurol. 2018, 265, 2893–2905. [Google Scholar] [CrossRef]
  291. Berezowska, M.; Coe, S.; Dawes, H. Effectiveness of Vitamin D Supplementation in the Management of Multiple Sclerosis: A Systematic Review. Int. J. Mol. Sci. 2019, 20, 1301. [Google Scholar] [CrossRef]
  292. Chen, Z.; Peng, C.; Mei, J.; Zhu, L.; Kong, H. Vitamin D Can Safely Reduce Asthma Exacerbations among Corticosteroid-Using Children and Adults with Asthma: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutr. Res. 2021, 92, 49–61. [Google Scholar] [CrossRef]
  293. Etemadifar, M.; Janghorbani, M. Efficacy of High-Dose Vitamin D3 Supplementation in Vitamin D Deficient Pregnant Women with Multiple Sclerosis: Preliminary Findings of a Randomized-Controlled Trial. Iran. J. Neurol. 2015, 14, 67–73. [Google Scholar]
  294. Thouvenot, E.; Laplaud, D.; Lebrun-Frenay, C.; Derache, N.; Le Page, E.; Maillart, E.; Froment-Tilikete, C.; Castelnovo, G.; Casez, O.; Coustans, M.; et al. High-Dose Vitamin D in Clinically Isolated Syndrome Typical of Multiple Sclerosis: The D-Lay MS Randomized Clinical Trial. JAMA 2025, 333, 1413. [Google Scholar] [CrossRef] [PubMed]
  295. Majak, P.; Olszowiec-Chlebna, M.; Smejda, K.; Stelmach, I. Vitamin D Supplementation in Children May Prevent Asthma Exacerbation Triggered by Acute Respiratory Infection. J. Allergy Clin. Immunol. 2011, 127, 1294–1296. [Google Scholar] [CrossRef] [PubMed]
  296. Nanzer, A.M.; Chambers, E.S.; Ryanna, K.; Freeman, A.T.; Colligan, G.; Richards, D.F.; Timms, P.M.; Martineau, A.R.; Griffiths, C.J.; Corrigan, C.J.; et al. The Effects of Calcitriol Treatment in Glucocorticoid-Resistant Asthma. J. Allergy Clin. Immunol. 2014, 133, 1755–1757.e4. [Google Scholar] [CrossRef] [PubMed]
  297. Wang, M.; Liu, M.; Wang, C.; Xiao, Y.; An, T.; Zou, M.; Cheng, G. Association between Vitamin D Status and Asthma Control: A Meta-Analysis of Randomized Trials. Respir. Med. 2019, 150, 85–94. [Google Scholar] [CrossRef]
  298. Pojsupap, S.; Iliriani, K.; Sampaio, T.Z.A.L.; O’Hearn, K.; Kovesi, T.; Menon, K.; McNally, J.D. Efficacy of High-Dose Vitamin D in Pediatric Asthma: A Systematic Review and Meta-Analysis. J. Asthma 2015, 52, 382–390. [Google Scholar] [CrossRef]
  299. Niu, H.; He, H.; Zhao, Z.; Lu, X.; Zhao, G. Asthmatic Patients with Vitamin D Deficiency: Can Vitamin D Supplementation Make a Difference. Technol. Health Care 2024, 32, 3985–4008. [Google Scholar] [CrossRef]
  300. Riverin, B.D.; Maguire, J.L.; Li, P. Vitamin D Supplementation for Childhood Asthma: A Systematic Review and Meta-Analysis. PLoS ONE 2015, 10, e0136841. [Google Scholar] [CrossRef]
  301. Andjelkovic, Z.; Vojinovic, J.; Pejnovic, N.; Popovic, M.; Dujic, A.; Mitrovic, D.; Pavlica, L.; Stefanovic, D. Disease Modifying and Immunomodulatory Effects of High Dose 1 Alpha (OH) D3 in Rheumatoid Arthritis Patients. Clin. Exp. Rheumatol. 1999, 17, 453–456. [Google Scholar]
  302. Zhang, J.; Chen, Y.; Li, H.; Li, H. Effects of Vitamin D on Thyroid Autoimmunity Markers in Hashimoto’s Thyroiditis: Systematic Review and Meta-Analysis. J. Int. Med. Res. 2021, 49, 03000605211060675. [Google Scholar] [CrossRef]
  303. Gregoriou, E.; Mamais, I.; Tzanetakou, I.; Lavranos, G.; Chrysostomou, S. The Effects of Vitamin D Supplementation in Newly Diagnosed Type 1 Diabetes Patients: Systematic Review of Randomized Controlled Trials. Rev. Diabet. Stud. 2017, 14, 260–268. [Google Scholar] [CrossRef]
  304. Najjar, L.; Sutherland, J.; Zhou, A.; Hyppönen, E. Vitamin D and Type 1 Diabetes Risk: A Systematic Review and Meta-Analysis of Genetic Evidence. Nutrients 2021, 13, 4260. [Google Scholar] [CrossRef] [PubMed]
  305. Nascimento, B.F.; Moreira, C.F.F.; Da Fonseca, E.R.; Fedeszen, P.M.K.; De Paula, T.P.; De Sena, A.S.S.; De Almeida, N.F.A.; Bandeira Filho, O.C.D.S.; Curval, D.R.; Padilha, P.D.C. Effects of Vitamin D Supplementation on Glycemic Control of Children and Adolescents with Type 1 Diabetes Mellitus: A Systematic Review. J. Pediatr. Endocrinol. Metab. 2022, 35, 973–988. [Google Scholar] [CrossRef] [PubMed]
  306. Yu, J.; Sharma, P.; Girgis, C.M.; Gunton, J.E. Vitamin D and Beta Cells in Type 1 Diabetes: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 14434. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The immune-mediated and anti-inflammatory actions of vitamin D from bench to bedside.
Figure 1. The immune-mediated and anti-inflammatory actions of vitamin D from bench to bedside.
Ijms 26 04798 g001
Figure 2. Synthesis and mechanism of action of vitamin D on bone metabolism and, in particular, on innate and adaptive immunity cells. Transcription factors FOXP3 (Forkhead box P3), GATA3, TBX21 (T-bet), RUNX1, RORγt, and NF-kb (protein complex functioning as a transcription factor) with its isoform p65. P35 and P40 are subunits of IL-12. VDRE is a type of DNA sequence found in the promoter region of vitamin D-regulated genes.
Figure 2. Synthesis and mechanism of action of vitamin D on bone metabolism and, in particular, on innate and adaptive immunity cells. Transcription factors FOXP3 (Forkhead box P3), GATA3, TBX21 (T-bet), RUNX1, RORγt, and NF-kb (protein complex functioning as a transcription factor) with its isoform p65. P35 and P40 are subunits of IL-12. VDRE is a type of DNA sequence found in the promoter region of vitamin D-regulated genes.
Ijms 26 04798 g002
Figure 3. Mechanism of action of vitamin D on monocytes and macrophages. TLRs expressed on monocytes’ and macrophages’ surfaces recognize components of bacteria, fungi, and viruses, upregulating the expression of VDR and CYP27B1. CYP27B1 metabolizes 25-hydroxyvitamin D into calcitriol and binds to VDR. The activated VDR forms a heterodimer with RXR, which induces the synthesis of cathelicidin, β-defensin 2 that promotes bacterial and viral autophagy. 25(OH)D: 25-hydroxyvitamin D; 1,25(OH)2D: 1-25-Dihydroxyvitamin D, NF-kb: Nuclear Factor Kappa light chain-enhancer of activated B cells; IL-1R: interleukin-1 receptor; NOD2: Nucleotide-binding oligomerization domain-containing protein 2; TLR: Toll-Like Receptor; VDR: vitamin D receptor.
Figure 3. Mechanism of action of vitamin D on monocytes and macrophages. TLRs expressed on monocytes’ and macrophages’ surfaces recognize components of bacteria, fungi, and viruses, upregulating the expression of VDR and CYP27B1. CYP27B1 metabolizes 25-hydroxyvitamin D into calcitriol and binds to VDR. The activated VDR forms a heterodimer with RXR, which induces the synthesis of cathelicidin, β-defensin 2 that promotes bacterial and viral autophagy. 25(OH)D: 25-hydroxyvitamin D; 1,25(OH)2D: 1-25-Dihydroxyvitamin D, NF-kb: Nuclear Factor Kappa light chain-enhancer of activated B cells; IL-1R: interleukin-1 receptor; NOD2: Nucleotide-binding oligomerization domain-containing protein 2; TLR: Toll-Like Receptor; VDR: vitamin D receptor.
Ijms 26 04798 g003
Figure 4. Mechanism of action of vitamin D on dendritic cells and lymphocytes. In particular, calcitriol promotes the expression of the anti-inflammatory cytokine IL-10, and it suppresses IL-12 production, which determines the differentiation of T helper cells into Th1 cells. Conversely, the anti-inflammatory cytokine IL-10 promotes the differentiation of regulatory T cells. Moreover, calcitriol promotes Th2-type differentiation and the secretion of IL-4, IL-5, and IL-10, anti-inflammatory cytokines. 1,25(OH)2D: 1-25-Dihydroxyvitamin D; IL-2: interLeukin-12; IL-10: interLeukin-10; Th1: T Helper 1 lymphocytes; Th2: T Helper 2 lymphocytes; T reg: T regulatory cells; TGF-β: transforming growth factor beta; TNF-α: Tumor Necrosis Factor alpha.
Figure 4. Mechanism of action of vitamin D on dendritic cells and lymphocytes. In particular, calcitriol promotes the expression of the anti-inflammatory cytokine IL-10, and it suppresses IL-12 production, which determines the differentiation of T helper cells into Th1 cells. Conversely, the anti-inflammatory cytokine IL-10 promotes the differentiation of regulatory T cells. Moreover, calcitriol promotes Th2-type differentiation and the secretion of IL-4, IL-5, and IL-10, anti-inflammatory cytokines. 1,25(OH)2D: 1-25-Dihydroxyvitamin D; IL-2: interLeukin-12; IL-10: interLeukin-10; Th1: T Helper 1 lymphocytes; Th2: T Helper 2 lymphocytes; T reg: T regulatory cells; TGF-β: transforming growth factor beta; TNF-α: Tumor Necrosis Factor alpha.
Ijms 26 04798 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Argano, C.; Torres, A.; Orlando, V.; Cangialosi, V.; Maggio, D.; Pollicino, C.; Corrao, S. Molecular Insight into the Role of Vitamin D in Immune-Mediated Inflammatory Diseases. Int. J. Mol. Sci. 2025, 26, 4798. https://doi.org/10.3390/ijms26104798

AMA Style

Argano C, Torres A, Orlando V, Cangialosi V, Maggio D, Pollicino C, Corrao S. Molecular Insight into the Role of Vitamin D in Immune-Mediated Inflammatory Diseases. International Journal of Molecular Sciences. 2025; 26(10):4798. https://doi.org/10.3390/ijms26104798

Chicago/Turabian Style

Argano, Christiano, Alessandra Torres, Valentina Orlando, Virginia Cangialosi, Dalila Maggio, Chiara Pollicino, and Salvatore Corrao. 2025. "Molecular Insight into the Role of Vitamin D in Immune-Mediated Inflammatory Diseases" International Journal of Molecular Sciences 26, no. 10: 4798. https://doi.org/10.3390/ijms26104798

APA Style

Argano, C., Torres, A., Orlando, V., Cangialosi, V., Maggio, D., Pollicino, C., & Corrao, S. (2025). Molecular Insight into the Role of Vitamin D in Immune-Mediated Inflammatory Diseases. International Journal of Molecular Sciences, 26(10), 4798. https://doi.org/10.3390/ijms26104798

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