Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder associated with immune dysregulation, skin barrier dysfunction, and microbial dysbiosis characterized by Staphylococcus aureus overcolonization and reduced bacterial diversity. Beyond its classical role in calcium homeostasis, Vitamin D (VD) influences skin immunity and microbial composition. This review summarizes current knowledge on VD metabolism, its immunological pathways in AD, and its interactions with the skin microbiome. Recent evidence positions the skin as an active immunological organ rather than a passive barrier. Commensal bacteria such as Staphylococcus epidermidis not only inhibit pathogens by producing bacteriocins and modulins but also generate ceramides and short-chain fatty acids (SCFAs) that stabilize the lipid barrier. Moreover, dermal fibroblasts and preadipocytes produce antimicrobial peptides, while resident γδ T cells release growth factors like fibroblast growth factor 7 (FGF7), linking host defense with tissue regeneration. VD modulates AD by suppressing T helper 2 cells/T helper 17 cell responses, enhancing regulatory T cell development, inducing antimicrobial peptides, and strengthening skin and gut barrier integrity. Its interaction with the microbiome and pathways such as SCFA and aryl hydrocarbon receptor (AhR) signaling supports its potential as an adjunctive therapy in AD management. Evidence from mechanistic studies and animal models suggests that VD supplementation may modulate inflammation and microbial diversity. Clinical implications, therapeutic perspectives, and future research directions highlight the potential of VD as a therapeutic adjunct in AD management.
1. Introduction
Atopic dermatitis (AD) is a multifactorial chronic disease characterized by impaired epidermal barrier function [,]. It manifests as recurrent inflammatory skin lesions accompanied by pruritus [], which significantly reduce patients’ quality of life, particularly in moderate-to-severe forms of the disease. Approximately 50% of adult patients with AD report experiencing social exclusion []. Sleep disturbances are common, and additional burdens include time-consuming skin care routines and dietary modifications [,]. Although some patients experience long periods of remission, in others, the disease presents as persistently active chronic skin inflammation []. The pathogenesis of AD is multifactorial, involving epidermal barrier dysfunction, dysregulated immune responses, and alterations in the skin microbiome []. A hallmark of allergic diseases, including AD, is elevated serum immunoglobulin E (IgE) levels []. Patients show enhanced type 2 immune responses, with increased production of interleukin- 4 (IL-4), IL-5, IL-13, and IL-31, along with reduced expression of filaggrin and antimicrobial peptides (AMPs) in the skin [,,]. Cluster of differentiation 4 positive (CD4+) T helper type 2 (Th2) cells play a central role in this process, as excessive activation of these cells amplifies the inflammatory cascade typical of AD [,]. Cytokines produced by Th22 and Th17 cells may also contribute to disease pathogenesis in certain patients []. A healthy skin microbiome is essential for maintaining cutaneous homeostasis, but in AD, microbial diversity is markedly reduced [,]. Up to 70% of patients exhibit increased colonization by Staphylococcus aureus, which correlates with disease severity [,]. In adults with AD, changes in the skin microbiome include not only the dominance of Staphylococcus aureus but also a significant decrease in microbial diversity, which correlates with the severity of clinical symptoms assessed using the Scoring Atopic Dermatitis (SCORAD) and Eczema Area and Severity Index (EASI) scales [,]. The role of Staphylococcus epidermidis appears to be phase-dependent: during remission, it supports microbial balance and suppresses Staphylococcus aureus, whereas during flares, it may be outcompeted or represented by pro-inflammatory strains [,].
Genetic studies have revealed that both well-known genes such as filaggrin (FLG) and rare variants affecting signaling pathways (e.g., signal transducer and activator of transcription 6 (STAT6), janus kinase 1 (JAK1)) are implicated in AD pathogenesis []. According to one hypothesis, FLG gene variants may have provided an adaptive function by facilitating vitamin D (VD) synthesis in populations living in regions with low sunlight exposure [].
The skin of patients with AD typically demonstrates increased pH, greater permeability, and reduced intercellular cohesion, all of which promote bacterial overgrowth and facilitate penetration of irritants []. Symptom exacerbation is frequently observed during winter months in regions with limited sun exposure, when cutaneous VD synthesis is at its lowest [,]. Indeed, reduced serum VD concentrations have been documented in patients with AD, particularly in those with moderate to severe disease [,]. The skin expresses the vitamin D receptor (VDR), the activation of which is pivotal for regulating the production of human cathelicidin antimicrobial peptide (LL-37), an antimicrobial peptide integral to innate host defense []. Moreover, VD metabolism within keratinocytes promotes their proliferation and differentiation, thereby contributing to barrier repair and maintenance of epidermal integrity [,]. Genetic polymorphisms in the VDR gene have been associated with susceptibility to AD [,,]. In recent years, studies have shown that VD supplementation may beneficially modulate the microbiome, reducing AD symptom severity [,].
The aim of this review is to analyze the impact of VD supplementation on skin microbiome modulation in patients with AD.
AD is a chronic inflammatory skin disorder characterized by a complex confluence of genetic predisposition, epidermal barrier dysfunction, immune dysregulation, and microbial imbalance. Mutations in the FLG gene and decreased levels of ceramides and antimicrobial peptides impair the skin’s barrier integrity, facilitating allergen penetration and microbial colonization, particularly by Staphylococcus aureus.
This barrier impairment triggers a predominant Th2-type immune response with elevated IL-4, IL-13, and IgE levels, while reduced microbial diversity further amplifies inflammation. Together, these mechanisms establish a vicious cycle linking the barrier, immune system, and microbiome in AD pathogenesis.
2. Materials and Methods
For this review, a systematic search for scientific publications on the role of VD in AD and its effects on both the skin and gut microbiome was conducted in the PubMed, Scopus, and Web of Science databases, covering studies published until September 2025.
A combination of the following keywords was used: “vitamin D”, “atopic dermatitis”, “skin microbiome”, “gut-skin axis”, “VDR”, and “Th17/Treg balance”. Original articles, review papers, meta-analyses, and both randomized and observational clinical studies published in English were included.
The analysis focused on studies involving patients with AD, as well as animal and cell models providing mechanistic insights. Non- full-text papers, conference abstracts, and studies unrelated to the topic were excluded (Figure 1).
Figure 1.
PRISMA flow diagram for study selection.
The selected data were categorized according to thematic domains: immunomodulation, barrier function, microbiome alterations, and molecular mechanisms.
3. Vitamin D Metabolism and Skin Immune Mechanisms
3.1. Vitamin D Metabolism
VD refers to a group of fat-soluble steroid compounds that play a pivotal role in calcium homeostasis and the proper functioning of the immune system []. Although VD can be obtained from dietary sources, such as fish, meat, eggs, chocolate, and mushrooms (Figure 2) [], achieving the recommended daily intake (600–800 IU/day) through diet alone is difficult. VD deficiency is common worldwide, particularly among populations living in temperate and polar regions, where annual sunlight exposure is limited. It is estimated that approximately 40% of the European population is VD deficient []. Thus, cutaneous synthesis under sunlight exposure remains the primary source for meeting daily requirements. Studies indicate that 35 min of daily sun exposure is sufficient to generate about 1000 IU of VD, which is adequate to maintain appropriate serum concentrations [].
Figure 2.
Synthesis and dietary sources of vitamin D. Under UVB (280–320 nm), 7-dehydrocholesterol in the skin is converted into previtamin D3 and then into cholecalciferol (VD3). VD3 is also obtained from animal-based foods such as fish, eggs, meat and chocolate, while ergocalciferol (VD2) originates from plant sources, mainly mushrooms.
VD is obtained from the diet in two forms: ergocalciferol (VD2), which is derived from plant-based foods and fungi, and cholecalciferol (VD3), which is derived from animal-based products. Both forms are biologically inactive and require enzymatic conversion to their active metabolites. However, approximately 90% of the body’s VD pool originates from endogenous cutaneous synthesis, with only about 10% obtained from dietary sources [], underscoring the importance of regular sunlight exposure. Cholecalciferol is synthesized in the skin upon exposure to ultraviolet B radiation (UVB) with wavelengths between 280 and 320 nm. This radiation induces the conversion of 7-dehydrocholesterol (7-DHC), which is present in the stratum spinosum and stratum granulosum of the epidermis, into cholecalciferol (Figure 1). When sunlight containing UVB reaches uncovered skin, it triggers the opening of the B-ring in the 7-DHC molecule, forming an intermediate compound known as previtamin D3. This compound then undergoes spontaneous thermal isomerization to produce VD3 (Figure 3) [].
Figure 3.
Conversion of 7-dehydrocholesterolaryl (7-DHC) to previtamin D under UVB and the spontaneous isomerization of previtamin D to cholecalciferol (VD3).
Dietary forms of VD are absorbed in the small intestine by enterocytes through passive diffusion, as well as with the assistance of cholesterol transport proteins such as Niemann-Pick C1-like protein 1 (NPC1L1), scavenger receptor class B type I (SR-BI), and CD36 [,]. Following absorption, they are transported into the lymphatic system and subsequently into the bloodstream. In circulation, they are predominantly bound to Vitamin D-binding protein (DBP); although to a lesser extent, they are also carried by albumin or present in a free form. Within the liver, VD undergoes enzymatic hydroxylation and bioactivation to 25-(OH)D3 (25-hydroxycholecalciferol, calcifediol) and 25-(OH)D2 (25-hydroxyergocalciferol, calcidiol) (Figure 4) [], mediated by the enzyme 25-hydroxylase encoded by the CYP2R1 gene. This product serves as the primary indicator of VD status in the body and is routinely measured in clinical practice to assess serum VD levels. However, it exhibits only weak biological activity.
Figure 4.
Enzymatic hydroxylation of ergocalciferol (VD2) and cholecalciferol (VD3) to 25-hydroxycholecalciferol and 25-hydroxyergocalciferol in the liver.
Subsequently, 25(OH)D bound to DBP and albumin is transported to the kidneys. There, under the action of the enzyme 1α-hydroxylase, calcifediol and calcidiol are converted into calcitriol (1,25-dihydroxycholecalciferol, 1,25(OH)2D3) and ercalcitriol (1,25-dihydroxyergocalciferol, 1,25(OH)2D2), respectively (Figure 5). Of these two metabolites, calcitriol exhibits the higher biological activity and is considered the most active form of VD [].
Figure 5.
Conversion of calcifediol and calcidiol to calcitriol (1,25(OH)2D3) and ercalcitriol (1,25(OH)2D2) in the kidneys under the action of α1-hydroxylase.
The active form of VD then enters target cells, where, by binding to nuclear VDR, a transcription factor, it regulates gene expression through transcriptional activation and transrepression of specific target genes [] (Figure 6).
Figure 6.
Transport and metabolism of vitamin D2 (ergocalciferol) and D3 (cholecalciferol). Vitamins D2 and D3 are absorbed in the intestine via passive diffusion and transporters (NPC1L1, SR-BI, and CD36) and enter the bloodstream through the lymphatic system. In the liver, they are hydroxylated by 25-hydroxylase to form 25-hydroxyvitamin D2 and D3. These metabolites are further hydroxylated in the kidneys by 1α-hydroxylase to produce the biologically active forms, 1,25-dihydroxyvitamin D2 and D3.
Mutations in genes encoding enzymes of the VD synthesis pathway are an important cause of functional VD deficiency, which may occur even in the presence of normal laboratory values. Mutations or deletions of the CYP2R1 gene are associated with VD-dependent rickets type 1b. Although this is a relatively rare disorder, defective alleles encoding 25-hydroxylase occur with a frequency of 3 in 1000 births in the Caucasian population []. Patients with VD-dependent rickets type 1b require supplementation with calcifediol and calcium []. Another clinically relevant gene is CYP27B1, which encodes 1α-hydroxylase, the enzyme responsible for the conversion of calcifediol and calcidiol to calcitriol and ercalcitriol. Mutations in CYP27B1 cause VD-dependent rickets type 1a (also referred to as pseudovitamin D deficiency rickets). Although biochemical testing may show normal or even elevated concentrations of 1,25(OH)2D3, the inability to generate its active form in the kidneys results in clinical manifestations of VD deficiency in early childhood []. Treatment involves supplementation with calcitriol and phosphate; early diagnosis enables good disease control and complete resolution of symptoms. In the case of mutations in the VDR gene, which encodes the nucVD receptor, patients develop hypocalcemic rickets resistant to VD. This disorder is characterized by reduced tissue responsiveness to VD, and one of its hallmark clinical features is near-total alopecia [].
3.2. Skin Immune Mechanisms
The skin is the largest organ of the human body [], serving as a key barrier that prevents microbial invasion. All anatomical layers of the skin contribute to antimicrobial defense through interlayer interactions or layer-specific mechanisms []. Breaches in skin integrity under non-sterile conditions may result in wound infections and, in severe cases, sepsis and death if adequate prophylactic antibiotic therapy is not initiated.
The skin consists of three layers: the outer epidermis, the dermis, and the subcutaneous tissue []. The epidermis is a stratified squamous keratinized epithelium in which keratinocytes are tightly interconnected by desmosomes, hemidesmosomes, tight junctions (TJs), and adherens junctions. These structures ensure impermeability to pathogens and maintain skin homeostasis []. The epidermis is colonized by a unique microbiome composed predominantly of commensal bacteria, viruses, and fungi, which vary between individuals []. These organisms primarily utilize lipids secreted by sebaceous glands located in the dermis, preventing their use by pathogenic bacteria attempting to colonize the skin. Common epidermal residents include Staphylococcus epidermidis and Cutibacterium acnes, which exert protective effects by limiting the growth of pathogenic bacteria such as Staphylococcus aureus and Streptococcus pyogenes, both of which are associated with skin disease [].They help maintain an acidic pH, which makes the skin a harsh environment for many pathogens and supports its structural integrity []. Moreover, commensal bacteria play a crucial role in preventing excessive inflammation and helping regulate immune responses [].
Commensal bacteria perform several antimicrobial functions, including the production of bacteriocins, ribosomally synthesized proteins that bind to receptors on the cell walls of susceptible species, inhibiting their growth or inducing cell death []. Another antimicrobial mechanism involves phenol-soluble modulins, amphipathic cytolytic molecules that act synergistically with bacteriocins. Staphylococcus epidermidis-derived modulins are active against Staphylococcus aureus and group A streptococci []. Commensal microbes also contribute maintaining the skin’s acidic pH by producing short-chain fatty acids (SCFAs), including butyric and propionic acids []. This acidic environment inhibits pathogen overgrowth while favoring beneficial residents such as Cutibacterium acnes and Staphylococcus epidermidis. These bacteria also produce sphingomyelinase, which hydrolyzes ceramides on the skin surface into free fatty acids and cholesterol. This activity further lowers skin pH and supports lipid barrier integrity, protecting against pathogenic colonization and excessive dryness []. Disturbances in skin pH balance, colonization by pathogens, and failure of antimicrobial defenses are strongly associated with skin diseases, including AD [].
The dermis is the thickest layer of the skin, containing blood vessels, sebaceous and sweat glands, immune cells, and nerve endings. This layer ensures proper thermoregulation, participates in tactile sensation, nourishes the epidermis, and serves as an immunological barrier against microorganisms. It harbors numerous immune cells, including T lymphocytes, mast cells, tissue-resident macrophages (TRMs), and dendritic cells. Fibroblasts within the dermis play multiple crucial roles in antimicrobial defense and immune regulation. The latest advances in bioengineering demonstrate the potential of bioactive gelatin nanofibers containing lumbrokinase as a novel strategy for improving wound healing. Their mechanism of action is primarily due to their anti-inflammatory (lowering the secretion of IL-6 and TNF-α) and fibroblast-stimulating effects, and they have achieved promising results in in vitro models [].
Fibroblasts synthesize pro-inflammatory cytokines such as IL-6, interferon-β (IFN-β), and tumor necrosis factor alpha (TNF-α), as well as chemokines like C-X-C motif chemokine ligand 1 (CXCL1) [], and secrete growth factors and metalloproteinases that support tissue repair after infection []. Fibroblasts can also differentiate into preadipocytes, which produce the antimicrobial peptide cathelicidin, thereby protecting the skin against deep bacterial penetration []. Recent studies also highlighted the significant role of this peptide in antifungal defense against Candida albicans. Metabolic products of this fungus induce inflammatory responses through the FGFR-MEK-ERK pathway, illustrating the multifaceted contribution of dermal fibroblasts to antimicrobial defense []. TRMs play a critical role in skin immunity through phagocytosis, antigen presentation to naïve T lymphocytes [], and secretion of chemokines that recruit neutrophils, including C-X-C motif chemokine receptor 2 (CXCR2) ligands []. Dermal T lymphocytes comprise multiple populations, including resident memory T cells, CD4+ helper T cells, CD8+ cytotoxic T cells, and γδ T cells []. CD4+ memory T cells mount rapid recall responses and secrete pro-inflammatory cytokines, such as TNF-α and IL-22, which are essential for rapid pathogen neutralization []. CD4+ helper T lymphocytes coordinate immune processes by secreting specific cytokines and chemokines that regulate inflammation and activate other immune cells, including macrophages, neutrophils, and B lymphocytes []. Another important component of antimicrobial defense are γδ T cells, which produce pro-inflammatory cytokines and growth factors such as fibroblast growth factor (FGF7), thereby supporting epithelial regeneration and wound healing []. They also exert direct cytotoxic effects through the release of perforin and granzymes, inducing apoptosis in target cells [].
The subcutaneous tissue plays an important role in defense against pathogens. It not only serves as an insulating layer, protecting deeper organs from heat loss and mechanical injury, but also acts as a reservoir of immune cells []. Adipocytes within this layer secrete cytokines, adipokines, and antimicrobial peptides that contribute directly to pathogen neutralization [].
In summary, the skin constitutes a key antimicrobial barrier, which not only provides mechanical protection according to its integrity but also serves as a reservoir of immune cells and a site of multiple immunological reactions that safeguard the body against pathogen invasion.
4. The Role of Vitamin D in the Pathogenesis and Treatment of Atopic Dermatitis/Immune Pathways Linking the Microbiota and Atopic Dermatitis
For more than a decade, the potential impact of VD on inflammatory processes and the severity of AD have been investigated. Through multiple regulatory mechanisms within the immune system, VD exerts beneficial effects on the course of the disease and improves patients’ quality of life []. The positive role of VD in AD is mediated by several mechanisms.
- Inhibition of Th2 polarization: By binding to the VDR in the nucleus of T lymphocytes, VD downregulates the transcription factor GATA3 [], which is critical for the polarization of naive CD4+ T cells into Th2 cells. This reduces the production of cytokines (IL-4, IL-5, and IL-13) essential for Th2 polarization, thereby preventing excessive pathological Th2 differentiation, which is a hallmark of autoimmune diseases, including AD [].
- Promotion of regulatory T cell (Treg) differentiation: VD enhances the expression of the transcription factor forkhead box P3 (FOXP3), the master regulator of Treg development, in naive T cells []. This leads to both increased Treg differentiation and suppression of Th17 differentiation, since FOXP3 inhibits the expression of RORγt, the transcription factor required for Th17 lineage commitment [].
- Induction of AMPs: In AD, 70–90% of lesions are colonized by S. aureus, which exacerbates inflammation and impairs barrier function []. VD increases the production of AMPs, including LL-37 [] and human β-defensin 4 (DEFB-4) [], which exert strong antimicrobial effects against S. aureus. By upregulating genes encoding AMPs, VD enhances their production and limits the pathogenic role of S. aureus in AD [].
- Upregulation of epidermal structural proteins: VD indirectly increases the expression of the key epidermal proteins filaggrin, involucrin, and loricrin by suppressing IL-4 and IL-13, which otherwise downregulate their synthesis []. A proper balance of these proteins ensures epidermal barrier integrity and contributes to improved AD outcomes [] (Figure 7).
Figure 7. Mechanisms of Vitamin D (VD) action and their impact on atopic dermatitis (AD). VD, by binding to the VDR, inhibits GATA3 expression, leading to a decrease in IL-4, IL-5, and IL-13 production and thereby reducing Th2 cell polarization. Consequently, the reduction in IL-4 and IL-13 levels enhances the expression of epidermal structural proteins (loricrin, filaggrin, and involucrin) which strengthens the integrity of the skin barrier. Vitamin D also stimulates FOXP3 expression, promoting the differentiation of T lymphocytes into Tregs. These cells secrete IL-10, which reinforces tight junctions by upregulating the expression of claudins and occludins. At the same time, FOXP3 suppresses the transcription of the RORγt gene, resulting in reduced differentiation of Th17 cells. Furthermore, vitamin D induces the production of AMPs (LL-37, DEFB-4, and HBD-2), which inhibit the growth of pathogenic microorganisms, including Staphylococcus aureus, thereby alleviating the clinical symptoms of AD.
The gut microbiota is an integral modifier of AD progression through its systemic effects. Beneficial bacteria such as Bifidobacterium, Roseburia, and Faecalibacterium promote the production of SCFAs []. SCFAs exert anti-inflammatory effects in allergic and autoimmune diseases by sequentially inducing polymorphonuclear myeloid-derived suppressor cells and Tregs []. These findings have fueled interest in therapeutic modulation of the gut microbiota, including fecal microbiota transplantation, as a potential future intervention for AD []. A healthy gut microbiota also reduces AD severity by enhancing AMP production in the skin. SCFAs produced by commensals act as histone deacetylase inhibitors in keratinocytes, increasing histone acetylation at AMP gene promoters. This results in upregulated transcription of AMPs such as LL-37 and human β-defensin 2 (HBD-2) [], leading to reduced S. aureus colonization and attenuated skin inflammation []. VD is also indispensable for maintaining both gut and skin barrier integrity. Active VD binds to VDR in intestinal epithelial cells, upregulating TJs proteins including claudins, occludins, and zonula occludens (ZO) proteins []. These proteins are essential for barrier function, as they prevent the translocation of pathogens and support the growth of beneficial SCFA-producing bacteria. Importantly, the gut microbiota itself modulates VDR expression. Reduced VDR expression associated with pathogenic colonization is implicated in the pathogenesis of inflammatory bowel diseases such as IBS and celiac disease []. Conversely, proper VDR expression in keratinocytes ensures intercellular TJs, preserving epidermal barrier integrity and limiting AD severity []. This highlights the crucial role of the gut microbiota not only in intestinal barrier function but also in systemic processes, including skin barrier maintenance, emphasizing the complexity of AD pathogenesis and the need for multi-targeted therapeutic strategies. Another important mechanism is the microbiota-driven regulation of cytokine balance. A healthy microbiota promotes anti-inflammatory IL-10 production via Treg induction []. IL-10 dampens excessive immune responses, reduces inflammation, and enhances the expression of TJ proteins such as claudins and occludins []. IL-17 production is also regulated by the gut microbiota. SCFAs suppress excessive IL-17 release from Th17 cells. Although physiological levels of IL-17 promote AMP synthesis [], overproduction amplifies pro-inflammatory cytokines and chemokines, increases metalloproteinase activity (damaging the epidermis), and impairs Treg activity [].
Another key cytokine influencing the epidermal barrier is IFN-γ, the levels of which can be reduced by SCFAs []. Excess IFN-γ suppresses the synthesis of barrier proteins (involucrin, filaggrin, and loricrin) [] and inhibits ceramide production, which is essential for a functional lipid barrier []. IFN-γ is also a major mediator of skin inflammation in AD. The gut microbiota regulates signaling through the aryl hydrocarbon receptor (AhR), a transcription factor expressed in the gut and skin. AhR modulates detoxification, cell proliferation, and immune responses []. Its agonists include aromatic hydrocarbons, polychlorinated biphenyls, and dioxins, which activate detoxification pathways via cytochrome P450 genes []. Importantly, AhR ligands are also derived from tryptophan metabolism by gut bacteria such as Lactobacillus, Bacteroides, Escherichia coli, and Clostridium, yielding indole-3-aldehyde (IAld), indole-3-acetic acid (IAA), and kynurenine []. These ligands activate AhR, leading to enhanced intestinal and skin barrier integrity, increased production of epidermal structural proteins, and the promotion of anti-inflammatory Tregs [,]. Tolerogenic dendritic cells (tol-DCs) are another crucial component of immune modulation. These cells maintain immune tolerance and suppress inflammation []. SCFAs influence tol-DCs epigenetically, promoting their tolerogenic phenotype. This contributes to proper immune responses to food antigens, mitigates intestinal inflammation (e.g., in Crohn’s disease), and supports Treg induction []. Taken together, the interplay between gut microbiota, their metabolites, and VD highlights the importance of beneficial gut bacteria in maintaining skin health and limiting inflammation under pathological conditions. These interactions also open new therapeutic perspectives for managing inflammatory skin diseases, including AD.
5. Evidence from Animal Models and Mechanistic Studies
Animal models of AD provide valuable tools for precisely investigating the effects of VD on inflammation and epidermal barrier function. The classical model using a VD analog (calcipotriol/MC903) involves inducing an “AD-like” phenotype in mice (eosinophilic infiltration, type 2 immune dominance, and pruritus), mainly through the induction of TSLP (thymic stromal lymphopoietin) in keratinocytes, highlighting that excessive local VDR activation may initiate inflammation [,,,,]. This phenomenon does not contradict the immunoregulatory role of the VD-VDR axis demonstrated in many studies, where systemic activation/supplementation of VD promotes normalization of innate and adaptive responses, in addition to supporting wound healing and epidermal homeostasis [,,,]. In an NC/Nga mouse model (spontaneous AD), topical administration of calcitriol alleviated lesion severity, reduced transepidermal water loss (TEWL), and restored tight junction integrity. Increased expression of filaggrin and loricrin, reduced IL-13/IL-33, and enhanced β-defensin expression were observed. These findings were confirmed in a Dermatophagoides farinae allergen-induced model, which provided further evidence of improvements in barrier function and inflammatory parameters [].
Subsequent studies elucidated inflammatory mechanisms triggered by MC903, including activation of the TSLP → dendritic cell/Th2 axis, the involvement of IL-33, and nodal points such as mechanistic target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor-reactive oxygen species (HIF-ROS). Pharmacologic or genetic inhibition of these pathways (via rapamycin, regulatory-associated protein of mTOR knockout (Raptor-KO), or DMOG) reduced inflammation, eosinophilia, and type 2 markers [,,,].
Regarding MC903 model modulation, independent groups demonstrated that modification of innate immune co-signals, e.g., toll-like receptor 3 (TLR3) activation by poly(I:C), enhances TSLP responses and the AD-like phenotype, confirming the model’s sensitivity to environmental stimuli [,,]. Although the results of human interventional trials remain heterogeneous, mechanistic studies have shown that VD supplementation increases AMP expression in the skin (LL-37), thereby improving control over S. aureus and other pathogens—a central element in AD pathophysiology [,,,]. VDR knockout models display impaired epidermal homeostasis (alopecia, defective keratinocyte proliferation/differentiation, and barrier dysfunction) and altered immune responses. Some VDR effects are independent of 1,25(OH)2D3, reflecting the receptor’s role as a transcription factor [,,]. Reviews of studies on epithelial barrier biology further confirm that the VDR axis regulates tight junction proteins and maintains barrier integrity [,]. In AD patients, reduced expression of claudin-1 (CLDN1) and other TJ components correlates with higher TEWL and increased susceptibility to infections; experimental manipulation of CLDN1 in models supports its causal role in inflammation and barrier dysfunction [,,].
Aquaporin-3 (AQP3), a regulator of water and glycerol transport in keratinocytes, also influences hydration and epidermal differentiation; its modulation has been linked to barrier properties and inflammatory pathways [,,].
Taken together, data from animal models and ex vivo/in vitro experiments indicate that proper VD/VDR signaling
- (I).
- strengthens barrier structure (filaggrin, loricrin, and TJ proteins);
- (II).
- limits Th2/Th17 inflammation and alarmins (TSLP/IL-33);
- (III).
- enhances antimicrobial defense (AMPs, especially LL-37).
In certain models, however, excessive local VDR activation (MC903) may paradoxically trigger type 2 inflammation, emphasizing the importance of dose, route of administration, and inflammatory context [,,,,].
From a clinical perspective, reviews and meta-analyses underline the association between low 25(OH)D levels and AD severity, as well as the potential benefits of supplementation, though randomized controlled trial (RCT) results remain inconsistent, depending on population, dose, and duration [,,].
In summary, a wide range of experimental models—pharmacological (MC903), spontaneous (NC/Nga), allergen-induced, VDR knockout mice, and ex vivo studies—enable a detailed investigation of VD effects on inflammation and epidermal barrier integrity. Table 1 below summarizes the key features of these models, their methods of induction, and the main observations regarding the VD/VDR axis.
Table 1.
Animal and cellular models of AD used to investigate the role of VD/VDR in inflammation and epidermal barrier function.
6. Vitamin D as a Modulator of the Skin Microbiome in Atopic Dermatitis
VD is a critical modulator of the cutaneous microbiome, and VD deficiency is associated with the exacerbation of inflammatory skin diseases, including AD []. VD increases the diversity of the gut microbiota, limits bacterial translocation, and fosters conditions favorable for the growth of Firmicutes, Bifidobacterium, and Lactobacillus while constraining pro-inflammatory Proteobacteria []. This supports microbial diversity, intestinal barrier integrity, and the production of beneficial SCFAs. VD therefore indirectly influences the maintenance of a healthy gut and skin microbiome, leading to a reduction in the intensity of the inflammatory processes occurring in AD.
Perturbations of the gut microbiome in early childhood are an important factor in the development of inflammatory skin diseases. Breastfeeding influences the maintenance of a healthy gut microbiota: compared with infants fed formula, exclusively breastfed infants exhibit greater microbial diversity [], higher levels of SCFA-producing bacteria (including Bifidobacterium and Lactobacillus), and lower levels of pathogenic taxa, such as Clostridioides and members of the Enterobacteriaceae family [,]. Studies also indicate that VD supports endothelial function by inhibiting the production of inflammatory cytokines [] (Figure 8). A meta-analysis of 39 studies assessing antibiotic exposure during pregnancy and early childhood found that maternal antibiotic use increased the risk of AD in offspring by 9%, whereas antibiotic use in early childhood increased the risk by 22% []. These findings underscore the importance of avoiding antibiotics during pregnancy and in early life, periods of heightened vulnerability to immune-mediated diseases often linked to gut dysbiosis []. Collectively, these data highlight the central role of VD in the diet: VD- and probiotic-rich nutritional patterns can optimize gut microbiota composition, reduce skin inflammation, and strengthen both cutaneous and intestinal barriers, potentially mitigating allergic and autoimmune diseases [,]. Clinical studies further suggest therapeutic benefits of VD supplementation, reporting a moderate and statistically significant reduction in AD severity (SMD = −0.41, 95% CI: −0.67 to −0.16, I2 = 58%, p < 0.01) []. Among individuals supplementing >1000 IU/day, the effect was statistically significant, whereas lower doses did not yield significant therapeutic benefit. These observations support a potential therapeutic role of VD supplementation in AD [].
Figure 8.
Vitamin D modulates gut microbiota composition, reduces inflammation, and enhances intestinal and skin barrier integrity. This diagram shows how vitamin D supplementation increases microbial diversity (↑ Firmicutes, Bifidobacterium, and Lactobacillus), promotes SCFA production, and decreases Proteobacteria and inflammatory cytokines. These effects improve barrier function and alleviate the clinical symptoms of atopic dermatitis (itching, erythema, and lichenification), leading to lower SCORAD, EASI, and TIS scores.
Changes in the gut microbiome occurring under the influence of VD supplementation have a positive effect on the course of AD and help reduce the severity of persistent symptoms such as erythema, pruritus, and skin lichenification []. Studies indicate that individuals with AD who supplement with VD experience a reduction in symptom severity according to the SCORAD, EASI, and Three Item Severity (TIS) scales [,]. The complexity of the therapeutic effects of VD is illustrated in Figure 8.
7. Potential Therapeutic Approaches, Clinical Implications, and Future Directions
In adult patients with moderate-to-severe AD, monitoring serum 25(OH)D levels is recommended []. VD concentrations in these patients are typically lower than in healthy populations and correlate with disease severity []. Serum 25(OH)D concentration is the established marker of VD status. The most effective supplementation strategy is daily oral cholecalciferol, which remains the preferred form. Other analogs (calcifediol, calcitriol, and alfacalcidol) and parenteral administration are reserved for specific indications []. Currently, no universal guidelines define optimal serum 25(OH)D levels; however, most clinical studies consider 30–60 ng/mL to be adequate, <30 ng/mL to indicate deficiency, and <20 ng/mL to indicate severe deficiency [,]. Some evidence suggests that levels below 50 ng/mL may be insufficient []. Higher VD concentrations (≥50 ng/mL) appear to support optimal immune responses, and in autoimmune diseases such as psoriasis and asthma, optimal levels may reach ≥80 ng/mL, raising the possibility of similar targets for AD, though further clinical studies are needed [].
Because dietary habits and sunlight exposure strongly influence VD metabolism, establishing universal supplementation regimens remains challenging. Individualized strategies accounting for geographic, environmental, and lifestyle factors may be required []. Dosing should also be adjusted for body mass index (BMI); in obesity, doses may need to be doubled or even tripled compared with standard regimens [,].
In a study by Borzutzky et al., children received weekly VD supplementation (8000–16,000 IU depending on age) for six weeks. This regimen did not improve SCORAD scores, symptoms, or AD biomarkers compared with placebo, although both daily and weekly dosing effectively increased serum VD levels. Symptom improvement was primarily correlated with rises in serum 25(OH)D, with the greatest benefit seen in children with severe baseline deficiency []. Pediatric studies administering 1000–1600 IU/day for 1–3 months reported reductions in SCORAD and EASI scores and clinical improvement [].
According to current guidelines, the upper safe daily intake for most adults is 4000 IU, and higher doses may cause complications []. VD supplementation can restore epidermal integrity and enhance microbiome diversity by limiting S. aureus overgrowth []. Adverse effects are rare, and toxicity usually occurs only when serum 25(OH)D exceeds 150 ng/mL [,,]. Therefore, serum monitoring is advised to individualize dosing and minimize adverse events []. Notably, in about half of patients, discontinuation of supplementation leads to disease relapse, suggesting that VD may act more as a preventive measure than as a therapeutic agent in preventing flares [].
The use of calcipotriol, a VD3 analog, is well established in psoriasis and chronic hand eczema but has not yet been validated for AD [,]. Topical creams containing VD analogs at appropriate concentrations may improve lesional skin []. Due to their immunomodulatory and barrier-enhancing properties, these treatments remain a promising research avenue requiring further clinical trials. Although evidence is still limited, the use of topical VD analogs is a potentially valuable therapeutic option, particularly in combination with barrier-repair strategies. In adults with moderate AD, 6–8 weeks of narrowband UVB therapy increased serum VD levels, reduced EASI, SCORAD, Patient-Oriented Eczema Measure (POEM), and Dermatology Life Quality Index (DLQI) scores, and improved microbiome diversity in lesional skin []. These findings suggest that combining oral VD supplementation with UVB phototherapy may be a promising therapeutic strategy. Another potential direction is combining VD with probiotics, particularly Lactobacillus and Bifidobacterium strains, which may synergistically reduce inflammation and promote microbiome restoration [,]. Given its low cost, supplementation has high clinical value []. Future research should focus on identifying VDR polymorphisms, which may allow for dose optimization based on genetic variants and receptor activity []. CYP24A1, the gene encoding the enzyme responsible for calcitriol degradation, has clinical relevance, as increased activity may lower serum VD levels [,]. Elevated CYP24A1 expression has been observed in inflamed skin, suggesting that inflammation itself alters VD metabolism. This illustrates the need for studies investigating personalized supplementation tailored to dynamic metabolic pathways []. Low maternal VD levels during the first trimester of pregnancy are associated with an increased risk of AD in offspring []. Thus, supplementation during pregnancy may help prevent AD development in children [,]. However, evidence remains inconclusive, underscoring the need for further investigation []. Importantly, most studies have had short follow-up periods, emphasizing the necessity of long-term trials to evaluate both potential adverse effects and sustained clinical benefits, including reduced AD severity. Translational implications derived from current evidence on VD in AD are shown in Table 2.
Table 2.
Translational implications derived from current evidence on Vitamin D in atopic dermatitis.
8. Strengths and Limitations
This review comprehensively addresses the role of VD in the pathogenesis and therapy of atopic dermatitis, integrating evidence related to immune modulation, epidermal barrier function, and the gut–skin microbiome axis. The work is distinguished by its up-to-date references, clear structure, and translational perspective linking basic and clinical findings, which enhances its scientific and practical relevance.
The available studies show significant heterogeneity in patient populations, methods of assessing the microbiome, and VD status, which limits comparability across studies. There is also a lack of clear data regarding optimal dosage, duration, and the safety of long-term VD supplementation, as well as its interaction with other therapeutic modalities used in atopic dermatitis. Further clinical studies are required to standardize treatment recommendations and clarify the impact of VD supplementation on the microbiome and disease outcomes.
9. Conclusions
VD plays an important role in the pathogenesis of AD, acting on the immune system, the epithelial barrier, and the microbiota. VD supplementation may help alleviate AD symptoms by modulating Th2/Th17 pathways and supporting Treg activity. Evidence from clinical and animal studies shows beneficial effects on keratinocyte differentiation, antimicrobial peptide production, and strengthening of the skin barrier. Mechanistically, these effects are mediated by the VDR and downstream pathways involved in immune regulation and skin homeostasis. The interactions between VD and the microbiome suggest a potential role in restoring immune and barrier homeostasis in AD. In the gut microbiota, SCFA-dependent mechanisms and activation of the AhR appear to be critical, while in the skin microbiome, processes are primarily driven by increased production of AMPs and stabilization of the lipid barrier. Despite promising results, optimal dosing, treatment duration, and combination approaches remain insufficiently defined. Further large-scale clinical and experimental studies are needed to confirm the efficacy of VD supplementation and develop standardized clinical guidelines.
Author Contributions
Conceptualization K.B. and A.S.; software K.B. and M.S.; validation K.B. and B.P.; formal analysis B.P. and M.S.; investigation B.P. and L.S.; resources A.S. and K.K.; data curation M.S. and K.K.; writing—original draft preparation K.B., B.P., M.S., L.S. and K.K.; writing—review and editing A.S. and K.K.; visualization B.P. and L.S.; supervision A.S.; project administration A.S. and K.B.; funding acquisition A.S. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Data Availability Statement
We used PubMed, ScienceDirect, and Google Scholar databases to screen articles for this review. We did not report any data.
Acknowledgments
During the preparation of this manuscript, the authors used ChatGPT (GPT-5, OpenAI, San Francisco, USA, 2025) for the purposes of generating schematic scientific figures. The authors have reviewed and edited the output and take full responsibility for the content of this publication.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
The following abbreviations are used in this manuscript:
| 1,25(OH)2D3 | 1,25-Dihydroxycholecalciferol, calcitriol |
| 1,25(OH)2D2 | 1,25-Dihydroxyergocalciferol, ercalcitriol |
| 25(OH)D | 25-Hydroxyvitamin D |
| 7-DHC | 7-dehydrocholesterol |
| 1,25(OH)2D | 1,25-Dihydroxyvitamin D |
| AD | Atopic dermatitis |
| AhR | Aryl hydrocarbon receptor |
| AMPs | Antimicrobial peptides |
| AQP3 | Aquaporin 3 |
| BMI | Body mass index |
| CD | Cluster of differentiation |
| CLDN1 | Claudin-1 |
| CXCL1 | C-X-C motif chemokine ligand 1 |
| CXCR2 | C-X-C motif chemokine receptor 2 |
| DBP | Vitamin D-binding protein |
| DEFB-4 | Human β-defensin 4 |
| DLQI | Dermatology Life Quality Index |
| DMOG | Dimethyloxalylglycine |
| EASI | Eczema Area and Severity Index |
| FGF7 | Fibroblast growth factor 7 |
| FLG | Filaggrin |
| FOXP3 | Forkhead box P3 |
| HBD-2 | Human β-defensin 2 |
| HIF-ROS | Hypoxia-inducible factor—reactive oxygen species |
| IAld | Indole-3-aldehyde |
| IAA | Indole-3-acetic acid |
| IFN | Interferon |
| Ig | Immunoglobulin |
| IL | Interleukin |
| JAK1 | Janus kinase 1 |
| LL-37 | Human cathelicidin antimicrobial peptide |
| MC903 (Calcipotriol) | Vitamin D analog used experimentally in dermatology |
| mTORC1 | Mechanistic target of rapamycin complex 1 |
| NPC1L1 | Niemann-Pick C1-like protein 1 |
| POEM | Patient-Oriented Eczema Measure |
| RCT | Randomized controlled trial |
| Raptor-KO | Regulatory-associated protein of mTOR knockout |
| SCFA | Short-chain fatty acids |
| SCORAD | Scoring Atopic Dermatitis |
| SR-BI | Scavenger receptor class B type I |
| STAT6 | Signal transducer and activator of transcription 6 |
| TEWL | Transepidermal water loss |
| Th | T helper cells |
| TIS | Three-Item Severity |
| TJs | Tight junctions |
| TLR3 | Toll-like receptor 3 |
| tol-DCs | Tolerogenic dendritic cells |
| TNF-α | Tumor necrosis factor alpha |
| TRMs | Tissue-resident macrophages |
| Treg | Regulatory T cells |
| TSLP | Thymic stromal lymphopoietin |
| UVB | Ultraviolet B radiation |
| VD | Vitamin D |
| VD2 | Vitamin D2 (ergocalciferol) |
| VD3 | Vitamin D3 (cholecalciferol) |
| VDR | Vitamin D receptor |
| ZO | Zonula occludens |
References
- Borrego-Ruiz, A.; Borrego, J.J. Nutritional and Microbial Strategies for Treating Acne, Alopecia, and Atopic Dermatitis. Nutrients 2024, 16, 3559. [Google Scholar] [CrossRef] [PubMed]
- Khalil, N.; Walton, J.; Roberts, N.; Hussain, K. Viral infections in atopic dermatitis. Clin. Exp. Dermatol. 2024, 50, 46–55. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Li, G.; Guo, Q.; Yang, Y.; Yang, J.; Feng, X.; Yao, Z. Allergens in Atopic Dermatitis. Clin. Rev. Allergy Immunol. 2025, 68, 11. [Google Scholar] [CrossRef] [PubMed]
- Cao, L.; Su, J.; Tian, F.; Zhou, Y.; Liu, S.; Lou, F. Risk of depression in patients with atopic dermatitis: An updated systematic review and meta-analysis of children, adolescent and adult groups. J. Paediatr. Child Health 2024, 60, 640–647. [Google Scholar] [CrossRef]
- Fitzmaurice, W.; Silverberg, N.B. Long-Term Impact of Atopic Dermatitis on Quality of Life. Dermatol. Clin. 2024, 42, 549–557. [Google Scholar] [CrossRef]
- Chatrath, S.; Lei, D.; Yousaf, M.; Chavda, R.; Gabriel, S.; Silverberg, J.I. Longitudinal course and predictors of depressive symptoms in atopic dermatitis. J. Am. Acad. Dermatol. 2022, 87, 582–591. [Google Scholar] [CrossRef]
- Guttman-Yassky, E.; Renert-Yuval, Y.; Brunner, P.M. Atopic dermatitis. Lancet 2025, 405, 583–596. [Google Scholar] [CrossRef]
- Joshi, A.A.; Vocanson, M.; Nicolas, J.F.; Wolf, P.; Patra, V. Microbial derived antimicrobial peptides as potential therapeutics in atopic dermatitis. Front. Immunol. 2023, 14, 1125635. [Google Scholar] [CrossRef]
- Tokura, Y.; Hayano, S. Subtypes of atopic dermatitis: From phenotype to endotype. Allergol. Int. 2022, 71, 14–24. [Google Scholar] [CrossRef]
- Rusu, E.; Enache, G.; Cursaru, R.; Alexescu, A.; Radu, R.; Onila, O.; Cavallioti, T.; Rusu, F.; Posea, M.; Jinga, M.; et al. Prebiotics and probiotics in atopic dermatitis. Exp. Ther. Med. 2019, 18, 926–931. [Google Scholar] [CrossRef]
- Fyhrquist, N.; Yang, Y.; Karisola, P.; Alenius, H. Endotypes of atopic dermatitis. J. Allergy Clin. Immunol. 2025, 156, 24–40.e4. [Google Scholar] [CrossRef]
- Wienholtz, N.K.F.; Vestergaard, C.; Deleuran, M.; Drljevic-Nielsen, A.; Andersson, A.M. Atopisk eksem [Atopic dermatitis]. Ugeskr Laeger 2025, 187, V10240722. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.; Li, D.; Feng, Z.; Zhu, X.; Yang, F.; Zhang, B.; Hu, M.; Wang, Y.; Feng, H.; Yu, Y.; et al. Diterpenoid DGT alleviates atopic dermatitis-like responses in vitro and in vivo via targeting IL-4Rα. Biomed. Pharmacother. 2024, 179, 117321. [Google Scholar] [CrossRef] [PubMed]
- Afshari, M.; Kolackova, M.; Rosecka, M.; Čelakovská, J.; Krejsek, J. Unraveling the skin; a comprehensive review of atopic dermatitis, current understanding, and approaches. Front. Immunol. 2024, 15, 1361005. [Google Scholar] [CrossRef] [PubMed]
- Nevid, M.; Boguniewicz, M. Current and Emerging Biologics for Atopic Dermatitis. Immunol. Allergy Clin. N. Am. 2024, 44, 577–594. [Google Scholar] [CrossRef]
- Harris-Tryon, T.A.; Grice, E.A. Microbiota and maintenance of skin barrier function. Science 2022, 376, 940–945. [Google Scholar] [CrossRef]
- Gallagher, K.; Halperin-Goldstein, S.; Paller, A.S. New treatments in atopic dermatitis update. Ann. Allergy Asthma Immunol. 2025, 135, 498–510.e10. [Google Scholar] [CrossRef]
- Elizalde-Jiménez, I.G.; Ruiz-Hernández, F.G.; Carmona-Cruz, S.A.; Pastrana-Arellano, E.; Aquino-Andrade, A.; Romo-González, C.; Arias-de la Garza, E.; Álvarez-Villalobos, N.A.; García-Romero, M.T. Global Antimicrobial Susceptibility Patterns of Staphylococcus aureus in Atopic Dermatitis: A Systematic Review and Meta-Analysis. JAMA Dermatol. 2024, 160, 1171–1181. [Google Scholar] [CrossRef]
- Qian, X.; Tong, M.; Zhang, T.; Li, Q.; Hua, M.; Zhou, N.; Zeng, W. IL-24 promotes atopic dermatitis-like inflammation through driving MRSA-induced allergic responses. Protein Cell 2025, 16, 188–210. [Google Scholar] [CrossRef]
- Saheb Kashaf, S.; Harkins, C.P.; Deming, C.; Joglekar, P.; Conlan, S.; Holmes, C.J.; NISK Comparative Sequencing Program; Almeida, A.; Finn, R.D.; Segre, J.A.; et al. Staphylococcal diversity in atopic dermatitis from an individual to a global scale. Cell Host Microbe 2023, 31, 578–592.e6. [Google Scholar] [CrossRef]
- Demessant-Flavigny, A.L.; Connétable, S.; Kerob, D.; Moreau, M.; Aguilar, L.; Wollenberg, A. Skin microbiome dysbiosis and the role of Staphylococcus aureus in atopic dermatitis in adults and children: A narrative review. J. Eur. Acad. Dermatol. Venereol. 2023, 37 (Suppl. S5), 3–17. [Google Scholar] [CrossRef]
- Meledathu, S.; Naidu, M.P.; Brunner, P.M. Update on atopic dermatitis. J. Allergy Clin. Immunol. 2025, 155, 1124–1132. [Google Scholar] [CrossRef]
- Paternoster, L. Genetic landscape of atopic dermatitis. Curr. Opin. Allergy Clin. Immunol. 2024, 24, 409–415. [Google Scholar] [CrossRef] [PubMed]
- Thyssen, J.P.; Bikle, D.D.; Elias, P.M. Evidence that loss-of-function filaggrin gene mutations evolved in Northern Europeans to favor intracutaneous vitamin D3 production. Evol. Biol. 2014, 41, 388–396. [Google Scholar] [CrossRef] [PubMed]
- Criado, P.R.; Miot, H.A.; Bueno-Filho, R.; Ianhez, M.; Criado, R.F.J.; de Castro, C.C.S. Update on the pathogenesis of atopic dermatitis. An. Bras. Dermatol. 2024, 99, 895–915. [Google Scholar] [CrossRef] [PubMed]
- Sidbury, R.; Sullivan, A.F.; Thadhani, R.I.; Camargo, C.A. Randomized controlled trial of vitamin D supplementation for winter related atopic dermatitis in Boston: A pilot study. Br. J. Dermatol. 2008, 159, 245–247. [Google Scholar] [CrossRef]
- Sullivan, E.; Firnhaber, J. Atopic and Contact Dermatitis. Prim. Care 2025, 52, 553–566. [Google Scholar] [CrossRef]
- Bath-Hextall, F.J.; Jenkinson, C.; Humphreys, R.; Williams, H.C. Dietary supplements for established atopic eczema. Cochrane Database Syst. Rev. 2012, 2015, CD005205. [Google Scholar] [CrossRef]
- 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]
- Jimenez-Sanchez, M.; Celiberto, L.S.; Yang, H.; Sham, H.P.; Vallance, B.A. The gut-skin axis: A bi-directional, microbiota-driven relationship with therapeutic potential. Gut Microbes 2025, 17, 2473524. [Google Scholar] [CrossRef]
- Grieco, T.; Paolino, G.; Moliterni, E.; Chello, C.; Sernicola, A.; Egan, C.G.; Morelli, M.; Nannipieri, F.; Battaglia, S.; Accoto, M.; et al. Transcriptomic profiling of lesional and perilesional skin in atopic dermatitis suggests barrier dysfunction, inflammatory activation, and alterations to vitamin D metabolism. Int. J. Mol. Sci. 2025, 26, 6152. [Google Scholar] [CrossRef]
- Samanta, S. Vitamin D and immunomodulation in the skin: A useful affirmative nexus. Explor. Immunol. 2021, 1, 90–111. [Google Scholar] [CrossRef]
- 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]
- Li, Q.; Chan, H. Vitamin D and skin disorders: Bridging molecular insights to clinical innovations. Mol. Med. 2025, 31, 259. [Google Scholar] [CrossRef] [PubMed]
- Przechowski, K.; Krawczyk, M.N.; Krasowski, R.; Pawliczak, R.; Kleniewska, P. Vitamin D and atopic dermatitis—A mere correlation or a real supportive treatment option? Nutrients 2025, 17, 2582. [Google Scholar] [CrossRef]
- Udompataikul, M.; Huajai, S.; Chalermchai, T.; Taweechotipatr, M.; Kamanamool, N. The effects of oral vitamin D supplement on atopic dermatitis: A clinical trial with Staphylococcus aureus colonization determination. J. Med. Assoc. Thai 2015, 98 (Suppl. S9), S23–S30. [Google Scholar]
- Cabalín, C.; Pérez-Mateluna, G.; Iturriaga, C.; Camargo, C.A.J.r.; Borzutzky, A. Oral vitamin D modulates the epidermal expression of the vitamin D receptor and cathelicidin in children with atopic dermatitis. Arch. Dermatol. Res. 2023, 315, 761–770. [Google Scholar] [CrossRef]
- Delrue, C.; Speeckaert, M.M. Vitamin D and vitamin D-binding protein in health and disease. Int. J. Mol. Sci. 2023, 24, 4642. [Google Scholar] [CrossRef]
- Cashman, K.D.; Kiely, M. Chapter 58-Vitamin D and Food Fortification. W: Feldman & Pike’s Vitamin D; Elsevier: Amsterdam, The Netherlands, 2024; pp. 135–160. [Google Scholar] [CrossRef]
- Crafa, A.; Cannarella, R.; Cannarella, V.; Condorelli, R.A.; La Vignera, S.; Calogero, A.E. Retrospective real world study on vitamin D supplementation: Looking for the most effective molecule and its frequency of use. Clin. Nutr. 2025, 47, 265–274. [Google Scholar] [CrossRef]
- Wu, S.E.; Chen, W.L. Moderate sun exposure is the complementor in insufficient vitamin D consumers. Front. Nutr. 2022, 9, 832659. [Google Scholar] [CrossRef]
- Kallioğlu, M.A.; Sharma, A.; Kallioğlu, A.; Kumar, S.; Khargotra, R.; Singh, T. UV index-based model for predicting synthesis of (pre-)vitamin D3 in the Mediterranean basin. Sci. Rep. 2024, 14, 3541. [Google Scholar] [CrossRef] [PubMed]
- Reboul, E. Proteins involved in fat-soluble vitamin and carotenoid transport across the intestinal cells: New insights from the past decade. Prog. Lipid Res. 2023, 92, 101166. [Google Scholar] [CrossRef] [PubMed]
- Kiourtzidis, M. Modifiers of Absorption, Tissue Distribution and Activation of Vitamin D: The Role of NPC1L1, SR-B1, CD36, and ABC-G5/G8 Transporters. Ph.D. Thesis, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany, 2023. Available online: https://d-nb.info/134023386X/34 (accessed on 3 September 2025).
- Bendotti, G.; Biamonte, E.; Leporati, P.; Goglia, U.; Ruggeri, R.M.; Gallo, M. Vitamin D supplementation: Practical advice in different clinical settings. Nutrients 2025, 17, 783. [Google Scholar] [CrossRef] [PubMed]
- Kampka, Z.; Czapla, D.; Wojakowski, W.; Stanek, A. Vitamin D supplementation in heart failure—Confusion without a cause? Nutrients 2025, 17, 1839. [Google Scholar] [CrossRef]
- Kise, S.; Iijima, A.; Nagao, C.; Okada, T.; Mano, H.; Nishikawa, M.; Ikushiro, S.; Kanemoto, Y.; Kato, S.; Nakanishi, T.; et al. Functional analysis of vitamin D receptor (VDR) using adenovirus vector. J. Steroid Biochem. Mol. Biol. 2023, 230, 106275. [Google Scholar] [CrossRef]
- Casella, A.; Long, C.; Zhou, J.; Lai, M.; O’Lear, L.; Caplan, I.; Levine, M.A.; Roizen, J.D. Differential frequency of CYP2R1 variants across populations reveals pathway selection for vitamin D homeostasis. J. Clin. Endocrinol. Metab. 2020, 105, 1302–1315. [Google Scholar] [CrossRef]
- Baroncelli, G.I.; Comberiati, P.; Aversa, T.; Baronio, F.; Cassio, A.; Chiarito, M.; Cosci o di Coscio, M.; De Sanctis, L.; Di Iorgi, N.; Faienza, M.F.; et al. Diagnosis, treatment, and management of rickets: A position statement from the Bone and Mineral Metabolism Group of the Italian Society of Pediatric Endocrinology and Diabetology. Front. Endocrinol. 2024, 15, 1383681. [Google Scholar] [CrossRef]
- Wang, L.K.; Shanmugasundaram, M.; Cooney, E.; Lee, P.D.K. Siblings with vitamin D-dependent rickets type 1A: Importance of genetic testing and a review of genotype-phenotype correlations. Am. J. Med. Genet. A 2024, 194, e63780. [Google Scholar] [CrossRef]
- Bin Rubaian, N.F.; Al-Awam, B.S.; Aljohani, S.M.; Almuhaidib, S.R. Alopecia with vitamin D-dependent rickets type 2A: A case report. Clin. Cosmet. Investig. Dermatol. 2024, 17, 13–16. [Google Scholar] [CrossRef]
- Jiao, Q.; Zhi, L.; You, B.; Wang, G.; Wu, N.; Jia, Y. Skin homeostasis: Mechanism and influencing factors. J. Cosmet. Dermatol. 2024, 23, 1518–1526. [Google Scholar] [CrossRef]
- Kwiecien, K.; Zegar, A.; Jung, J.; Brzoza, P.; Kwitniewski, M.; Godlewska, U.; Grygier, B.; Kwiecinska, P.; Morytko, A.; Cichy, J. Architecture of antimicrobial skin defense. Cytokine Growth Factor Rev. 2019, 49, 70–84. [Google Scholar] [CrossRef]
- Agarwal, S.; Krishnamurthy, K. Histology, Skin. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- Zwirner, J.; Hammer, N. Anatomy and physiology of the skin. In Scars; Nischwitz, S.P., Kamolz, L.P., Branski, L.K., Eds.; Springer: Cham, Switzerland, 2024; pp. 1–16. [Google Scholar] [CrossRef]
- MacGibeny, M.A.; Adjei, S.; Pyle, H.; Bunick, C.G.; Ghannoum, M.; Grada, A.; Harris-Tryon, T.; Tyring, S.K.; Kong, H.H. The human skin microbiome in health. J. Am. Acad. Dermatol. 2025, 93, 329–336. [Google Scholar] [CrossRef] [PubMed]
- Ahle, C.M.; Stødkilde, K.; Poehlein, A.; Bömeke, M.; Streit, W.R.; Wenck, H.; Reuter, J.H.; Hüpeden, J.; Brüggemann, H. Interference and co-existence of staphylococci and Cutibacterium acnes within the healthy human skin microbiome. Commun. Biol. 2022, 5, 923. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Peng, G.; Abudouwanli, A.; Yang, M.; Sun, Q.; Zhao, W.; Ikeda, A.; Tan, Y.; Ma, L.; Ogawa, H.; et al. The interaction between the skin microbiome and antimicrobial peptides within the epidermal immune microenvironment: Bridging insights into atopic dermatitis. Allergol. Int. 2025. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.E.; Zheng, P.; Ye, S.Z.; Ma, X.; Liu, E.; Pang, Y.B.; He, Q.Y.; Zhang, Y.X.; Li, W.Q.; Zeng, J.H.; et al. Microbiome: Role in Inflammatory Skin Diseases. J. Inflamm. Res. 2024, 17, 1057–1082. [Google Scholar] [CrossRef]
- Puls, J.-S.; Winnerling, B.; Power, J.J.; Krüger, A.M.; Brajtenbach, D.; Johnson, M.; Bilici, K.; Camus, L.; Fließwasser, T.; Schneider, T.; et al. Staphylococcus epidermidis bacteriocin A37 kills natural competitors with a unique mechanism of action. ISME J. 2024, 18, wrae044. [Google Scholar] [CrossRef]
- Severn, M.M.; Horswill, A.R. Staphylococcus epidermidis and its dual lifestyle in skin health and infection. Nat. Rev. Microbiol. 2023, 21, 97–111. [Google Scholar] [CrossRef]
- Lima, R.D.; Hajiarbabi, K.; Ng, B.D.; Sood, A.; Ferreira, R.B.R. Skin-associated commensal microorganisms and their metabolites. J. Appl. Microbiol. 2025, 136, lxaf111. [Google Scholar] [CrossRef]
- Zheng, Y.; Hunt, R.L.; Villaruz, A.E.; Fisher, E.L.; Liu, R.; Liu, Q.; Cheung, G.Y.C.; Li, M.; Otto, M. Commensal Staphylococcus epidermidis contributes to skin barrier homeostasis by generating protective ceramides. Cell Host Microbe 2022, 30, 301–313.e9. [Google Scholar] [CrossRef]
- Wang, W.-L.; Lai, Y.-H.; Huang, C.-H.; Lai, J.-Y.; Yao, C.-H. Lumbrokinase-containing gelatin nanofibers with multiple bioactivities for effective skin wound healing. Mater. Today Bio. 2025, 32, 101713. [Google Scholar] [CrossRef]
- Boraldi, F.; Lofaro, F.D.; Bonacorsi, S.; Mazzilli, A.; Garcia-Fernandez, M.; Quaglino, D. The role of fibroblasts in skin homeostasis and repair. Biomedicines 2024, 12, 1586. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Duan, Z.; Chen, X.; Li, M. The immune function of dermal fibroblasts in skin defence against pathogens. Exp. Dermatol. 2023, 32, 1326–1333. [Google Scholar] [CrossRef] [PubMed]
- Fleckner, M.; Döhmen, N.K.; Salz, K.; Christophers, T.; Windolf, J.; Suschek, C.V.; Oezel, L. Exposure of primary human skin fibroblasts to carbon dioxide-containing solution significantly reduces TGF-β-induced myofibroblast differentiation in vitro. Int. J. Mol. Sci. 2024, 25, 13013. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Duan, Z.; Zeng, R.; Yang, L.; Liu, W.; Liu, Y.; Yao, Q.; Chen, X.; Zhang, L.-J.; Li, M. Antimicrobial peptide-producing dermal preadipocytes defend against Candida albicans skin infection via the FGFR-MEK-ERK pathway. PLoS Pathog. 2023, 19, e1011754. [Google Scholar] [CrossRef]
- Lee, S.H.; Sacks, D.L. Resilience of dermis resident macrophages to inflammatory challenges. Exp. Mol. Med. 2024, 56, 2105–2112. [Google Scholar] [CrossRef]
- Guan, F.; Wang, R.; Yi, Z.; Luo, P.; Liu, W.; Xie, Y.; Liu, Z.; Xia, Z.; Zhang, H.; Cheng, Q. Tissue macrophages: Origin, heterogeneity, biological functions, diseases and therapeutic targets. Signal Transduct. Target. Ther. 2025, 10, 93. [Google Scholar] [CrossRef]
- Liu, G.; Wang, Z.; Li, S. Heterogeneity and plasticity of tissue-resident memory T cells in skin diseases and homeostasis: A review. Front. Immunol. 2024, 15, 1378359. [Google Scholar] [CrossRef]
- Ruchti, F.; LeibundGut-Landmann, S. New insights into immunity to skin fungi shape our understanding of health and disease. Parasite Immunol. 2023, 45, e12948. [Google Scholar] [CrossRef]
- Reider, I.E.; Lin, E.; Krouse, T.E.; Parekh, N.J.; Nelson, A.M.; Norbury, C.C. γδ T cells mediate a requisite portion of a wound healing response triggered by cutaneous poxvirus infection. Viruses 2024, 16, 425. [Google Scholar] [CrossRef]
- Lujan, R.A.; Pei, L.; Shannon, J.P.; Dábilla, N.; Dolan, P.T.; Hickman, H.D. Widespread and dynamic expression of granzyme C by skin-resident antiviral T cells. Front. Immunol. 2023, 14, 1236595. [Google Scholar] [CrossRef]
- Ziadlou, R.; Pandian, G.N.; Hafner, J.; Akdis, C.A.; Stingl, G.; Maverakis, E.; Brüggen, M.C. Subcutaneous adipose tissue: Implications in dermatological diseases and beyond. Allergy 2024, 79, 3310–3325. [Google Scholar] [CrossRef]
- Guan, J.; Wu, C.; He, Y.; Lu, F. Skin-associated adipocytes in skin barrier immunity: A mini-review. Front. Immunol. 2023, 14, 1116548. [Google Scholar] [CrossRef] [PubMed]
- Erfanian, S.; Jafaripour, S.; Jokar, M.H.; Sedighi, S.; Jahromi, A.S.; Razavi, F.; Zargar, M.F.; Moradzadeh, M. The effect of vitamin D on GATA3 gene expression in peripheral blood mononuclear cells in allergic asthma. Adv. Respir. Med. 2022, 90, 118–124. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Kim, S.; Lee, S.K.; Jeong, S.-Y.; Jung, M.; Han, S.-D.; Lee, J.; You, H.; Kim, H.; Kim, T.M. Amelioration of atopic dermatitis via suppression of Th2 receptor expression by miR-22-3p and HSP70 in extracellular vesicles. J. Allergy Clin. Immunol. 2025, 155, AB222. [Google Scholar] [CrossRef]
- Li, D.; Ma, X.; Zhang, W.; Zhong, P.; Li, M.; Liu, S. Impact of vitamin D3 supplementation on motor functionality and the immune response in Parkinson’s disease patients with vitamin D deficiency. Sci. Rep. 2025, 15, 25154. [Google Scholar] [CrossRef]
- Cartes-Velásquez, R.; Vera, A.; Torres-Quevedo, R.; Medrano-Díaz, J.; Pérez, A.; Muñoz, C.; Carrillo-Bestagno, H.; Nova-Lamperti, E. The immunomodulatory role of vitamin D in regulating the Th17/Treg balance and epithelial-mesenchymal transition: A hypothesis for gallbladder cancer. Nutrients 2024, 16, 4134. [Google Scholar] [CrossRef]
- Wang, Z.; Hülpüsch, C.; Traidl-Hoffmann, C.; Reiger, M.; Schloter, M. Understanding the role of Staphylococcus aureus in atopic dermatitis: Strain diversity, microevolution, and prophage influences. Front. Med. 2024, 11, 1480257. [Google Scholar] [CrossRef]
- Aidoukovitch, A.; Bankell, E.; Svensson, D.; Nilsson, B.O. Vitamin D triggers hCAP18/LL-37 production: Implications for LL-37-induced human osteoblast cytotoxicity. Biochem. Biophys. Res. Commun. 2024, 712–713, 149962. [Google Scholar] [CrossRef]
- Bishop, L.; Ismailova, A.; Dimeloe, S.; Hewison, M.; White, J.H. Vitamin D and immune regulation: Antibacterial, antiviral, anti-inflammatory. JBMR Plus 2020, 5, e10405. [Google Scholar] [CrossRef]
- Artusa, P.; White, J.H. Vitamin D and its analogs in immune system regulation. Pharmacol. Rev. 2025, 77, 100032. [Google Scholar] [CrossRef]
- Napolitano, M.; di Vico, F.; Ruggiero, A.; Fabbrocini, G.; Patruno, C. The hidden sentinel of the skin: An overview on the role of interleukin-13 in atopic dermatitis. Front. Med. 2023, 10, 1165098. [Google Scholar] [CrossRef]
- Moosbrugger-Martinz, V.; Leprince, C.; Méchin, M.C.; Simon, M.; Blunder, S.; Gruber, R.; Dubrac, S. Revisiting the roles of filaggrin in atopic dermatitis. Int. J. Mol. Sci. 2022, 23, 5318. [Google Scholar] [CrossRef]
- Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-chain fatty-acid-producing bacteria: Key components of the human gut microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.T.; Chiu, C.J.; Tsai, C.Y.; Lee, Y.-R.; Liu, W.-L.; Chuang, H.-L.; Huang, M.-T. Short-chain fatty acids ameliorate allergic airway inflammation via sequential induction of PMN-MDSCs and Treg cells. J. Allergy Clin. Immunol. Glob. 2023, 2, 100163. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.H.; Kim, K.; Kim, W. Gut microbiota restoration through fecal microbiota transplantation: A new atopic dermatitis therapy. Exp. Mol. Med. 2021, 53, 907–916. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.F.; Shao, J.H.; Liao, Y.T.; Wang, L.-N.; Jia, Y.; Dong, P.-J.; Liu, Z.-Z.; He, D.-D.; Li, C.; Zhang, X. Regulation of short-chain fatty acids in the immune system. Front. Immunol. 2023, 14, 1186892. [Google Scholar] [CrossRef]
- Kim, K.; Jang, H.; Kim, E.; Kim, H.; Sung, G.Y. Recent advances in understanding the role of the skin microbiome in the treatment of atopic dermatitis. Exp. Dermatol. 2023, 32, 2048–2061. [Google Scholar] [CrossRef]
- Sun, J.; Zhang, Y.G. Vitamin D receptor influences intestinal barriers in health and disease. Cells 2022, 11, 1129. [Google Scholar] [CrossRef]
- Wang, J.; Mei, L.; Hao, Y.; Xu, Y.; Yang, Q.; Dai, Z.; Yang, Y.; Wu, Z.; Ji, Y. Contemporary perspectives on the role of vitamin D in enhancing gut health and its implications for preventing and managing intestinal diseases. Nutrients 2024, 16, 2352. [Google Scholar] [CrossRef]
- Grieco, T.; Moliterni, E.; Paolino, G.; Chello, C.; Sernicola, A.; Egan, C.G.; Nannipieri, F.; Battaglia, S.; Accoto, M.; Tirotta, E.; et al. Association between vitamin D receptor polymorphisms, tight junction proteins and clinical features of adult patients with atopic dermatitis. Dermatol. Pract. Concept. 2024, 14, e2024214. [Google Scholar] [CrossRef]
- Nguyen, H.D.; Aljamaei, H.M.; Stadnyk, A.W. The production and function of endogenous interleukin-10 in intestinal epithelial cells and gut homeostasis. Cell Mol. Gastroenterol. Hepatol. 2021, 12, 1343–1352. [Google Scholar] [CrossRef]
- Sun, L.; Wang, L.; Moore, B.B.; Zhang, S.; Xiao, P.; Decker, A.M.; Wang, H.-L. IL-17: Balancing protective immunity and pathogenesis. J. Immunol. Res. 2023, 2023, 3360310. [Google Scholar] [CrossRef]
- Davydova, A.; Kurochkina, Y.; Goncharova, V.; Vorobyeva, M.; Korolev, M. The interleukin-17 cytokine family: Role in development and progression of spondyloarthritis, current and potential therapeutic inhibitors. Biomedicines 2023, 11, 1328. [Google Scholar] [CrossRef]
- Porbahaie, M.; Hummel, A.; Saouadogo, H.; Coelho, R.M.L.; Savelkokoul, H.F.J.; Teodorowicz, M.; van Neerven, R.J.J. Short-chain fatty acids inhibit the activation of T lymphocytes and myeloid cells and induce innate immune tolerance. Benef. Microbes 2023, 14, 401–419. [Google Scholar] [CrossRef]
- Hatano, Y.; Elias, P.M. “Outside-to-inside,” “inside-to-outside,” and “intrinsic” endogenous pathogenic mechanisms in atopic dermatitis: Keratinocytes as the key functional cells involved in both permeability barrier dysfunction and immunological alterations. Front. Immunol. 2023, 14, 1239251. [Google Scholar] [CrossRef] [PubMed]
- Berdyshev, E. Skin lipid barrier: Structure, function and metabolism. Allergy Asthma Immunol. Res. 2024, 16, 445–461. [Google Scholar] [CrossRef] [PubMed]
- Bahman, F.; Choudhry, K.; Al-Rashed, F.; Al-Mulla, F.; Sindhu, S.; Ahmad, R. Aryl hydrocarbon receptor: Current perspectives on key signaling partners and immunoregulatory role in inflammatory diseases. Front. Immunol. 2024, 15, 1421346. [Google Scholar] [CrossRef] [PubMed]
- Rakateli, L.; Huchzermeier, R.; van der Vorst, E.P.C. AhR, PXR and CAR: From xenobiotic receptors to metabolic sensors. Cells 2023, 12, 2752. [Google Scholar] [CrossRef]
- Han, H.; Safe, S.; Jayaraman, A.; Chapkin, R.S. Diet-host-microbiota interactions shape aryl hydrocarbon receptor ligand production to modulate intestinal homeostasis. Annu. Rev. Nutr. 2021, 41, 455–478. [Google Scholar] [CrossRef]
- Wang, M.; Guo, J.; Hart, A.L.; Li, J.V. Indole-3-aldehyde reduces inflammatory responses and restores intestinal epithelial barrier function partially via aryl hydrocarbon receptor (AhR) in experimental colitis models. J. Inflamm. Res. 2023, 16, 5845–5864. [Google Scholar] [CrossRef]
- Kim, J.E.; Kim, H.S. Microbiome of the skin and gut in atopic dermatitis (AD): Understanding the pathophysiology and finding novel management strategies. J. Clin. Med. 2019, 8, 444. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Xu, Y.; Wang, L.; Lin, J.; Xu, C.; Zhao, X.; Zhang, H. TolDC restores the balance of Th17/Treg via aryl hydrocarbon receptor to attenuate colitis. Inflamm. Bowel Dis. 2024, 30, 1546–1555. [Google Scholar] [CrossRef] [PubMed]
- Tan, J.; Taitz, J.; Sun, S.M.; Langford, L.; Ni, D.; Macia, L. Your regulatory T cells are what you eat: How diet and gut microbiota affect regulatory T cell development. Front. Nutr. 2022, 9, 878382. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Hener, P.; Zhang, Z.; Kato, S.; Metzger, D.; Chambon, P. Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proc. Natl. Acad. Sci. USA 2006, 103, 11736–11741. [Google Scholar] [CrossRef]
- Landheer, J.; Giovannone, B.; Sadekova, S.; Tjabringa, S.; Hofstra, C.; Dechering, K.; Bruijnzeel-Koomen, C.; Chang, C.; Ying, Y.; de Waal Malefyt, R.; et al. TSLP is differentially regulated by vitamin D3 and cytokines in human skin. Immun. Inflamm. Dis. 2015, 3, 32–43. [Google Scholar] [CrossRef]
- Sakamoto, K.; Nagao, K. Mouse models for atopic dermatitis. Curr. Protoc. 2023, 3, e709. [Google Scholar] [CrossRef]
- Hoshino, Y.; Kirima, K.; Arichika, N.; Kakumoto, Y.; Shibamori, M.; Matsumoto, S.; Hiyama, H. Long-term application of MC903 in mice prolongs the characteristic symptoms of atopic dermatitis, such as inflammation, skin barrier dysfunction, and itching. Exp. Anim. 2025, 74, 276–285. [Google Scholar] [CrossRef]
- Choi, J.; Sutaria, N.; Roh, Y.S.; Bordeaux, Z.; Alphonse, M.P.; Kwatra, S.G.; Kwatra, M.M. Translational relevance of mouse models of atopic dermatitis. J. Clin. Med. 2021, 10, 613. [Google Scholar] [CrossRef]
- Antal, A.S.; Dombrowski, Y.; Koglin, S.; Ruzicka, T.; Schaubre, J. Impact of vitamin D3 on cutaneous immunity. J. Investig. Dermatol. 2011, 131, 194–204. [Google Scholar] [CrossRef]
- Bikle, D.D. Vitamin D and the skin: Physiology and pathophysiology. Rev. Endocr. Metab. Disord. 2012, 13, 3–19. [Google Scholar] [CrossRef]
- Hata, T.R.; Kotol, P.; Jackson, M.; Nguyen, M.; Paik, A.; Udall, D.; Kanada, K.; Yamasaki, K.; Alexandrescu, D.; Gallo, R.L. Administration of oral vitamin D induces cathelicidin production in atopic individuals. J. Allergy Clin. Immunol. 2008, 122, 829–831. [Google Scholar] [CrossRef] [PubMed]
- Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef] [PubMed]
- Umehara, Y.; Trujillo-Paez, J.V.; Yue, H.; Peng, G.; Nguyen, H.L.T.; Okumura, K.; Ogawa, H.; Niyonsaba, F. Calcitriol, an active form of vitamin D3, mitigates skin barrier dysfunction in atopic dermatitis NC/Nga mice. Int. J. Mol. Sci. 2023, 24, 9347. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.; Lee, K.; Oh, K. mTORC1 deficiency prevents the development of MC903-induced atopic dermatitis through the downregulation of type 2 inflammation. Int. J. Mol. Sci. 2023, 24, 5968. [Google Scholar] [CrossRef]
- Gupta, A.; Song, M.H.; Youn, D.H.; Ku, D.; Sasidharan Nair, V.; Oh, K. Prolyl hydroxylase inhibition protects against murine MC903-induced skin inflammation by downregulating TSLP. Front. Immunol. 2024, 15, 1330011. [Google Scholar] [CrossRef]
- Wan, H.; Yang, H.; Wei, M.; Chen, W. Polyinosinic:polycytidylic acid aggravates calcipotriol-induced atopic dermatitis-like skin lesions in mice by increasing the expression of thymic stromal lymphopoietin. Ann. Transl. Med. 2022, 10, 209. [Google Scholar] [CrossRef]
- Alam, M.J.; Xie, L.; Yap, Y.A.; Robert, R. A mouse model of MC903-induced atopic dermatitis. Curr. Protoc. 2023, 3, e695. [Google Scholar] [CrossRef]
- Reinholz, M.; Ruzicka, T.; Schauber, J. Vitamin D and its role in allergic disease. Clin. Exp. Allergy 2012, 42, 817–826. [Google Scholar] [CrossRef]
- Gallieni, M.; Cozzolino, M.; Fallabrino, G.; Pasho, S.; Olivi, L.; Brancaccio, D. Vitamin D: Physiology and pathophysiology. Int. J. Artif. Organs 2009, 32, 87–94. [Google Scholar] [CrossRef]
- Pike, J.; Meyer, M.B. The vitamin D receptor: New paradigms for the regulation of gene expression by 1, 25-dihydroxyvitamin D3. Endocrinol. Metab. Clin. 2010, 39, 255–269. [Google Scholar] [CrossRef]
- Luderer, H.F.; Demay, M.B. The vitamin D receptor, the skin and stem cells. J. Steroid Biochem. Mol. Biol. 2010, 121, 314–316. [Google Scholar] [CrossRef]
- Guo, Y.; Li, X.; Geng, C.; Song, S.; Xie, X.; Wang, C. Vitamin D receptor involves in the protection of intestinal epithelial barrier function via up-regulating SLC26A3. J. Steroid Biochem. Mol. Biol. 2023, 227, 106231. [Google Scholar] [CrossRef]
- De Benedetto, A.; Rafaels, N.M.; McGirt, L.Y.; Ivanov, A.I.; Georas, S.N.; Cheadle, C.; Berger, A.E.; Zhang, K.; Vidyasagar, S.; Yoshida, T.; et al. Tight junction defects in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2010, 127, 773–786. [Google Scholar] [CrossRef]
- Bergmann, S.; von Buenau, B.; Vidal-Y-Sy, S.; Haftek, M.; Wladykowski, E.; Houdek, P.; Lezius, S.; Duplan, H.; Bäsler, K.; Dähnhardt-Pfeiffer, S.; et al. Claudin-1 decrease impacts epidermal barrier function in atopic dermatitis lesions dose-dependently. Sci. Rep. 2020, 145, 1708–1719. [Google Scholar] [CrossRef] [PubMed]
- Xia, Y.; Cao, H.; Zheng, J.; Chen, L. Claudin-1 Mediated Tight Junction Dysfunction as a Contributor to Atopic March. Front Immunol. 2022, 13, 927465. [Google Scholar] [CrossRef] [PubMed]
- Bollag, W.B.; Aitkens, L.; White, J.; Hyndman, K.A. Aquaporin-3 in the epidermis: More than skin deep. Am. J. Physiol. Cell Physiol. 2020, 318, C1144–C1153. [Google Scholar] [CrossRef] [PubMed]
- Hara-Chikuma, M.; Verkman, A.S. Aquaporin-3 functions as a glycerol transporter in mammalian skin. Biol. Cell 2005, 97, 479–486. [Google Scholar] [CrossRef]
- Tricarico, P.M.; Mentino, D.; De Marco, A.; Del Vecchio, C.; Garra, S.; Cazzato, G.; Foti, C.; Crovella, S.; Calamita, G. Aquaporins are one of the critical factors in the disruption of the skin barrier in inflammatory skin diseases. Int. J. Mol. Sci. 2022, 23, 4020. [Google Scholar] [CrossRef]
- Wang, S.-H.; Zuo, Y.-G. Thymic stromal lymphopoietin in cutaneous immune-mediated diseases. Front. Immunol. 2021, 12, 698522. [Google Scholar] [CrossRef]
- Kim, M.J.; Kim, S.-N.; Lee, Y.W.; Choe, Y.B.; Ahn, K.J. Vitamin D status and efficacy of supplementation in atopic dermatitis: A systematic review and meta-analysis. Nutrients 2016, 8, 789. [Google Scholar] [CrossRef]
- Çiçek, F.; Köle, M.T. Evaluation of the impact of serum vitamin D levels on the scoring atopic dermatitis index in pediatric atopic dermatitis. Children 2023, 10, 1522. [Google Scholar] [CrossRef]
- McCarthy, R.L.; Tawfik, S.S.; Theocharopoulos, I.; Atkar, R.; McDonald, B.; Dhoat, S.; Hughes, A.; Thomas, B.R.; O’Toole, E.A. Vitamin D deficiency and atopic dermatitis severity in a Bangladeshi population living in East London: A cross-sectional study. Skin Health Dis. 2024, 4, e358. [Google Scholar] [CrossRef] [PubMed]
- Tabassum, A.; Ali, A.; Zahedi, F.D.; Ismail, N.A.S. Immunomodulatory role of vitamin D on gut microbiome in children. Biomedicines 2023, 11, 1441. [Google Scholar] [CrossRef] [PubMed]
- Pantazi, A.C.; Nori, W.; Kassim, M.A.K.; Balasa, A.L.; Mihai, C.M.; Chisnoiu, T.; Mihai, L.; Ungureanu, A.; Frecus, C.E.; Chirila, S.I.; et al. Gut microbiota profile and atopic dermatitis in the first year of life. J. Med. Life 2024, 17, 948–952. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Li, Z.; Zhang, W.; Zhang, C.; Zhang, Y.; Mei, H.; Zhuo, N.; Wang, H.; Wang, L.; Wu, D. Comparison of gut microbiota in exclusively breast-fed and formula-fed babies: A study of 91 term infants. Sci. Rep. 2020, 10, 15792. [Google Scholar] [CrossRef]
- Davis, E.C.; Castagna, V.P.; Sela, D.A.; Hillard, M.A.; Lindberg, S.; Mantis, N.J.; Seppo, A.E.; Järvinen, K.M. Gut microbiome and breast-feeding: Implications for early immune development. J. Allergy Clin. Immunol. 2022, 150, 523–534. [Google Scholar] [CrossRef]
- Ojaroodi, A.F.; Jafarnezhad, F.; Eskandari, Z.; Keramat, S.; Stanek, A. Recent updates and advances in the association between vitamin D deficiency and risk of thrombotic disease. Nutrients 2024, 17, 90. [Google Scholar] [CrossRef]
- Zhao, H.; Luo, Y.; Li, W.; Jiang, C.; Jin, E.; Xu, Z. Association between antibiotic exposure and childhood atopic dermatitis: A systematic review and meta-analysis. EClinicalMedicine 2025, 84, 103296. [Google Scholar] [CrossRef]
- Tang, H.; Li, W.; Xu, Y.; Zhou, Y.; Hamblin, M.R.; Wen, X. Gut microbiota modulation: A key determinant of atopic dermatitis susceptibility in children. Front. Microbiol. 2025, 16, 1549895. [Google Scholar] [CrossRef]
- Thangamuni, A.; Fathimathul Harshiba, H.; Muhammed Rafi, N.; Nitol, A.; Mohan, J.; Korrapati, N. Beauty from within: A comprehensive review on interplay between gut health and skin. CosmoDerma 2024, 4, 97. [Google Scholar] [CrossRef]
- 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 2023, 11, 274. [Google Scholar] [CrossRef]
- Amestejani, M.; Seyed Salehi, B.; Vasigh, M.; Sobhkhiz, A.; Karami, M.; Alinia, H.; Kamrava, S.K.; Shamspour, N.; Ghalehbaghi, B.; Heshmatzade Behzadi, A. Vitamin D supplementation in the treatment of atopic dermatitis: A clinical trial study. J. Drugs Dermatol. 2012, 11, 327–330. [Google Scholar]
- Park, J.S.; Kim, S.; Sol, I.S.; Lee, K.S.; Park, S.; Yang, H.J.; Lee, E. Effect of vitamin D on the treatment of atopic dermatitis with consideration of heterogeneities: Meta-analysis of randomized controlled trials. Allergy Asthma Immunol. Res. 2023, 15, 262–270. [Google Scholar] [CrossRef]
- Vaughn, A.R.; Foolad, N.; Maarouf, M.; Tran, K.A.; Shi, V.Y. Micronutrients in atopic dermatitis: A systematic review. J. Altern. Complement. Med. 2019, 25, 567–577. [Google Scholar] [CrossRef]
- Giustina, A.; Bilezikian, J.P.; Adler, R.A.; Banfi, G.; Bikle, D.D.; Binkley, N.C.; Bollerslev, J.; Bouillon, R.; Brandi, M.L.; Casanueva, F.F.; et al. Consensus statement on vitamin D status assessment and supplementation: Whys, whens, and hows. Endocr. Rev. 2024, 45, 625–654. [Google Scholar] [CrossRef] [PubMed]
- Ziada, S.; Wishahe, A.; Mabrouk, N.; Sahtout, S. Vitamin D deficiency and oral health: A systematic review of literature. BMC Oral Health 2025, 25, 468. [Google Scholar] [CrossRef] [PubMed]
- Venter, F.C.; Ghitea, T.C.; Venter, A.N.; El-Kharoubi, A.F.; El-Kharoubi, M.; Ghitea, E.C.; Ghitea, M.C.; Venter, A. Correlation between vitamin D deficiency (25(OH)D3) and the severity of purulent oropharyngeal infections. J. Clin. Med. 2025, 14, 2410. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Cao, Y. Association between atopic dermatitis with hyperparathyroidism not mediated by vitamin D in the United States (NHANES 2005–2006). Arch. Dermatol. Res. 2024, 317, 100. [Google Scholar] [CrossRef]
- Grant, W.B.; Wimalawansa, S.J.; Pludowski, P.; Cheng, R.Z. Vitamin D: Evidence-based health benefits and recommendations for population guidelines. Nutrients 2025, 17, 277. [Google Scholar] [CrossRef]
- Ohya, Y. Prevention of atopic dermatitis: What are we missing? Allergy Asthma Immunol. Res. 2025, 17, 433–446. [Google Scholar] [CrossRef]
- Pludowski, P.; Marcinowska-Suchowierska, E.; Togizbayev, G.; Belaya, Z.; Grant, W.B.; Pilz, S.; Holick, M.F. Daily and weekly “high doses” of cholecalciferol for the prevention and treatment of vitamin D deficiency for obese or multi-morbidity and multi-treatment patients requiring multi-drugs—A narrative review. Nutrients 2024, 16, 2541. [Google Scholar] [CrossRef] [PubMed]
- Adeloye, D.; Song, P.; Zhu, Y.; Campbell, H.; Sheikh, A.; Rudan, I.; NIHR RESPIRE Global Respiratory Health Unit. Global, regional, and national prevalence of, and risk factors for, chronic obstructive pulmonary disease (COPD) in 2019: A systematic review and modelling analysis. Lancet Respir. Med. 2022, 10, 447–458. [Google Scholar] [CrossRef] [PubMed]
- Borzutzky, A.; Iturriaga, C.; Pérez-Mateluna, G.; Cristi, F.; Cifuentes, L.; Silva-Valenzuela, S.; Vera-Kellet, C.; Cabalin, C.; Hoyos-Bachiloglu, R.; Navarrete-Dechent, C.; et al. Effect of weekly vitamin D supplementation on the severity of atopic dermatitis and type 2 immunity biomarkers in children: A randomized controlled trial. J. Eur. Acad. Dermatol. Venereol. 2024, 38, 1760–1768. [Google Scholar] [CrossRef] [PubMed]
- 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. J. Immunol. Res. 2022, 2022, 9407888. [Google Scholar] [CrossRef]
- Vasdeki, D.; Tsamos, G.; Dimakakos, E.; Patriarcha, V.; Koufakis, T.; Kotsa, K.; Cholewka, A.; Stanek, A. Vitamin D Supplementation: Shedding Light on the Role of the Sunshine Vitamin in the Prevention and Management of Type 2 Diabetes and Its Complications. Nutrients 2024, 16, 3651. [Google Scholar] [CrossRef]
- Wimalawansa, S.J. Physiology of vitamin D—Focusing on disease prevention. Nutrients 2024, 16, 1666. [Google Scholar] [CrossRef]
- Chauhan, K.; Shahrokhi, M.; Huecker, M.R. Vitamin D. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK441912/ (accessed on 3 September 2025).
- Connell, T.; Seidler, K.; Neil, J. Cathelicidin expression in the pathogenesis of atopic dermatitis and the therapeutic potential of vitamin D. Nutr. Res. 2025, 139, 113–123. [Google Scholar] [CrossRef]
- Rizzoli, R. Vitamin D supplementation: Upper limit for safety revisited? Aging Clin. Exp. Res. 2021, 33, 19–24. [Google Scholar] [CrossRef]
- Diotallevi, F.; Campanati, A.; Martina, E.; Radi, G.; Paolinelli, M.; Marani, A.; Molinelli, E.; Candelora, M.; Taus, M.; Galeazzi, T.; et al. The role of nutrition in immune-mediated, inflammatory skin disease: A narrative review. Nutrients 2022, 14, 591. [Google Scholar] [CrossRef]
- Port, L.R.; Brunner, P.M. Management of atopic hand dermatitis. Dermatol. Clin. 2024, 42, 619–623. [Google Scholar] [CrossRef]
- 1Yang, L.; Zhang, Y.; Wu, J.; Wang, L.; Liu, S.; Zhou, L.; Zhang, J.; Li, C. Calcipotriol inhibits the proliferation of psoriasis HaCaT cells by activating the ferroptosis pathway. Acta Histochem. 2025, 127, 152274. [Google Scholar] [CrossRef]
- Vähävihu, K.; Ala-Houhala, M.; Peric, M.; Karisola, P.; Kautiainen, H.; Hasan, T.; Snellman, E.; Alenius, H.; Schauber, J.; Reunala, T. Narrowband ultraviolet B treatment improves vitamin D balance and alters antimicrobial peptide expression in skin lesions of psoriasis and atopic dermatitis. Br. J. Dermatol. 2010, 163, 321–328. [Google Scholar] [CrossRef]
- Lossius, A.H.; Sundnes, O.; Ingham, A.C.; Edslev, S.M.; Bjornholt, J.V.; Lilje, B.; Bradley, M.; Asad, S.; Haraldsen, G.; Skytt-Andersen, P.; et al. Shifts in the Skin Microbiota after UVB Treatment in Adult Atopic Dermatitis. Dermatology 2022, 238, 109–120. [Google Scholar] [CrossRef]
- ClinicalTrials.gov. Vitamin D Treatment Effect for Atopic Dermatitis in Children; Identifier NCT05523986. U.S. National Library of Medicine. Available online: https://clinicaltrials.gov/study/NCT05523986 (accessed on 3 September 2025).
- Navarro-López, V.; Ramírez-Boscá, A.; Ramón-Vidal, D.; Ruzafa-Costas, B.; Genovés-Martínez, S.; Chenoll-Cuadros, E.; Carrión-Gutiérrez, M.; Horga de la Parte, J.; Prieto-Merino, D.; Codoñer-Cortés, F.M. Effect of oral administration of a mixture of probiotic strains on SCORAD index and use of topical steroids in young patients with moderate atopic dermatitis: A randomized clinical trial. JAMA Dermatol. 2018, 154, 37–43. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.W.; Ong, M.T.; Man, G.C.; Yeung, Y.M.; He, X.; Choi, B.C.; Ng, J.P.; Mok, D.K.; Lam, T.P.; Yung, P.S. The effectiveness of vitamin D supplementation in patients with end-stage knee osteoarthritis: Study protocol for a double-blinded, randomized controlled trial. PLoS ONE 2024, 19, e0309610. [Google Scholar] [CrossRef] [PubMed]
- González-Tarancón, R.; Goñi-Ros, N.; Salvador-Rupérez, E.; Hernández-Martín, Á.; Izquierdo-Álvarez, S.; Puzo-Foncillas, J.; Gilaberte-Calzada, Y. Association between VDR and CYP24A1 polymorphisms, atopic dermatitis, and biochemical lipid and vitamin D profiles in Spanish population: Case-control study. JMIR Dermatol. 2023, 6, e39567. [Google Scholar] [CrossRef] [PubMed]
- Meyer, M.B.; Lee, S.M.; Towne, J.M.; Cichanski, S.R.; Kaufmann, M.; Jones, G.; Pike, J.W. In vivo contribution of Cyp24a1 promoter vitamin D response elements. Endocrinology 2024, 165, bqae134. [Google Scholar] [CrossRef]
- Milan, K.L.; Ramkumar, K.M. Regulatory mechanisms and pathological implications of CYP24A1 in vitamin D metabolism. Pathol. Res. Pract. 2024, 264, 155684. [Google Scholar] [CrossRef]
- Brustad, N.; Wang, T.; Chen, L.; Kaiser, H.; Gomes, B.; Klein, A.; Vahman, N.; Skov, L.; Stokholm, J.; Schoos, A.M.; et al. Effect of prenatal high-dose vitamin D on childhood atopic dermatitis is modified by maternal cotinine metabolome: A secondary analysis of a randomized clinical trial. J. Am. Acad. Dermatol. 2025. [Google Scholar] [CrossRef]
- Bukvić Mokos, Z.; Tomić Krsnik, L.; Harak, K.; Marojević Tomić, D.; Tešanović Perković, D.; Vukojević, M. Vitamin D in the prevention and treatment of inflammatory skin diseases. Int. J. Mol. Sci. 2025, 26, 5005. [Google Scholar] [CrossRef]
- Camargo, C.A.; Ganmaa, D.; Sidbury, R.; Erdenedelger, K.; Radnaakhand, N.; Khandsuren, B. Randomized trial of vitamin D supplementation for winter-related atopic dermatitis in children. J. Allergy Clin. Immunol. 2014, 134, 831–835.e1. [Google Scholar] [CrossRef]
- Patchen, B.K.; Best, C.M.; Boiteau, J.; Solvik, B.S.; Vonderschmidt, A.; Xu, J.; Cohen, R.T.; Cassano, P.A. Vitamin D supplementation in pregnant or breastfeeding women or young children for preventing asthma. Cochrane Database Syst. Rev. 2025, 8, CD013396. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).