Next Article in Journal
An Artificial Intelligence Approach for the Kinodynamically Feasible Trajectory Planning of a Car-like Vehicle
Previous Article in Journal
A Comparative Study of Advanced Oxidation Processes for the Removal of the Antibiotic Sulfadoxine from Water—Transformation Products and Toxicity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review on the Beneficial Roles of Vitamin D in Skin Health as a Bio-Functional Ingredient in Nutricosmetic, Cosmeceutical, and Cosmetic Applications

by
Sofia Neonilli A. Papadopoulou
1,†,
Elena A. Anastasiou
1,†,
Theodora Adamantidi
1,†,
Anna Ofrydopoulou
1,
Sophia Letsiou
2 and
Alexandros Tsoupras
1,*
1
Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
2
Department of Biomedical Sciences, University of West Attica, Ag. Spiridonos St. Egaleo, 12243 Athens, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2025, 15(2), 796; https://doi.org/10.3390/app15020796
Submission received: 1 December 2024 / Revised: 12 January 2025 / Accepted: 13 January 2025 / Published: 15 January 2025

Abstract

:

Featured Application

The utilization and beneficial roles of vitamin D as a bioactive ingredient with applications in health-promoting functional cosmetics and skincare products, offering anti-inflammatory and anti-aging protection against psoriasis; dermatitis; acne; and other inflammation-related skin disorders.

Abstract

Vitamin D, also called the “sunshine” vitamin, has gained great attention recently due to the observed high percentage of the worldwide population being deficient in this essential bioactive vitamin. Primarily, vitamin D was known for its important role in bone health. Nevertheless, recent research has shown its importance for the brain, heart, muscles, immune system, and skin health, due to its distinct bio-functionality in almost every tissue in the human body. Therefore, its deficiency has been highly correlated with multiple diseases, including skin and dermatologically associated ones. Moreover, different methodologies are applied to synthesize vitamin D, while the main vitamin D sources in human plasma levels and the factors that can cause adverse modifications are multiple. Further research upon vitamin D has exhibited its notable role against skin diseases, such as psoriasis, atopic dermatitis, vitiligo, acne, and rosacea. In this article, a critical review of the most relevant and significant information regarding the relationship between vitamin D and skin health is thoroughly conducted, while emphasis is given to its potential uses and benefits in several cosmetic applications. Current status, limitations, and future perspectives of such a potent bioactive are also extensively discussed.

1. Introduction

Vitamin D’s history dates back to the 17th century when a rickets’ disease outbreak shocked and negatively impacted the global population [1,2]. In 1645, Daniel Whistler was the first to describe the term “Rickets”, known widely as the hallmark of vitamin D deficiency [2,3]. However, scientists for almost three centuries were not capable of acknowledging vitamin D’s potential as a significant protective, preventative, and therapeutic agent against this disease [4,5]. Kurt Huldschinsky proposed in the year of 1919 that ultraviolet B (UVB) rays were sufficient enough to end the suffering and eventually heal rickets disease [6,7]. During the same year, dietary vitamin D deficiency was recognized as another factor by Sir Edwars Mellanby (1920), especially the deficiency of a vital component mostly traced in cod liver oil. In 1922, following the same scientific patterns, E. V. McCollum, after performing multiple animal studies over the years, managed to distinguish the several biological activities between the fat-soluble vitamins D and A, and ultimately, introduced the name “vitamin D” [8,9,10,11].
Despite the fact that vitamin D was deemed as an important compound for bone and calcium metabolism, its chemical structure was not yet uniformly established among scientists. Thereby, during the year of 1937, Windaus and Bock collaborated upon the chemical characterization of vitamins D2 (ergocalciferol) and D3 (cholecalciferol) [12,13,14]. More specifically, vitamin D2 was laboratory-generated through the UV irradiation of the plant/yeast steroid ergosterol, while vitamin D3 was formulated via UV irradiation exposure of 7-dehydrocholesterol. Interestingly, the newly emerged vitamin D3 component was pointed out as the antirachitic constituent in cod liver oil [15]. Such notable findings marked the antirachitic vitamin D as a steroid, and more precisely a bioactive secosteroid. The year 1937 was when Nicolayson experimentally proved that vitamin D is essential for successful calcium (Ca2+) absorption, while 16 years later, it was also confirmed by this scientist that a low-calcium-based diet could increase Ca absorption [4].
The isolation of 25-hydroxy vitamin D3 (25(OH)D3) by DeLuca (1968) as well as the notable discovery of the vitamin D receptor (VDR) by Mark Haussler (1969) paved the way for the uncovering and chemical characterization of 1α,25-dihydroxy–vitamin D3 (1α,25(OH)2D3; otherwise, calcitriol) by Hector DeLuca and Anthony Norman in 1971 [16,17,18]. Validated research findings demonstrated that 1α,25(OH)2D3 may interact with the VDR and modulate gene transcription in various cell types, an observation further supported by a scientific discovery regarding the human (as well as other) genomes, enabling gene regulation by 1α,25(OH)2D3 that generally impacts genes throughout the genome [19]. Adversely to DeLuca’s confirmation that the skin partakes in the synthesis of vitamin D in 1978, other researchers during this period associated vitamin D with multiple disease risks, a cause–effect relationship, globally addressed in many preclinical studies, clinical studies, and trials [20,21,22,23]. Historically, the “transcription era” marked the initial focus on vitamin D’s molecular and cellular mechanisms (1971–2000). This was followed by the “association era”, where researchers examined the link between vitamin D levels and various health outcomes (1980–2010). The “genomics era” emerged in the beginning of the 21st century (2000–present), emphasizing the exploration of genetic variations influencing vitamin D’s metabolism. Over the decades, clinical trials primarily centered on bone health and vitamin D’s role in preventing conditions like osteoporosis and rickets (1970–2010). More recently, the scientific focus has shifted to clinical trials investigating vitamin D’s non-skeletal effects, including its role modulating immune function, reducing inflammation, and managing chronic diseases and manifestations, such as cancer and cardiovascular disorders (Figure 1) [24].
Vitamin D is a naturally occurring, fat-soluble photoproduct found in both plants and human and animal skin. Vitamin D2 is present in plants, while vitamin D3 is traced mainly in the skin of humans and animals. The predominant difference between the two lies in the additional double bond in vitamin D3 [25]. Both forms of vitamin D exhibit similar effects, and potency, and follow similar metabolic procedures [4]. Vitamin D is synthesized when exposed to UVB radiation in fungi, plants, animal skin, and human skin. At this point, it is important to mention that other forms of vitamin D exist, such as D1, D4, and D5, but are of little significance to humans [26]. Chemically, vitamin D shares a structure with cholesterol, consisting of four rings, labeled A, B, C, and D. A broken B ring in cholesterol results in the formation of vitamin D [24]. Since its discovery, vitamin D has been integrated into numerous applications, for several medical, cosmetic, and cosmeceutical purposes [10]. Furthermore, vitamin D has also been implicated in skin aging processes [27,28]. Many cells, particularly skin cells, can produce both vitamin D and its active form, which exerts distinct biological effects based on the target cell. This observation led to classifying vitamin D as a fat-soluble prohormone and its active form as a hormone [29]. The active complex of vitamin D3, 1α,25(OH)2D3, plays a significant role as a regulator in a plethora of biochemical cascades and homeostasis systems [30].
The discovery of vitamin D’s astonishing properties sparked the development of a wide range of skincare products, cosmetic formulations, and systemic and topical analogs. Vitamin D’s anti-inflammatory [25,30], anti-melanoma [31,32], anti-cancer [33], anti-UV [25], anti-aging [25,27,34], and antioxidant properties [25,35] have been harnessed against various diseases or conditions, including psoriasis [36,37,38], atopic dermatitis [39,40,41], vitiligo [42,43], acne [44,45,46], and rosacea [47]. This review explores vitamin D’s structural, chemical, biological, and metabolic roles, highlighting its unique activity and supporting its increasing use in the cosmeceutical field. In addition, the review addresses vitamin D’s role in different skin-related conditions, the controversies surrounding its impact, and suggestions for future research mainly within the cosmetic domain. A timeline summarizing key milestones in vitamin D research is presented in Figure 1.

2. Methods

During requisite literature review, various databases were investigated including Scopus, Science Direct, MDPI, PubMed, and Google Scholar. The following broad combination of keywords was used, “vitamin D”, “cosmetics”, “applications”, “skin health”, “vitamin D2”, “vitamin D3”, “calcitriol”, “cholecalciferol”, “ergocalciferol”, “vitamin D deficiency”, “structure”, “calcium metabolism”, “bone metabolism”, “cosmetic formulations”, “topical analogues”, “skin cosmetics”, “conditions”, “skin disorders”, “acne”, “rosacea”, “atopic dermatitis”, “vitiligo”, “psoriasis”, “anti-inflammatory”, “anti-melanoma”, “antioxidant”, “anti-cancer”, “anti-UV”, and “anti-aging”, with the use of combinations of these keywords by using the AND and/or OR terms.
The research process was conducted during May–November 2024 regarding the last 5–7 years and the selection criteria and filtering features were established by delving into the available metadata from the pre-mentioned databases. This timeframe was chosen deliberately, with the aim of addressing the latest information on our topic and including recently published data, aligning with our main objective: pointing out the current knowledge status, presenting the latest research findings and comparing them to past ones, recognizing all existent limitations, and suggesting future ways to overcome them.
Each selected study was included due to following several pre-established criteria, such as being exclusively research-based articles, including eligible reviews and clinical trials from the aforementioned valid scientific databases. Moreover, all research articles must have been written in the peer-reviewed English language. Additionally, all included publications should have been published between 2018 and 2024. At this point, it must be pointed out that a few important articles published prior to the year of 2018, but followed the inclusion criteria and enclosed important data that were not mentioned in more recent paper reviews, were as well included in our study. If a research paper was in accordance with the above criteria, with a view to evaluate the quality and relevance of each included article, all articles were then thoroughly assessed concerning their title, abstract, and keywords utilized. Subsequently, all remaining articles were extensively analyzed in order to confirm whether they met all predefined inclusion criteria and incorporated relevant information for our review. This article cites predominantly valid academic papers, press releases from research institutes and universities, authentic clinical trials, observational studies, meta-analyses, experimental-founded studies, and well-grounded scientific reviews and peer-reviewed papers. All extracted data were synthesized collectively by all authors, and all conflicting findings were addressed collaboratively. Zotero 6.0.36 software was also utilized for citing the selected publications.
Articles retrieved from non-well-founded databases or unrelated to the topic were excluded along with duplicates or publications written in other languages rather than the English one. Conference papers, book chapters, government publications, theses, dissertations, technical reports, working papers, non-academic reports, internal documents, presentations, white papers, websites, newsletters, short surveys, previous publications, old reviews, newspaper or magazine articles, blog articles, open-content resources, and otherwise collaborative knowledge databases (e.g., Wikipedia) were also excluded. Articles with a low relevance score, very few citations, or a low impact factor and papers with methodological flaws and high bias, were excluded and deemed as the “gray literature” (Figure 2).

3. Vitamin D’s Structural Profile, Skincare Applications, and Functions

3.1. Chemical Structure and Types of Vitamin D

Vitamin D refers to a group of fat-soluble secosteroids essential for various bodily functions. Particularly, vitamin D plays an important role in calcium and phosphorus metabolism, which is crucial for maintaining bone health. The two primary forms of vitamin D are vitamin D2 (C28H44O) and vitamin D3 (C27H44O) (Figure 3). Both forms derive from different sources, and their own individual chemical structures, and exhibit distinct biological activities, as vitamin D2 is present in plants and yeast while vitamin D3 derives from animal sources. Vitamin D’s chemical structure is mainly depicted as a steroid molecule. Sex hormones, adrenal hormones, and cholesterol are placed amongst the many chemical molecules that make up the huge family known as steroids. Interestingly, a unique feature of vitamin D’s structure is its secosteroid nucleus, which resembles a steroid nucleus but lacks one of its rings. Four rings designated as A, B, C, and D make up the secosteroid nucleus as important components of the cyclopentanoperhydrophenanthrene skeleton. The B ring especially is broken at the 9, 10 carbon–carbon bond with a view to form pre-vitamin D. Small differences comprise the molecular structures of vitamin D2 and D3. Specifically, the methyl group at carbon 24 and a double bond located between carbons 22 and 23 are present in vitamin D2. In comparison to vitamin D3, the methyl group at carbon 24 is absent and only one bond is found between carbon 22 and 23. These structural variations are primarily responsible for their stability and metabolism within the human body and impact the sequence of many biochemical processes [48,49]. The two primary forms of vitamin D are depicted in Figure 3 and Figure 4.
Sunlight is the most significant and natural source of vitamin D. Sunlight’s UVB rays cause the skin’s 7-dehydrocholesterol to automatically transform into provitamin D3, which subsequently changes into vitamin D3. Latitude, season, time of day, skin pigmentation, age, and usage of sunscreen are only a few of the variables that might affect the skin’s ability to produce vitamin D3 [50]. In the case of vitamin D2, the main sources of vitamin D2 are fungus and plants formed by subjecting ergosterol—a sterol present in plant and fungal cell membranes—to ultraviolet radiation [51]. As vitamin D3 reaches the liver, it is converted by the aid of enzymes to 25(OH)D3. 25(OH)D3 is then transformed to 1α,25(OH)2D3 in the kidneys, and this process will be explained in more detail in the following section (Figure 2) [30]. Vitamin D3 is also present in animal-based foods like fatty fish (salmon, mackerel, and sardines), cod liver oil, egg yolks, and fortified dairy products (20%). For those at risk of deficiency, and especially people dealing with limited sun exposure, older adults, and those with specific medical problems that influence their metabolism of vitamin D, vitamin D supplements are vital (Figure 3) [52].

3.2. Absorption of Vitamin D from Skincare Products

Vitamin D is incorporated in skincare products offering a wide range of beneficial effects. However, it is important to note that topical vitamin D application does not replace the need for its systematic consumption, which is essential for overall health. Vitamin D3 is more frequently incorporated in skincare formulations, improving the appearance of fine lines and wrinkles, decreasing inflammation, and enhancing skin moisture. During the winter, when skin is typically drier and more prone to irritation, skincare products with vitamin D3 are quite helpful. Different skincare formulas enable personalized routines that target certain requirements. These products focus on key goals such as improving skin hydration, reducing inflammation, and enhancing overall skin health by promoting and providing better texture, tone, and resilience [53].
Calcipotriol, also known as calcipotriene, is commercially available in several products under the brand names Dovonex® and Diavonex®, with comparable efficacy reported to that of calcitriol, due to its identical affinity for VDRs. Calcipotriol has significant impact on the immunosuppression of inflammatory mediators and melanin formation enhancement [54]. Multi-hyaluronic antioxidant hydration serums [55], multivitamin supplies including vitamin D [56], vitamin D3 orally administered coating tablets [57,58], oral capsules [59], softgel formulations [60], topical calcineurin [61], cosmetic serums destined for facial skin improvement [62], and vitamin D3–phytochemical complex formulations [63] have been clinically approved and utilized in several acne, vitiligo, atopic dermatitis, psoriasis, rosacea, and anti-aging treatment strategies globally, and have offered multiple beneficial outcomes.
Vitamin D as a natural photoproduct that is mainly administrated either as an oral supplement (e.g., tablet, syrup, soft gel capsules, and other supplements) or in an injectable dosage form is connected with many beneficial outcomes for the body [25,64]. Nanotechnology has revolutionized medicine by encouraging targeted cellular drug delivery, enhancing the bioavailability of encapsulated vitamin D and its derivatives, and promoting several health-associated benefits. Organic, and especially lipid-based, nanocarriers for the delivery of vitamin D and its analogs have been extensively utilized over the past few years, stabilizing this hormone, improving its solubility, enhancing its absorption, and enabling and bettering targeted cellular release in specific tissues. Such innovative systems have overcome several adverse effects of oral administration including poor permeability in epithelial barriers and solubility [65]. Furthermore, photodynamic therapy supplemented with vitamin D3 has increased 6–30% of the response of this treatment against squamous cell carcinomas (SCCs), actinic keratosis, and psoriasis, in a plethora of breast cancer cell lines and animal models [66]. For instance, topical vitamin D formulations enhanced with cholecalciferol, calcipotriol, or tacalcitol have been confirmed to stimulate melanin and melanogenesis during narrowband UV phototherapy (NB-UVB), in therapeutic treatments against vitiligo [54,67]. However, in conjunction with psoralen long-wave UV radiation (PUVA) and monochromatic excimer light (MEL) vitiligo treatments, less beneficial effects have been recorded, in contrast to vitamin D combined with corticosteroid formulations, where vitamin D minimizes the adverse impact of corticosteroids and offers superior efficacy and vitamin D microneedle provision [54,68,69].
Interestingly, recent study outcomes confirmed that tumor cells of various origins are capable of expressing VDRs and enabling restrained growth and cancer cell arrest/apoptosis. An optimized vitamin D3 nanospray formula had fewer drips, fast drying time, and long-term storage capacity, and maintained bioavailability and biological stability, as well as a faster coverage of the administration time, in less delivery time and enhanced efficacy [70]. Finally, a recent clinical trial revealed that vitamin D supplementation was interconnected with a notable chronic pruritus severity reduction, as well as a decrease in the skin lesion area, and inflammatory cytokine levels including tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), and high-sensitivity C-reactive protein (hs-CRP) [71]. Furthermore, carboxymethyl-cellulose-based hydrogels integrated with cellulose nanocrystals loaded with vitamin D have been pointed out as great vitamin D carriers for controlled and targeted drug delivery, mainly in cases of vitamin D deficiency [72].
The absorption of vitamin D in the skin entails different biochemical processes such as penetration through the stratum corneum, diffusion through the epidermis, and interaction with keratinocytes, which lead to the activation of the vitamin D form. The initial part of vitamin D absorption in skin is its penetration through the stratum corneum—the skin’s outermost layer. This layer functions as a barrier to various substances and is made up of dead keratinocytes embedded in a lipid matrix. Vitamin D’s lipophilicity facilitates its pass through this layer’s lipid matrix. Moreover, the product’s formulation can increase skin permeability through this lipid matrix, by containing suitable ingredients [53].
The second step of the absorption process is vitamin D diffusion via the viable epidermis where the viable keratinocytes have a significant role in metabolizing and utilizing vitamin D. Vitamin D interacts with keratinocytes once it has penetrated the epidermis, while the nuclear VDR that mediates the biological effects of vitamin D is also present. Vitamin D binds to the VDR, leading to its activation, a situation that allows the VDR to control and mediate the production of genes related to immunological responses, cell division, and proliferation, effectively [73].
The keratinocytes, the main cells found in the epidermis, act as enzymatic machinery converting vitamin D from its inactive to its active form. For instance, keratinocytes are capable of transforming vitamin D3 into 1α,25(OH)2D3, its active metabolite, which is essential for vitamin D3’s skin regulatory actions [73,74]. Consequently, various biological incidences arise from the activation of VDR in keratinocytes, including immune response regulation that reportedly lowers inflammation and promotes wound healing, and cell differentiation stimulation, responsible for fortifying the skin barrier [73,74].

3.3. Topical Vitamin D Analogs

Calcitriol (1α,25(OH)2D3) is widely recognized as the active form of vitamin D, which is essential for skin cell differentiation, the inhibition of proliferation, and local immunomodulation [75]. In 1985, after using oral vitamin D3 supplements, a patient with severe psoriasis and osteoporosis symptoms displayed remarkable improvement [76]. However, in the following ten years, an interactive bridge between vitamin D3 supplementation and psoriasis was unfortunately not achieved. Furthermore, managing occurring side effects such as nephrocalcinosis, nephrolithiasis, hypercalcemia, hypercalciuria, and decreased bone mineral density became an extremely difficult task for the scientific community [77]. Nonetheless, such pitfalls urged the investigation of vitamin D substitutes with the aim to prevent all negative consequences. As a consequence, topical calcipotriene (calcipotriol) foam was approved by the US Food and Drug Administration in 1993 for the treatment of mild-to-moderate psoriasis. Additionally, more than 15 years later, a topical 1α,25(OH)2D3 ointment was approved for mild-to-moderate psoriasis treatment. Since then, topical vitamin D analogs—either alone or in conjunction with topical corticosteroids—have become a crucial component in psoriasis treatment strategies. These vitamin D analogs are currently commonly integrated in topical therapeutic interventions, minimizing the reliance on steroid use [78,79,80].
Topical vitamin D analogs directly provide a convenient, safe, and generally well-tolerated therapeutic alternative and examples of such therapeutic means are depicted in Table 1 [81]. Although exclusively authorized for psoriasis, they have demonstrated effectiveness in treating numerous other dermatological conditions [81].

3.4. Indicative Beneficial and Adverse Functions of Vitamin D in the Human Body

Vitamin D plays a pivotal role in maintaining calcium and phosphorus homeostasis, which is essential for bone mineralization. It promotes the absorption of calcium and phosphorus from the gut, reabsorption of calcium in the kidneys, and mobilization of calcium from bones. These actions ensure adequate levels of calcium and phosphorus for the formation and maintenance of healthy bones and teeth. As widely supported, a deficiency in vitamin D might lead to bone disorders such as rickets in children and osteomalacia in adults, characterized by soft and weak bones [87]. Hence, vitamin D-based treatment is believed to reduce inflammation and boost the ability of monocytes and macrophages, two types of white blood cells (leukocytes), vital to the immune defense and responsible for combating pathogens. By controlling the synthesis of antimicrobial peptides and adjusting immune cell activity, vitamin D contributes to the prevention and management of infections and autoimmune disorders [50]. Moreover, it has been demonstrated to control cell division and proliferation, and prevent specific malignancies’ spread by interfering with cancerous cells’ proliferation and induction of their differentiation [88]. Vitamin D’s role mainly lies in controlling cell growth, but also acting as a defense mechanism, by balancing the skin’s high rate of cell turnover [50].
Vitamin D3 and its receptor (VDR) are ultimately crucial in maintaining skin health [89]. Primarily, it is essential for the proper functioning and integrity of the skin barrier [90]. By facilitating the differentiation of keratinocytes, namely skin cells that basically form the outermost layer, vitamin D aids in maintaining a robust epidermal barrier able to protect an organism against infections and environmental damage [91,92]. Additionally, vitamin D modulates cell growth and differentiation, a vital function in conditions characterized by abnormal cell proliferation, such as psoriasis or even skin cancer, and also supports wound healing, by promoting cell repair and regeneration [21,93,94]. Moreover, this hormone exhibits its anti-inflammatory profile, by reducing inflammation and contributing to managing conditions like eczema [95,96]. Overall, vitamin D’s impact on the skin’s health underscores its importance in maintaining skin barrier function, regulating cell growth, and enhancing the skin’s natural defense mechanisms [87].
However, apart from vitamin D’s beneficial impact, some adverse effects do exist but can be easily managed. Hypervitaminosis D results from excessive intake of vitamin D supplements, leading to elevated serum calcium levels (hypercalcemia) and associated complications [97]. Pointedly, high 1α,25(OH)2D3 increases intestinal calcium absorption; thus, elevated calcium and phosphate levels may induce the supersaturation of urine, promoting the formation of calcium oxalate or calcium phosphate crystals, the primary components of renal calculi (kidney stones). Prolonged hypercalcemia can unfortunately lead to nephrocalcinosis, a condition marked by calcium deposition in renal tissue, impairing kidney function [97,98,99,100,101]. Moreover, such a vitamin D increase and regulation failure for individuals poses several risk factors for individuals with predisposing conditions including chronic kidney disease (CKD), hyperparathyroidism, or sarcoidosis [102,103,104]. In CKD especially, impaired kidney function reduces the conversion of 25(OH)D3 to its active form, often requiring supplementation. Over-supplementation may adversely worsen hyperphosphatemia and accelerate vascular calcification, increasing the risk of CVD complications, while high vitamin D doses may also suppress the parathyroid hormone (PTH), leading to adynamic bone disease [30,103,104]. Moreover, in autoimmune disorders like sarcoidosis or tuberculosis, macrophages may autonomously produce 1α,25(OH)2D3, independent of PTH regulation, where in such cases, vitamin D supplementation may exacerbate hypercalcemia and its complications [102,105].
Hyper-elevated levels of vitamin D have additionally many other negative influences in several chronic diseases. For instance, hypercalcemia-induced vascular classification initiated by excess vitamin D may contribute to calcium deposition in arteries, leading to arterial stiffness, hypertension, risk of arrythmias, and several other cardiovascular-associated disorders (CVDs) [22,106]. Some studies also suggest that excessive vitamin D may contribute to certain cancers’ progression, such as prostate cancer, although evidence is mixed. Over-vitamin D supplementation could disrupt cellular signaling pathways involved in apoptosis and proliferation [107]. In addition to the previous complications, chronic hypervitaminosis may lead to bone resorption, because of increased PTH suppression and osteosclerosis (excessive bone hardening) or brittleness, from dysregulated calcium homeostasis [108,109,110]. Finally, some negative, hypercalcemia-related neurological symptoms like confusion, lethargy, and in severe cases such as comas have been recorded, but research is still required for fully comprehending this association [97,111].

4. Skin Biology, Vitamin D Synthesis, and Its Impacting Factors

4.1. General Structure and Function of Skin

The largest space-consuming organ in the human body is the skin, which has an average surface area of 2 m² in humans and is the body’s main line of defense against harmful substances, ultraviolet light, microbes, and physical trauma. It acts as a complex barrier and interfaces between the internal and external environment of the body. In addition, it serves as an excretory, sensory, and immunologically active organ and partakes in the regulation of the body’s temperature [112]. The epidermis, dermis, and hypodermis are namely the three main layers that make up the skin [113]. The outermost layer, known as the epidermis, is made up of a stratified squamous epithelium that is mostly composed of keratinocytes [114]. Layers of keratinocytes move from the stratum basale to the stratum corneum, where they mature into corneocytes and eventually shed to form the epidermis that performs various barrier functions [113,115]. Corneocytes in the stratum corneum then are enclosed in a lipid matrix that resists shearing forces and permits water loss, functioning as a “bricks and mortar” system. In the stratum corneum, lipids produced by lamellar bodies constitute a vital barrier, controlled by calcium concentrations [112]. At this point, it must be referred to that fibroblasts inside a collagen–elastin matrix, blood arteries, lymphatic vessels, nerve terminals, and skin appendages like hair follicles and glands are all located in the dermis, a connective tissue layer that lies underneath the epidermis. Adipose tissue, which fabricates the hypodermis, serves as insulation, a cushioning agent, and the starting point for blood arteries that continue into the dermis [114].
Many vital tasks necessary for preserving health and homeostasis are carried out by the skin [116]. In addition to reducing water loss and preserving hydration, skin serves as a barrier of defense, protecting internal organs from environmental danger such as infections, toxins, and physical damage [117,118,119]. By producing perspiration and modifying blood flow, the skin plays a crucial part in thermoregulation [120]. Being a sensitive organ, it has receptors for pain, pressure, touch, and temperature changes, all of which enable one’s body to react to outside stimuli. Another essential role of the skin involves, among many other immune responses, the one due to Langerhans cells that constitute the immunological defense, deciphering skin’s ability to recognize and counteract infections. Furthermore, exposure to UVB rays stimulates the skin to synthesize vitamin D, necessary for bone health and calcium absorption, while the skin aids in the excretion of body wastes such as salts and urea through its appendages, the sweat glands. Such distinct roles demonstrate how important the skin is as a dynamic, multipurpose organ for defense, control, and general physiological homeostasis [115].

4.2. Synthesis and Metabolism of Vitamin D in Human Skin

Pre-vitamin D3 is created via a photochemical reaction involving provitamin D3, also known as 7-dehydrocholestrol (7-DHC), which is a precursor mostly present in the basal and spinous cell layers of the skin’s epidermis [121]. More specific to this is the stratum basale, comprising stem and transient amplifying cells that generate the upper cells, which in turn produce keratins K5 and K15 and proliferation markers such as β-catenin (CTNNB), cyclin D1, and the GLI1 gene. After migrating to the spinous layer, the cells produce different keratins, namely K1 and K10, as well as enzymes and precursors implicated in cornified envelope formation, namely involucrin and transglutaminase-K. Filaggrin and loricrin protein expressions mark the beginning of further migration into the stratum granulosum, thus contributing to cornified envelope generation. Such cells additionally partake in lipid production, including lipids packaged into lamellar bodies, and are injected into the intracellular spaces between the stratum granulosum and stratum corneum with a view to waterproof the permeability barrier. Interestingly, lamellar bodies contain antimicrobial peptides like cathelicidin formulated in the stratum granulosum, providing protection against invasive organisms. VDR and CYP27B1 expression is the highest in the stratum basale, where the mediator of RNA polymerase (MED1) and the steroid receptor coactivator-3 (SRC-3) are two major VDR action coregulators. On the one hand, MED1 expressed mainly in the stratum basale and spinosum facilitates VDR’s modulation of proliferation at early stages of differentiation, and stratum granulosum SRC-3 accelerates the VDR’s regulation of terminal differentiation [122].
Along with its effects on the kidneys, calcitriol bound to the vitamin D binding protein also affects target organs that express VDRs, such as the bone, gut, and parathyroid (PTH) gland, via both genomic and non-genomic pathways. Geographical, seasonal, and environmental factors including pollution, sun exposure, month, location, or altitude heavily affect the amount of vitamin D generated and the time required for this procedure [123]. Under the predominant impact of UVB radiation (280–320 nm), 7-DHC is quickly converted to pre-vitamin D3 (pre-D3) in the skin, and then pre-D3 rearranges under heat (thermal) exposure to generate vitamin D3. By converting pre-D3 into the physiologically inert substances tachysterol and lumisterol through continued UVB exposure, harmful D3 buildup is avoided. Lumisterol and tachysterol are reverted back to pre-D3 in dark conditions (Figure 5) [30]. Thus, skin pigmentation and UV intensity impact this process; melanin absorbs UV radiation, lowering the efficiency of D3 generation in people with darker skin, which helps explain why these target groups demonstrate lower 25(OH)D3 levels in clinical tests [124]. At the end of this procedure, vitamin D3 enters the bloodstream and is transported to the liver, after binding to the VDR [30,121].
Skin pigmentation is influenced by the melanin pigment, as well as seasonal aspects, UVR’s impact, tanning, and genetic adaptation capabilities. Direct skin sun exposure to sun rays and the need to improve thermoregulation also led humans to gradually evolve the skin pigment over time. Interestingly, skin pigmentation not only acts as a shield against UVR damage, but also as an important defensive mechanism against heat dissipation and vitamin D deficiency, and thus, an evolutionary balancing aspect between vitamin D synthesis, vitamin D’s activities, and UVR protection [125].
Seasonal differences may also have an impact on D3 production in areas far from the equator; vitamin D3 levels affected by melanin’s activity are higher during summer, and lower during winter levels. Furthermore, younger age, darker skin type, not using vitamin D supplementation, not consuming oily fish, and living in specific low-UV regions are all predicting factors of vitamin D deficiency, poor myoskeletal health, and chronic health issues [126]. Such observation comes in accordance with research studies highlighting that the risk of rickets and osteomalacia is higher in completely clothed Bedouin people under similar sunshine conditions than in less covered cultures, suggesting that clothing based either on personal preferences or religious practices and sunscreens may limit D3 production. Conflicting studies, however, proved that the use of sunscreen during summer allows sufficient sunlight to be received, enables vitamin D, and does not interfere with vitamin D deficiency [127,128]. The skin produces vitamin D3 in a safe and controlled manner thanks to photoconversion procedures and environmental influences [129]. Of note, ozone has an excellent function in mitigating hazardous UVC rays that pass through the stratosphere, and hence, preserves skin health [130].
Following its production, the metabolism of vitamin D3 is a procedure first conducted in the liver and then in the kidneys. Vitamins deriving especially from food bind to the VDR, with this complex reaching the liver and being converted to 25(OH)D3. This compound, as it reaches the kidney, is then enzymatically converted by 25(OH)D3-1α-hydroxylase (CYP27B1) and several other cofactors like nicotinamide adenine dinucleotide phosphate (NADPH), magnesium, and interaction with other degrading enzymes of vitamin D metabolites, like CYP3A4, to two distinct bioactives. More specifically, through a hydroxylation in the 1α (A ring) position, 25(OH)D3 is converted to 1α,25(OH)2D3 and through the hydroxylation in the 24th position, 25(OH)D3 is directly converted to 24,25-dihydroxyvitamin D3 (24,25(OH)2D3). The first conversion leads to increased PTH and fibroblast growth factor-23 (FGF-23) levels and decreased phosphorus (P) (phosphate excretion by the kidneys) and Ca levels (calcium resorption from bones), while the second conversion, in contrast, leads to elevated P and Ca (due to renal reabsorption of phosphate) levels (Figure 5) [30]. Ultimately, 1α,25(OH)2D3 promotes calcium and phosphate absorption, while 24,25(OH)2D3 inhibits excessive Ca absorption, preserving balance [30].
However, excessive 1α,25(OH)2D3 may lead to conditions like hypercalcemia and hyperphosphatemia, contributing to complications such as vascular classification, skeletal anomalies, and renal calculi [131,132]. Furthermore, in CKD patients, impaired 1α-hydrolase activity results in decreased 1α,25(OH)2D3 levels, leading to secondary hyperparathyroidism and bone mineral disorders [30,103]. On this note, a deficiency in 25-hydroxylase, otherwise CYP2R1 deficiency, induces vitamin D-dependent rickets type 1B [133,134]; CYP27B1 mutations are associated with vitamin D-dependent rickets type 1A [133,135], and polymorphisms in CYP24A1 may cause hypercalcemia [133,136].
Besides producing vitamin D3, keratinocytes also convert it into its active form, 1α,25(OH)2D3, through the enzymes vitamin D-25 hydroxylase CYP27A and CYP27B1. Unlike most cells, which rely on the kidneys for circulating 1α,25(OH)2D3, keratinocytes are able to operate this entire mechanism to perform this conversion. Higher quantities of CYP27B1 are expressed by keratinocytes than by kidney cells, further suggesting that the skin produces 1α,25(OH)2D3 primarily for local application rather than systemic circulation. Such local production enhances innate immunity, increasing the expression of antimicrobial peptides like cathelicidin, and modulates the adaptive immune system, promoting T cells and reducing inflammation activity. This local activation occurs even under conditions where circulating levels of 1α,25(OH)2D3 might be low due to impaired kidney function such as in CKD [30,103]. Via a different mechanism than the one in the kidney, the PTH promotes the creation of 1α,25(OH)2D3 in keratinocytes, since they do not respond to cycle adenosine monophosphate (cAMP) and do not own the PTH receptor. This makes keratinocyte-driven 1α,25(OH)2D3 generation independent of systemic PTH control, highlighting a local, rather than a systemic, role and a preventative effect against hyperproliferating conditions like psoriasis [129].
Additionally, 1α,25(OH)2D3 regulates its own levels within keratinocytes by inducing 25(OH)D3-24-hydroxylase (CYP24A), which breaks down 1α,25(OH)2D3 and prevents excess levels. This feedback mechanism ensures that little 1α,25(OH)2D3 enters the bloodstream from the skin. The activity of CYP27B1 in keratinocytes varies according to their differentiation and is the highest in undifferentiated cells and in the stratum basale of the epidermis. Elevations in factors like TNF-α and interferon-gamma (IFN-γ), in conditions like eczema and psoriasis, enhance 1α,25(OH)2D3 production in response to UV light and skin barrier disruption, increasing CYP27B1 expression. This compensatory mechanism aids in restoring epidermal barrier function and reducing inflammation. Furthermore, 1α,25(OH)2D3 controls its own levels in keratinocytes by stimulating CYP24A, an enzyme capable of degrading 1α,25(OH)2D3 and preventing overabundance. This feedback system ensures that a very small amount of 1α,25(OH)2D3 can leave the skin and enter the bloodstream. Moreover, when keratinocytes differentiate, CYP27B1 activity changes. Accordingly, the highest activity is recorded in undifferentiated cells and the stratum basale of the epidermis. TNF-α and IFN-γ are two pro-inflammatory factors that increase CYP27B1 expression, by mainly enhancing the generation of 1α,25(OH)2D3 in response to UV radiation and the disruption of the epidermal barrier [129].
As mentioned previously, via a hydroxylation in the 1α (A ring) position, vitamin D3 is first transformed into 25(OH)D3 in the liver, and then is enzymatically converted to 1α,25(OH)2D3 in the kidney. 1α,25(OH)2D3 binds to the VDRs located both on the cell surface (membrane bound) and inside the immune cell’s cytoplasm (intracellular), and thus, activates both innate and adaptive immune cells [30,137,138,139].
Concerning the innate immune cells and specifically dendritic cells (DCs), 1α,25(OH)2D3 is responsible for downregulating the major histocompatibility complex (MHC) class II’s expression, and as a result, antigen recognition and DCs’ activation are induced. When DCs are activated, the vast activity of T-lymphocytes (T cells) is triggered and vice versa, while 1α,25(OH)2D3 suppresses pro-inflammatory cytokines’ (i.e., IL–2) production and encourages anti-inflammatory cytokine generation [30]. Regarding macrophages/monocytes, contrastingly, the 1α,25(OH)2D3-VDR complex stimulates such leukocytes to produce antibiotic peptides including β-defensin 2 and cathelicidins and enhances via epigenetic programming their immunological memory and differentiation. Furthermore, as of its role in adaptive immunity, 1α,25(OH)2D3 participates in the reduction in T-helper cells’ differentiation into T-helper 1 and T-helper 17 (Th1 and Th17, respectively), the upregulation of Th2 cells’ differentiation, the suppression of pro-inflammatory cytokine production, and the enhancement of anti-inflammatory cytokines’ (i.e., IL-10) generation [30]. T cell-derived pro-inflammatory cytokines are finally responsible for inducing B-lymphocytes’ (B cells) differentiation. T cells’ suppression leads subsequently to an indirect downregulation of B cells’ maturation and hence, activity, while direct B cell differentiation and maturation into memory and plasma cells is also encouraged (Figure 6) [30,36,138,139].

4.3. Factors That Affect the Synthesis of Vitamin D

Many factors affect the process by which the skin makes vitamin D when exposed to UV light, known as cutaneous vitamin D synthesis. Maintaining appropriate levels of vitamin D is critical for immune system performance, bone health, and general wellbeing. This mechanism makes this possible. The amount of UV radiation, skin type, location, season, time of day, and use of sun protection are some of the major variables that affect how well vitamin D is produced in the skin. The main factor influencing the synthesis of vitamin D in the skin is the UV light intensity. As mentioned previously, 7-DHC in the skin is changed by UVB radiation to pre-vitamin D3, which is then transformed into vitamin D3. UVB’s radiation intensity, in general, varies with height, latitude, and cloud cover, among other factors. For instance, UVB radiation is stronger near the equator and at high altitudes. On the other hand, UVB intensity drops in the winter and in areas distant from the equator, where vitamin D synthesis is reduced [140].
The effectiveness of vitamin D production is significantly influenced by skin type as well. The pigment melanin, which gives skin its color, absorbs UV rays and lessens their ability to penetrate the skin’s deeper layers. As a result, those with darker skin display higher melanin levels, and are less able to synthesize vitamin D than people with lighter skin. Skin types are categorized on the Fitzpatrick scale from I (very light) to VI (extremely dark), with darker skin types needing more sun exposure to generate the same amount of vitamin D in contrast to lighter skin types [141]. In addition, geographical differences in solar UVB exposure have a significant impact on vitamin D production. More direct sunshine is received year-round in areas near the equator, which promotes steady vitamin D synthesis. Higher-latitude regions, on the other hand, undergo notable seasonal fluctuations. For instance, there may be insufficient UVB sunlight during the winter in northern latitudes like Scandinavia or Canada, which increases the risk of vitamin D deficiency. UVB exposure is also notably influenced by day and season. During the summer, when the sun is at its maximum point, UVB radiation is more intense. The angle of sunlight decreases the amount of UVB rays that reach the skin in the winter or in the early morning and late afternoon. For efficient vitamin D synthesis, one must understand the idea of the “solar noon”, or the time when UVB rays are at their strongest. Hence, exposure to the sun at midday, when UVB radiation is at its highest, is ultimately more effective in producing vitamin D in individuals [142].

5. Vitamin D Health-Promoting Activities

5.1. Anti-Melanoma and Anti-Cancer Activities

Melanoma is a cancerous condition during which melanocytes—the neutral crest-originated cells responsible for producing melanin (the pigment that gives skin its color)—undergo a transformation, becoming malignant [31,37]. Ultraviolet radiation (UVR), chemical and biological mediators, including hormonal and non-hormonal regulators, and genetic and molecular modulators are examples of some factors that can regulate the melanogenic activity and action of melanocytes [37,143]. The World Health Organization (WHO) classifies melanoma based on etiology, and more specifically whether it is connected to sun exposure or not, with the former stratified into low- and high-cumulative solar damage (CSD) of the skin. Examples non-related to sun exposure etiology are mucosal, acral, uveal, and Spitzoid melanomas; melanomas developing in blue or congenital nevi; and uncommon melanomas in the central nervous system (CNS) [144,145].
Melanoma is a lethal type of cancer, with a poor prognosis especially when the disease is spread to other body areas. One of the most common types of melanomas is cutaneous melanoma, which has a constantly increasing annual incident rate. Such a type mainly appears in older people, but it has been found in younger people as well. There are a few current treatment schemes, which are unfortunately expensive, and are followed by multiple side effects. Thus, the need for new treatment methods is vastly increasing [32].
It is important to understand how vitamin D may regulate a variety of biological pathways. In more detail, responsible for both the genomic and non-genomic regulation of several biological processes is the binding of 1α,25(OH)2D3 to the VDR in the target tissues. As widely accepted, the VDR belongs to the nuclear type of receptor family, which contains ligand-activated transcription factors, and nearly all tissues and cells, including skin cells, are able to express it. According to Podgorska et al. [146], their research indicated that there is an inverse relationship between melanoma progression and VDR expression, proposing that VDR may function as a gene that suppresses melanoma tumors [146]. It is generally acknowledged that vitamin D compounds hold great promise for both the prevention and treatment of cancer, due to their antiproliferative properties towards specific cancer types, predominantly by promoting differentiation and suppressing proliferation [147]. This observation has been confirmed by results deriving from clinical trials on different melanoma types and by epidemiological and preclinical evidence [32,148]. Moreover, both molecular and clinicopathological studies have pointed out a connection between vitamin D signaling abnormalities and the development and the possible outcome of melanoma. As a consequence, successful treatment for melanoma may involve sufficient vitamin D signaling and vitamin D-based interventions [32].
According to Shariev et al. [147], in the human melanoma cell lines used in the investigation, 1α,25(OH)2D3 dramatically decreased cell viability and raised caspase levels. Usually during melanoma, the phosphatase and tensin homologue (PTEN), which is a tumor suppressor, is mutated. In this study, results showed an increase in the PTEN levels and a reported downregulation of both the protein kinase B (AKT) pathway and its downstream effectors. Thus, 1α,25(OH)2D3 is a potent factor able to efficiently target PTEN and to reduce melanoma cell viability [147].

5.2. Anti-UV Activity

UVR is a major physical agent that causes skin damage and carcinogenesis through wavelength-dependent processes, such as the chromophore-based absorption of UVB radiation, reactive oxygen species (ROS), and UVA reactive nitrogen species production (RNS) [149,150]. Normally, ROS are synthesized in cells endogenously, in a decreased amount, as part of the normal signaling process [25]. UVB can damage DNA directly via forming cytotoxic cyclobutane-pyrimidine dimers (CPDs), 6-4 pyrimidine photoproducts (6-4PPs), and 8-hydroxy-2′-deoxyguanosine (8-OH-DG), along with the contribution of the generated ROS [25,151]. Additionally, 8-OH-DG, mutations occurring at non-pyrimidine sites, and the so-called “dark CPD” (dCPD) are created by UVA exposure. Half of the total produced CPDs are formulated by dCPD, which often develop well after UVA exposure and are dependent on melanin [25]. Crucial in the development of cutaneous carcinogenesis are CPDs and 6-4PP, as they involve the alteration of the p53 gene, a key tumor suppressor [152]. Important defense mechanisms against mutations and photo-carcinogenesis include predominantly the nucleotide excision repair (NER) system, the nuclear factor E2-related factor 2 (Nrf2) with its associated downstream antioxidant components, and the p53 signaling cascade [153].
Vitamin D3 and its active forms have well-documented photoprotective properties [154,155]. Furthermore, the oral administration of high dosages of vitamin D3 can cure UVB-induced skin damage by reducing inflammation and promoting barrier repair mechanisms [156]. Slominski et al. [150] reported that vitamin D3 prohormone 1α,25(OH)2D3 and low-calcemic chemically produced derivatives of vitamin D3 and 1α,25(OH)2L3 offer great protection against UVR skin damage and cutaneous carcinogenesis [150]. The anti-cancer, anti-melanoma, and anti-UV effects of vitamin D in indicative clinical trials are analyzed in detail in Table 1.

5.3. Wound-Healing Activity

Wound healing is a compilated biological process in which the injured skin is trying to be repaired [157]. Until now, there have been a variety of reagents used in the treatment of such skin wounds. Notably, the greatest of all has been proven to be vitamin D [89]. It is important to mention that increased immunological problems and increased susceptibility to infections are linked to vitamin D deficiency [158].
Epithelial-to-mesenchymal transition (EMT) plays a crucial role in the process of wound healing. Re-epithelialization is one of the vital steps involved in wound-healing processes, in which the migration and proliferation of epidermal keratinocytes take place and surround the lesion [159]. Studies have shown that EMT takes place in keratinocytes at the site of wounds, during re-epithelization [160]. EMT’s onset may be due to tumor growth factor β (TGF-β), a cytokine highly implicated in wound healing [161].
Many processes related to wound healing, such as inflammatory and immune reactions, as well as the proliferation and differentiation of keratinocytes and fibroblasts, may be influenced by vitamin D [162,163,164]. Furthermore, calcium signaling is essential for normal wound healing, where vitamin D has a significant impact on calcium metabolism and vice versa [165]. To demonstrate this delay in wound healing, Oda et al. employed a mouse model missing both the calcium-sensing receptor (CaSR) and the VDR [166]. Studies have also shown that calcipotriol is able to reduce redness, thickening, or other types of discomforts caused by different skin diseases [167]. According to Wang et al., calcipotriol has been shown to have positive effects on the process of wound healing both in vivo and in vitro, as it can promote skin keratinocyte migration and wound healing via suppressing the Hippo signaling pathway [157].

5.4. Antioxidant Activity

Vitamin D was first recognized as an antioxidant in 1993, and since then, research has clarified its antioxidant function mechanisms [35]. Interestingly, vitamin D enhances the expression of antioxidant genes and inhibits oxidative stress pathways, while reducing the accumulation of harmful molecules like advanced glycation end-products. Vitamin D, like other antioxidants, plays a crucial role in neutralizing ROS and RNS, which are major mediators of oxidative stress [168]. This oxidative damage contributes to skin aging, inflammation, and various skin diseases’ onset. Antioxidants aid in maintaining skin integrity by reducing ROS and RNS levels, thus protecting cellular structures like lipids, proteins, and DNA from oxidative damage [35].
The vitamin D hormone contributes to neutralizing oxidative stress by activating the Nrf2 pathway, a critical/master regulator of the antioxidant response [168]. This activation subsequently enhances the expression of antioxidant enzymes, which protect against oxidative damage in skin cells [34]. Nrf2 also induces the action of cytoprotective genes essential for redox balance and detoxification, which partake in protecting against various diseases and aging. Moreover, vitamin D increases total antioxidant capacity by raising superoxide levels [169]. Additionally, vitamin D plays a protective role in mitochondria by regulating ROS production and preventing lipid peroxidation, a crucial activity for the insurance of cellular integrity. A deficiency in vitamin D can lead to increased ROS and oxidative damage, especially in mitochondria. By activating the Nrf2 pathway, vitamin D helps mitigate mitochondrial damage, reduces oxidative stress, and favorably supports the regulation of several inflammatory responses [169].
Human skin naturally produces vitamin D upon exposure to UV rays. This hormone serves as a regulator in maintaining skin homeostasis by influencing various cellular processes, like proliferation, differentiation, and apoptosis processes of skin cells. As an antioxidant, vitamin D can mitigate the damage caused by excessive UV exposure, which leads to ROS formation. By reducing this UV-induced oxidative stress, vitamin D aids in preventing skin damage, inflammation, and even skin cancer development [170].
Furthermore, vitamin D enhances the expression of antioxidant enzymes, such as GSH, which generally neutralizes ROS. Furthermore, it boosts the skin’s immune responses and enhances the activity of enzymes such as superoxide dismutase (SOD) that disarms superoxide radicals, with great efficacy. Such a protective intervention is crucial in maintaining the skin’s barrier function and preventing conditions like eczema, psoriasis, and acne [170]. Vitamin D’s antioxidant properties extend to repairing oxidative damage by modulating DNA repair mechanisms [171]. It also ensures the removal or repair of damaged DNA segments caused by ROS, further preventing mutations and carcinogenesis’ onset in skin cells [170,171]. Studies have highlighted the broader antioxidant framework that includes other vitamins like E and C, flavonoids, and polyphenols, which work synergistically and collaboratively to protect ones’ skin from oxidative stress, and thus the overall skin health [170].

5.5. Anti-Inflammatory Activity

Vitamin D’s anti-inflammatory qualities are essential for healthy skin maintenance, as it keeps inflammation under control by regulating both innate and adaptive immune responses and subsequently ensures the skin’s safety against illness, while efficiently fostering healing and barrier function. Because of this, vitamin D plays a crucial role in the treatment and prevention of inflammation-related skin disorders [27,172]. Vitamin D exhibits anti-inflammatory properties through its active metabolite 1α,25(OH)2D3, which binds to the VDR present in various skin cells such as keratinocytes, fibroblasts, and immune cells like macrophages and dendritic cells [30]. This interaction not only regulates calcium homeostasis, but also modulates immune responses, by primarily reducing inflammation and promoting skin barrier integrity [27,172].
Reducing pro-inflammatory cytokines is one of vitamin D’s most significant impacts observed in several inflammation-related disorders [30,173]. Regarding skin inflammation, 1α,25(OH)2D3 suppresses cytokines implicated in initiating inflammatory reactions, such as interleukins 2 and 17 (IL-2 and IL-17), as well as TNF-α. Reduced inflammation is linked to this suppression, which alters the immune response from a pro-inflammatory Th1/Th17 state to a more tolerogenic Th2 state [172]. This alteration is notably helpful in the treatment of inflammatory skin conditions including acne, eczema, and psoriasis, because these cytokines are major pathogens in these conditions [128,129,130]. Additionally, vitamin D stimulates the production of regulatory T cells (T-regs), essential for promoting the immunological tolerance maintenance and avoidance of exaggerated inflammatory responses [172]. T-regs also aid in reducing skin inflammation by producing anti-inflammatory cytokines like IL-10. Vitamin D contributes to maintain a more regulated immunological milieu by increasing T-reg activity, which actively induces in turn the prevention of chronic inflammation that could possibly cause skin damage [172].
To further reduce inflammation, vitamin D also aids in the regulation of innate immune responses in the skin [172]. Vitamin D, in fact, reinforces the skin’s defense mechanisms against infections, by directly stimulating antimicrobial peptides such as cathelicidin and β-defensins, while also lowering inflammatory reactions [172,174]. Such dual function emphasizes how crucial vitamin D is for preserving skin homeostasis [27,172]. Beyond its direct immune-modulatory effects, vitamin D also plays a role in inhibiting oxidative stress, which is often a trigger for inflammation. By reducing the levels of ROS in the skin, vitamin D minimizes oxidative damage that can lead to chronic inflammatory states. This antioxidant action works in synergy with immune-modulatory functions, to protect the skin from both internal and external inflammatory stimuli [27].

5.6. Anti-Aging Activity

An intricate process of both internal and external variables contributes effortlessly to skin aging. Like all other organs, the physiological, morphological, and functional characteristics of the skin gradually deteriorate with age. In general, aging is a multifactorial process, as a natural and genetically predisposed phenomenon [27]. More specifically, genetically predetermined intrinsic aging, commonly called chronological aging, is driven by genetic factors and the passage of time and starts around the mid-20s [28]. It entails a slower synthesis of collagen and elastin, two proteins necessary to preserve skin firmness and suppleness, as well as a progressive reduction in the skin’s capacity to regenerate. As a result, the skin thins, loses volume and elasticity, and develops fine lines and wrinkles, whereas dryness and a lifeless complexion result from cellular turnover deceleration, where the skin becomes more brittle and less able to preserve moisture [27].
Conversely, extrinsic aging results from environmental factors such as pollution, UV radiation, and also lifestyle choices like smoking and bad dietary habits [175]. The main cause of photoaging is UV radiation, and particularly UVA rays that induce collagen fibers to break down and aberrant elastin to be produced, which results in deeper wrinkle formation, a leathery texture, and pigmentation changes like age spots or hyperpigmentation [27]. Additionally, UV radiation produces ROS that expedite further extracellular matrix (ECM) damage, which accelerates the aging process of the skin by degrading collagen and elevating the expression of enzymes like matrix metalloproteinases (MMPs). Additionally, air pollution, smoking, and oxidative stress are in accordance with the breakdown of collagen and elastin, worsening skin texture and elasticity. Smoking reportedly reduces blood flow of the skin, depriving it of oxygen and nutrients, while pollution introduces free radicals that damage skin cells [28,175]. Chronic stress, poor diet, and sleep deprivation can all hasten these consequences by interfering with skin regeneration processes [27]. As a sequel, the skin becomes drier and thinner due to collagen loss [28,175,176].
The impairment of the skin’s collagen partakes in a condition called dermatoporosis [177]. Normally, young skin contains more than 75% of collagen [178,179]. As collagen is the protein of the skin responsible for strength and elasticity, aging leads to thinning and fragility. Studies have shown that vitamin D can act as a stimulating agent for the synthesis of collagen that promotes skin’s thickening and resilience [177]. Also, vitamin D influences the collagen turnover, by repairing and replacing collagen, reducing collagenase, and stimulating collagen production. Collagenase is particularly an enzyme that breaks down collagen [27]. Thus, vitamin D acts equally both as an antioxidant and an anti-aging agent, with great efficacy and promising outcomes [25].
Vitamin D participates dexterously in slowing the aging process, acting as a powerful “anti-aging shield” at both a cellular and a molecular level. Its anti-aging effects are attributed to its regulatory functions in immune modulation, inflammation control, and the maintenance of skin homeostasis. 1α,25(OH)2D3 impacts a wide range of biological processes, indisputably crucial for healthy aging [27,34].
Cellular senescence is another hallmark of aging, characterized by the cessation of cell division and the secretion of inflammatory mediators, known as the senescence-associated secretory phenotype (SASP). As people age, senescent cells proliferate and contribute to tissue malfunction and persistent inflammation [34]. It has been demonstrated that vitamin D prevents the accumulation of senescent cells and modulates pathways involved in cell cycle regulation, in order to counteract cellular senescence [34,180]. Vitamin D reduces the expression of senescence markers and inflammatory mediators in aging cells, aiding in maintaining cellular function and tissue integrity. This protective effect extends to various tissues including the skin, where vitamin D prevents at a great extent the breakdown of collagen and elastin [34].
One of the main contributors to aging is mitochondrial malfunction. The energy-producing organelles in cells, namely mitochondria, lose productibility with age, which increases the production of ROS and decreases energy output. This leads to weariness, cellular damage, and a general deterioration in tissue function. Vitamin D improves mitochondrial function and lowers oxidative stress, which subsequently enhances mitochondrial protection [34]. Moreover, it promotes mitochondrial biogenesis, a process that creates new mitochondria organelles and keeps cells healthy and able to produce energy as we age [34]. The anti-inflammatory, antioxidant, and anti-aging effects of vitamin D in several indicative clinical trials and studies are presented in detail in Table 2.

6. Skin Conditions and Evidence of the Beneficial Role of Vitamin D

Vitamin D’s immunomodulatory actions are generally mediated via its binding to the VDR, present in keratinocytes and immune cells. Such interaction influences gene expression, resulting in a reduced generation of pro-inflammatory cytokines (e.g., IL-6, IL-17, and TNF-α), increased T-reg action and production of anti-inflammatory IL-10, and enhanced antimicrobial peptide expression, thus boosting immunity [139,182,183]. Newly emerged technological vitamin D applications, photodynamic therapy (PDT) exploitation, melanoma or non-melanoma skin cancer prevention, and vitamin D combination treatments with corticosteroids or calcineurin inhibitors have shown superior outcomes in reducing inflammatory skin disease symptoms [31,64,184,185]. Concerning several disorders, including psoriasis, atopic dermatitis, vitiligo, acne vulgaris, and acne rosacea, vitamin D has been successfully implicated and integrated in many cosmetic, cosmeceutical, and nutraceutical/nutricosmetic applications, and some clinical trials and vitamin D-based applications will be further discussed in the following sections. All mentioned dermatological disorders connected with vitamin D levels, are presented in Table 3.

6.1. Psoriasis

Psoriasis is characterized as a chronic, inflammation-related skin disease that affects approximately 2–3% of the worldwide population, causing significant morbidity [186]. The pathogenesis of this disease is not fully understood yet, but research has shown that it is linked to the impairment of immune cells in the skin. More specifically, T cells are the ones possessing a crucial role in the development of psoriasis [89,187].
Psoriatic fibroblasts were demonstrated to be somewhat resistant to the antiproliferative actions of 1α,25(OH)2D3 in 1985 [188]. This observation triggered the outbreak of theories suggesting that oral vitamin D, and subsequently its topical preparations, would be useful in treating hyperproliferative skin conditions. While the introduction of topical vitamin D analogs notably altered the treatment of psoriasis, the oral use of vitamin D3 as a therapeutic failed due to the risk of hypercalcemia [37]. Vitamin D analogs such as calcipotriol are widely used to manage psoriasis plaques, as they regulate keratinocyte proliferation and differentiation. Vitamin D suppresses Th1- and Th17-mediated inflammation and reduces pro-inflammatory cytokines like IL-17 and TNF-α [36,189,190,191].
Numerous investigations have revealed extremely decreased serum vitamin D levels in psoriasis patients; however, the significance of this discovery is still quite blurry. It is recommended that patients with low serum vitamin D levels should be provided with oral vitamin D3 supplements, so as to avoid bone mineral density loss and comorbidities associated with psoriasis [192], given that a diet high in vitamin D is believed to place a positive impact on various comorbidities (i.e., obesity, diabetes mellitus, metabolic syndrome, cardiovascular events) [193]. The cornerstone of treatment for mild to severe psoriasis is topical vitamin D analogs, either alone or in conjunction with topical corticosteroids, especially betamethasone [81]. Especially the use of topical calcipotriol has been proven as a safe treatment in cases of localized plaque psoriasis (Table 3) [37,194].
Table 3. Dermatological Disorders connected with Vitamin D levels.
Table 3. Dermatological Disorders connected with Vitamin D levels.
DiseaseType of Vitamin D’s AssociationReferences
Psoriasis
  • Chronic inflammation and immune system dysregulation (specifically T cells), linked to vitamin D deficiency and associated with various comorbidities (e.g., obesity, diabetes, cardiovascular events), which responded to vitamin D analogs (topical treatment), effectively
[37,89,186,187,188]
Atopic dermatitis (AD)
  • Increased prevalence at higher latitudes, due to reduced sun exposure and decreased vitamin D production
  • Characterized by epidermal barrier dysfunction and immune system dysregulation, and linked to low serum vitamin D levels (especially in moderate to severe AD and lighter skin types)
  • Topical vitamin D analogs are contraindicated, as they can exacerbate the condition by inducing TSLP-related inflammatory responses
[38,39,40,41,195,196,197,198,199]
Vitiligo
  • Autoimmune pigmentary disorder, initiated by melanocyte destruction, leading to depigmented patches
  • Vitamin D plays a crucial role in restoring melanocyte function, through modulating T cell activation, but its effectiveness (both oral and topical) in treatment remains inconsistent
  • The combination of topical vitamin D analogs with corticosteroids or calcineurin inhibitors enhances repigmentation; however, a combined use with phototherapy is not recommended
[42,43,89,200]
Acne vulgaris
  • Common inflammatory skin disorder, related to immune response targeting Propionibacterium acnes
  • Vitamin D suppresses Th17 differentiation induced by P. acnes, being a potent candidate for controlling acne
  • Sebocytes respond to 1α,25OH2D3, indicating that vitamin D analogs may be beneficial in acne management
[44,89]
Acne rosacea
  • A chronic skin disorder, primarily affecting the face, that is associated with elevated serum vitamin D levels, potentially indicating a link between increased vitamin D and the subsequent decrease in the risk development of rosacea
[201]

6.2. Atopic Dermatitis

Greater geographic latitudes display greater rates of atopic dermatitis (AD), which is correlated with reduced sun exposure and a decreased generation of vitamin D [195]. AD is a chronic, recurrent inflammatory disease, characterized by pruritus and eczematous lesions, that affects approximately 20% of children and 7.2% of adults, globally [41]. The primary cause of AD is the malfunction in the synthesis of the epidermal barrier and a change in the cutaneous immune system, which impacts mainly filaggrin [196]. Topical vitamin D enhances skin barrier function by promoting keratinocyte differentiation and the formation of antimicrobial peptides like cathelicidin, therefore decreasing Staphylococcus aureus colonization, which is a common aggravating AD factor [202,203].
Vitamin D is known to regulate the integrity and permeability of the epidermal barrier, although its exact role in this process is still unknown [38]. There has been a long debate concerning the efficacy of oral vitamin D as an AD supplement. Low serum vitamin D levels, especially during winter, have been linked in several studies to the progression and severity of the illness. However, the exact role that vitamin D plays in the pathophysiology of AD remains unclear [197]. Serum vitamin D levels were shown to be considerably lower in moderate and severe AD than in mild AD, in a recent study conducted in Spain, but this link was only significant for patients with light Fitzpatrick skin types (study involving 134 children with AD and 105 control samples, without AD) [40].
To sum up, there is no clear evidence pointing out the importance of dietary vitamin D as an AD treatment [39]. Further investigation should be conducted to prove the importance of vitamin D supplementation in such types of patients. Conversely, animal investigations using topical vitamin D analogs have been claimed to cause AD, mostly via activating TSLP in keratinocytes [198]. TSLP acts as a switch for the T-helper 2 (Th2) allergic inflammation, a common characteristic of AD, and is necessary for the induction of pro-inflammatory chemokines—mostly IL-4, IL-13, and IL-31 in AD (Table 3) [199].

6.3. Vitiligo

Vitiligo is a pigmentary disorder, initially induced by the breakdown of functioning melanocytes in the epidermis, which formulate, as a result, well-defined, depigmented patches or macules of varying sizes and forms [89]. Vitamin D acts as protection in the epidermal melanin unit and contributes to restoring melanocyte integrity, through many ways like the activation, proliferation, and migration of melanocytes, and pigmentation cascades. Such properties derive from their ability to modulate T cell activation, which is probably correlated with melanocyte disappearance in vitiligo [89].
Vitiligo is an autoimmune skin disorder, and due to various vitamin D favorable effects, oral vitamin D supplements could be used in vitiligo therapy. However, multiple studies with incompatible results regarding the efficacy of oral vitamin D in treating vitiligo must not be overlooked [89,200]. A confliction also exists about topical vitamin D analogs’ ineffectiveness [200]. Useful in enhancing clinical repigmentation is combining topical vitamin D analogs with topical corticosteroids or the topical calcineurin inhibitor [43]. Furthermore, it is not advised to combine vitamin D analogs with phototherapy [42]. Narrowband UVB phototherapy combined with topical vitamin D has shown increased efficacy in repigmentation, as compared to phototherapy alone, by triggering melanogenesis and regulating immune responses in vitiligo cases (Table 3) [54,204,205].

6.4. Acne Vulgaris and Acne Rosacea

Acne vulgaris is one of the most common skin disorders affecting many people worldwide. Acne vulgaris’ pathogenesis relates to inflammation, mainly enhanced by immune response and specifically targets Propionibacterium acnes [89]. Recent study outcomes have shown that P. acnes is a strong Th17 inducer and that 1α,25(OH)2D3 suppresses Th17 differentiation generated by P. acne, which may be useful in controlling acne [44]. Plus, it was demonstrated that sebocytes are 1α,25(OH)2D3-responsive target cells, suggesting that vitamin D analogs might be useful in acne management, as vitamin D deficiency is pronounced in acne vulgaris patients [44,46]. Vitamin D has pleiotropic effects in preventing acne vulgaris lesions, through directly inhibiting cell division, decreasing sebum secretion and pore blockage, and impeding Cutibacterium acnes’ growth [206].
Similarly, another study suggested that in patients with rosacea, a high level of serum vitamin D was traced. Rosacea is a chronic skin disorder that mainly affects an individual’s face and is generally recognized by symptoms including flushing or long-term redness, especially in the nose and cheeks. Study findings have suggested that elevated serum 1α,25(OH)2D3 may be correlated with a lower risk of incidence and further development of rosacea [201,207]. However, the link between vitamin D levels and rosacea remains inconclusive, necessitating further research to clarify its therapeutic role [47]. Vitamin D’s anti-inflammatory actions may aid in mitigating redness and pustule formation in rosacea, while considering acne vulgaris, vitamin D reduces sebum formation and inhibits Cutibacterium acnes-induced inflammation (Table 3) [201,207,208].

7. Vitamin D in Cosmetics and Cosmeceuticals

Vitamin D has become an important part of dermatology because of its many advantages concerning skin health [209]. Its usefulness in different topical formulations, its underlying mechanisms of action, and its therapeutic efficacy in treating skin disorders have been brought to light by current scientific breakthroughs. Vitamin D is already used in skincare products in a variety of ways, each designed to meet unique dermatological requirements [81,124,210]. This hormone can, for instance, be found in creams, which are made to provide specific troubled skin areas with high concentrations of bioactive substances. When treating illnesses like psoriasis and AD, where localized treatment is vital, cosmetic formulations involving vitamin D have reportedly exhibited great results [121]. Because of their thick texture, creams can be applied deeply into the layers of the afflicted skin. In contrast to creams, lotions are designed with a lower viscosity and are appropriate for daily use. Lotions are therefore perfect for efficiently covering bigger body areas. Because of their thinner texture, vitamin D may be applied consistently, offering broad coverage. Moreover, sprays offer a practical way to get vitamin D into difficult-to-reach areas, providing a non-greasy substitute for creams and lotions, which makes them useful for people looking for a simple and fast application technique [121].
Ointments work well on excessively dry or irritated skin because of their thick, occlusive texture [211]. Such mixtures are dedicated to improving the absorption of vitamin D by creating a barrier, which prolongs the feeling of comfort and hydration. Because of their propensity to stay on the skin for a long time period, vitamin D has plenty of time to enter the body and initiate its function. Lighter options are provided by serums that are frequently mixed with different active components, which in turn, due to their composition, are less residual and enable tailored vitamin D administration [27]. Because of their quick absorption, serums are very helpful for targeted therapy regions. Vitamin D can also be found in infused oils mixed with ones, to increase their moisturizing capabilities, enhance their particularly helpful profile for people with dry skin types, and offer several therapeutic effects that improve the overall skin health. Numerous anti-aging and photoprotective effects towards human skin health, of vitamin D3 and its active metabolites, derive from their utilization in cosmetic formulations [27]. These positive outcomes are accomplished by immunomodulation, which also regulates keratinocyte proliferation and differentiation so as to provide the epidermal barrier the ability to preserve skin homeostasis. Furthermore, vitamin D products trigger antioxidative reactions, prevent DNA damage, and promote DNA repair to reduce the risk of cancer’s onset and premature skin aging. The activities of lumisterol metabolites are indeed comparable [27].
It is important to mention that the most frequently utilized type of vitamin D in skincare is vitamin D3. Because of its higher efficacy and stability, vitamin D3 is preferred in skincare products over vitamin D2. When it comes to increasing and/or preserving vitamin D levels in the body, vitamin D3 is notably superior to vitamin D2, while being more stable, ensuring that the medicine sustains its potency over time. Furthermore, because of its stability and efficacy, vitamin D3 is the recommended ingredient in a variety of skincare formulations, such as oils, serums, ointments, lotions, and creams, as it keeps its effectiveness in a variety of formulations and blends in well with other active components [121,212]. In addition, calcipotriol and 1α,25(OH)2D3, two forms of vitamin D3, are widely used to treat skin diseases including psoriasis. By slowing down the skin cells’ fast proliferation, calcipotriol, namely a synthetic derivative of vitamin D3, is topically applied to treat the symptoms of psoriasis. Plus, the active 1α,25(OH)2D3 is used as well in a plethora of medical applications, due to its strong potency against skin diseases [121].
Vitamin D is categorized as an active ingredient in medicine. The European Union prohibits the use of vitamin D3 and D2 in cosmetic formulations (No. 335 in Annex II of Regulation (EC) No. 1223/2009) [213]. Nevertheless, in the CosIng database, a vitamin D precursor, 7-dehydrocholesterol, is characterized as an emulsion-stabilizing, skin-conditioning, and viscosity-controlling ingredient [214]. Vitamin D provitamins are allowed to be used in cosmetic and cosmeceutical formulations. Notably, the European Cosmetics Directive explicitly bans the use of both ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3). However, this does not apply to provitamins that are licensed in the US, for their utilization in skincare products [25,215]. Several examples of products utilizing vitamin D in the form of provitamins exist, including anti-aging day and night creams, serums, and many other formulations deriving from numerous global companies. Moisturizers enriched with vitamin D improve hydration and reduce transepidermal water loss, benefiting dry and sensitive skin [216]; vitamin D-based anti-aging serums regulate collagen synthesis and reduce oxidative stress [34]; and topical vitamin D formulations may regulate melanogenesis, addressing melasma and uneven skin tone [217,218].
As mentioned before, a whole series of interesting effects in the skin are described in the context of vitamin D. To begin with, calcitriol has an impact on keratinocyte development and differentiation, playing a crucial function in the therapy of psoriasis [219]. In parallel, the skin’s calcium gradients are properly maintained by calcitriol [25]. Stimulation for the formation of anti-microbially effective peptides, like defensins and cathelicidins, is another function activated by 1α,25(OH)2D3. The impact of such peptides is interesting in the context of inflammatory processes in neurodermatitis cases [220]. Furthermore, calcitriol prolongs the self-protection of the skin during the UVB radiation exposure, by stimulating heat shock proteins. Lastly, several studies [221,222,223] describe a repigmentation in vitiligo cases, after the application of calcitriol or vitamin D analogous products. In Table 4, various cosmetic and cosmeceutical formulations are described.

7.1. Mechanism of Action

The mechanism of action of vitamin D when applied topically to the skin consists of several complex biological processes that contribute to its therapeutic effects [74,81].
Topical vitamin D formulations mostly act locally within the skin in contrast to systemic vitamin D ones, which are converted to their active form in the liver and kidneys. After being applied topically, vitamin D in the form of calcitriol or its analogs interacts with particular skin cellular targets, to produce its advantageous effects. One of the main mechanisms of action involves the VDR, which is expressed in various skin cells including keratinocytes, fibroblasts, and immune cells. The primary mode of action is via its VDR interaction, where the N-terminal DNA-binding domain of the VDR transcription factor binds to vitamin D response elements (VDREs) on DNA, the C-terminal ligand-binding domain, and the hinge region that joins these transcription factors’ three primary domains. After binding to the ligand 1α,25(OH)2D3, its 12 helices in the ligand-binding domain undergo conformational changes that ultimately enable the domain to interact with coactivators and its heterodimer partner, which is usually the RXR [74].
VDR attaches itself to VDREs, which are primarily direct repeats of hexanucleotides, spaced three nucleotides apart (DR3 call II gene motifs). This binding enhances the recruitment of several coregulatory complexes necessary for the genomic VDR action. Due to the gene- and cell-specific nature of these complexes, precise control over gene expression is possible. Coactivators with histone acetyltransferase activity, such as the non-receptor, protein-tyrosine kinase (SRC) family, and corepressors with histone deacetylase activity, like the silencing mediator of retinoic acid and the thyroid hormone receptor (SMRT), as well as the nuclear receptor corepressor (NCoR), are examples of coregulators. Methyltransferases, demethylases, adenosine triphosphate (ATP)-ase-containing nucleosomal remodeling complexes, and elements of the mediator complex interacting with RNA polymerase II are other interesting factors linked to the VDR cascade [74].
This multifaceted procedure encourages keratinocytes to differentiate and proliferate, a necessary action for preserving the skin barrier. By upregulating genes involved in keratinocyte differentiation and cornification, vitamin D strengthens the skin’s protective layer and promotes the development of a functioning stratum corneum. Furthermore, topically applying vitamin D promotes the synthesis of antimicrobial peptides such as cathelicidins, which are essential for the skin’s innate immune response, as they support the skin’s defensive processes and aid in the fight against infections. Vitamin D helps the skin keep itself healthy and fight off infections, by improving the synthesis of cathelicidins [37]. Additionally, using vitamin D topically shields the skin from harm caused by UV rays. Antioxidant qualities of vitamin D contribute, in fact, to lessening oxidative stress acceleration by the UV light. The advantages of vitamin D in preserving skin health and averting UV-induced premature aging are consequently more and more clear [37].
The anti-inflammatory properties of vitamin D play a critical role in its skin activity [27,37,230]. By upregulating anti-inflammatory cytokines and downregulating pro-inflammatory ones, vitamin D highly participates in the immune response. This skin-correlated activity is especially helpful in the treatment of inflammatory skin diseases like psoriasis, since it reportedly reduces inflammation and excessive skin cell turnover, mainly by inhibiting T cell proliferation and further regulating the formation and repair of skin cells [37]. Through the facilitation of cellular processes involved in tissue repair and regeneration, it aids in the healing of wounds. Topical vitamin D application also promotes keratinocyte migration and proliferation to the wound site, hastening the closure of wounds and aiding in the repair of injured skin [37]. Vitamin D provides, additionally, defense against UV-induced oxidative stress, due to its antioxidant qualities of effectively neutralizing free radicals, and thus, aids in the prevention of skin damage and premature aging and the promotion of the overall skin resilience and health [37].
Skin health is significantly impacted by vitamin D, and its therapeutic advantages are mediated through several pathways [25]. By maintaining the epidermal barrier and facilitating the development of epidermal keratinocytes, vitamin D encourages the skin to function as a protective barrier, less vulnerable to infection and environmental harm, towards a potential injury and keratinocytes maturing properly [25,27]. Furthermore, this hormone plays a critical role in controlling the proliferation of cells in the epidermis, as it regulates aberrant cell growth, which is important in conditions like psoriasis with increased rates of cell turnover [25]. Finally, vitamin D helps to promote skin health during the skin renewal process, relieves the symptoms of skin-associated comorbidities, normalizes epidermal growth, and enhances cell proliferation as well as repair activities, functions that make this hormone a useful tool in the treatment of wounds and skin injuries, since it primarily speeds up the healing process of injured skin [25].

7.2. Efficacy of Vitamin D

Clinical trials and studies have highlighted the efficacy of vitamin D in managing various skin conditions like psoriasis, wound healing, different types of acne, and UV-induced damage [231,232,233,234]. While these findings are promising, ongoing research is essential so as to fully understand the optimal use and value of vitamin D in skincare. The treatment of psoriasis, a condition marked by thick, scaly patches caused by a rapid turnover of skin cells, is listed among the most investigated uses of vitamin D and its analogs [37,57,231,235,236,237]. One particularly useful synthetic version of vitamin D is calcipotriene. An experiment based on the utilization of calcipotriene exhibited a vast improvement in psoriasis symptoms, especially in lowering plaque thickness and scaling [37]. These results have been validated by other similar research that highlighted the efficacy of topical vitamin D analogs, towards lowering psoriatic plaques without any recorded side effects or adverse observations [238].
The interesting function of vitamin D in wound healing has also been evaluated in several clinical trials [30,94,232,239]. Effective wound healing depends on its role in cell differentiation and proliferation. Vitamin D supplementation has been pointed out both to enhance the wound-healing rates and to shorten the acquired healing time [240]. Recent research outcomes by Q. Chen et al. [239] have demonstrated vitamin D’s crucial role in maintaining skin barrier integrity and reducing the symptoms of chloasma. The findings clearly highlighted that vitamin D, whether applied topically or administrated orally, reduced the chloasma severity and enhanced the wound-healing process [239].
Another topic receiving vast attention has been the use of vitamin D in acne treatment and, for example, in treating acne vulgaris [45,233,241]. Topical vitamin D formulations were observed to reduce inflammation and acne lesions; however, the effects were not as consistent as with other, already widely utilized acne therapeutic interventions [45]. Additionally, vitamin D aids in minimizing UV ray damage and potentially slowing down the skin aging process, while considering photoprotection, vitamin D’s antioxidant qualities could lessen the UV-accelerated oxidative damage [45]. According to research based on how vitamin D affects skin aging, adequate levels of vitamin D in the body may contribute to keeping one’s skin supple and minimizing wrinkles’ appearance, although more studies are required, so as to further validate such benefits [25,45,193].

8. Discussion—Challenges and Controversies

The use of vitamin D in cosmetic products has garnered noteworthy interest owing to its possible advantages for the skin’s wellbeing, such as better wound healing, improved barrier function, and anti-inflammatory properties [242,243,244]. However, issues and disagreements have emerged, regarding the inclusion of vitamin D in cosmetic products, that must be resolved. Stability, bioavailability, efficacy, regulatory difficulties, and possible interactions with other components are only a few of the challenges, where especially vitamin D’s low stability poses a major obstacle for its use in cosmetic products. When exposed to light, heat, and air, vitamin D is prone to deterioration, chiefly in its active forms (D2 and D3), while over time this instability may cause the product’s efficacy to decline. Cosmeticians and cosmetic industries, as a result, need to take great caution when creating such formulations, to shield the vitamin D content from any external elements that may degrade it. Stabilizing chemicals and encapsulation techniques are common approaches engaged to improve vitamin D’s stability in cosmetic products. It is difficult to guarantee whether vitamin D cosmetic formulations will benefit our skin, while also owning longer shelf life, and this is the main reason why further investigation upon improving both efficacy and longevity issues is highly necessitated [245].
Another point of contention is the bioavailability of vitamin D when applied topically [54,246,247]. Although the systemic benefits of vitamin D are well established, its efficacy in topical preparations remains uncertain. The composition of the substance and the skin state might affect how well it absorbs and implements vitamin D. Studies have claimed that topical vitamin D may not have a substantial impact on systemic vitamin D levels but may enhance localized effects like lowering inflammation and enhancing skin hydration [216,248]. This limitation raises questions about the overall effectiveness of vitamin D in cosmetics and whether it can provide safe, substantial benefits. Regulatory issues also complicate its cosmeceutical use. Different countries have established varying regulations regarding the use of vitamin D in skincare products, as, for example, while some countries have allowed the inclusion of vitamin D at certain concentrations, others have imposed stricter guidelines [249]. Additionally, concerns about its potential for adverse reactions or interactions with other cosmetic ingredients have recently been raised [250]. The safety profile of vitamin D at high concentrations or when used in combination with other active ingredients, needs to be conclusively and thoroughly assessed [249].
Vitamin D incorporation into cosmetic formulas can be rather expensive, as maintaining its stability, efficacy, and proper function is vital. This may result in higher retail prices, potentially limiting the availability of certain products. Furthermore, market acceptance is also crucial, due to the fact that while many customers recognize vitamin D’s benefits, others may still be skeptical of its effectiveness when applied topically, given its well-established advantages when ingested or produced directly after sun exposure. An additional point of contention is how vitamin D interacts with other active chemicals utilized in cosmetic formulations, such as retinoids, peptides, or antioxidants. While some combinations might increase the product’s overall efficacy, others might cause negative side effects or lessen the final product’s benefits. In order to guarantee that vitamin D stays effective, and that the formulation does not cause unforeseen side effects, understanding these interactions takes substantial research and testing. Consumer education is essential in addressing misconceptions about the role of vitamin D in skincare. There is often confusion regarding the differences between vitamin D’s systemic effects and its topical benefits. Educating consumers about realistic expectations and the specific benefits of topical vitamin D is hence of great importance [245].
The role of vitamin D in skincare is still being investigated from a scientific perspective. So as to completely understand the mechanisms via which topical vitamin D works and to identify the best formulations and doses regarding different skin diseases, further research is required, and current cosmetic formulations may need to be adjusted as new findings emerge. Because scientific research is constantly evolving, it can influence market trends and product development, creating uncertainty for both consumers and manufacturers. Despite the challenges, the use of vitamin D in cosmeceuticals holds significant promise for improving skin health and addressing various dermatological concerns. However, the complexities and controversies surrounding its incorporation must be carefully navigated. Factors including stability, bioavailability, regulatory compliance, cost, and potential interactions with other ingredients are all key elements in shaping vitamin D’s skincare profile. As research progresses and consumer awareness expands, addressing and overcoming these challenges will be crucial for maximizing the benefits of vitamin D, while ensuring both safety and efficacy in cosmeceuticals [245].

9. Future Directions and Research

The application of vitamin D in cosmetics is an area of growing interest due to its potential benefits for skin health. However, to fully realize and harness these benefits, future research and development must focus on several key areas, including optimizing formulation techniques, enhancing bioavailability, comprehending the mechanisms of action, evaluating long-term safety, and exploring innovative delivery systems.
Future research should focus on advancing formulation techniques to improve the stability and efficacy of vitamin D in cosmetics. Vitamin D, particularly in its active forms, is vulnerable to degradation from UV, heat, and air exposure. Innovative encapsulation methods, such as nanoencapsulation or liposomal delivery systems, could provide enhanced protection against environmental factors, thereby extending the shelf life and stability of vitamin D in cosmeceuticals [251]. Moreover, developing new stabilizing agents and excipients that preserve vitamin D’s potency, without also compromising the product’s texture and feel, is crucial, as this will not only improve the quality of vitamin D-based cosmetics, but also ensure their maximum benefits [251]. Additionally, vitamin D could be a key ingredient in orally administered nutricosmetics, designed to improve skin health and appearance. Future research may focus and explore optimal formulations, combining vitamin D with other skin-beneficial nutrients, including collagen, hyaluronic acid, omega-3 fatty acids, polyphenols, biotin, and antioxidants, able to enhance skin hydration, elasticity, and overall vitality [252,253,254,255,256,257,258]. Moreover, the synergistic activity of antioxidants like vitamin C and E with vitamin D [25,254,259], or niacinamide–vitamin D [260] combinations, could offer several cosmeceutical and nutricosmetic benefits against dermatological and skin health conditions.
Sunscreens, a type of cosmetic product, can provide protection to the skin against UV and dermatologists recommend their use [261]. It is unfortune to mention that some of these cosmetic preparations contain chemicals, something that is not safe and can even lead to systemic absorption [262]. To add, not all sunscreens offer protection against UVA, pollution, infrared light (IR), or blue light and so as to produce vitamin D, UVB is necessary. More specifically, the role of vitamin D in mitigating oxidative stress could be harnessed in anti-pollution skincare formulations. Plus, as blue light exposure from screens becomes a growing concern, combining vitamin D with other protective agents may aid in counteracting its effects, promoting healthier skin [263]. There are currently no recommended sunscreen usage guidelines or ideal sun protection agents [264]. New active compounds, stable and safe to add to cosmetic compositions [265] that may also function as antioxidant agents preventing photoaging like vitamin D, are therefore needed [266].
One major challenge in the topical application of vitamin D remains as its bioavailability [267]. Enhancing the skin’s capacity to absorb and utilize topical vitamin D should be a primary research goal. By comparing the effectiveness of various vitamin D forms, such as vitamin D3 and its metabolites, in topical applications, researchers may be able to identify the most suitable variants for topical use. Moreover, its delivery and absorption could also be improved by researching synergistic effects with other compounds, such as penetration enhancers or advanced vehicle systems that enhance skin permeability. Future research on the optimal vitamin D content for various skin types and conditions will help create products tailored to specific dermatological needs [267]. Beyond traditional encapsulation, emerging encapsulation techniques like solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), and stimuli-responsive carriers could offer superior protection, and sustained release, and target both oral and topical vitamin D delivery, while improving stability, enhancing bioavailability, minimizing potent degradation, and responding to environmental changes (e.g., pH, temperature, UV exposure) [65,268].
Optimizing vitamin D’s use in cosmetics also requires a deeper understanding of its molecular and cellular actions. Scientists should focus on clarifying the exact pathways through which vitamin D impacts skin health, encompassing its effects on cell differentiation, proliferation, and barrier function. Examining the role of coregulatory proteins and VDR in these processes will provide critical insights into how vitamin D works and how its effects can be maximized in cosmetic formulations. Also, exploring how vitamin D interacts with other signaling pathways, like those connected to oxidative stress and inflammation, may reveal novel therapeutic targets and improve product development. Safety is of paramount importance when formulating cosmetic products, and long-term research is required to evaluate vitamin’s D efficacy. Future studies could explore as well how vitamin D affects melanogenesis and pigmentation pathways. Such product development could lead in successfully targeting pigmentation disorders, like melasma, or developments aiming to even out skin tone (skin tone regulation) [25,269,270].
The skin microbiome plays a vital role in maintaining skin health. Therefore, studies could focus on investigating how vitamin D influences the skin microbiome and whether combining vitamin D with prebiotics, probiotics, or postbiotics in cosmetic, cosmeceutical, and nutricosmetic formulations can lead to enhanced skin barrier function and resilience against environmental stressors [271,272,273]. While vitamin D is essential for overall health, excessive use of high-concentration products over extended periods may have adverse effects. Research should hence focus on long-term safety evaluations, such as potential skin reactions, systemic absorption, and combinations with other topical or systemic treatments. Rigorous clinical trials and post-market surveillance are necessary to guarantee that consumers are not exposed to risks from vitamin D-based cosmetics and that any potential side effects are promptly identified and addressed [269].
Innovative delivery systems offer promising avenues for enhancing the effectiveness of vitamin D in skincare. Research into new delivery methods, such as microneedle systems (e.g., patches), microemulsions, smart skin patches, bioengineered vitamin D precursors, nanoencapsulation carriers, photonic delivery, quantum dot systems, or hydrogel matrices, could enable more efficient and controlled release of vitamin D into the skin. These advanced systems may enhance the penetration and targeting of vitamin D to specific skin layers where it can exert its beneficial effects [72,274,275,276,277,278]. Exploring combination therapies that integrate vitamin D with other active ingredients like antioxidants or anti-inflammatory agents could amplify the overall efficacy of cosmetic products. Although the number of studies describing the emerging technologies appear large at first glance and various strategies have been covered, they are all at an early research stage [64].
As personalized skincare continues to grow in popularity, future studies should investigate vitamin D’s role in customized skincare uses. Individual responses to vitamin D are influenced by genetic and environmental factors; a better understanding of these variables might result in more individualized and efficient treatments. Studying the impact of genetic variations in vitamin D’s metabolism on skin health and sensitivity to topical treatments, as well as how factors like skin type, age, and lifestyle affect vitamin D-based cosmetics, will aid in the development of more tailored and customized products, towards several skin conditions [279]. Additionally, during the rise in machine learning, and artificial intelligence (AI)-driven skincare diagnostics, vitamin D products could be tailored to an individual’s needs. AI tools are able to analyze genetic markers, skin type, and environmental factors, with a view to recommend personalized vitamin D products, ensuring maximum efficacy for different users [280,281]. Three-dimensional bioprinting could revolutionize personalized vitamin D cosmetic formulations, as custom-printed patches or masks, for instance, can deliver precise, individual-specific dosages [282].
Further clinical evidence is essential to support the benefits of vitamin D in cosmetics/cosmeceuticals. Large-scale, carefully monitored clinical trials should assess the effects of vitamin D on various skin disorders, like eczema, acne, and aging [283]. The widely known vitamin D benefits in promoting cell differentiation and proliferation suggest its potential use in wound-healing and scar management applications. Future cosmeceuticals could incorporate vitamin D for post-surgical or post-operative skincare, burn recovery, and minimizing scar appearance, effectively [25,284,285,286,287]. Moreover, as vitamin D plays a significant role in collagen synthesis and photoaging prevention, further studies could elaborate on its interaction with peptides, growth factors, or retinoids to develop comprehensive anti-aging solutions. Such products may target wrinkles, fine lines, and loss of elasticity, improving overall skin health and barrier function [25,34,63]. Cross-disciplinary research, involving dermatology and endocrinology, could offer deeper insights into the systemic and topical effects of vitamin D on skin health. Such an interesting approach may lead to innovations in treating chronic skin conditions, such as psoriasis and atopic dermatitis, while improving general skin vitality and wellness [288,289].
In addition to clinical evaluation, these trials should incorporate patient-reported outcomes and quality of life assessments. Robust clinical data will help advocate for the use of vitamin D in skincare products and guide both consumers and healthcare professionals in making rational decisions. Consumer education is crucial for the successful integration of vitamin D into cosmeceuticals. Prospective investigations ought to concentrate on strategies aiming to augment consumers’ comprehension of the benefits and constraints of vitamin D in skincare. This entails developing educational campaigns that define the differences between systemic and topical vitamin D, as well as establishing reasonable performance standards for such products. Educating consumers about proper usage, possible interactions with other products, and the significance of a thorough skincare routine is believed to be a major key in integrating vitamin D in the cosmetic, cosmeceutical, and nutricosmetic field [283]. Furthermore, future vitamin D-based cosmetics can involve the exploration of plant-derived precursors or lab-synthesized alternatives, which could ensure that such products align with vegan, cruelty-free, and environmentally sustainable practices [290,291]. Finally, as vitamin D applications in cosmetics and nutricosmetics expand, global regulatory alignment, ensuring consistent guidelines and transparency in product labeling for its safe and effective use across regions, will be crucial [290,291].
Ultimately, the future of vitamin D in cosmetics or cosmeceuticals holds significant promise, but advancing in this field requires addressing several key research and development areas. Enhancing formulation techniques, improving bioavailability, understanding molecular mechanisms of action, ensuring safety, exploring innovative delivery systems, investigating personalized skincare solutions, expanding clinical evidence, and even addressing consumer education are all critical aspects to advancing vitamin D’s role in skincare. By tackling these challenges and leveraging new scientific insights, researchers and industry professionals can unlock vitamin D’s full potential in cosmetics, leading mainly to improved skin health and overall wellness [283].

10. Conclusions

Vitamin D has a long history dating back to the 17th century, with its most notable forms being vitamin D2 and D3. Vitamin D can be absorbed through the skin in multiple ways, namely penetration through the stratum corneum, diffusion through the epidermis, interaction with keratinocytes, and local metabolism to its active form. Research indicates that topical vitamin D analogs provide great therapeutic benefits, contributing mostly to healthy bones and teeth, immune function, and skin health against diseases like psoriasis. Factors such as UV light exposure and the specific skin type may notably influence vitamin D synthesis in human skin.
A clear relationship indeed exists between vitamin D and skin health. On one hand, the skin serves as a source of vitamin D, and on the other, vitamin D plays an important role in maintaining skin health, with its deficiency leading to dermatological disorders. Preserving optimal vitamin D levels involves multiple factors, and at this point it must be highlighted that sunny climates do not always guarantee sufficient vitamin D production. Based on available research, the supplementation of vitamin D is recommended to maintain normal serum levels and therefore prevent the negative effects of deficiency.
Vitamin D’s confirmed unique anti-inflammatory, anti-melanoma, anti-cancer, anti-UV, anti-aging, and antioxidant properties have ushered the onset of the production of several skincare products, including cosmetic systemic and topical formulations and analogs. This hormone’s astonishing structural, chemical, and biological profile has been extensively exploited against conditions like psoriasis and atopic dermatitis, and less utilized considering vitiligo, acne (vulgaris), and rosacea. Consequently, further research is required to evaluate its role in different skin-related conditions, addressing all arousing controversies and potential positive or side effects, and thus ensuring optimal results in the cosmeceutical field. Vitamin D holds great promise as a natural bioactive constituent, and continuous research and innovation will be essential to unlock its full skincare potential, towards safer, more targeted, and more personalized treatment solutions for a range of dermatological needs and several existent skin types.

Author Contributions

Conceptualization, A.T.; methodology, A.T.; software, All Authors; validation, A.T.; investigation, A.T., T.A., S.N.A.P. and E.A.A.; writing—original draft preparation, A.T., T.A., S.N.A.P. and E.A.A.; writing—review and editing, A.T., A.O., S.L. and T.A.; visualization, A.T.; supervision, A.T.; project administration, A.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Holick, M.F. Resurrection of Vitamin D Deficiency and Rickets. J. Clin. Investig. 2006, 116, 2062–2072. [Google Scholar] [CrossRef] [PubMed]
  2. Pettifor, J.M. Nutritional Rickets: Deficiency of Vitamin D, Calcium, or Both? Am. J. Clin. Nutr. 2004, 80, 1725S–1729S. [Google Scholar] [CrossRef] [PubMed]
  3. Ellis, H. Daniel Whistler: English Physician Who Published the First Book on Rickets in 1645. Br. J. Hosp. Med. 2019, 80, 51. [Google Scholar] [CrossRef] [PubMed]
  4. Jones, G. 100 Years of Vitamin D: Historical Aspects of Vitamin D. Endocr. Connect. 2022, 11, e210594. [Google Scholar] [CrossRef] [PubMed]
  5. O’Riordan, J.L.H.; Bijvoet, O.L.M. Rickets before the Discovery of Vitamin D. BoneKEy Rep. 2014, 3, 478. [Google Scholar] [CrossRef] [PubMed]
  6. Rajakumar, K.; Thomas, S.B. Reemerging Nutritional Rickets: A Historical Perspective. Arch. Pediatr. Adolesc. Med. 2005, 159, 335. [Google Scholar] [CrossRef] [PubMed]
  7. Huldschinsky, K. Heilung von Rachitis durch künstliche Höhensonne. DMW—Dtsch. Med. Wochenschr. 1919, 45, 712–713. [Google Scholar] [CrossRef]
  8. Hawgood, B.J. Sir Edward Mellanby (1884–1955) GBE KCB FRCP FRS: Nutrition Scientist and Medical Research Mandarin. J. Med. Biogr. 2010, 18, 150–157. [Google Scholar] [CrossRef]
  9. Carpenter, K.J.; Zhao, L. Forgotten Mysteries in the Early History of Vitamin D. J. Nutr. 1999, 129, 923–927. [Google Scholar] [CrossRef]
  10. McCollum, E.V.; Simmonds, N.; Becker, J.E.; Shipley, P.G. Studies on Experimental Rickets. J. Biol. Chem. 1922, 53, 293–312. [Google Scholar] [CrossRef]
  11. Mellanby, E. An Experimental Investigation on Rickets. Lancet 1919, 193, 407–412. [Google Scholar] [CrossRef]
  12. DeLuca, H.F. History of the Discovery of Vitamin D and Its Active Metabolites. BoneKEy Rep. 2014, 3, 479. [Google Scholar] [CrossRef] [PubMed]
  13. Fernando, D.S.; Graciela, E.C. The Saga of Vitamin D Discovery. Anat. Physiol. Biochem. Int. J. 2023, 6, 555685. [Google Scholar] [CrossRef]
  14. Windans, A.; Bock, F. Über Das Provitamin Aus Dem Sterin Der Schweineschwarte. Biol. Chem. 1936, 245, 168–170. [Google Scholar] [CrossRef]
  15. Copping, A.M. Origin of Vitamin D in Cod-Liver Oil: Vitamin D Content of Zooplankton. Biochem. J. 1934, 28, 1516–1520. [Google Scholar] [CrossRef] [PubMed]
  16. Holick, M.F.; Schnoes, H.K.; DeLuca, H.F.; Gray, R.W.; Boyle, I.T.; Suda, T. Isolation and Identification of 24,25-Dihydroxycholecalciferol, a Metabolite of Vitamin D3 Made in the Kidney. Biochemistry 1972, 11, 4251–4255. [Google Scholar] [CrossRef]
  17. Haussler, M.R.; Norman, A.W. Chromosomal Receptor for a Vitamin D Metabolite. Proc. Natl. Acad. Sci. USA 1969, 62, 155–162. [Google Scholar] [CrossRef]
  18. Norman, A.W.; Myrtle, J.F.; Miogett, R.J.; Nowicki, H.G.; Williams, V.; Popjaák, G. 1,25-Dihydroxycholecalciferol: Identification of the Proposed Active Form of Vitamin D3 in the Intestine. Science 1971, 173, 51–54. [Google Scholar] [CrossRef] [PubMed]
  19. Carlberg, C. Molecular Endocrinology of Vitamin D on the Epigenome Level. Mol. Cell. Endocrinol. 2017, 453, 14–21. [Google Scholar] [CrossRef]
  20. Grant, W.B. Epidemiology of Disease Risks in Relation to Vitamin D Insufficiency. Prog. Biophys. Mol. Biol. 2006, 92, 65–79. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, S. Epidemiology of Vitamin D in Health and Disease. Nutr. Res. Rev. 2009, 22, 188–203. [Google Scholar] [CrossRef] [PubMed]
  22. Nardin, M.; Verdoia, M.; Nardin, S.; Cao, D.; Chiarito, M.; Kedhi, E.; Galasso, G.; Condorelli, G.; De Luca, G. Vitamin D and Cardiovascular Diseases: From Physiology to Pathophysiology and Outcomes. Biomedicines 2024, 12, 768. [Google Scholar] [CrossRef] [PubMed]
  23. Cavalier, E.; Makris, K.; Heijboer, A.C.; Herrmann, M.; Souberbielle, J.-C. Vitamin D: Analytical Advances, Clinical Impact, and Ongoing Debates on Health Perspectives. Clin. Chem. 2024, 70, 1104–1121. [Google Scholar] [CrossRef] [PubMed]
  24. Fleet, J.C.; Shapses, S.A. Vitamin D. In Present Knowledge in Nutrition; Elsevier: Amsterdam, The Netherlands, 2020; pp. 93–114. ISBN 978-0-323-66162-1. [Google Scholar]
  25. Janjetovic, Z.; Slominski, A.T. Promising Functions of Novel Vitamin D Derivatives as Cosmetics: A New Fountain of Youth in Skin Aging and Skin Protection. Cosmetics 2024, 11, 37. [Google Scholar] [CrossRef]
  26. Bogh, M.K.B.; Schmedes, A.V.; Philipsen, P.A.; Thieden, E.; Wulf, H.C. Vitamin D Production after UVB Exposure Depends on Baseline Vitamin D and Total Cholesterol but Not on Skin Pigmentation. J. Investig. Dermatol. 2010, 130, 546–553. [Google Scholar] [CrossRef]
  27. Bocheva, G.; Slominski, R.M.; Slominski, A.T. The Impact of Vitamin D on Skin Aging. Int. J. Mol. Sci. 2021, 22, 9097. [Google Scholar] [CrossRef]
  28. Tuohimaa, P. Vitamin D and Aging. J. Steroid Biochem. Mol. Biol. 2009, 114, 78–84. [Google Scholar] [CrossRef] [PubMed]
  29. Courbebaisse, M.; Cavalier, E. Vitamin D in 2020: An Old Pro-Hormone with Potential Effects beyond Mineral Metabolism. Nutrients 2020, 12, 3378. [Google Scholar] [CrossRef]
  30. Adamantidi, T.; Maris, G.; Altantsidou, P.; Tsoupras, A. Anti-Inflammatory Benefits of Vitamin D and Its Analogues Against Glomerulosclerosis and Kidney Diseases. Sclerosis 2024, 2, 217–265. [Google Scholar] [CrossRef]
  31. Slominski, R.M.; Kim, T.-K.; Janjetovic, Z.; Brożyna, A.A.; Podgorska, E.; Dixon, K.M.; Mason, R.S.; Tuckey, R.C.; Sharma, R.; Crossman, D.K.; et al. Malignant Melanoma: An Overview, New Perspectives, and Vitamin D Signaling. Cancers 2024, 16, 2262. [Google Scholar] [CrossRef] [PubMed]
  32. Brożyna, A.A.; Hoffman, R.M.; Slominski, A.T. Relevance of Vitamin D in Melanoma Development, Progression and Therapy. Anticancer. Res. 2020, 40, 473–489. [Google Scholar] [CrossRef]
  33. Seraphin, G.; Rieger, S.; Hewison, M.; Capobianco, E.; Lisse, T.S. The Impact of Vitamin D on Cancer: A Mini Review. J. Steroid Biochem. Mol. Biol. 2023, 231, 106308. [Google Scholar] [CrossRef]
  34. Fantini, C.; Corinaldesi, C.; Lenzi, A.; Migliaccio, S.; Crescioli, C. Vitamin D as a Shield against Aging. Int. J. Mol. Sci. 2023, 24, 4546. [Google Scholar] [CrossRef] [PubMed]
  35. Wiseman, H. Vitamin D Is a Membrane Antioxidant Ability to Inhibit Iron-dependent Lipid Peroxidation in Liposomes Compared to Cholesterol, Ergosterol and Tamoxifen and Relevance to Anticancer Action. FEBS Lett. 1993, 326, 285–288. [Google Scholar] [CrossRef] [PubMed]
  36. 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] [PubMed]
  37. 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]
  38. 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]
  39. Van Der Schaft, J.; Ariens, L.F.M.; Bruijnzeel-Koomen, C.A.F.M.; De Bruin-Weller, M.S. Serum Vitamin D Status in Adult Patients with Atopic Dermatitis: Recommendations for Daily Practice. J. Am. Acad. Dermatol. 2016, 75, 1257–1259. [Google Scholar] [CrossRef] [PubMed]
  40. Sanmartin, R.; Pardos, C.; Doste, D.; Aguilera, J.; Alijarde, R.; Jesús Agón-Banzo, P.; García-Malinis, A.J.; Puzo, J.; Hernández-Martín, Á.; Gilaberte, Y. The Association between Atopic Dermatitis and Serum 25-hydroxyvitamin D in Children: Influence of Sun Exposure, Diet, and Atopy Features—A Cross-sectional Study. Pediatr. Dermatol. 2020, 37, 294–300. [Google Scholar] [CrossRef]
  41. Silverberg, J.I. Public Health Burden and Epidemiology of Atopic Dermatitis. Dermatol. Clin. 2017, 35, 283–289. [Google Scholar] [CrossRef] [PubMed]
  42. Taieb, A.; Alomar, A.; Böhm, M.; Dell’Anna, M.L.; De Pase, A.; Eleftheriadou, V.; Ezzedine, K.; Gauthier, Y.; Gawkrodger, D.J.; Jouary, T.; et al. Guidelines for the Management of Vitiligo: The European Dermatology Forum Consensus: EDF Vitiligo Guidelines. Br. J. Dermatol. 2013, 168, 5–19. [Google Scholar] [CrossRef] [PubMed]
  43. Karagüzel, G.; Sakarya, N.P.; Bahadır, S.; Yaman, S.; Ökten, A. Vitamin D Status and the Effects of Oral Vitamin D Treatment in Children with Vitiligo: A Prospective Study. Clin. Nutr. ESPEN 2016, 15, 28–31. [Google Scholar] [CrossRef]
  44. Agak, G.W.; Qin, M.; Nobe, J.; Kim, M.-H.; Krutzik, S.R.; Tristan, G.R.; Elashoff, D.; Garbán, H.J.; Kim, J. Propionibacterium Acnes Induces an IL-17 Response in Acne Vulgaris That Is Regulated by Vitamin A and Vitamin D. J. Investig. Dermatol. 2014, 134, 366–373. [Google Scholar] [CrossRef]
  45. Alhetheli, G.; Elneam, A.I.A.; Alsenaid, A.; Al-Dhubaibi, M. Vitamin D Levels in Patients with and without Acne and Its Relation to Acne Severity: A Case-Control Study. Clin. Cosmet. Investig. Dermatol. 2020, 13, 759–765. [Google Scholar] [CrossRef] [PubMed]
  46. Iqbal, T.; Asim, S.A.; Bhatti, S.; Sajid, M.; Mirza, R.; Huma, Z. Association of Vitamin D with Moderate to Severe Acne Vulgaris. J. Coll. Physicians Surg. Pak. 2023, 33, 527–530. [Google Scholar] [CrossRef] [PubMed]
  47. Algarin, Y.A.; Pulumati, A.; Jaalouk, D.; Tan, J.; Nouri, K. The Role of Vitamins and Nutrients in Rosacea. Arch. Dermatol. Res. 2024, 316, 142. [Google Scholar] [CrossRef] [PubMed]
  48. Kutner, A.; Brown, G. Vitamins D: Relationship between Structure and Biological Activity. Int. J. Mol. Sci. 2018, 19, 2119. [Google Scholar] [CrossRef]
  49. Chiellini, G.; DeLuca, H.F. The Importance of Stereochemistry on the Actions of Vitamin D. Curr. Top. Med. Chem. 2011, 11, 840–859. [Google Scholar] [CrossRef] [PubMed]
  50. DeLuca, H.F. Overview of General Physiologic Features and Functions of Vitamin D. Am. J. Clin. Nutr. 2004, 80, 1689S–1696S. [Google Scholar] [CrossRef] [PubMed]
  51. Jasinghe, V.J.; Perera, C.O. Distribution of Ergosterol in Different Tissues of Mushrooms and Its Effect on the Conversion of Ergosterol to Vitamin D2 by UV Irradiation. Food Chem. 2005, 92, 541–546. [Google Scholar] [CrossRef]
  52. Benedik, E. Sources of Vitamin D for Humans. Int. J. Vitam. Nutr. Res. 2022, 92, 118–125. [Google Scholar] [CrossRef] [PubMed]
  53. Bikle, D.D. Vitamin D and the Skin. J. Bone Miner. Metab. 2010, 28, 117–130. [Google Scholar] [CrossRef] [PubMed]
  54. Al-Smadi, K.; Ali, M.; Alavi, S.E.; Jin, X.; Imran, M.; Leite-Silva, V.R.; Mohammed, Y. Using a Topical Formulation of Vitamin D for the Treatment of Vitiligo: A Systematic Review. Cells 2023, 12, 2387. [Google Scholar] [CrossRef]
  55. Pilz, S.; Putz-Bankuti, C.; Gaksch, M.; Spindelboeck, W.; Haselberger, M.; Rainer, F.; Posch, A.; Kreuzer, P.; Stojakovic, T.; Stadlbauer, V.; et al. Effects of Vitamin D Supplementation on Serum 25-Hydroxyvitamin D Concentrations in Cirrhotic Patients: A Randomized Controlled Trial. Nutrients 2016, 8, 278. [Google Scholar] [CrossRef] [PubMed]
  56. Ponti, L.; Gabutti, L.; Faré, P.B.; Janett, S.; Bianchetti, M.G.; Schulz, P.J.; Lava, S.A.G.; Agostoni, C.; Milani, G.P. Vitamin D Supply of Multivitamins Commercialized Online by Amazon in Western and Southern Europe: A Labeling Analysis. Nutrients 2023, 15, 326. [Google Scholar] [CrossRef] [PubMed]
  57. Stanescu, A.M.A.; Simionescu, A.A.; Diaconu, C.C. Oral Vitamin D Therapy in Patients with Psoriasis. Nutrients 2021, 13, 163. [Google Scholar] [CrossRef]
  58. Tóth, B.E.; Takács, I.; Kádár, K.; Mirani, S.; Vecsernyés, M.; Lakatos, P. Safety and Efficacy of Loading Doses of Vitamin D: Recommendations for Effective Repletion. Pharmaceuticals 2024, 17, 1620. [Google Scholar] [CrossRef]
  59. Iwanowski, T.; Kołkowski, K.; Nowicki, R.J.; Sokołowska-Wojdyło, M. Etiopathogenesis and Emerging Methods for Treatment of Vitiligo. Int. J. Mol. Sci. 2023, 24, 9749. [Google Scholar] [CrossRef]
  60. Solnier, J.; Chang, C.; Zhang, Y.; Kuo, Y.C.; Du, M.; Roh, Y.S.; See, J.; Brix, J.; Gahler, R.J.; Green, T.; et al. A Comparison and Safety Evaluation of Micellar versus Standard Vitamin D3 Oral Supplementation in a Randomized, Double-Blind Human Pilot Study. Nutrients 2024, 16, 1573. [Google Scholar] [CrossRef]
  61. Galluccio, G.; D’Onghia, M.; Malvaso, D.; Lazzeri, L.; Cinotti, E.; Rubegni, G.; Rubegni, P.; Calabrese, L. Advances in the Pathogenesis and Treatment of Rosacea: A Phenotype-Based Therapeutic Approach. Cosmetics 2024, 11, 11. [Google Scholar] [CrossRef]
  62. Rusic, D.; Ivic, M.; Slugan, A.; Leskur, D.; Modun, D.; Durdov, T.; Vukovic, D.; Bukic, J.; Bozic, J.; Seselja Perisin, A. Pilot Study on the Effects of a Cosmetic Serum Containing Niacinamide, Postbiotics and Peptides on Facial Skin in Healthy Participants: A Randomized Controlled Trial. Life 2024, 14, 1677. [Google Scholar] [CrossRef]
  63. Santa, K.; Kumazawa, Y.; Watanabe, K.; Nagaoka, I. The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects. Int. J. Mol. Sci. 2024, 25, 2125. [Google Scholar] [CrossRef] [PubMed]
  64. Glowka, E.; Stasiak, J.; Lulek, J. Drug Delivery Systems for Vitamin D Supplementation and Therapy. Pharmaceutics 2019, 11, 347. [Google Scholar] [CrossRef] [PubMed]
  65. Aggeletopoulou, I.; Kalafateli, M.; Geramoutsos, G.; Triantos, C. Recent Advances in the Use of Vitamin D Organic Nanocarriers for Drug Delivery. Biomolecules 2024, 14, 1090. [Google Scholar] [CrossRef]
  66. Mazur, A.; Koziorowska, K.; Dynarowicz, K.; Aebisher, D.; Bartusik-Aebisher, D. Vitamin D and Vitamin D3 Supplementation during Photodynamic Therapy: A Review. Nutrients 2022, 14, 3805. [Google Scholar] [CrossRef]
  67. McCullough, P.J.; McCullough, W.P.; Lehrer, D.; Travers, J.B.; Repas, S.J. Oral and Topical Vitamin D, Sunshine, and UVB Phototherapy Safely Control Psoriasis in Patients with Normal Pretreatment Serum 25-Hydroxyvitamin D Concentrations: A Literature Review and Discussion of Health Implications. Nutrients 2021, 13, 1511. [Google Scholar] [CrossRef] [PubMed]
  68. Kittaneh, M.; Qurt, M.; Malkieh, N.; Naseef, H.; Muqedi, R. Preparation and Evaluation of Vitamin D3 Supplementation as Transdermal Film-Forming Solution. Pharmaceutics 2022, 15, 39. [Google Scholar] [CrossRef] [PubMed]
  69. Vasdeki, D.; Tsamos, G.; Dimakakos, E.; Patriarcheas, 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]
  70. Yan, X.; Lu, E.; Song, Z.; Wu, Y.; Sha, X. Development and In Vivo Evaluation of a Novel Vitamin D3 Oral Spray Delivery System. Pharmaceutics 2023, 16, 25. [Google Scholar] [CrossRef]
  71. Li, C.-P.; Huang, S.-C.; Hsiao, Y.; Tsai, R.-Y. Evaluating the Role of Vitamin D in Alleviating Chronic Pruritus: A Meta-Analysis. Int. J. Mol. Sci. 2024, 25, 9983. [Google Scholar] [CrossRef] [PubMed]
  72. Silva, N.D.C.; Silva, C.J.T.; Gonçalves, M.P.; Borsagli, F.G.L.M. Carboxymethyl-Cellulose-Based Hydrogels Incorporated with Cellulose Nanocrystals Loaded with Vitamin D for Controlled Drug Delivery. Processes 2024, 12, 1437. [Google Scholar] [CrossRef]
  73. Holick, M.F. Vitamin D Deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  74. Bikle, D.D. Vitamin D Metabolism, Mechanism of Action, and Clinical Applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef] [PubMed]
  75. Bikle, D.D. Vitamin D and the Skin: Physiology and Pathophysiology. Rev. Endocr. Metab. Disord. 2012, 13, 3–19. [Google Scholar] [CrossRef]
  76. Morimoto, S.; Kumahara, Y. A Patient with Psoriasis Cured by 1 Alpha-Hydroxyvitamin D3. Med. J. Osaka Univ. 1985, 35, 51–54. [Google Scholar] [PubMed]
  77. Ingram, M.A.; Jones, M.B.; Stonehouse, W.; Jarrett, P.; Scragg, R.; Mugridge, O.; Von Hurst, P.R. Oral Vitamin D3 Supplementation for Chronic Plaque Psoriasis: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Dermatol. Treat. 2018, 29, 648–657. [Google Scholar] [CrossRef] [PubMed]
  78. Lebwohl, M.; Yoles, A.; Lombardi, K.; Lou, W. Calcipotriene Ointment and Halobetasol Ointment in the Long-Term Treatment of Psoriasis: Effects on the Duration of Improvement. J. Am. Acad. Dermatol. 1998, 39, 447–450. [Google Scholar] [CrossRef] [PubMed]
  79. Jalili, A.; Lebwohl, M.; Stein Gold, L.; Andersen, S.B.; Jensen, K.L.; Pink, A.E.; Segaert, S.; Berg, P.; Calzavara-Pinton, P.G.; De La Cueva Dobao, P.; et al. Itch Relief in Patients with Psoriasis: Effectiveness of Calcipotriol plus Betamethasone Dipropionate Foam. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 709–717. [Google Scholar] [CrossRef] [PubMed]
  80. Del Rosso, J.; Friedlander, S.F. Corticosteroids: Options in the Era of Steroid-Sparing Therapy. J. Am. Acad. Dermatol. 2005, 53, S50–S58. [Google Scholar] [CrossRef]
  81. Alia, E.; Kerr, P.E. Vitamin D: Skin, Sunshine, and Beyond. Clin. Dermatol. 2021, 39, 840–846. [Google Scholar] [CrossRef] [PubMed]
  82. Wasiewicz, T.; Szyszka, P.; Cichorek, M.; Janjetovic, Z.; Tuckey, R.; Slominski, A.; Zmijewski, M. Antitumor Effects of Vitamin D Analogs on Hamster and Mouse Melanoma Cell Lines in Relation to Melanin Pigmentation. Int. J. Mol. Sci. 2015, 16, 6645–6667. [Google Scholar] [CrossRef] [PubMed]
  83. Indra, A.K.; Castaneda, E.; Antal, M.C.; Jiang, M.; Messaddeq, N.; Meng, X.; Loehr, C.V.; Gariglio, P.; Kato, S.; Wahli, W.; et al. Malignant Transformation of DMBA/TPA-Induced Papillomas and Nevi in the Skin of Mice Selectively Lacking Retinoid-X-Receptor α in Epidermal Keratinocytes. J. Investig. Dermatol. 2007, 127, 1250–1260. [Google Scholar] [CrossRef] [PubMed]
  84. Janjetovic, Z.; Brozyna, A.A.; Tuckey, R.C.; Kim, T.-K.; Nguyen, M.N.; Jozwicki, W.; Pfeffer, S.R.; Pfeffer, L.M.; Slominski, A.T. High Basal NF-κB Activity in Nonpigmented Melanoma Cells Is Associated with an Enhanced Sensitivity to Vitamin D3 Derivatives. Br. J. Cancer 2011, 105, 1874–1884. [Google Scholar] [CrossRef] [PubMed]
  85. Cunningham, T.J.; Tabacchi, M.; Eliane, J.-P.; Tuchayi, S.M.; Manivasagam, S.; Mirzaalian, H.; Turkoz, A.; Kopan, R.; Schaffer, A.; Saavedra, A.P.; et al. Randomized Trial of Calcipotriol Combined with 5-Fluorouracil for Skin Cancer Precursor Immunotherapy. J. Clin. Investig. 2016, 127, 106–116. [Google Scholar] [CrossRef] [PubMed]
  86. Slominski, A.T.; Janjetovic, Z.; Kim, T.-K.; Wasilewski, P.; Rosas, S.; Hanna, S.; Sayre, R.M.; Dowdy, J.C.; Li, W.; Tuckey, R.C. Novel Non-Calcemic Secosteroids That Are Produced by Human Epidermal Keratinocytes Protect against Solar Radiation. J. Steroid Biochem. Mol. Biol. 2015, 148, 52–63. [Google Scholar] [CrossRef]
  87. Goltzman, D. Functions of Vitamin D in Bone. Histochem. Cell Biol. 2018, 149, 305–312. [Google Scholar] [CrossRef]
  88. Kulling, P.M.; Olson, K.C.; Olson, T.L.; Feith, D.J.; Loughran, T.P. Vitamin D in Hematological Disorders and Malignancies. Eur. J. Haematol. 2017, 98, 187–197. [Google Scholar] [CrossRef] [PubMed]
  89. Mostafa, W.Z.; Hegazy, R.A. Vitamin D and the Skin: Focus on a Complex Relationship: A Review. J. Adv. Res. 2015, 6, 793–804. [Google Scholar] [CrossRef] [PubMed]
  90. Oda, Y.; Uchida, Y.; Moradian, S.; Crumrine, D.; Elias, P.M.; Bikle, D.D. Vitamin D Receptor and Coactivators SRC2 and 3 Regulate Epidermis-Specific Sphingolipid Production and Permeability Barrier Formation. J. Investig. Dermatol. 2009, 129, 1367–1378. [Google Scholar] [CrossRef] [PubMed]
  91. Hawker, N.P.; Pennypacker, S.D.; Chang, S.M.; Bikle, D.D. Regulation of Human Epidermal Keratinocyte Differentiation by the Vitamin D Receptor and Its Coactivators DRIP205, SRC2, and SRC3. J. Investig. Dermatol. 2007, 127, 874–880. [Google Scholar] [CrossRef] [PubMed]
  92. Bikle, D.D.; Pillai, S. Vitamin D, Calcium, and Epidermal Differentiation. Endocr. Rev. 1993, 14, 3–19. [Google Scholar] [CrossRef] [PubMed]
  93. Gisondi, P.; Rossini, M.; Di Cesare, A.; Idolazzi, L.; Farina, S.; Beltrami, G.; Peris, K.; Girolomoni, G. Vitamin D Status in Patients with Chronic Plaque Psoriasis: Vitamin D Serum Levels in Patients with Psoriasis. Br. J. Dermatol. 2012, 166, 505–510. [Google Scholar] [CrossRef] [PubMed]
  94. Smith, K.; Hewlings, S. Correlation between Vitamin D Levels and Hard-to-Heal Wounds: A Systematic Review. J. Wound Care 2021, 30, S4–S10. [Google Scholar] [CrossRef] [PubMed]
  95. Heimbeck, I.; Wjst, M.; Apfelbacher, C.J. Low Vitamin D Serum Level Is Inversely Associated with Eczema in Children and Adolescents in Germany. Allergy 2013, 68, 906–910. [Google Scholar] [CrossRef] [PubMed]
  96. Mutgi, K.; Koo, J. Update on the Role of Systemic Vitamin D in Atopic Dermatitis. Pediatr. Dermatol. 2013, 30, 303–307. [Google Scholar] [CrossRef] [PubMed]
  97. Galior, K.; Grebe, S.; Singh, R. Development of Vitamin D Toxicity from Overcorrection of Vitamin D Deficiency: A Review of Case Reports. Nutrients 2018, 10, 953. [Google Scholar] [CrossRef] [PubMed]
  98. Letavernier, E.; Daudon, M. Vitamin D, Hypercalciuria and Kidney Stones. Nutrients 2018, 10, 366. [Google Scholar] [CrossRef]
  99. Messa, P.; Castellano, G.; Vettoretti, S.; Alfieri, C.M.; Giannese, D.; Panichi, V.; Cupisti, A. Vitamin D and Calcium Supplementation and Urolithiasis: A Controversial and Multifaceted Relationship. Nutrients 2023, 15, 1724. [Google Scholar] [CrossRef] [PubMed]
  100. Hu, H.; Zhang, J.; Lu, Y.; Zhang, Z.; Qin, B.; Gao, H.; Wang, Y.; Zhu, J.; Wang, Q.; Zhu, Y.; et al. Association between Circulating Vitamin D Level and Urolithiasis: A Systematic Review and Meta-Analysis. Nutrients 2017, 9, 301. [Google Scholar] [CrossRef]
  101. Garunkstiene, R.; Levuliene, R.; Cekuolis, A.; Cerkauskiene, R.; Drazdiene, N.; Liubsys, A. A Prospective Study of Nephrocalcinosis in Very Preterm Infants: Incidence, Risk Factors and Vitamin D Intake in the First Month. Medicina 2024, 60, 1910. [Google Scholar] [CrossRef]
  102. Gwadera, Ł.; Białas, A.J.; Kumor-Kisielewska, A.; Miłkowska-Dymanowska, J.; Majewski, S.; Piotrowski, W.J. Calcium, Phosphate, and Vitamin D Status in Patients with Sarcoidosis—Associations with Disease Activity and Symptoms. J. Clin. Med. 2023, 12, 4745. [Google Scholar] [CrossRef] [PubMed]
  103. Brandenburg, V.; Ketteler, M. Vitamin D and Secondary Hyperparathyroidism in Chronic Kidney Disease: A Critical Appraisal of the Past, Present, and the Future. Nutrients 2022, 14, 3009. [Google Scholar] [CrossRef] [PubMed]
  104. Bover, J.; Massó, E.; Gifre, L.; Alfieri, C.; Soler-Majoral, J.; Fusaro, M.; Calabia, J.; Rodríguez-Pena, R.; Rodríguez-Chitiva, N.; López-Báez, V.; et al. Vitamin D and Chronic Kidney Disease Association with Mineral and Bone Disorder: An Appraisal of Tangled Guidelines. Nutrients 2023, 15, 1576. [Google Scholar] [CrossRef] [PubMed]
  105. Mamadapur, V.K.; Nagaraju, S.; Prabhu, M.M. Comparative Study of Vitamin D Levels in Newly Diagnosed Tuberculosis and a Normal Population. Medicina 2024, 60, 685. [Google Scholar] [CrossRef] [PubMed]
  106. Jensen, N.S.; Wehland, M.; Wise, P.M.; Grimm, D. Latest Knowledge on the Role of Vitamin D in Hypertension. Int. J. Mol. Sci. 2023, 24, 4679. [Google Scholar] [CrossRef]
  107. Dallavalasa, S.; Tulimilli, S.V.; Bettada, V.G.; Karnik, M.; Uthaiah, C.A.; Anantharaju, P.G.; Nataraj, S.M.; Ramashetty, R.; Sukocheva, O.A.; Tse, E.; et al. Vitamin D in Cancer Prevention and Treatment: A Review of Epidemiological, Preclinical, and Cellular Studies. Cancers 2024, 16, 3211. [Google Scholar] [CrossRef] [PubMed]
  108. Phiri, C.B.; Davis, C.R.; Grahn, M.; Gannon, B.M.; Kokinos, B.P.; Crenshaw, T.D.; Tanumihardjo, S.A. Vitamin D Maintains Growth and Bone Mineral Density against a Background of Severe Vitamin A Deficiency and Moderate Toxicity in a Swine Model. Nutrients 2024, 16, 2037. [Google Scholar] [CrossRef] [PubMed]
  109. Wintermeyer, E.; Ihle, C.; Ehnert, S.; Stöckle, U.; Ochs, G.; De Zwart, P.; Flesch, I.; Bahrs, C.; Nussler, A. Crucial Role of Vitamin D in the Musculoskeletal System. Nutrients 2016, 8, 319. [Google Scholar] [CrossRef]
  110. Tallon, E.; Macedo, J.P.; Faria, A.; Tallon, J.M.; Pinto, M.; Pereira, J. Can Vitamin D Levels Influence Bone Metabolism and Osseointegration of Dental Implants? An Umbrella Review. Healthcare 2024, 12, 1867. [Google Scholar] [CrossRef]
  111. Cui, X.; Eyles, D.W. Vitamin D and the Central Nervous System: Causative and Preventative Mechanisms in Brain Disorders. Nutrients 2022, 14, 4353. [Google Scholar] [CrossRef] [PubMed]
  112. Hwa, C.; Bauer, E.A.; Cohen, D.E. Skin Biology: Systems That Enhance Drug Delivery—Skin Biology. Dermatol. Ther. 2011, 24, 464–470. [Google Scholar] [CrossRef] [PubMed]
  113. Gruber, F.; Kremslehner, C.; Eckhart, L.; Tschachler, E. Cell Aging and Cellular Senescence in Skin Aging—Recent Advances in Fibroblast and Keratinocyte Biology. Exp. Gerontol. 2020, 130, 110780. [Google Scholar] [CrossRef] [PubMed]
  114. Woo, W. Skin Structure and Biology. In Imaging Technologies and Transdermal Delivery in Skin Disorders; Xu, C., Wang, X., Pramanik, M., Eds.; Wiley: Hoboken, NJ, USA, 2019; pp. 1–14. ISBN 978-3-527-34460-4. [Google Scholar]
  115. Proksch, E.; Brandner, J.M.; Jensen, J. The Skin: An Indispensable Barrier. Exp. Dermatol. 2008, 17, 1063–1072. [Google Scholar] [CrossRef]
  116. Ibrahim, A.A.E.; Bagherani, N.; Smoller, B.R.; Reyes-Baron, C.; Bagherani, N. Functions of the Skin. In Atlas of Dermatology, Dermatopathology and Venereology; Smoller, B., Bagherani, N., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–11. ISBN 978-3-319-45134-3. [Google Scholar]
  117. Grice, E.A.; Segre, J.A. The Skin Microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef] [PubMed]
  118. Kubo, A.; Ishizaki, I.; Kubo, A.; Kawasaki, H.; Nagao, K.; Ohashi, Y.; Amagai, M. The Stratum Corneum Comprises Three Layers with Distinct Metal-Ion Barrier Properties. Sci. Rep. 2013, 3, 1731. [Google Scholar] [CrossRef] [PubMed]
  119. Nguyen, A.V.; Soulika, A.M. The Dynamics of the Skin’s Immune System. Int. J. Mol. Sci. 2019, 20, 1811. [Google Scholar] [CrossRef] [PubMed]
  120. Park, J.-H.; Lee, J.-W.; Kim, Y.-C.; Prausnitz, M.R. The Effect of Heat on Skin Permeability. Int. J. Pharm. 2008, 359, 94–103. [Google Scholar] [CrossRef]
  121. Relhan, V.; Goel, K.; Kochhar, A.; Garg, V.; Wadhwa, B. Vitamin D and Skin Diseases: A Review. Indian J. Dermatol. Venereol. Leprol. 2015, 81, 344. [Google Scholar] [CrossRef] [PubMed]
  122. Bikle, D.D. The Role of the Vitamin D Receptor in the Epidermal Stem Cell Response to Wounding. Receptors 2024, 3, 397–407. [Google Scholar] [CrossRef]
  123. Veleva, B.I.; Caljouw, M.A.A.; Van Der Steen, J.T.; Mertens, B.J.A.; Chel, V.G.M.; Numans, M.E. The Effect of Ultraviolet B Irradiation Compared with Oral Vitamin D Supplementation on the Well-Being of Nursing Home Residents with Dementia: A Randomized Controlled Trial. Int. J. Environ. Res. Public Health 2020, 17, 1684. [Google Scholar] [CrossRef]
  124. Lehmann, B.; Querings, K.; Reichrath, J. Vitamin D and Skin: New Aspects for Dermatology. Exp. Dermatol. 2004, 13, 11–15. [Google Scholar] [CrossRef] [PubMed]
  125. Mosca, S.; Morrone, A. Human Skin Pigmentation: From a Biological Feature to a Social Determinant. Healthcare 2023, 11, 2091. [Google Scholar] [CrossRef] [PubMed]
  126. Vearing, R.M.; Hart, K.H.; Charlton, K.; Probst, Y.; Blackbourn, D.J.; Ahmadi, K.R.; Lanham-New, S.A.; Darling, A.L. Vitamin D Status of the British African-Caribbean Residents: Analysis of the UK Biobank Cohort. Nutrients 2021, 13, 4104. [Google Scholar] [CrossRef] [PubMed]
  127. Feketea, G.M.; Bocsan, I.C.; Tsiros, G.; Voila, P.; Stanciu, L.A.; Zdrenghea, M. Vitamin D Status in Children in Greece and Its Relationship with Sunscreen Application. Children 2021, 8, 111. [Google Scholar] [CrossRef]
  128. Herdea, A.; Marie, H.; Ionescu, A.; Sandu, D.-M.; Pribeagu, S.-T.; Ulici, A. Vitamin D Deficiency—A Public Health Issue in Children. Children 2024, 11, 1061. [Google Scholar] [CrossRef] [PubMed]
  129. Bikle, D.D. Vitamin D Metabolism and Function in the Skin. Mol. Cell. Endocrinol. 2011, 347, 80–89. [Google Scholar] [CrossRef] [PubMed]
  130. Letsiou, S.; Koldiri, E.; Beloukas, A.; Rallis, E.; Kefala, V. Deciphering the Effects of Different Types of Sunlight Radiation on Skin Function: A Review. Cosmetics 2024, 11, 80. [Google Scholar] [CrossRef]
  131. Dickerson, R.N.; Turner, S.C.; Holmes, W.L.; Van Matre, E.T.; Swanson, J.M.; Byerly, S.; Filiberto, D.M.; Fischer, P.E. Reduction in Hypercalcemia Following Readjustment of Target Serum 25-Hydroxy Vitamin D Concentration during Cholecalciferol Therapy in Vitamin D-Deficient Critically Ill Patients. Nutrients 2022, 14, 1650. [Google Scholar] [CrossRef] [PubMed]
  132. Sivagurunathan, U.; Izquierdo, M.; Tseng, Y.; Prabhu, P.A.J.; Zamorano, M.J.; Robaina, L.; Domínguez, D. Effects of the Interaction between Dietary Vitamin D3 and Vitamin K3 on Growth, Skeletal Anomalies, and Expression of Bone and Calcium Metabolism-Related Genes in Juvenile Gilthead Seabream (Sparus Aurata). Animals 2024, 14, 2808. [Google Scholar] [CrossRef]
  133. Biasucci, G.; Donini, V.; Cannalire, G. Rickets Types and Treatment with Vitamin D and Analogues. Nutrients 2024, 16, 416. [Google Scholar] [CrossRef] [PubMed]
  134. Fronczek, M.; Strzelczyk, J.K.; Biernacki, K.; Salatino, S.; Osadnik, T.; Ostrowska, Z. New Variants of the Cytochrome P450 2R1 (CYP2R1) Gene in Individuals with Severe Vitamin D-Activating Enzyme 25(OH)D Deficiency. Biomolecules 2021, 11, 1867. [Google Scholar] [CrossRef]
  135. Toral López, J.; Candia Tenopala, C.; Reyes Mosqueda, A.D.; Fonseca Sánchez, M.Á.; González Huerta, L.M. A Novel Compound Nonsense Variant in CYP27B1 Causes an Atypical Form of Vitamin D-Dependent Rickets Type 1A: A Case Report of Two Siblings in a Mexican Family. Diseases 2024, 12, 248. [Google Scholar] [CrossRef] [PubMed]
  136. Bizerea-Moga, T.O.; Chisavu, F.; Ilies, C.; Olah, O.; Marginean, O.; Gafencu, M.; Doros, G.; Stroescu, R. Phenotype of Idiopathic Infantile Hypercalcemia Associated with the Heterozygous Pathogenic Variant of SLC34A1 and CYP24A1. Children 2023, 10, 1701. [Google Scholar] [CrossRef] [PubMed]
  137. 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] [PubMed]
  138. Ao, T.; Kikuta, J.; Ishii, M. The Effects of Vitamin D on Immune System and Inflammatory Diseases. Biomolecules 2021, 11, 1624. [Google Scholar] [CrossRef] [PubMed]
  139. Martens, P.-J.; Gysemans, C.; Verstuyf, A.; Mathieu, C. Vitamin D’s Effect on Immune Function. Nutrients 2020, 12, 1248. [Google Scholar] [CrossRef]
  140. Holick, M.F. Sunlight and Vitamin D for Bone Health and Prevention of Autoimmune Diseases, Cancers, and Cardiovascular Disease. Am. J. Clin. Nutr. 2004, 80, 1678S–1688S. [Google Scholar] [CrossRef] [PubMed]
  141. Webb, A.R. Who, What, Where and When—Influences on Cutaneous Vitamin D Synthesis. Prog. Biophys. Mol. Biol. 2006, 92, 17–25. [Google Scholar] [CrossRef]
  142. Kimlin, M.G. Geographic Location and Vitamin D Synthesis. Mol. Asp. Med. 2008, 29, 453–461. [Google Scholar] [CrossRef] [PubMed]
  143. Slominski, R.M.; Sarna, T.; Płonka, P.M.; Raman, C.; Brożyna, A.A.; Slominski, A.T. Melanoma, Melanin, and Melanogenesis: The Yin and Yang Relationship. Front. Oncol. 2022, 12, 842496. [Google Scholar] [CrossRef] [PubMed]
  144. Elder, D.E.; Bastian, B.C.; Cree, I.A.; Massi, D.; Scolyer, R.A. The 2018 World Health Organization Classification of Cutaneous, Mucosal, and Uveal Melanoma: Detailed Analysis of 9 Distinct Subtypes Defined by Their Evolutionary Pathway. Arch. Pathol. Lab. Med. 2020, 144, 500–522. [Google Scholar] [CrossRef] [PubMed]
  145. Long, G.V.; Swetter, S.M.; Menzies, A.M.; Gershenwald, J.E.; Scolyer, R.A. Cutaneous Melanoma. Lancet 2023, 402, 485–502. [Google Scholar] [CrossRef] [PubMed]
  146. Podgorska, E.; Kim, T.-K.; Janjetovic, Z.; Urbanska, K.; Tuckey, R.C.; Bae, S.; Slominski, A.T. Knocking out the Vitamin D Receptor Enhances Malignancy and Decreases Responsiveness to Vitamin D3 Hydroxyderivatives in Human Melanoma Cells. Cancers 2021, 13, 3111. [Google Scholar] [CrossRef]
  147. Shariev, A.; Painter, N.; Reeve, V.E.; Haass, N.K.; Rybchyn, M.S.; Ince, F.A.; Mason, R.S.; Dixon, K.M. PTEN: A Novel Target for Vitamin D in Melanoma. J. Steroid Biochem. Mol. Biol. 2022, 218, 106059. [Google Scholar] [CrossRef]
  148. Piotrowska, A.; Wierzbicka, J.; Kwiatkowska, K.; Chodyński, M.; Kutner, A.; Żmijewski, M.A. Antiproliferative Activity of Side-Chain Truncated Vitamin D Analogs (PRI-1203 and PRI-1204) against Human Malignant Melanoma Cell Lines. Eur. J. Pharmacol. 2020, 881, 173170. [Google Scholar] [CrossRef] [PubMed]
  149. Kleszczyński, K.; Bilska, B.; Stegemann, A.; Flis, D.J.; Ziolkowski, W.; Pyza, E.; Luger, T.A.; Reiter, R.J.; Böhm, M.; Slominski, A.T. Melatonin and Its Metabolites Ameliorate UVR-Induced Mitochondrial Oxidative Stress in Human MNT-1 Melanoma Cells. Int. J. Mol. Sci. 2018, 19, 3786. [Google Scholar] [CrossRef] [PubMed]
  150. Slominski, A.T.; Chaiprasongsuk, A.; Janjetovic, Z.; Kim, T.-K.; Stefan, J.; Slominski, R.M.; Hanumanthu, V.S.; Raman, C.; Qayyum, S.; Song, Y.; et al. Photoprotective Properties of Vitamin D and Lumisterol Hydroxyderivatives. Cell Biochem. Biophys. 2020, 78, 165–180. [Google Scholar] [CrossRef] [PubMed]
  151. Drigeard Desgarnier, M.-C.; Rochette, P.J. Enhancement of UVB-Induced DNA Damage Repair after a Chronic Low-Dose UVB Pre-Stimulation. DNA Repair 2018, 63, 56–62. [Google Scholar] [CrossRef]
  152. Raad, H.; Serrano-Sanchez, M.; Harfouche, G.; Mahfouf, W.; Bortolotto, D.; Bergeron, V.; Kasraian, Z.; Dousset, L.; Hosseini, M.; Taieb, A.; et al. NADPH Oxidase-1 Plays a Key Role in Keratinocyte Responses to UV Radiation and UVB-Induced Skin Carcinogenesis. J. Investig. Dermatol. 2017, 137, 1311–1321. [Google Scholar] [CrossRef] [PubMed]
  153. Jeayeng, S.; Wongkajornsilp, A.; Slominski, A.T.; Jirawatnotai, S.; Sampattavanich, S.; Panich, U. Nrf2 in Keratinocytes Modulates UVB-Induced DNA Damage and Apoptosis in Melanocytes through MAPK Signaling. Free Radic. Biol. Med. 2017, 108, 918–928. [Google Scholar] [CrossRef]
  154. Dixon, K.M.; Deo, S.S.; Norman, A.W.; Bishop, J.E.; Halliday, G.M.; Reeve, V.E.; Mason, R.S. In Vivo Relevance for Photoprotection by the Vitamin D Rapid Response Pathway. J. Steroid Biochem. Mol. Biol. 2007, 103, 451–456. [Google Scholar] [CrossRef]
  155. Mason, R.S.; Sequeira, V.B.; Dixon, K.M.; Gordon-Thomson, C.; Pobre, K.; Dilley, A.; Mizwicki, M.T.; Norman, A.W.; Feldman, D.; Halliday, G.M.; et al. Photoprotection by 1α,25-Dihydroxyvitamin D and Analogs: Further Studies on Mechanisms and Implications for UV-Damage. J. Steroid Biochem. Mol. Biol. 2010, 121, 164–168. [Google Scholar] [CrossRef] [PubMed]
  156. Scott, J.F.; Das, L.M.; Ahsanuddin, S.; Qiu, Y.; Binko, A.M.; Traylor, Z.P.; Debanne, S.M.; Cooper, K.D.; Boxer, R.; Lu, K.Q. Oral Vitamin D Rapidly Attenuates Inflammation from Sunburn: An Interventional Study. J. Investig. Dermatol. 2017, 137, 2078–2086. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, D.; Lin, L.; Lei, K.; Zeng, J.; Luo, J.; Yin, Y.; Li, Y.; Zhang, L.; Nie, X.; Zuo, D.; et al. Vitamin D3 Analogue Facilitates Epithelial Wound Healing through Promoting Epithelial-Mesenchymal Transition via the Hippo Pathway. J. Dermatol. Sci. 2020, 100, 120–128. [Google Scholar] [CrossRef] [PubMed]
  158. Lee, J.; Zhao, H.; Fenta, Y.; Kita, H.; Kumar, R.; Juhn, Y.J. Serum 25-Hydroxyvitamin D Is Associated with Enhanced Pneumococcal Antibody Levels in Individuals with Asthma. Allergy Asthma Proc. 2011, 32, 445–452. [Google Scholar] [CrossRef] [PubMed]
  159. Haensel, D.; Dai, X. Epithelial-to-mesenchymal Transition in Cutaneous Wound Healing: Where We Are and Where We Are Heading. Dev. Dyn. 2018, 247, 473–480. [Google Scholar] [CrossRef] [PubMed]
  160. Thiery, J.P.; Acloque, H.; Huang, R.Y.J.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef] [PubMed]
  161. Behm, B.; Babilas, P.; Landthaler, M.; Schreml, S. Cytokines, Chemokines and Growth Factors in Wound Healing. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 812–820. [Google Scholar] [CrossRef] [PubMed]
  162. Bikle, D.D. Role of Vitamin D and Calcium Signaling in Epidermal Wound Healing. J. Endocrinol. Investig. 2022, 46, 205–212. [Google Scholar] [CrossRef]
  163. 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] [PubMed]
  164. Ismailova, A.; White, J.H. Vitamin D, Infections and Immunity. Rev. Endocr. Metab. Disord. 2022, 23, 265–277. [Google Scholar] [CrossRef]
  165. Hudson, L.; Begg, M.; Wright, B.; Cheek, T.; Jahoda, C.A.B.; Reynolds, N.J. Dominant Effect of Gap Junction Communication in Wound-induced Calcium-wave, NFAT Activation and Wound Closure in Keratinocytes. J. Cell. Physiol. 2021, 236, 8171–8183. [Google Scholar] [CrossRef]
  166. Oda, Y.; Hu, L.; Nguyen, T.; Fong, C.; Tu, C.; Bikle, D.D. Combined Deletion of the Vitamin D Receptor and Calcium-Sensing Receptor Delays Wound Re-Epithelialization. Endocrinology 2017, 158, 1929–1938. [Google Scholar] [CrossRef] [PubMed]
  167. Umar, M.; Sastry, K.S.; Al Ali, F.; Al-Khulaifi, M.; Wang, E.; Chouchane, A.I. Vitamin D and the Pathophysiology of Inflammatory Skin Diseases. Skin Pharmacol. Physiol. 2018, 31, 74–86. [Google Scholar] [CrossRef] [PubMed]
  168. Berridge, M.J. Vitamin D, Reactive Oxygen Species and Calcium Signalling in Ageing and Disease. Philos. Trans. R. Soc. B Biol. Sci. 2016, 371, 20150434. [Google Scholar] [CrossRef] [PubMed]
  169. Vázquez-Lorente, H.; Herrera-Quintana, L.; Jiménez-Sánchez, L.; Fernández-Perea, B.; Plaza-Diaz, J. Antioxidant Functions of Vitamin D and CYP11A1-Derived Vitamin D, Tachysterol, and Lumisterol Metabolites: Mechanisms, Clinical Implications, and Future Directions. Antioxidants 2024, 13, 996. [Google Scholar] [CrossRef] [PubMed]
  170. Parcheta, M.; Świsłocka, R.; Orzechowska, S.; Akimowicz, M.; Choińska, R.; Lewandowski, W. Recent Developments in Effective Antioxidants: The Structure and Antioxidant Properties. Materials 2021, 14, 1984. [Google Scholar] [CrossRef]
  171. Nair-Shalliker, V.; Armstrong, B.K.; Fenech, M. Does Vitamin D Protect against DNA Damage? Mutat. Res. Mol. Mech. Mutagen. 2012, 733, 50–57. [Google Scholar] [CrossRef]
  172. L Bishop, E.; Ismailova, A.; Dimeloe, S.; Hewison, M.; White, J.H. Vitamin D and Immune Regulation: Antibacterial, Antiviral, Anti-Inflammatory. JBMR Plus 2021, 5, e10405. [Google Scholar] [CrossRef] [PubMed]
  173. Verouti, S.N.; Tsoupras, A.B.; Alevizopoulou, F.; Demopoulos, C.A.; Iatrou, C. Paricalcitol Effects on Activities and Metabolism of Platelet Activating Factor and on Inflammatory Cytokines in Hemodialysis Patients. Int. J. Artif. Organs 2013, 36, 87–96. [Google Scholar] [CrossRef]
  174. Schauber, J.; Gallo, R.L. Antimicrobial Peptides and the Skin Immune Defense System. J. Allergy Clin. Immunol. 2008, 122, 261–266. [Google Scholar] [CrossRef] [PubMed]
  175. Farage, M.A.; Miller, K.W.; Elsner, P.; Maibach, H.I. Intrinsic and Extrinsic Factors in Skin Ageing: A Review. Int. J. Cosmet. Sci. 2008, 30, 87–95. [Google Scholar] [CrossRef] [PubMed]
  176. Bolke, L.; Schlippe, G.; Gerß, J.; Voss, W. A Collagen Supplement Improves Skin Hydration, Elasticity, Roughness, and Density: Results of a Randomized, Placebo-Controlled, Blind Study. Nutrients 2019, 11, 2494. [Google Scholar] [CrossRef] [PubMed]
  177. Romano, F.; Serpico, D.; Cantelli, M.; Di Sarno, A.; Dalia, C.; Arianna, R.; Lavorgna, M.; Colao, A.; Di Somma, C. Osteoporosis and Dermatoporosis: A Review on the Role of Vitamin D. Front. Endocrinol. 2023, 14, 1231580. [Google Scholar] [CrossRef]
  178. Cole, M.A.; Quan, T.; Voorhees, J.J.; Fisher, G.J. Extracellular Matrix Regulation of Fibroblast Function: Redefining Our Perspective on Skin Aging. J. Cell Commun. Signal. 2018, 12, 35–43. [Google Scholar] [CrossRef] [PubMed]
  179. Arseni, L.; Lombardi, A.; Orioli, D. From Structure to Phenotype: Impact of Collagen Alterations on Human Health. Int. J. Mol. Sci. 2018, 19, 1407. [Google Scholar] [CrossRef] [PubMed]
  180. Sosa-Díaz, E.; Hernández-Cruz, E.Y.; Pedraza-Chaverri, J. The Role of Vitamin D on Redox Regulation and Cellular Senescence. Free Radic. Biol. Med. 2022, 193, 253–273. [Google Scholar] [CrossRef]
  181. Choudhary, V.; Choudhary, M.; Bollag, W.B. Exploring Skin Wound Healing Models and the Impact of Natural Lipids on the Healing Process. Int. J. Mol. Sci. 2024, 25, 3790. [Google Scholar] [CrossRef]
  182. Prtina, A.; Rašeta Simović, N.; Milivojac, T.; Vujnić, M.; Grabež, M.; Djuric, D.; Stojiljković, M.P.; Soldat Stanković, V.; Čolić, M.J.; Škrbić, R. The Effect of Three-Month Vitamin D Supplementation on the Levels of Homocysteine Metabolism Markers and Inflammatory Cytokines in Sera of Psoriatic Patients. Biomolecules 2021, 11, 1865. [Google Scholar] [CrossRef]
  183. 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]
  184. Mazur, A.; Koziorowska, K.; Dynarowicz, K.; Aebisher, D. Vitamin D Supplementation and Photodynamic Therapy. Biol. Life Sci. Forum 2022, 12, 28. [Google Scholar] [CrossRef]
  185. Efird, J.T.; Anderson, E.J.; Jindal, C.; Redding, T.S.; Thompson, A.D.; Press, A.M.; Upchurch, J.; Williams, C.D.; Choi, Y.M.; Suzuki, A. The Interaction of Vitamin D and Corticosteroids: A Mortality Analysis of 26,508 Veterans Who Tested Positive for SARS-CoV-2. Int. J. Environ. Res. Public Health 2021, 19, 447. [Google Scholar] [CrossRef] [PubMed]
  186. Fitch, E.; Harper, E.; Skorcheva, I.; Kurtz, S.E.; Blauvelt, A. Pathophysiology of Psoriasis: Recent Advances on IL-23 and Th17 Cytokines. Curr. Rheumatol. Rep. 2007, 9, 461–467. [Google Scholar] [CrossRef]
  187. Cai, Y.; Fleming, C.; Yan, J. New Insights of T Cells in the Pathogenesis of Psoriasis. Cell. Mol. Immunol. 2012, 9, 302–309. [Google Scholar] [CrossRef]
  188. MacLaughlin, J.A.; Gange, W.; Taylor, D.; Smith, E.; Holick, M.F. Cultured Psoriatic Fibroblasts from Involved and Uninvolved Sites Have a Partial but Not Absolute Resistance to the Proliferation-Inhibition Activity of 1,25-Dihydroxyvitamin D3. Proc. Natl. Acad. Sci. USA 1985, 82, 5409–5412. [Google Scholar] [CrossRef] [PubMed]
  189. Fenercioglu, A.K. The Anti-Inflammatory Roles of Vitamin D for Improving Human Health. Curr. Issues Mol. Biol. 2024, 46, 13514–13525. [Google Scholar] [CrossRef] [PubMed]
  190. Bastyte, D.; Tamasauskiene, L.; Stakaitiene, I.; Briede, K.; Ugenskiene, R.; Valiukeviciene, S.; Gradauskiene, B. Relation of T Cell Profile with Vitamin D Receptor and Vitamin D-Binding Protein Gene Polymorphisms in Atopy. Int. J. Mol. Sci. 2024, 25, 9021. [Google Scholar] [CrossRef]
  191. Gallo, D.; Baci, D.; Kustrimovic, N.; Lanzo, N.; Patera, B.; Tanda, M.L.; Piantanida, E.; Mortara, L. How Does Vitamin D Affect Immune Cells Crosstalk in Autoimmune Diseases? Int. J. Mol. Sci. 2023, 24, 4689. [Google Scholar] [CrossRef]
  192. Hambly, R.; Kirby, B. The Relevance of Serum Vitamin D in Psoriasis: A Review. Arch. Dermatol. Res. 2017, 309, 499–517. [Google Scholar] [CrossRef] [PubMed]
  193. Navarro-Triviño, F.J.; Arias-Santiago, S.; Gilaberte-Calzada, Y. Vitamin D and the Skin: A Review for Dermatologists. Actas Dermo-Sifiliográficas Engl. Ed. 2019, 110, 262–272. [Google Scholar] [CrossRef] [PubMed]
  194. Mason, A.; Mason, J.; Cork, M.; Hancock, H.; Dooley, G. Topical Treatments for Chronic Plaque Psoriasis: An Abridged Cochrane Systematic Review. J. Am. Acad. Dermatol. 2013, 69, 799–807. [Google Scholar] [CrossRef] [PubMed]
  195. Fenner, J.; Silverberg, N.B. Oral Supplements in Atopic Dermatitis. Clin. Dermatol. 2018, 36, 653–658. [Google Scholar] [CrossRef] [PubMed]
  196. Leung, D.Y.; Bieber, T. Atopic Dermatitis. Lancet 2003, 361, 151–160. [Google Scholar] [CrossRef] [PubMed]
  197. 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]
  198. Li, M.; Hener, P.; Zhang, Z.; Ganti, K.P.; Metzger, D.; Chambon, P. Induction of Thymic Stromal Lymphopoietin Expression in Keratinocytes Is Necessary for Generating an Atopic Dermatitis upon Application of the Active Vitamin D3 Analogue MC903 on Mouse Skin. J. Investig. Dermatol. 2009, 129, 498–502. [Google Scholar] [CrossRef] [PubMed]
  199. 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]
  200. Watabe, A.; Yamasaki, K.; Asano, M.; Kanbayashi, Y.; Nasu-Tamabuchi, M.; Terui, H.; Furudate, S.; Kakizaki, A.; Tsuchiyama, K.; Kimura, Y.; et al. Efficacy of Oral Cholecalciferol on Rhododendrol-induced Vitiligo: A Blinded Randomized Clinical Trial. J. Dermatol. 2018, 45, 456–462. [Google Scholar] [CrossRef] [PubMed]
  201. Ekiz, Ö.; Balta, İ.; Şen, B.B.; Dikilitaş, M.C.; Özuğuz, P.; Rifaioğlu, E.N. Vitamin D Status in Patients with Rosacea. Cutan. Ocul. Toxicol. 2014, 33, 60–62. [Google Scholar] [CrossRef] [PubMed]
  202. Chrobak-Chmiel, D.; Golke, A.; Kwiecień, E.; Biegańska, M.J.; Dembele, K.; Dziekiewicz-Mrugasiewicz, M.; Czopowicz, M.; Kizerwetter-Świda, M.; Rzewuska, M. Is Vitamin D3 a Worthy Supplement Protecting against Secondary Infections in Dogs with Atopic Dermatitis? Pathogens 2023, 12, 145. [Google Scholar] [CrossRef] [PubMed]
  203. Liu, A.; Garrett, S.; Hong, W.; Zhang, J. Staphylococcus Aureus Infections and Human Intestinal Microbiota. Pathogens 2024, 13, 276. [Google Scholar] [CrossRef] [PubMed]
  204. AL-smadi, K.; Imran, M.; Leite-Silva, V.R.; Mohammed, Y. Vitiligo: A Review of Aetiology, Pathogenesis, Treatment, and Psychosocial Impact. Cosmetics 2023, 10, 84. [Google Scholar] [CrossRef]
  205. Elmelid, A.; Vandikas, M.S.; Gillstedt, M.; Alsterholm, M.; Osmancevic, A. The Effect of Phototherapy on Systemic Inflammation Measured with Serum Vitamin D-Binding Protein and hsCRP in Patients with Inflammatory Skin Disease. Int. J. Mol. Sci. 2024, 25, 8632. [Google Scholar] [CrossRef] [PubMed]
  206. Podgórska, A.; Puścion-Jakubik, A.; Markiewicz-Żukowska, R.; Gromkowska-Kępka, K.J.; Socha, K. Acne Vulgaris and Intake of Selected Dietary Nutrients—A Summary of Information. Healthcare 2021, 9, 668. [Google Scholar] [CrossRef]
  207. Mao, R.; Zhou, G.; Jing, D.; Liu, H.; Shen, M.; Li, J. Vitamin D Status, Vitamin D Receptor Polymorphisms, and the Risk of Incident Rosacea: Insights from Mendelian Randomization and Cohort Study in the UK Biobank. Nutrients 2023, 15, 3803. [Google Scholar] [CrossRef]
  208. Kim, H.J.; Kim, Y.H. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. Int. J. Mol. Sci. 2024, 25, 5302. [Google Scholar] [CrossRef]
  209. Jasso-Olivares, J.; Diaz-Gonzalez, J.M.; Miteva, M. Horizontal and Vertical Sections of Scalp Biopsy Specimens from Dermatomyositis Patients with Scalp Involvement. J. Am. Acad. Dermatol. 2018, 78, 1178–1184. [Google Scholar] [CrossRef] [PubMed]
  210. Kechichian, E.; Ezzedine, K. Vitamin D and the Skin: An Update for Dermatologists. Am. J. Clin. Dermatol. 2018, 19, 223–235. [Google Scholar] [CrossRef] [PubMed]
  211. Mayba, J.N.; Gooderham, M.J. A Guide to Topical Vehicle Formulations. J. Cutan. Med. Surg. 2018, 22, 207–212. [Google Scholar] [CrossRef]
  212. Heaney, R.P.; Recker, R.R.; Grote, J.; Horst, R.L.; Armas, L.A.G. Vitamin D3 Is More Potent Than Vitamin D2 in Humans. J. Clin. Endocrinol. Metab. 2011, 96, E447–E452. [Google Scholar] [CrossRef]
  213. Regulation (EC) No 1223/2009 of the European Parliament and of the Council of 30 November 2009 on Cosmetic Products. Available online: https://health.ec.europa.eu/system/files/2016-11/cosmetic_1223_2009_regulation_en_0.pdf (accessed on 20 December 2024).
  214. CosIng—Cosmetics—GROWTH—European Commission. Available online: https://ec.europa.eu/growth/tools-databases/cosing/details/74131 (accessed on 9 January 2025).
  215. Vitamins in Cosmetics—Special Actives. Available online: https://dermaviduals.de/english/publications/special-actives/vitamins-in-cosmetics.html (accessed on 11 January 2025).
  216. Russell, M. Assessing the Relationship between Vitamin D3 and Stratum Corneum Hydration for the Treatment of Xerotic Skin. Nutrients 2012, 4, 1213–1218. [Google Scholar] [CrossRef] [PubMed]
  217. Speeckaert, R.; Bulat, V.; Speeckaert, M.M.; Van Geel, N. The Impact of Antioxidants on Vitiligo and Melasma: A Scoping Review and Meta-Analysis. Antioxidants 2023, 12, 2082. [Google Scholar] [CrossRef] [PubMed]
  218. Carlberg, C. Vitamin D and Pigmented Skin. Nutrients 2022, 14, 325. [Google Scholar] [CrossRef] [PubMed]
  219. Kircik, L. Efficacy and Safety of Topical Calcitriol 3 Microg/g Ointment, a New Topical Therapy for Chronic Plaque Psoriasis. J. Drugs Dermatol. JDD 2009, 8, s9–s16. [Google Scholar]
  220. Segaert, S. Vitamin D Regulation of Cathelicidin in the Skin: Toward a Renaissance of Vitamin D in Dermatology? J. Investig. Dermatol. 2008, 128, 773–775. [Google Scholar] [CrossRef] [PubMed]
  221. Parsad, D.; Saini, R.; Verma, N. Combination of PUVAsol and Topical Calcipotriol in Vitiligo. Dermatology 1998, 197, 167–170. [Google Scholar] [CrossRef]
  222. Khullar, G.; Kanwar, A.J.; Singh, S.; Parsad, D. Comparison of Efficacy and Safety Profile of Topical Calcipotriol Ointment in Combination with NB-UVB vs. NB-UVB Alone in the Treatment of Vitiligo: A 24-week Prospective Right–Left Comparative Clinical Trial. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 925–932. [Google Scholar] [CrossRef]
  223. Gargoom, A.M.; Duweb, G.A.; Elzorghany, A.H.; Benghazil, M.; Bugrein, O.O. Calcipotriol in the Treatment of Childhood Vitiligo. Int. J. Clin. Pharmacol. Res. 2004, 24, 11–14. [Google Scholar] [PubMed]
  224. Holick, M.F. Skin as the Site of Vitamin D Synthesis and Target Tissue for 1,25-Dihydroxyvitamin D3: Use of Calcitriol (1,25-Dihydroxyvitamin D3) for Treatment of Psoriasis. Arch. Dermatol. 1987, 123, 1677. [Google Scholar] [CrossRef] [PubMed]
  225. Patel, P.; Pal, R.; Butani, K.; Singh, S.; Prajapati, B.G. Nanomedicine-Fortified Cosmeceutical Serums for The Mitigation of Psoriasis and Acne. Nanomedicine 2023, 18, 1769–1793. [Google Scholar] [CrossRef]
  226. Alsaqr, A.; Rasoully, M.; Musteata, F.M. Investigating Transdermal Delivery of Vitamin D3. AAPS PharmSciTech 2015, 16, 963–972. [Google Scholar] [CrossRef] [PubMed]
  227. Kim, G.K. The Rationale behind Topical Vitamin d Analogs in the Treatment of Psoriasis: Where Does Topical Calcitriol Fit In? J. Clin. Aesthetic Dermatol. 2010, 3, 46–53. [Google Scholar]
  228. Marty, J.; Lafforgue, C.; Grossiord, J.; Soto, P. Rheological Properties of Three Different Vitamin D Ointments and Their Clinical Perception by Patients with Mild to Moderate Psoriasis. J. Eur. Acad. Dermatol. Venereol. 2005, 19, 7–10. [Google Scholar] [CrossRef] [PubMed]
  229. Kolodny, E.H.; Mumford, R.A. Arylsulfatases A and B in Metachromatic Leukodystrophy and Maroteaux-Lamy Syndrome: Studies with 4-Methylumelliferyl Sulfate. Adv. Exp. Med. Biol. 1976, 68, 239–251. [Google Scholar] [CrossRef] [PubMed]
  230. Bickers, D.R.; Athar, M. Oxidative Stress in the Pathogenesis of Skin Disease. J. Investig. Dermatol. 2006, 126, 2565–2575. [Google Scholar] [CrossRef]
  231. Jenssen, M.; Furberg, A.-S.; Jorde, R.; Wilsgaard, T.; Danielsen, K. Effect of Vitamin D Supplementation on Psoriasis Severity in Patients with Lower-Range Serum 25-Hydroxyvitamin D Levels: A Randomized Clinical Trial. JAMA Dermatol. 2023, 159, 518. [Google Scholar] [CrossRef] [PubMed]
  232. Razzaghi, R.; Pourbagheri, H.; Momen-Heravi, M.; Bahmani, F.; Shadi, J.; Soleimani, Z.; Asemi, Z. The Effects of Vitamin D Supplementation on Wound Healing and Metabolic Status in Patients with Diabetic Foot Ulcer: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Diabetes Complicat. 2017, 31, 766–772. [Google Scholar] [CrossRef]
  233. Wang, M.; Zhou, Y.; Yan, Y. Vitamin D Status and Efficacy of Vitamin D Supplementation in Acne Patients: A Systematic Review and Meta-analysis. J. Cosmet. Dermatol. 2021, 20, 3802–3807. [Google Scholar] [CrossRef]
  234. Moukayed, M.; Grant, W.B. The Roles of UVB and Vitamin D in Reducing Risk of Cancer Incidence and Mortality: A Review of the Epidemiology, Clinical Trials, and Mechanisms. Rev. Endocr. Metab. Disord. 2017, 18, 167–182. [Google Scholar] [CrossRef]
  235. Näslund-Koch, C.; Skov, L. New Insights in the Complex Relationship between Psoriasis and Vitamin D…and, Not to Forget, Obesity! Br. J. Dermatol. 2024, 190, 611–612. [Google Scholar] [CrossRef] [PubMed]
  236. Nedoszytko, B.; Owczarczyk-Saczonek, A.; Krasowska, D.; Szczerkowska-Dobosz, A. New Insights into Psoriasis. Int. J. Mol. Sci. 2022, 23, 12851. [Google Scholar] [CrossRef] [PubMed]
  237. 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]
  238. Trémezaygues, L.; Reichrath, J. Vitamin D Analogs in the Treatment of Psoriasis: Where Are We Standing and Where Will We Be Going? Dermato-Endocrinology 2011, 3, 180–186. [Google Scholar] [CrossRef] [PubMed]
  239. Chen, Q.; Liu, L.; Zhang, Y. Vitamin D and Wound Healing: Assessing Skin Barrier Function and Implications for Chloasma Treatment. Int. Wound J. 2024, 21, e14541. [Google Scholar] [CrossRef] [PubMed]
  240. Siregar, F.D.; Hidayat, W. The Role of Vitamin D on the Wound Healing Process: A Case Series. Int. Med. Case Rep. J. 2023, 16, 227–232. [Google Scholar] [CrossRef] [PubMed]
  241. Ahmed Mohamed, A.; Salah Ahmed, E.M.; Abdel-Aziz, R.T.A.; Eldeeb Abdallah, H.H.; El-Hanafi, H.; Hussein, G.; Abbassi, M.M.; El Borolossy, R. The Impact of Active Vitamin D Administration on the Clinical Outcomes of Acne Vulgaris. J. Dermatol. Treat. 2021, 32, 756–761. [Google Scholar] [CrossRef]
  242. Paul, S.; Kaushik, R.; Chawla, P.; Upadhyay, S.; Rawat, D.; Akhtar, A. Vitamin-D as a Multifunctional Molecule for Overall Well-Being: An Integrative Review. Clin. Nutr. ESPEN 2024, 62, 10–21. [Google Scholar] [CrossRef] [PubMed]
  243. Oda, Y.; Hu, L.; Nguyen, T.; Fong, C.; Zhang, J.; Guo, P.; Bikle, D.D. Vitamin D Receptor Is Required for Proliferation, Migration, and Differentiation of Epidermal Stem Cells and Progeny during Cutaneous Wound Repair. J. Investig. Dermatol. 2018, 138, 2423–2431. [Google Scholar] [CrossRef]
  244. 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]
  245. Heaney, R.P. Vitamin D: Criteria for Safety and Efficacy. Nutr. Rev. 2008, 66, S178–S181. [Google Scholar] [CrossRef] [PubMed]
  246. Sawarkar, S.; Ashtekar, A. Transdermal Vitamin D Supplementation—A Potential Vitamin D Deficiency Treatment. J. Cosmet. Dermatol. 2020, 19, 28–32. [Google Scholar] [CrossRef]
  247. Harada, S.; Horisawa, E.; Kano, S.; Sugibayashi, K. Formulation Study of Topically Applied O/W Lotion Containing Vitamin D3 Derivative, Focusing on Skin Permeability of the Drug. Drug Dev. Ind. Pharm. 2011, 37, 917–925. [Google Scholar] [CrossRef] [PubMed]
  248. Usategui, A.; Criado, G.; Del Rey, M.J.; Faré, R.; Pablos, J.L. Topical Vitamin D Analogue Calcipotriol Reduces Skin Fibrosis in Experimental Scleroderma. Arch. Dermatol. Res. 2014, 306, 757–761. [Google Scholar] [CrossRef]
  249. Bikle, D.D. Vitamin D Regulation of Immune Function. Curr. Osteoporos. Rep. 2022, 20, 186–193. [Google Scholar] [CrossRef] [PubMed]
  250. Pan, S.; Sivanathan, S.; Kiepe, G.; Kiepe, T.; Germann, N. Candidate Formulations for a Sustainable Lipstick Supplemented with Vitamin D3: Effects of Wax Type and Concentration on Material Properties. Ind. Eng. Chem. Res. 2021, 60, 2027–2040. [Google Scholar] [CrossRef]
  251. Khalil, K.; Arnold, N.; Seiger, E. Chronic Eyelid Edema and Xerophthalmia Secondary to Periorbital Hyaluronic Acid Filler Injection. J. Cosmet. Dermatol. 2020, 19, 824–826. [Google Scholar] [CrossRef] [PubMed]
  252. Trifan, D.F.; Tirla, A.G.; Mos, C.; Danciu, A.; Bodog, F.; Manole, F.; Ghitea, T.C. Involvement of Vitamin D3 in the Aging Process According to Sex. Cosmetics 2023, 10, 114. [Google Scholar] [CrossRef]
  253. Januszewski, J.; Forma, A.; Zembala, J.; Flieger, M.; Tyczyńska, M.; Dring, J.C.; Dudek, I.; Świątek, K.; Baj, J. Nutritional Supplements for Skin Health—A Review of What Should Be Chosen and Why. Medicina 2023, 60, 68. [Google Scholar] [CrossRef] [PubMed]
  254. Konstantinidi, M.; Stoupi, S.; Anastasiou, I.; Nikolaos, V.Z.; Tsakotos, G.; Koutelidakis, A.E. Effect of Vitamin C, D3, Ca Supplements and Olive Paste Enriched with Mountain Tea on Health Biomarkers in Postmenopausal Women with Osteopenia or Osteoporosis: A Prospective Interventional Study. Appl. Sci. 2024, 14, 5610. [Google Scholar] [CrossRef]
  255. Porcaro, G.; Laganà, A.S.; Neri, I.; Aragona, C. The Association of High-Molecular-Weight Hyaluronic Acid (HMWHA), Alpha Lipoic Acid (ALA), Magnesium, Vitamin B6, and Vitamin D Improves Subchorionic Hematoma Resorption in Women with Threatened Miscarriage: A Pilot Clinical Study. J. Clin. Med. 2024, 13, 706. [Google Scholar] [CrossRef]
  256. Brennan Laing, B.; Cavadino, A.; Ellett, S.; Ferguson, L. Effects of an Omega-3 and Vitamin D Supplement on Fatty Acids and Vitamin D Serum Levels in Double-Blinded, Randomized, Controlled Trials in Healthy and Crohn’s Disease Populations. Nutrients 2020, 12, 1139. [Google Scholar] [CrossRef]
  257. Le Jan, D.; Siliman Misha, M.; Destrumelle, S.; Terceve, O.; Thorin, C.; Larcher, T.; Ledevin, M.; Desfontis, J.-C.; Betti, E.; Mallem, Y. Omega-3 Fatty Acid and Vitamin D Supplementations Partially Reversed Metabolic Disorders and Restored Gut Microbiota in Obese Wistar Rats. Biology 2024, 13, 1070. [Google Scholar] [CrossRef] [PubMed]
  258. Thompson, K.G.; Kim, N. Dietary Supplements in Dermatology: A Review of the Evidence for Zinc, Biotin, Vitamin D, Nicotinamide, and Polypodium. J. Am. Acad. Dermatol. 2021, 84, 1042–1050. [Google Scholar] [CrossRef] [PubMed]
  259. Mavar, M.; Sorić, T.; Bagarić, E.; Sarić, A.; Matek Sarić, M. The Power of Vitamin D: Is the Future in Precision Nutrition through Personalized Supplementation Plans? Nutrients 2024, 16, 1176. [Google Scholar] [CrossRef]
  260. Joshi, M.; Hiremath, P.; John, J.; Ranadive, N.; Nandakumar, K.; Mudgal, J. Modulatory Role of Vitamins A, B3, C, D, and E on Skin Health, Immunity, Microbiome, and Diseases. Pharmacol. Rep. 2023, 75, 1096–1114. [Google Scholar] [CrossRef]
  261. Guan, L.L.; Lim, H.W.; Mohammad, T.F. Sunscreens and Photoaging: A Review of Current Literature. Am. J. Clin. Dermatol. 2021, 22, 819–828. [Google Scholar] [CrossRef] [PubMed]
  262. Panico, A.; Serio, F.; Bagordo, F.; Grassi, T.; Idolo, A.; De Giorgi, M.; Guido, M.; Congedo, M.; De Donno, A. Skin Safety and Health Prevention: An Overview of Chemicals in Cosmetic Products. J. Prev. Med. Hyg. 2019, 60, E50. [Google Scholar] [CrossRef] [PubMed]
  263. Grant, W.B. Reply to “The Five Paradoxes of Vitamin D and the Importance of Sunscreen Protection”. Clin. Pediatr. 2013, 52, 994. [Google Scholar] [CrossRef] [PubMed]
  264. Young, A.R.; Narbutt, J.; Harrison, G.I.; Lawrence, K.P.; Bell, M.; O’Connor, C.; Olsen, P.; Grys, K.; Baczynska, K.A.; Rogowski-Tylman, M.; et al. Optimal Sunscreen Use, during a Sun Holiday with a very High Ultraviolet Index, Allows Vitamin D Synthesis without Sunburn. Br. J. Dermatol. 2019, 181, 1052–1062. [Google Scholar] [CrossRef]
  265. Manela-Azulay, M.; Bagatin, E. Cosmeceuticals Vitamins. Clin. Dermatol. 2009, 27, 469–474. [Google Scholar] [CrossRef]
  266. Karimi, N. Approaches in Line with Human Physiology to Prevent Skin Aging. Front. Physiol. 2023, 14, 1279371. [Google Scholar] [CrossRef] [PubMed]
  267. Ye, S.; Matthan, N.R.; Lamon-Fava, S.; Solano-Aguilar, G.; Turner, J.R.; Walker, M.E.; Chai, Z.; Lakshman, S.; Chen, C.; Dawson, H.; et al. Colon Transcriptome Is Modified by a Dietary Pattern/Atorvastatin Interaction in the Ossabaw Pig. J. Nutr. Biochem. 2021, 90, 108570. [Google Scholar] [CrossRef] [PubMed]
  268. Vieira, E.F.; Souza, S. Formulation Strategies for Improving the Stability and Bioavailability of Vitamin D-Fortified Beverages: A Review. Foods 2022, 11, 847. [Google Scholar] [CrossRef] [PubMed]
  269. Yamada-Kanazawa, S.; Kajihara, I.; Kobayashi, A.; Watanabe, C.; Ihn, H. Infliximab Improved the Refractory Cutaneous Involvement in a Patient with Dermatomyositis. Dermatol. Ther. 2019, 32, e12859. [Google Scholar] [CrossRef]
  270. Jones, P.; Lucock, M.; Veysey, M.; Beckett, E. The Vitamin D–Folate Hypothesis as an Evolutionary Model for Skin Pigmentation: An Update and Integration of Current Ideas. Nutrients 2018, 10, 554. [Google Scholar] [CrossRef] [PubMed]
  271. 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]
  272. 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] [PubMed]
  273. Al-Khaldy, N.S.; Al-Musharaf, S.; Aljazairy, E.A.; Hussain, S.D.; Alnaami, A.M.; Al-Daghri, N.; Aljuraiban, G. Serum Vitamin D Level and Gut Microbiota in Women. Healthcare 2023, 11, 351. [Google Scholar] [CrossRef]
  274. Bryson, W.G.; McCormack, A.C.; Plowman, J.E.; Grosvenor, A.J.; Murphy, C.J.; Nagase, S.; Itou, T.; Koike, K. Improved Two-dimensional Electrophoretic Mapping of Japanese Human Hair Proteins; Application to Curved and Straight Japanese Human Hairs; and Protein Identification by MALDI MS and MS/MS Quadrupole Time-of-flight Mass Spectrometry. Int. J. Cosmet. Sci. 2020, 42, 346–358. [Google Scholar] [CrossRef]
  275. Peñuñuri-Pacheco, N.; Moreno-García, Y.A.; González-Ríos, H.; Astiazarán-García, H.; López-Franco, Y.L.; Tortoledo-Ortiz, O.; Pérez-Báez, A.J.; Dávila-Ramírez, J.L.; Lizardi-Mendoza, J.; Valenzuela-Melendres, M. Optimization of the Encapsulation of Vitamin D3 in Oil in Water Nanoemulsions: Preliminary Application in a Functional Meat Model System. Foods 2024, 13, 2842. [Google Scholar] [CrossRef] [PubMed]
  276. Akkam, Y.; Rababah, T.; Costa, R.; Almajwal, A.; Feng, H.; Laborde, J.E.A.; Abulmeaty, M.M.; Razak, S. Pea Protein Nanoemulsion Effectively Stabilizes Vitamin D in Food Products: A Potential Supplementation during the COVID-19 Pandemic. Nanomaterials 2021, 11, 887. [Google Scholar] [CrossRef]
  277. Crintea, A.; Dutu, A.G.; Sovrea, A.; Constantin, A.-M.; Samasca, G.; Masalar, A.L.; Ifju, B.; Linga, E.; Neamti, L.; Tranca, R.A.; et al. Nanocarriers for Drug Delivery: An Overview with Emphasis on Vitamin D and K Transportation. Nanomaterials 2022, 12, 1376. [Google Scholar] [CrossRef] [PubMed]
  278. Du, M.; Chang, C.; Zhang, X.; Zhang, Y.; Radford, M.J.; Gahler, R.J.; Kuo, Y.C.; Wood, S.; Solnier, J. Designing Vitamin D3 Formulations: An In Vitro Investigation Using a Novel Micellar Delivery System. Nutraceuticals 2023, 3, 290–305. [Google Scholar] [CrossRef]
  279. Zhao, F.; Jia, Z.; Feng, Y.; Li, Z.; Feng, J. Circular RNA Circ_0079593 Enhances Malignant Melanoma Progression by the Regulation of the miR-573/ABHD2 Axis. J. Dermatol. Sci. 2021, 102, 7–15. [Google Scholar] [CrossRef] [PubMed]
  280. Wang, C.-K.; Chang, C.-Y.; Chu, T.-W.; Liang, Y.-J. Using Machine Learning to Identify the Relationships between Demographic, Biochemical, and Lifestyle Parameters and Plasma Vitamin D Concentration in Healthy Premenopausal Chinese Women. Life 2023, 13, 2257. [Google Scholar] [CrossRef] [PubMed]
  281. Patino-Alonso, C.; Gómez-Sánchez, M.; Gómez-Sánchez, L.; Sánchez Salgado, B.; Rodríguez-Sánchez, E.; García-Ortiz, L.; Gómez-Marcos, M.A. Predictive Ability of Machine-Learning Methods for Vitamin D Deficiency Prediction by Anthropometric Parameters. Mathematics 2022, 10, 616. [Google Scholar] [CrossRef]
  282. Xie, Y.; Liu, Q.; Zhang, W.; Yang, F.; Zhao, K.; Dong, X.; Prakash, S.; Yuan, Y. Advances in the Potential Application of 3D Food Printing to Enhance Elderly Nutritional Dietary Intake. Foods 2023, 12, 1842. [Google Scholar] [CrossRef] [PubMed]
  283. Darrigade, A.; Dendooven, E.; Mangodt, E.; Van Dyck, F.; Aerts, O. Airborne Allergic Contact Dermatitis by Proxy Caused by ‘Poppers’. Contact Dermat. 2021, 84, 212–214. [Google Scholar] [CrossRef]
  284. Płomiński, J.; Grzybowski, R.; Fiedorowicz, E.; Sienkiewicz-Szłapka, E.; Rozmus, D.; Król-Grzymała, A.; Jarmołowska, B.; Kordulewska, N.; Cieślińska, A. Vitamin D Metabolic Pathway Components in Orthopedic Patientes—Systematic Review. Int. J. Mol. Sci. 2022, 23, 15556. [Google Scholar] [CrossRef] [PubMed]
  285. Horas, K.; Hoxha, M.; Heinz, T.; Jakuscheit, A.; List, K.; Maier, G.S.; Weißenberger, M.; Rudert, M. Prevalence and Risk Factors of Vitamin D Deficiency in Patients Scheduled to Undergo Revision Arthroplasty of the Hip, Knee and Shoulder—Data from a Single-Centre Analysis. Nutrients 2024, 16, 3060. [Google Scholar] [CrossRef] [PubMed]
  286. Shields, B.A.; Nakakura, A.M. Nutrition Considerations for Burn Patients: Optimizing Recovery and Healing. Eur. Burn J. 2023, 4, 537–547. [Google Scholar] [CrossRef] [PubMed]
  287. Chen, L.-R.; Yang, B.-S.; Chang, C.-N.; Yu, C.-M.; Chen, K.-H. Additional Vitamin and Mineral Support for Patients with Severe Burns: A Nationwide Experience from a Catastrophic Color-Dust Explosion Event in Taiwan. Nutrients 2018, 10, 1782. [Google Scholar] [CrossRef] [PubMed]
  288. Makrantonaki, E.; Zouboulis, C.C. Association of Acne Tarda with Endocrinological Disorders. Dermato 2022, 2, 109–120. [Google Scholar] [CrossRef]
  289. Cammisa, I.; Zona, M.; Guerriero, C.; Cipolla, C.; Rigante, D. Skin Sceneries of Thyroid Disorders and Impact of Thyroid on Different Skin Diseases: A Scoping Review Focused on Pediatric Patients. Children 2024, 11, 1488. [Google Scholar] [CrossRef]
  290. Varzakas, T.; Antoniadou, M. A Holistic Approach for Ethics and Sustainability in the Food Chain: The Gateway to Oral and Systemic Health. Foods 2024, 13, 1224. [Google Scholar] [CrossRef] [PubMed]
  291. Bruins, M.J.; Létinois, U. Adequate Vitamin D Intake Cannot Be Achieved within Carbon Emission Limits Unless Food Is Fortified: A Simulation Study. Nutrients 2021, 13, 592. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Historical timeline on significant findings in vitamin D research over time. (Parts of this figure were obtained by https://thenounproject.com/ (accessed on 29 December 2024)).
Figure 1. Historical timeline on significant findings in vitamin D research over time. (Parts of this figure were obtained by https://thenounproject.com/ (accessed on 29 December 2024)).
Applsci 15 00796 g001
Figure 2. Article selection, inclusion, and exclusion methodology.
Figure 2. Article selection, inclusion, and exclusion methodology.
Applsci 15 00796 g002
Figure 3. Metabolism pathway of vitamin D3. (Parts of this figure were obtained by https://smart.servier.com/ (accessed on 29 December 2024) and structures from PubChem: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)).
Figure 3. Metabolism pathway of vitamin D3. (Parts of this figure were obtained by https://smart.servier.com/ (accessed on 29 December 2024) and structures from PubChem: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)).
Applsci 15 00796 g003
Figure 4. The two common forms of vitamin D (D2, D3), their origin, and their differences. (Parts of this figure were obtained by https://smart.servier.com/ (accessed on 29 December 2024)).
Figure 4. The two common forms of vitamin D (D2, D3), their origin, and their differences. (Parts of this figure were obtained by https://smart.servier.com/ (accessed on 29 December 2024)).
Applsci 15 00796 g004
Figure 5. The production of vitamin D3 from 7–DHC in the skin epidermis. (Reproduced from [30], while the structures were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)), and Molview (https://molview.org/ (accessed on 29 December 2024)).
Figure 5. The production of vitamin D3 from 7–DHC in the skin epidermis. (Reproduced from [30], while the structures were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)), and Molview (https://molview.org/ (accessed on 29 December 2024)).
Applsci 15 00796 g005
Figure 6. Vitamin D3’s 25-hydroxylation metabolism and the anti-inflammatory and pro-inflammatory roles of vitamin d3 and its active form calcitriol in immune response. (Reproduced from [30], while all the structures utilized were obtained from the free databases PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)), and Molview (https://molview.org/ (accessed on 29 December 2024)).
Figure 6. Vitamin D3’s 25-hydroxylation metabolism and the anti-inflammatory and pro-inflammatory roles of vitamin d3 and its active form calcitriol in immune response. (Reproduced from [30], while all the structures utilized were obtained from the free databases PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 29 December 2024)), and Molview (https://molview.org/ (accessed on 29 December 2024)).
Applsci 15 00796 g006
Table 1. Anti-Melanoma, Anti-cancer, and Anti-UV Activities of Vitamin D.
Table 1. Anti-Melanoma, Anti-cancer, and Anti-UV Activities of Vitamin D.
Study HypothesisCosmetic ApplicationMain ActivityPathwayResultsReference
This study demonstrated the effects of secosteroidal analogs (1α,25(OH)2D3 and 25(OH)D3) and non-calcemic ones (20(OH)D3, calcipotriol, 21(OH)pD, pD, and 20(OH)pL) on proliferation, colony formation in both monolayer and soft agar, and mRNA and protein expression by melanoma cellsIn vivo testingAnti-melanoma
  • In primary cultures of Ab cells freshly isolated from solid tumors, moderate-melanin-pigmentation sensitized cells towards 1α,25(OH)2D3, 25(OH)D3, and the short side-chained 21(OH)pD analog were prepared
  • Moreover, two rodent (mouse and hamster) melanoma models were used
  • Inhibitory effects of vitamin D and lumisterol analogs were tested with a short side-chain, including pD, 20(OH)pL, and 20(OH)pD, on a colony in soft agar
  • The testing was performed on primary cell cultures using melanoma cells, isolated from transplantable Ab amelanotic tumors
  • Murine B16-f10 and hamster Bomirski Ab cell lines were utilized as models to study the effect of melanin pigmentation and its influence in the expression of genes
  • The last Western blot was performed to show the VDR synthesis as well as its translocation to the nucleus
  • Only 21(OH)pD showed antiproliferative effects
  • All three lumisterol analogs having side-chain pD, 20(OH)pL, and 20(OH)pD inhibited colony formation in soft agar
  • 1α,25(OH)2D3 decreased the ability of primary Ab cells to form colonies in soft agar assays, following also a dose-dependent manner and resulting in decreased size
  • Pigmentation increased the expression of VDR and co-receptors like the retinoid X receptor (RXR) at the mRNA level
  • 1α,25(OH)2D3 treatment increased the VDR and CYP24A1 mRNA levels in pigmented Ab cells compared to in the non-pigmented ones
  • The Western blot analysis showed the stimulation of the VDR translocation by 1α,25(OH)2D3
[82]
An investigation of the role of RXRα and its partners in mouse skin tumor formation and malignant progression upon topical mutagen dimethylbenzanthracene (DMBA)/12–O–tetradecanoyl phorbol–13–acetate (TPA) treatment was followed in this studyIn vivo testingAnti-cancer, anti-melanoma
  • The two-step chemical tumorigenesis model was used to study the 3 different stages, namely the initiation, promotion, and progression of mouse skin-originated tumors
  • Human squamous cell carcinoma (SCC) acts similarly to mouse chemically activated squamous cell tumors
  • The initiation takes place after a single topical application of DMBA. Promotion is achieved by a repeated topical application of a tumor promoter (i.e., terephthalic acid (TPA))
  • The application of a tumor promoter results in sustained hyperplasia and inflammation and then in a benign papilloma
  • The last stage is the progression where a conversion of the benign papilloma to invasive SCC occurs
  • Keratolytic RXRα has a key role in the formation and malignant transformation of DMBA/TPA-induced epidermal skin tumors
[83]
An examination of the role of the active forms of vitamin D in regulating the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) activity in melanoma cells that are dependent on melanin In vitro testingAnti-melanoma
  • Firstly, an examination of the induction of melanin pigmentation was performed to check if it can affect the VDR expression
  • Secondly, in order to test the subcellular location of the p65 NF-κB subunit in SKMEL-188 melanoma cells, the preparation of cytoplasmic and nuclear extracts with pigmented and non-pigmented melanoma cells took place
  • The further characterization of the activation of the NF-κB in melanoma cells required an immunofluorescent staining performance for p65 in both pigmented and non-pigmented SKMEL-188 cells
  • To determine which 20(OH)D3 or 1α,25(OH)2D3 affected NF-κB-driven transcriptional activity, pigmented and non-pigmented melanoma cells were transfected with a NF-κB-dependent luciferase reporter construct (NF-κB-p-Luc)
  • An electrophoretic mobility shift assay (EMSA) was used for the determination of the NF-κB DNA-binding activity by using an NF-κB oligonucleotide probe
  • An enzyme-linked immunosorbent assay (ELISA), a Western blot, and immunofluorescence were utilized so as to characterize in more detail the effect of secosteroids and the pigmentation of NF-κB activity and especially to examine p65 nuclear translocation
  • An analysis of biopsies of human melanoma cells was used to examine the reflection of their pigmentation
  • An inversely proportional relationship was found between the melanin content and the VDR protein expression
  • The analysis of the Western blot showed that in non-pigmented SKMEL-188 melanoma cells, a higher nuclear level of p65 was indeed traced
  • Intense p65 nuclear staining was concluded to be present in non-pigmented melanoma cells
  • On the other hand, in the pigmented ones, p65 was shown to be localized mainly in the cells’ cytoplasm
  • Both secosteroids, 20(OH)D3 and 1α,25(OH)2D3, were shown to reduce the NF-κB-driven transcriptional activity in non-pigmented melanoma cells in a time-dependent mechanism of action
  • Nuclear extracts prepared by non-pigmented SKMEL-188 melanoma cells showed a higher activation of NF-κB DNA-binding activity, compared to extracts prepared by pigmented cells
  • Treatment of non-pigmented cells with 20(OH)D3 resulted in a time-dependent inhibition of NF-κB-dependent DNA-binding activity, versus treatment of pigmented cells that had no effects on the NF-κB activation
  • When non-pigmented SKMEL-188 melanoma cells were treated with 20(OH)D3, the nuclear levels of p65 were decreased and the cytoplasmic levels increased, subsequently
  • When pigmented cells were treated either with 20(OH)D3 or with 1α,25(OH)2D3 intracellularly, the localization of p65 was not affected at all. Especially, p65 was mainly found in the nucleus of non-pigmented cells
  • In human biopsies, the results showed that pigmentation affects the activity of NF-κB as the intracellular distribution of p65 changes
  • Therefore, higher nuclear p65 levels were traced in non-pigmented than in pigmented melanoma cells
[84]
An investigation of skin cancer therapy in mouse models of skin carcinogenesis with the application of topical calcipotriol was conducted. An additional randomized, double-blind clinical trial investigation for testing the combination of 0.005% calcipotriol ointment together with 5% fluorouracil (5-FU) cream and its comparison with Vaseline and 5-FU used in the treatment of actinic keratoses on the face, scalp, and upper extremities was tested as wellIn vivo and in vitro testingAnti-cancer
  • First, calcipotriol was applied three times per week at the mouse’s skin-located carcinogenic site, so as to test the induction of the thymic stromal lymphopoietin (TSLP) in the skin
  • Secondly, the impact of short-term TSLP induction by calcipotriol was tested
  • As an experimental example, calcipotriol was used as a short pulse and in the starting stage of the tumor, in DMBA-TPA-treated animals
  • Calcipotriol was applied to the ears, every day for 3 days in a row
  • In order to test the human approach regarding the efficacy of TSLP induction as a cancer immunotherapeutic candidate, a hypothesis of a combination of calcipotriol and 5-FU was made
  • 5-FU used alone follows the recommended use of twice daily, for 2–4 weeks. This randomized, double-blind, clinical trial took place to examine the results after using a combination of 0.005% calcipotriol ointment (final concentration: 0.0025%) combined with 5% 5-FU cream (final concentration: 2.5%) twice, for 4 days
  • Topical calcipotriol blocked skin cancer via the TSLP expression in the skin
  • Increased serum levels of TSLP were the result of treatment with a short calcipotriol regimen, which led to a long-lasting anti-cancer effect, highlighting the effectiveness and the safety of this topical agent
  • The synergistic effect of calcipotriol and 5-FU was further supported due to an observed activation of the CD4+ T cell-mediated immunity cells and the subsequent protection against actinic keratoses and potential skin cancer onset
[85]
An investigation regarding the ability of novel non-calcemic secosteroids on how they could protect against solar radiation, which can damage human epidermal keratinocytes, melanocytes, and HaCaT keratinocytes, was conducted in this studyIn vitroAnti-melanoma
  • CYP11A1, including epidermal keratinocytes, can metabolize vitamin D3 and produce 20S(OH)D3 as the major metabolite and 20,23(OH)2D3 as the second major metabolite of the pathway
  • Smaller quantities of other mono-, di-, and tri-hydroxy-derivatives of vitamin D3 may also be produced
  • The first step was the confirmation that HaCaT keratinocytes can generate 20S(OH)D3 from vitamin D3 precursors, showing in this way that 20S(OH)D3 is present in the human epidermis in vivo
  • After that, a test regarding the protective effects of non-calcemic 20S(OH)D3 and 20,23(OH)2D3 against UVB-induced damage in comparison to 1α,25 (OH)2D3 and related sterols, in human epidermal keratinocytes and melanocytes, took place
  • A preliminary experiment followed, where the MTS test on HaCaT keratinocytes was held so as to make a comparison between the protective effects of 20S(OH)D3, its enantiomer 20R(OH)D3, and precursors 20S(OH)7DHC and D3 against UVB-induced cell death
  • To continue, 20S(OH)D3 and 20,23(OH)2D3, in comparison to 1α,25(OH)2D3, 20S(OH)7DHC, 7-hydroxycholestrol (7(OH)C), and cholesterol sulfate, were tested in order to check if they could weaken ROS production, followed by UVB exposure
  • A following test was conducted so as to check if vitamin D hydroxy-derivatives and 20S(OH)7DHC can scavenge H2O2, which was formed as a direct response to UVR
  • An additional test was also held so as to confirm the protective effects of 20S(OH)D3 on the UVB-induced production of nitric oxide (NO)
  • The effects of 20S(OH)D3, 20,23(OH)2D3, 20S(OH)7DHC, 1α,25(OH)2D3, and 25(OH)D3 on the relative glutathione (GSH) content were tested as well
  • An investigation on whether secosteroids can stimulate the expression of genes was additionally conducted
  • A final experiment that was held and was utilized to analyze the DNA damage was the Comet assay, where a visualization of the DNA migration was performed by fluorescence microscopy
  • 20S(OH)D3 could speed up the cell death induced by UVB
  • The preliminary experiment showed that only 20S(OH)D3 could weaken the cell death, which was induced from UVB
  • Also, they reported that the R enantiomer had a smaller potency compared to the S enantiomer
  • Therefore, the S enantiomer, the naturally occurring one, was selected for further experiments
  • All hydroxy-derivatives of vitamin D3, except for 7(OH)C and cholesterol sulfate, as well as 20S(OH) 7DHC, could help reduce the UVB-induced ROS production
  • When cells were treated with 20S(OH)D3, 20,23(OH)2D3, 20S(OH)7DHC, 1α,25(OH)2D3, or 25(OH)D3, there was a significant reduction in H2O2 in concentration after UVB exposure
  • Treatment of cells with 20S(OH)D3, 20,23(OH)2D3, 20S(OH)7DHC, 1α,25(OH)2D3, or 25(OH)D3 before UVB irradiation reduced NO production. Specifically, 20(OH)D3, 20,23(OH)2D3, 1α,25(OH)2D3, and 20(OH)7DHC strongly inhibited the NO generation
  • A significant reduction in the GSH content was observed as an aftermath of UVB exposure
  • Extensive DNA damage was observed, after the exposure of melanocytes to UVB radiation, while 20S(OH)D3, 20,23(OH)2D3, and 1α,25(OH)2D3 were found to be the most protective agents against UVB-induced damage
[86]
Table 2. The wound-healing, anti-inflammatory, antioxidant, and anti-aging activities of Vitamin D.
Table 2. The wound-healing, anti-inflammatory, antioxidant, and anti-aging activities of Vitamin D.
Study HypothesisCosmetic ApplicationMain ActivityExperimental Pathway—Review TargetResultsRef.
Vitamin D and calcium are critical for the activation, migration, and function of stem cells during wound healingIn vivoWound HealingMouse models with full-thickness skin biopsies (control and receptor-deficient) were utilized to assess healing, regarding the fact that vitamin D and calcium aid wound healing by activating VDR and CaSR receptor pathwaysDeletion of VDR and CaSR:
  • Delayed wound healing
  • Impairing re-epithelialization
  • Reducing stem cell activity
Findings suggested that vitamin D deficiency may cause chronic or delayed healing
[162]
Vitamin D modulates inflammation, supports keratinocyte function, and promotes tissue regeneration to improve wound healingIn vivoAnti-inflammatory Role in Wound HealingThe key pathway, Vitamin D–VDR signaling, regulates
  • The Wnt/β-catenin cascade for keratinocyte function and skin regeneration
  • Inflammation control, by reducing pro-inflammatory cytokines’ generation
  • Re-epithelialization, aiding keratinocyte migration and skin barrier repair, which was also this review’s predominant confirmation target
Vitamin D aids in successful wound healing, by reducing inflammation, promoting keratinocyte growth and migration, and accelerating re-epithelialization[181]
Vitamin D protects against aging by modulating immunity, reducing inflammation, and enhancing overall healthIn vivo and in vitroAnti-inflammatory and Anti-aging EffectsVitamin D–VDR signaling regulates immunity and reduces inflammation, while inhibiting NF-kB, and lowers inflammatory cytokine productionVitamin D contributes to
  • Reducing effectively chronic inflammation
  • Suppressing pro-inflammatory cytokines (e.g., TNF-α, IL-6)
  • Promoting Tregs and inhibiting pro-inflammatory Th1 and Th17, for immune tolerance
  • Delaying cellular aging by supporting mitochondrial function and DNA repair, while reducing oxidative stress
[34]
Vitamin D’s role as an antioxidant that modulates ROS in keratinocytes, while potentially influencing skin health/disease prevention by reducing oxidative stressIn vivoAnti-inflammatory and Antioxidant
  • Keratinocytes were used to examine vitamin D’s cellular impact—effects
  • The reduction in ROS was measured by the activity of antioxidant enzymes like glutathione peroxidase (GPx), which neutralizes harmful peroxides such as hydrogen peroxide (H2O2)
Vitamin D reduces the levels of ROS in keratinocytes, so as to protect cells from oxidative stress and to promote healthier skin, by mainly alleviating inflammation and damage caused by oxidative agents[170]
Vitamin D (especially its D3 metabolite) induces beneficial anti-aging and photoprotective effects on the skinIn vivoAnti-aging, Photoprotective (Anti-UV), Anti-inflammatory, Antioxidant
  • The pathways involved are the VDR and Nrf2
  • The Nrf2 pathway specifically is crucial for combating oxidative stress and providing photoprotection, while p53 activation supports further DNA repair
Vitamin D induces the activation of pathways that reduce ROS, inhibit DNA damage, and promote DNA repair, protecting the skin from premature aging and photoaging, mostly due to its antioxidant and anti-inflammatory profile [27]
Vitamin D can act as a protective agent against aging by regulating cellular processes such as oxidative stress, immune response, and mitochondrial functionIn vivo and in vitroAnti-aging, Photoprotective (Anti-UV), Anti-inflammatory, Antioxidant
  • The Nrf2-Keap1 pathway, activated by Vitamin D, combats oxidative stress
  • The NF-kB pathway, on the contrary, mainly reduces enhanced inflammation
  • Both pathways are essential for vitamin D’s anti-aging and other protective effects
Vitamin D has several anti-aging-associated properties:
  • It acts as an antioxidant via the Nrf2-Keap1 pathway
  • It reduces inflammation through the NF-kB pathway
  • It protects against mitochondrial dysfunction and ROS accumulation
  • Its metabolites support DNA repair, helping to reduce UV-induced skin damage
[34]
Table 4. Possible cosmetic and cosmeceutical formulations containing vitamin D.
Table 4. Possible cosmetic and cosmeceutical formulations containing vitamin D.
Cosmetic TypePurpose of Vitamin DEffectsExamplesReferences
MoisturizersEnhances hydration and skin barrier functionImprove moisture retention and sooth dry skinVitamin D3 creams, hydrating lotions[216]
Anti-aging productsSupports skin homeostasis, reduces oxidative stress, improves elasticityStimulate DNA repair, enhance collagen production, antioxidant and anti-photoaging property, reduce wrinklesD3 anti-aging serums [27,28,34,224]
Acne treatmentsReduces inflammation and promotes healingReduce skin sebum, clear acne scarsAcne serums with vitamin D[225]
Psoriasis treatmentsReduces inflammationHelp keratinocyte regenerationOintments[219,226,227,228]
CleansersMild exfoliation and hydrationCleanse without stripping natural oilsCleansers with vitamin D derivatives[121]
Scalp treatmentsImproves scalp health and prevents dandruffReduce flakiness, support hair follicle healthShampoos, hair oils, and scalp serums [210]
Hand creamsRepairs cracked skin, enhances hydrationImprove moisture retention and sooth dry skinHand creams with vitamin D[229]
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

Papadopoulou, S.N.A.; Anastasiou, E.A.; Adamantidi, T.; Ofrydopoulou, A.; Letsiou, S.; Tsoupras, A. A Comprehensive Review on the Beneficial Roles of Vitamin D in Skin Health as a Bio-Functional Ingredient in Nutricosmetic, Cosmeceutical, and Cosmetic Applications. Appl. Sci. 2025, 15, 796. https://doi.org/10.3390/app15020796

AMA Style

Papadopoulou SNA, Anastasiou EA, Adamantidi T, Ofrydopoulou A, Letsiou S, Tsoupras A. A Comprehensive Review on the Beneficial Roles of Vitamin D in Skin Health as a Bio-Functional Ingredient in Nutricosmetic, Cosmeceutical, and Cosmetic Applications. Applied Sciences. 2025; 15(2):796. https://doi.org/10.3390/app15020796

Chicago/Turabian Style

Papadopoulou, Sofia Neonilli A., Elena A. Anastasiou, Theodora Adamantidi, Anna Ofrydopoulou, Sophia Letsiou, and Alexandros Tsoupras. 2025. "A Comprehensive Review on the Beneficial Roles of Vitamin D in Skin Health as a Bio-Functional Ingredient in Nutricosmetic, Cosmeceutical, and Cosmetic Applications" Applied Sciences 15, no. 2: 796. https://doi.org/10.3390/app15020796

APA Style

Papadopoulou, S. N. A., Anastasiou, E. A., Adamantidi, T., Ofrydopoulou, A., Letsiou, S., & Tsoupras, A. (2025). A Comprehensive Review on the Beneficial Roles of Vitamin D in Skin Health as a Bio-Functional Ingredient in Nutricosmetic, Cosmeceutical, and Cosmetic Applications. Applied Sciences, 15(2), 796. https://doi.org/10.3390/app15020796

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