Next Article in Journal
Implementing Whole Genome Sequencing (WGS) in Clinical Practice: Advantages, Challenges, and Future Perspectives
Previous Article in Journal
Leucine Supplementation Prevents the Development of Skeletal Muscle Dysfunction in a Rat Model of HFpEF
Previous Article in Special Issue
What Does the Future Hold for Biomarkers of Response to Extracorporeal Photopheresis for Mycosis Fungoides and Sézary Syndrome?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin D in Cutaneous T-Cell Lymphoma

by
August-Witte Feentved Ødum
and
Carsten Geisler
*
The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
*
Author to whom correspondence should be addressed.
Cells 2024, 13(6), 503; https://doi.org/10.3390/cells13060503
Submission received: 21 February 2024 / Revised: 5 March 2024 / Accepted: 12 March 2024 / Published: 13 March 2024

Abstract

:
Cutaneous T-cell lymphoma (CTCL) is characterized by the proliferation of malignant T cells in inflamed skin lesions. Mycosis fungoides (MF)—the most common variant of CTCL—often presents with skin lesions around the abdomen and buttocks (“bathing suit” distribution), i.e., in skin areas devoid of sun-induced vitamin D. For decades, sunlight and vitamin D have been connected to CTCL. Thus, vitamin D induces apoptosis and inhibits the expression of cytokines in malignant T cells. Furthermore, CTCL patients often display vitamin D deficiency, whereas phototherapy induces vitamin D and has beneficial effects in CTCL, suggesting that light and vitamin D have beneficial/protective effects in CTCL. Inversely, vitamin D promotes T helper 2 (Th2) cell specific cytokine production, regulatory T cells, tolerogenic dendritic cells, as well as the expression of immune checkpoint molecules, all of which may have disease-promoting effects by stimulating malignant T-cell proliferation and inhibiting anticancer immunity. Studies on vitamin D treatment in CTCL patients showed conflicting results. Some studies found positive effects, others negative effects, while the largest study showed no apparent clinical effect. Taken together, vitamin D may have both pro- and anticancer effects in CTCL. The balance between the opposing effects of vitamin D in CTCL is likely influenced by treatment and may change during the disease course. Therefore, it remains to be discovered whether and how the effect of vitamin D can be tilted toward an anticancer response in CTCL.

1. Sunlight, Vitamin D, and Cutaneous T-Cell Lymphoma

Vitamin D is a fat-soluble secosteroid that comes in two forms, namely vitamin D2 (ergocalciferol) and vitamin D3 (cholecalciferol). These forms differ only in the side chain, and the biological activities are comparable [1]. Vitamin D3 is produced in the skin from 7-dehydrocholesterol [2], whereas vitamin D2 is produced in plants and fungi from ergosterol [3]. For decades, it has been known that sunlight elicits significant production of vitamin D3 in the skin. Solar ultraviolet B (UVB) radiation (280–315 nm) is absorbed by 7-dehydrocholesterol in the plasma membrane of keratinocytes, leading to its transformation to pre-vitamin D3, which is rapidly converted to vitamin D3 [4,5,6,7]. Vitamin D3 is ejected from the plasma membrane into the extracellular space and then drawn into the circulation by the vitamin D binding protein (DBP), which has high affinity for vitamin D3 [6,8]. Vitamin D3 is transported by DBP to the liver, where it is metabolized to 25(OH)D3 (calcifediol) by vitamin D 25-hydroxylases CYP2R1 and CYP27A1 [7]. 25(OH)D3 re-enters the circulation, where it is bound to DBP. In the kidney, 25(OH)D3 is converted to the active form of vitamin D, 1,25(OH)2D3 (calcitriol), by the 25(OH)D 1-α hydroxylase CYP27B1 [9,10,11].
Humans have a combination of vitamins D2 and D3 available to them derived from UV-exposed skin (vitamin D3), dietary intakes of vitamin D3-rich foods such as egg yolks and oily fish, fortified foods that generally have vitamin D2 fortification, and vitamin supplements that are available both as vitamins D2 and D3 [12]. The concentration of 25(OH)D (25(OH)D2 plus 25(OH)D3 (from here on D is D2 plus D3) in serum is considered the best parameter for the evaluation of the vitamin D status of a subject. Although the reliance on a single cutoff value to define vitamin D deficiency or insufficiency is problematic because of the wide individual variability of the functional effects of vitamin D [13,14], the Endocrine Society defines vitamin D deficiency as a 25(OH)D concentration below 50 nM (20 ng/mL) and vitamin D insufficiency as a 25(OH)D concentration of 51–72 nM (21–29 ng/mL) [12]. There is consensus that severe vitamin D deficiency (serum 25(OH)D concentration < 30 nM) should be corrected, whereas most guidelines recommend serum 25(OH)D concentrations of >50 nM for optimal bone health in older adults [15]. However, the causal link between vitamin D and many extra-skeletal outcomes remains unclear. The serum concentration of the active 1,25(OH)2D is approximately 1000-fold lower (60–110 pM, 25–46 pg/mL) than the 25(OH)D concentration, far below the effective concentration of 1,25(OH)2D applied in the majority of in vitro studies on the effects of vitamin D on immune cells. Thus, more than a 100-fold higher concentration of 1,25(OH)2D than the amount found in serum is often required to obtain an effect in most in vitro studies [16,17,18,19,20,21,22]. It has therefore been suggested that the level of circulating 1,25(OH)2D is too low to affect immune responses in vivo, and that sufficient levels are obtained by the local conversion of 25(OH)D to 1,25(OH)2D [23,24,25]. Accordingly, a number of other tissues than the kidney also express CYP27B1 and can produce 1,25(OH)2D [7,26].
In the skin, keratinocytes, dendritic cells (DCs), and macrophages express CYP27B1 and have the capacity to produce 1,25(OH)2D [11,25,26,27]. The production of 1,25(OH)2D in the skin could serve in regulating skin resident cells, including keratinocytes, immune cells, and, in special cases, malignant cells. Cutaneous T-cell lymphoma (CTCL) is characterized by the proliferation of malignant T cells in inflamed skin lesions that potentially could be affected by vitamin D. The most common variant of CTCL, mycosis fungoides (MF), often presents with lesions in skin areas devoid of sun-induced vitamin D around the abdomen and buttocks, the so-called “bathing suit” distribution. The characteristic distribution of MF has contributed to the idea that sunlight and vitamin D might have a preventive effect against CTCL [28,29,30]. The notion was further accentuated by the recent discovery of an association between vitamin D deficiency and the risk of disease progression in other types of lymphoma [31,32,33] and a recent case study on juvenile CTCL and hypovitaminosis D [34].

2. The Vitamin D Receptor

The cellular actions of 1,25(OH)2D are mediated by the vitamin D receptor (VDR), a ligand-dependent transcription factor belonging to the superfamily of nuclear receptors [35,36]. Following interaction with 1,25(OH)2D, the VDR dimerizes with the retinoid X receptor (RXR) and translocates to the nucleus, where it binds to vitamin D response elements (VDREs) in vitamin D responsive genes. Depending on the target gene, either coactivators or corepressors are attracted to the 1,25(OH)2D/VDR/RXR complexes to induce or repress gene transcription. Figure 1 gives a simplified overview of the vitamin D/VDR signaling pathway. For a more detailed description of vitamin D/VDR-mediated gene regulation, please see [10,37,38,39,40,41,42]. The highest concentration of VDR is found in the intestine, kidney, and bone involved in the maintenance of calcium homeostasis. However, the VDR has been found in many other tissues, including the skin, not involved in calcium homeostasis, suggesting that vitamin D affects many cellular processes beyond the regulation of calcium homeostasis [9,11,43].

3. Effect of 1,25(OH)2D on Cells in the Skin

Many skin cells, including keratinocytes and immune cells, express the VDR [9,11,44]. Acting through the VDR, 1,25(OH)2D regulates the proliferation of the basal layer of the epidermis and the sequential differentiation of keratinocytes [2,11,44,45,46]. The loss of VDR disrupts keratinocyte differentiation and epithelial barrier formation, impairs wound healing, and predisposes to cancer [47,48,49,50,51].
It has long been recognized that 1,25(OH)2D plays an important role in the regulation and function of the immune system [52,53,54,55]. Accordingly, various immune cells express the VDR [52,55,56]. 1,25(OH)2D affects the activation and differentiation of several types of immune cells via the VDR [57]. Among other effects, it promotes the differentiation of monocytes into macrophages and the expression of the antimicrobial peptides cathelicidin and beta-defensin [40,53,58]. In this way, 1,25(OH)2D plays an important role in the immune system’s defense against bacteria and in particular mycobacteria [40,53,58]. In support, patients with genetic defects in the VDR have a compromised cathelicidin response and increased susceptibility to infection with Mycobacterium tuberculosis and other mycobacteria [53,59]. 1,25(OH)2D has recently been ascribed a role in COVID-19 infections, but the clinical importance remains controversial [60]. In addition to its role in the antimicrobial defense, 1,25(OH)2D has also anti-inflammatory effects such as increasing the tolerogenic effect of macrophages and other antigen-presenting cells. This is achieved partly through the inhibition of tumor necrosis factor α (TNF-α) and interleukin (IL)-6 secretion, as well as a decreased expression of Toll-like receptor (TLR) molecules and the class II major histocompatibility complex (MHC II). Furthermore, 1,25(OH)2D can also promote a change toward a tolerogenic phenotype in DC by increasing the expression of inhibitory molecules such as programmed death ligand-1 (PD-L1) [61]. In this way, tolerogenic DC induces T-cell anergy and induction of tolerogenic cytokines (IL-10), i.e., a phenotype resembling that of DC in cutaneous T-cell lymphoma (CTCL) [62].
In the skin, T cells play important roles in protective immunity and the development of inflammatory and autoimmune diseases [63,64,65]. Several studies have examined the role of 1,25(OH)2D and the VDR in T-cell development, differentiation, and function. Studies of mice lacking the VDR and in humans with a defect VDR showed a normal number of CD4+ and CD8+ T cells including naturally occurring CD4+FoxP3+ regulatory T cells [66,67,68], suggesting that the 1,25(OH)2D and VDR are not required for the development of either of these T-cell types. However, other studies indicated that 1,25(OH)2D is required for the normal development and function of invariant natural killer T cells [69] and CD8αα+ intraepithelial lymphocytes [68]. Naïve T cells express no or very low levels of the VDR; however, T-cell activation leads to the rapid upregulation of the VDR [24,56,70,71]. In accordance, 1,25(OH)2D significantly affects T-cell function [53,72,73]. In CD4+ T cells, for instance, VDR ligation leads to the inhibition of interferon (IFN)γ, IL-17A, and IL-22 expression, while it results in an increased expression of IL-4, IL-5, and IL-13 characteristic of T helper cell type 2 (Th2) responses [24,25,52,53,74,75]. In parallel, 1,25(OH)2D favors the expansion of regulator T cells (Treg) and the expression of the regulatory cytokine IL-10, suggesting that 1,25(OH)2D has an additional role in protecting against overt immune reactions [16,52,53,55,76].
Strikingly, solar UV radiation was shown to induce immunosuppression, inhibiting sensibilization in a model of contact hypersensitivity induced by an epicutaneous application of contact allergens [77,78,79]. Soon, it became clear that UV-induced immunosuppression was antigen-specific and mediated by Treg cells, tolerogenic antigen-presenting cells, and IL-10 [80,81,82], and later, it turned out that a multitude of cell types and cytokines—both in the innate and adaptive immune systems as well as in skin stroma cells—were affected by sunlight and involved in the complex immunological events following exposure to UV radiation [81,83]. Keratinocytes—the most abundant cell type in the epidermis—respond to UV radiation in a multitude of ways that directly and indirectly stimulate inflammation, immunosuppression, and DNA damage repair [84]. In addition to vitamin D, keratinocytes synthesize and release other effector molecules, including cytokines and enzymes, following exposure to UVB radiation [84,85].

4. Overview of Cutaneous T-Cell Lymphoma

CTCL belongs to a type of lymphoma collectively called non-Hodgkin’s lymphoma, which is often subdivided into four subgroups: Sezary syndrome (SS), a severe leukemic variant; CD30 positive lymphoproliferative diseases; mycosis fungoides (MF); and finally non-MF variants [86]. A characteristic feature of CTCL is epidermotropism—i.e., the accumulation of malignant T cells to the outermost layer of the skin that is potentially exposed to UV light [87]. CTCL often remains limited to the skin over long periods of disease evolution, and progression with extra-cutaneous spread usually occurs only in advanced late stages.
The most common variant of CTCL, MF, displays a characteristic disease evolution initially with erythematous patches (the patch stage), which may evolve into more infiltrated plaques (the plaque stage) [86]. The patches and plaques usually persist over a long time, but in a subset of patients, the disease develops into large and often ulcerated tumors (the tumor stage) or generalized erythroderma. Skin lesions may be associated with severe pruritus. The prognosis in the patch and limited plaque stage is favorable, whereas the tumor stage and generalized erythroderma MF are aggressive and associated with a risk of spreading outside the skin (extra-cutaneous MF). The type and extent of skin involvement as well as the presence of extra-cutaneous disease are significant prognostic factors [86] guiding the selection and type of therapeutic approaches and the acceptable risk of side effects [88].
Except for allogeneic stem cell transplantation and localized radiotherapy in unilesional MF, there are no curative therapies [89,90]. The treatment of MF initially involves skin-directed therapies (phototherapy/photochemotherapy, topical corticosteroids, and nitrogen mustard), which are the main strategies for early-stage MF. In subjects with later-stage disease, both approved and unapproved agents, including immune modulators, single agent, or combination chemotherapy (methotrexate, retinoids, pegylated interferon, electrophoresis, targeted therapies, and whole-body electron irradiation) are used. Potentially curative treatments are usually only relevant in subpopulations of patients. For example, allogeneic stem cell transplantation is usually only suitable for younger and fit patients with severe disease [89,90,91], whereas most treatment regimens are stage-directed and mainly directed at relieving symptoms and comorbidity. Accordingly, there is an unmet medical need for novel adjuvant therapies that can be combined with and amplify the effect of current forms of treatment.
The etiology of CTCL is largely unknown, and the pathogenesis is far from understood, but both genetic, epigenetic, and environmental factors are believed to be involved [91,92,93,94,95,96]. Disease heterogeneity is a characteristic feature of CTCL [93,97,98,99,100,101] even at the single-cell level [100,102,103,104,105], which has hampered the unraveling of the etiology and pathogenesis and has been a complicating factor in the development of novel therapies. Although high sun exposure is associated with a lower risk of developing MF in Caucasians [106], and heliotherapy (the use of natural sunlight for the treatment—also named climate therapy) [107,108] and phototherapy/photochemotherapy have beneficial effects on CTCL skin lesions [84,109], other studies indicate that UV light is also a driver of mutations in CTCL [110,111,112,113]. Thus, a large part of the mutational burden in MF and SS was linked to a UV signature, which was also observed in CD4+ T cells isolated from the blood of SS patients [111,112,113]. In support, some data indicated an association between occupational sun exposure and MF [110]. Interestingly, abnormally strong skin reactions to UVA and UVB were observed in a third of MF patients, suggesting that an increased sensitivity to UV light could play a pathogenic role in MF [114]. Taken together, these findings suggest that sunlight and UV radiation play complex—even opposing—roles in the initiation/aggravation versus protection/inhibition of the disease. Given the great heterogeneity among CTCL patients, it is also possible that synthesis, turnover, and responses to vitamin D—and its role in pathogenesis—are influenced by disease heterogeneity.
Numerous studies have consistently shown that infections are prevalent and significantly contribute to the morbidity and mortality of CTCL, especially among patients in advanced stages [90,115,116,117,118,119,120,121,122]. In a Danish twin study, susceptibility to severe bacterial infections in CTCL patients was not genetically determined but rather a vulnerability, which developed gradually after the diagnosis had been established [123].
Recent research supports the idea that malignant T cells orchestrate changes in the tumor microenvironment (TME), resulting in the suppression of skin barrier proteins like filaggrin, thereby compromising the skin barrier [124]. Filaggrin components, known for their anti-Staphylococcus aureus (S. aureus) properties, are often depleted in CTCL, making skin lesions vulnerable to colonization, particularly by S. aureus, the most common pathogen in CTCL-related skin infections [116,121,125,126]. Importantly, the risk of skin colonization by S. aureus increases with disease progression, and between one-third and two-thirds of patients with advanced disease harbor S. aureus, a large proportion of which produce toxins such as staphylococcal enterotoxins (SEs) [121,125]. This colonization not only poses a risk for invasive infections but also appears to exacerbate disease activity and potentially contributes to drug resistance in malignant T cells [120,127,128].
Antibiotic therapy often provides temporary relief by reducing skin symptoms and improving disease status; however, the recurrence of S. aureus colonization is common [129,130,131,132,133]. Aggressive antibiotic treatment targeting toxin-producing S. aureus strains has shown promising results, leading to reductions in skin symptoms, disease activity, and the number of malignant T cells in skin lesions [130]. This supports the notion that S. aureus colonization plays a pivotal role in fueling disease progression in CTCL [120]. Because many patients harbor methicillin-resistant S. aureus (MRSA) [132], and lesional skin becomes recolonized shortly after the antibiotic treatment is halted [131], new (antibiotic-free) treatments are warranted, such as engineered bacteriophage-derived endolysins, which selectively kills enterotoxin-producing S. aureus isolated from CTCL skin lesions [134].
In addition to a compromised skin barrier as a likely port of entrance for S. aureus and other bacteria implicated in CTCL [124,135], there is also evidence suggesting a potential deficiency in antimicrobial defense mechanisms such as an impaired expression of antimicrobial peptides including cathelicidin and other molecules involved in the antibacterial defense in the skin [135,136,137,138]. As mentioned above, cathelicidin is a potent antimicrobial peptide, which targets an array of bacteria including S. aureus. As cathelicidin expression is compromised in lesional skin in CTCL patients [138] and is induced by vitamin D in healthy individuals [27,139], it is possible that a deficient expression is due to a deficient vitamin D synthesis in affected skin or, alternatively, a deficient response to vitamin D in CTCL skin lesions.

5. Vitamin D in CTCL

For decades, vitamin D has been suspected to play a role in the pathogenesis of CTCL [94]. Concerning the vitamin D nomenclature, it is known that it is the active form of vitamin D, 1,25(OH)2D, that exerts the biological effects of vitamin D. However, in the literature, the term vitamin D is generally used without discrimination between the various forms of vitamin D. We will follow the same nomenclature for vitamin D in this review if not otherwise stated. The most common variant, MF, often presents in light-protected skin areas devoid of sun-induced vitamin D, which early on prompted the hypothesis that sun-induced vitamin D expression could play a role in the repression of the disease, and inversely, that low levels of vitamin D were permissive for the development of MF [28,140]. In support of this idea, it was reported that a history of greater sun exposure was associated with some degree of protection from contracting some subtypes of non-Hodgkin lymphoma, including CTCL [141,142], and recently, high sun exposure was associated with a lower risk of developing early-stage MF in Caucasians [106]. This notion also gained some indirect support from the observations that the disease incidence increased with distance from the equator for several mature cutaneous T-cell neoplasms, including MF/Sezary syndrome [29], and the observation that greater body surface area involvement was inversely correlated with vitamin D levels in MF patients [28]. Likewise, the beneficial response to phototherapy in CTCL patients has also been ascribed to vitamin D [85], although a multitude of vitamin D-independent mechanisms may also be involved in the clinical effects of phototherapy [143,144]. In addition, vitamin D and vitamin D analogs seem to potentiate the in vitro effect of chemotherapeutic agents such as doxorubicin, sometimes used in the treatment of advanced-stage CTCL patients [145].
Genetic factors involving the VDR have previously been suggested to play a role in CTCL pathogenesis [146,147,148]. One study obtained evidence that a single-nucleotide polymorphism in the VDR-Fokl gene was associated with CTCL [146], whereas other studies did not find this association [147,148]. On the contrary, Incel et al. [147] reported that a single-nucleotide polymorphism in VDR Taq1 was associated with decreased susceptibility to MF. Notably, a recent study of CTCL in a larger cohort of twins found that the disease was not inherited and that genetic predisposition did not play a major role in CTCL [123].

6. Vitamin D Serum Levels in CTCL

A few studies have examined vitamin D levels in CTCL patients compared to the background population, patients with other cancers, and patients with benign inflammatory diseases [28,146,148,149,150]. Some studies reported significant reductions in the serum levels of vitamin D in CTCL patients when compared to the general population and/or matched controls [28,141,149,150], whereas others did not [148]. Notably, the serum levels of vitamin D in CTCL patients were comparable to the serum levels observed in comparable groups of patients with benign dermatoses such as atopic dermatitis and psoriasis [149]. Thus, a compromised vitamin D expression seemed to be due to the inflammatory process rather than the cancer per se.

7. Vitamin D in the Pathogenesis of CTCL

Some studies have addressed the expression of the VDR and the effect of vitamin D in malignant T cells, and they report that primary malignant T cells and T-cell lines express the VDR and respond to vitamin D in relevant doses [150,151]. Notably, vitamin D triggered apoptosis in both primary Sezary syndrome malignant T cells and an MF-derived, malignant T-cell line [150]. Moreover, vitamin D inhibited the spontaneous and induced expression of IL-22 mRNA and protein by malignant T cells [74]. The inhibition was mediated via a repressive vitamin D response element in the IL-22 promoter and independently of JAK/STAT signaling [74], which is constitutively active in malignant T-cell lines [152]. As IL-22 is a survival factor in malignant T cells and is believed to play a role in the pathogenesis through changes in the TME [153], these findings suggest a potential anticancer effect of vitamin D both directly through the inhibition of survival signals in malignant T cells and indirectly through the modulation of the TME (Figure 2, left side). Mrotzek et al. [150] examined the effect of vitamin D on the proliferation of an MF cell line treated with bexarotene routinely used in the treatment of CTCL patients [94] and found that vitamin D inhibited the proliferation of bexarotene-treated cells showing synergistic anticancer effects of vitamin D and bexarotene. In addition, vitamin D inhibits integrin-mediated adhesion by malignant T-cell lines and interactions with other cells in the TME [154], which are believed to promote the proliferation of malignant T cells [155]. Thus, vitamin D may inhibit disease activity via direct effects on the malignant T cell, an increase in sensitivity to treatment, and/or indirectly via the inhibition of interactions with the TME.
It is well established that CTCL patients have an increased susceptibility to bacterial skin infection, which is likely due to a compromised skin barrier and deregulated expression of antimicrobial peptides [124]. As mentioned above, vitamin D plays an important role in the expression of antimicrobial peptides as illustrated by an increased risk of bacterial infections in patients with severe vitamin D deficiency [156,157,158] and genetic conditions such as patients with loss-of-function mutations in the VDR [67]. Therefore, it is possible—but remains to be proven—that vitamin D deficiency could predispose to an increased risk of bacterial skin infections in CTCL patients, and inversely that vitamin D—alone or in combination with antibacterial proteins such as endolysins [134]—could have a preventive effect on bacterial infections in these patients. As bacteria such as enterotoxin-producing S. aureus seem to fuel disease activity and drug resistance in patients with advanced disease [128,130], vitamin D may inhibit disease activity via the induction of antimicrobial peptides (Figure 2, left side).
Most of the symptoms related to CTCL, such as itch, pain, and skin lesions, are the results of inflammatory processes [91,155]. As vitamin D has anti-inflammatory properties [72], it is possible that vitamin D could alleviate inflammation-associated symptoms. On the other hand, vitamin D also promotes a series of immunological changes, which could promote—rather than inhibit—symptoms and disease progression. First, vitamin D enhances Th2 immune responses [24,52,53,55]. As the Th2 cytokines IL-4 and IL-13 are growth factors for malignant T cells and induce the repression of filaggrin in the skin [124,159], vitamin D could indirectly enhance malignant proliferation, disease progression, and the deterioration of the skin barrier (Figure 2, right side). Furthermore, vitamin D inhibits Th1 responses and IL-2-driven proliferation while promoting Treg, tolerogenic macrophages, and dendritic cells, as well as the expression of immune checkpoint molecules such as PD-L1 and PD-L2 [16,21,160,161,162,163,164,165,166], suggesting that vitamin D may also inhibit anticancer immunity (Figure 2, right side). Taken together, these seemingly opposing effects described above indicate that vitamin D may have both beneficial and harmful effects depending on the context, suggesting a complex role of vitamin D in CTCL biology (Figure 2).

8. Effects of Vitamin D Treatment in CTCL

In an early case study, Scott-Mackie et al. [167] were the first to report on the beneficial effect of treatment with topical calcipotriol in a patient with low-grade angioimmunoblastic T-cell lymphoma. Shortly after, French et al. [168] reported that systemic therapy with calcipotriol in combination with acitretin was associated with a clearing of the skin lesions in a patient with CTCL, whereas topical treatment with calcipotriol had no effect. These studies prompted more investigations, but other researchers did not replicate these initial, positive findings. On the contrary, later case studies reported on disease aggravation and progression following treatment with vitamin D in combination with the retinoids acitretin [169] and isotretinoin [170]. Thus, in one patient, new cutaneous lesions and an extra-cutaneous lesion appeared and rapidly increased during treatment with acitretin and calcitriol [169], whereas three patients experienced rapidly increasing skin lesions and tumors following the initiation of treatment with calcitriol and isotretinoin [170], prompting warnings and caution in relation to the use of calcitriol in CTCL. Twenty years later, the first systematic prospective study addressing vitamin D status and the effect of vitamin D supplementation in a larger cohort of 311 CTCL patients was performed [28]. As mentioned above, most patients displayed vitamin D deficiency as judged by the serum levels of vitamin 25(OH)D, which was, however, not significantly different from that observed in comparable patients with other types of cancer. Importantly, the correction of vitamin D deficiency was successful in a large fraction of patients (27–50% depending on the vitamin D treatment regimen). Because the disease course and responses to standard CTCL therapy were similar among patients with corrected and persistently low levels of vitamin D, it was concluded that the correction of vitamin D deficiency did not affect clinical disease activity, response to treatment, or disease course in CTCL patients [28].

9. Phototherapy and Sunlight Modulate Skin Inflammation in Vitamin D Independent Ways

It was the common observation of a “bathing suit” distribution (i.e., skin areas usually protected from sunlight) of skin lesions in MF patients that led to the assumption that sunlight and phototherapy induced its effects through vitamin D production in the skin. However, sunlight and phototherapy affect the skin in multiple ways, several of which are independent of vitamin D. For instance, UV light stimulates DNA damage, platelet-activating factor, the activation of the aryl hydrocarbon receptor, and oxidative stress-related enzymes, all of which, like vitamin D, subsequently stimulate regulatory mechanisms via Treg and Langerhans cells and factors like IL-10, TGFβ, and IDO-1 [84,143,144]. Notably, phototherapy was recently shown to restore a deficient type I IFN production and enhance antitumor responses in MF, providing an alternative explanation for the effects of phototherapy in CTCL [171].

10. Concluding Remarks

The longstanding question concerning the role of vitamin D in the pathogenesis, treatment, and prevention of CTCL remains largely unresolved. Although some epidemiological data suggest a protective role of sunlight, and experimental studies indicate anticancer effects, other data suggest that vitamin D may also have tumor-promoting effects. Likewise, clinical studies provide opposing results or no effect at all. Taken together, these data indicate a complex and incompletely understood role of vitamin D in CTCL that warrants further investigation. Importantly, they do not provide a rationale for vitamin D therapy beyond the personalized treatment of vitamin D deficiency in individual patients.

Author Contributions

Conceptualization, A.-W.F.Ø. and C.G.; writing—original draft preparation, A.-W.F.Ø.; writing—review and editing, A.-W.F.Ø. and C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by LEO Fondet.

Acknowledgments

Thanks are due to Nicolaj Menné Bonefeld for technical support in preparing Figure 1.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tripkovic, L.; Lambert, H.; Hart, K.; Smith, C.P.; Bucca, G.; Penson, S.; Chope, G.; Hyppönen, E.; Berry, J.; Vieth, R.; et al. Comparison of vitamin D2 and vitamin D3 supplementation in raising serum 25-hydroxyvitamin D status: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2012, 95, 1357–1364. [Google Scholar] [CrossRef]
  2. Bikle, D.D. Vitamin D and the skin: Physiology and pathophysiology. Rev. Endocr. Metab. Disord. 2012, 13, 3–19. [Google Scholar] [CrossRef]
  3. Jäpelt, R.B.; Jakobsen, J. Vitamin D in plants: A review of occurrence, analysis, and biosynthesis. Front. Plant Sci. 2013, 4, 136. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. Holick, M.F.; Tian, X.Q.; Allen, M. Evolutionary importance for the membrane enhancement of the production of vitamin D3 in the skin of poikilothermic animals. Proc. Natl. Acad. Sci. USA 1995, 92, 3124–3126. [Google Scholar] [CrossRef]
  6. Holick, M.F.; MacLaughlin, J.A.; Doppelt, S.H. Regulation of cutaneous previtamin D3 photosynthesis in man: Skin pigment is not an essential regulator. Science 1981, 211, 590–593. [Google Scholar] [CrossRef]
  7. Bikle, D.D. Vitamin D metabolism, mechanism of action, and clinical applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef]
  8. Haddad, J.G.; Matsuoka, L.Y.; Hollis, B.W.; Hu, Y.Z.; Wortsman, J. Human plasma transport of vitamin D after its endogenous synthesis. J. Clin. Investig. 1993, 91, 2552–2555. [Google Scholar] [CrossRef] [PubMed]
  9. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, Molecular Mechanism of Action, and Pleiotropic Effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [PubMed]
  10. Pike, J.W.; Christakos, S. Biology and Mechanisms of Action of the Vitamin D Hormone. Endocrinol. Metab. Clin. N. Am. 2017, 46, 815–843. [Google Scholar] [CrossRef]
  11. Bikle, D.; Christakos, S. New aspects of vitamin D metabolism and action—Addressing the skin as source and target. Nat. Rev. Endocrinol. 2020, 16, 234–252. [Google Scholar] [CrossRef]
  12. Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef]
  13. Revez, J.A.; Lin, T.; Qiao, Z.; Xue, A.; Holtz, Y.; Zhu, Z.; Zeng, J.; Wang, H.; Sidorenko, J.; Kemper, K.E.; et al. Genome-wide association study identifies 143 loci associated with 25 hydroxyvitamin D concentration. Nat. Commun. 2020, 11, 1647. [Google Scholar] [CrossRef] [PubMed]
  14. Gospodarska, E.; Ghosh Dastidar, R.; Carlberg, C. Intervention Approaches in Studying the Response to Vitamin D3 Supplementation. Nutrients 2023, 15, 3382. [Google Scholar] [CrossRef] [PubMed]
  15. Bouillon, R.; Manousaki, D.; Rosen, C.; Trajanoska, K.; Rivadeneira, F.; Richards, J.B. The health effects of vitamin D supplementation: Evidence from human studies. Nat. Rev. Endocrinol. 2022, 18, 96–110. [Google Scholar] [CrossRef]
  16. Jeffery, L.E.; Burke, F.; Mura, M.; Zheng, Y.; Qureshi, O.S.; Hewison, M.; Walker, L.S.; Lammas, D.A.; Raza, K.; Sansom, D.M. 1,25-Dihydroxyvitamin D3 and IL-2 combine to inhibit T cell production of inflammatory cytokines and promote development of regulatory T cells expressing CTLA-4 and FoxP3. J. Immunol. 2009, 183, 5458–5467. [Google Scholar] [CrossRef]
  17. Palmer, M.T.; Lee, Y.K.; Maynard, C.L.; Oliver, J.R.; Bikle, D.D.; Jetten, A.M.; Weaver, C.T. Lineage-specific effects of 1,25-dihydroxyvitamin D3 on the development of effector CD4 T cells. J. Biol. Chem. 2011, 286, 997–1004. [Google Scholar] [CrossRef] [PubMed]
  18. Joshi, S.; Pantalena, L.C.; Liu, X.K.; Gaffen, S.L.; Liu, H.; Rohowsky-Kochan, C.; Ichiyama, K.; Yoshimura, A.; Steinman, L.; Christakos, S.; et al. 1,25-dihydroxyvitamin D3 ameliorates Th17 autoimmunity via transcriptional modulation of interleukin-17A. Mol. Cell. Biol. 2011, 31, 3653–3669. [Google Scholar] [CrossRef]
  19. Urry, Z.; Chambers, E.S.; Xystrakis, E.; Dimeloe, S.; Richards, D.F.; Gabrysova, L.; Christensen, J.; Gupta, A.; Saglani, S.; Bush, A.; et al. The role of 1alpha,25-dihydroxyvitamin D3 and cytokines in the promotion of distinct Foxp3+ and IL-10+ CD4+ T cells. Eur. J. Immunol. 2012, 42, 2697–2708. [Google Scholar] [CrossRef] [PubMed]
  20. Joseph, R.W.; Bayraktar, U.D.; Kim, T.K.; St John, L.S.; Popat, U.; Khalili, J.; Molldrem, J.J.; Wieder, E.D.; Komanduri, K.V. Vitamin D receptor upregulation in alloreactive human T cells. Hum. Immunol. 2012, 73, 693–698. [Google Scholar] [CrossRef]
  21. Müller, K.; Odum, N.; Bendtzen, K. 1,25-dihydroxyvitamin D3 selectively reduces interleukin-2 levels and proliferation of human T cell lines in vitro. Immunol. Lett. 1993, 35, 177–182. [Google Scholar] [CrossRef]
  22. Lemire, J.M.; Ince, A.; Takashima, M. 1,25-Dihydroxyvitamin D3 attenuates the expression of experimental murine lupus of MRL/l mice. Autoimmunity 1992, 12, 143–148. [Google Scholar] [CrossRef]
  23. Jeffery, L.E.; Wood, A.M.; Qureshi, O.S.; Hou, T.Z.; Gardner, D.; Briggs, Z.; Kaur, S.; Raza, K.; Sansom, D.M. Availability of 25-hydroxyvitamin D3 to APCs controls the balance between regulatory and inflammatory T cell responses. J. Immunol. 2012, 189, 5155–5164. [Google Scholar] [CrossRef] [PubMed]
  24. Kongsbak, M.; von Essen, M.R.; Levring, T.B.; Schjerling, P.; Woetmann, A.; Odum, N.; Bonefeld, C.M.; Geisler, C. Vitamin D-binding protein controls T cell responses to vitamin D. BMC Immunol. 2014, 15, 35. [Google Scholar] [CrossRef] [PubMed]
  25. Lopez, D.V.; Al-Jaberi, F.A.H.; Woetmann, A.; Ødum, N.; Bonefeld, C.M.; Kongsbak-Wismann, M.; Geisler, C. Macrophages Control the Bioavailability of Vitamin D and Vitamin D-Regulated T Cell Responses. Front. Immunol. 2021, 12, 722806. [Google Scholar] [CrossRef] [PubMed]
  26. Bikle, D.D.; Nemanic, M.K.; Gee, E.; Elias, P. 1,25-Dihydroxyvitamin D3 production by human keratinocytes. Kinetics and regulation. J. Clin. Investig. 1986, 78, 557–566. [Google Scholar] [CrossRef] [PubMed]
  27. Rode, A.K.O.; Kongsbak, M.; Hansen, M.M.; Lopez, D.V.; Levring, T.B.; Woetmann, A.; Odum, N.; Bonefeld, C.M.; Geisler, C. Vitamin D Counteracts Mycobacterium tuberculosis-Induced Cathelicidin Downregulation in Dendritic Cells and Allows Th1 Differentiation and IFNgamma Secretion. Front. Immunol. 2017, 8, 656. [Google Scholar] [CrossRef] [PubMed]
  28. Talpur, R.; Cox, K.M.; Hu, M.; Geddes, E.R.; Parker, M.K.; Yang, B.Y.; Armstrong, P.A.; Liu, P.; Duvic, M. Vitamin D deficiency in mycosis fungoides and Sézary syndrome patients is similar to other cancer patients. Clin. Lymphoma Myeloma Leuk. 2014, 14, 518–524. [Google Scholar] [CrossRef]
  29. van Leeuwen, M.T.; Turner, J.J.; Falster, M.O.; Meagher, N.S.; Joske, D.J.; Grulich, A.E.; Giles, G.G.; Vajdic, C.M. Latitude gradients for lymphoid neoplasm subtypes in Australia support an association with ultraviolet radiation exposure. Int. J. Cancer 2013, 133, 944–951. [Google Scholar] [CrossRef] [PubMed]
  30. Ramsay, D.L.; Lish, K.M.; Yalowitz, C.B.; Soter, N.A. Ultraviolet-B phototherapy for early-stage cutaneous T-cell lymphoma. Arch. Dermatol. 1992, 128, 931–933. [Google Scholar] [CrossRef]
  31. Odutola, M.K.; van Leeuwen, M.T.; Bruinsma, F.; Turner, J.; Hertzberg, M.; Seymour, J.F.; Prince, H.M.; Trotman, J.; Verner, E.; Roncolato, F.; et al. A Population-Based Family Case-Control Study of Sun Exposure and Follicular Lymphoma Risk. Cancer Epidemiol. Biomark. Prev. 2024, 33, 106–116. [Google Scholar] [CrossRef]
  32. Gao, F.; Zhang, T.; Liu, H.; Li, W.; Liu, X.; Qiu, L.; Li, L.; Zhou, S.; Qian, Z.; Dong, S.; et al. Risk factors for POD24 in patients with previously untreated follicular lymphoma: A systematic review and meta-analysis. Ann. Hematol. 2022, 101, 2383–2392. [Google Scholar] [CrossRef]
  33. Tao, Y.; Chen, H.; Zhou, Y.; Shi, Y. Meta-analysis of the prognostic and clinical value of serum 25-hydroxyvitamin D levels in previously untreated lymphoma. Future Oncol. 2021, 17, 1825–1838. [Google Scholar] [CrossRef] [PubMed]
  34. Tung-Hahn, E.; Mogilevskiy, V.; Black, E.; Morgan, M.; Tung, R. Pediatric primary cutaneous anaplastic large-cell lymphoma with associated hypovitaminosis D. Arch. Dermatol. Res. 2023, 316, 50. [Google Scholar] [CrossRef] [PubMed]
  35. Mangelsdorf, D.J.; Thummel, C.; Beato, M.; Herrlich, P.; Schutz, G.; Umesono, K.; Blumberg, B.; Kastner, P.; Mark, M.; Chambon, P.; et al. The nuclear receptor superfamily: The second decade. Cell 1995, 83, 835–839. [Google Scholar] [CrossRef] [PubMed]
  36. Bain, D.L.; Heneghan, A.F.; Connaghan-Jones, K.D.; Miura, M.T. Nuclear receptor structure: Implications for function. Annu. Rev. Physiol. 2007, 69, 201–220. [Google Scholar] [CrossRef] [PubMed]
  37. Haussler, M.R.; Whitfield, G.K.; Kaneko, I.; Haussler, C.A.; Hsieh, D.; Hsieh, J.C.; Jurutka, P.W. Molecular mechanisms of vitamin D action. Calcif. Tissue Int. 2013, 92, 77–98. [Google Scholar] [CrossRef]
  38. Nagpal, S.; Na, S.; Rathnachalam, R. Noncalcemic actions of vitamin D receptor ligands. Endocr. Rev. 2005, 26, 662–687. [Google Scholar] [CrossRef]
  39. Pike, J.W.; Meyer, M.B.; Bishop, K.A. Regulation of target gene expression by the vitamin D receptor—An update on mechanisms. Rev. Endocr. Metab. Disord. 2012, 13, 45–55. [Google Scholar] [CrossRef]
  40. Carlberg, C. Genomic signaling of vitamin D. Steroids 2023, 198, 109271. [Google Scholar] [CrossRef]
  41. Carlberg, C. Vitamin D and Its Target Genes. Nutrients 2022, 14, 1354. [Google Scholar] [CrossRef] [PubMed]
  42. Voltan, G.; Cannito, M.; Ferrarese, M.; Ceccato, F.; Camozzi, V. Vitamin D: An Overview of Gene Regulation, Ranging from Metabolism to Genomic Effects. Genes 2023, 14, 1691. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, Y.; Zhu, J.; DeLuca, H.F. Where is the vitamin D receptor? Arch. Biochem. Biophys. 2012, 523, 123–133. [Google Scholar] [CrossRef] [PubMed]
  44. Reichrath, J.; Zouboulis, C.C.; Vogt, T.; Holick, M.F. Targeting the vitamin D endocrine system (VDES) for the management of inflammatory and malignant skin diseases: An historical view and outlook. Rev. Endocr. Metab. Disord. 2016, 17, 405–417. [Google Scholar] [CrossRef] [PubMed]
  45. Hosomi, J.; Hosoi, J.; Abe, E.; Suda, T.; Kuroki, T. Regulation of terminal differentiation of cultured mouse epidermal cells by 1 alpha,25-dihydroxyvitamin D3. Endocrinology 1983, 113, 1950–1957. [Google Scholar] [CrossRef] [PubMed]
  46. Smith, E.L.; Walworth, N.C.; Holick, M.F. Effect of 1 alpha,25-dihydroxyvitamin D3 on the morphologic and biochemical differentiation of cultured human epidermal keratinocytes grown in serum-free conditions. J. Investig. Dermatol. 1986, 86, 709–714. [Google Scholar] [CrossRef] [PubMed]
  47. 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]
  48. Tu, C.L.; Crumrine, D.A.; Man, M.Q.; Chang, W.; Elalieh, H.; You, M.; Elias, P.M.; Bikle, D.D. Ablation of the calcium-sensing receptor in keratinocytes impairs epidermal differentiation and barrier function. J. Investig. Dermatol. 2012, 132, 2350–2359. [Google Scholar] [CrossRef]
  49. 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]
  50. Zinser, G.M.; Sundberg, J.P.; Welsh, J. Vitamin D3 receptor ablation sensitizes skin to chemically induced tumorigenesis. Carcinogenesis 2002, 23, 2103–2109. [Google Scholar] [CrossRef]
  51. Ellison, T.I.; Smith, M.K.; Gilliam, A.C.; MacDonald, P.N. Inactivation of the vitamin D receptor enhances susceptibility of murine skin to UV-induced tumorigenesis. J. Investig. Dermatol. 2008, 128, 2508–2517. [Google Scholar] [CrossRef] [PubMed]
  52. Peelen, E.; Knippenberg, S.; Muris, A.H.; Thewissen, M.; Smolders, J.; Tervaert, J.W.; Hupperts, R.; Damoiseaux, J. Effects of vitamin D on the peripheral adaptive immune system: A review. Autoimmun. Rev. 2011, 10, 733–743. [Google Scholar] [CrossRef] [PubMed]
  53. Kongsbak, M.; Levring, T.B.; Geisler, C.; von Essen, M.R. The vitamin D receptor and T cell function. Front. Immunol. 2013, 4, 148. [Google Scholar] [CrossRef] [PubMed]
  54. Charoenngam, N.; Holick, M.F. Immunologic Effects of Vitamin D on Human Health and Disease. Nutrients 2020, 12, 2097. [Google Scholar] [CrossRef] [PubMed]
  55. Baeke, F.; Takiishi, T.; Korf, H.; Gysemans, C.; Mathieu, C. Vitamin D: Modulator of the immune system. Curr. Opin. Pharmacol. 2010, 10, 482–496. [Google Scholar] [CrossRef] [PubMed]
  56. Baeke, F.; Korf, H.; Overbergh, L.; van Etten, E.; Verstuyf, A.; Gysemans, C.; Mathieu, C. Human T lymphocytes are direct targets of 1,25-dihydroxyvitamin D3 in the immune system. J. Steroid Biochem. Mol. Biol. 2010, 121, 221–227. [Google Scholar] [CrossRef] [PubMed]
  57. Carlberg, C.; Campbell, M.J. Vitamin D receptor signaling mechanisms: Integrated actions of a well-defined transcription factor. Steroids 2013, 78, 127–136. [Google Scholar] [CrossRef] [PubMed]
  58. Wimalawansa, S.J. Infections and Autoimmunity-The Immune System and Vitamin D: A Systematic Review. Nutrients 2023, 15, 3842. [Google Scholar] [CrossRef]
  59. Al-Jaberi, F.A.H.; Crone, C.G.; Lindenstrøm, T.; Arildsen, N.S.; Lindeløv, E.S.; Aagaard, L.; Gravesen, E.; Mortensen, R.; Andersen, A.B.; Olgaard, K.; et al. Reduced vitamin D-induced cathelicidin production and killing of Mycobacterium tuberculosis in macrophages from a patient with a non-functional vitamin D receptor: A case report. Front. Immunol. 2022, 13, 1038960. [Google Scholar] [CrossRef]
  60. Feentved Ødum, S.L.; Kongsbak-Wismann, M. Vitamin D and SARS-CoV-2. Basic Clin. Pharmacol. Toxicol. 2023, 133, 6–15. [Google Scholar] [CrossRef]
  61. Zhang, L.; Xia, C.Q. PD-1/PD-L1 Interaction Maintains Allogeneic Immune Tolerance Induced by Administration of Ultraviolet B-Irradiated Immature Dendritic Cells. J. Immunol. Res. 2016, 2016, 2419621. [Google Scholar] [CrossRef]
  62. Berger, C.L.; Hanlon, D.; Kanada, D.; Dhodapkar, M.; Lombillo, V.; Wang, N.; Christensen, I.; Howe, G.; Crouch, J.; El-Fishawy, P.; et al. The growth of cutaneous T-cell lymphoma is stimulated by immature dendritic cells. Blood 2002, 99, 2929–2939. [Google Scholar] [CrossRef] [PubMed]
  63. Sabat, R.; Wolk, K.; Loyal, L.; Döcke, W.D.; Ghoreschi, K. T cell pathology in skin inflammation. Semin. Immunopathol. 2019, 41, 359–377. [Google Scholar] [CrossRef] [PubMed]
  64. Nomura, T.; Kabashima, K.; Miyachi, Y. The panoply of αβT cells in the skin. J. Dermatol. Sci. 2014, 76, 3–9. [Google Scholar] [CrossRef]
  65. Pham, J.P.; Wark, K.J.L.; Woods, J.; Frew, J.W. Resident cutaneous memory T cells: A clinical review of their role in chronic inflammatory dermatoses and potential as therapeutic targets. Br. J. Dermatol. 2023, 189, 656–663. [Google Scholar] [CrossRef] [PubMed]
  66. Mathieu, C.; van Etten, E.; Gysemans, C.; Decallonne, B.; Kato, S.; Laureys, J.; Depovere, J.; Valckx, D.; Verstuyf, A.; Bouillon, R. In vitro and in vivo analysis of the immune system of vitamin D receptor knockout mice. J. Bone Miner. Res. 2001, 16, 2057–2065. [Google Scholar] [CrossRef] [PubMed]
  67. Al-Jaberi, F.A.H.; Kongsbak-Wismann, M.; Aguayo-Orozco, A.; Krogh, N.; Buus, T.B.; Lopez, D.V.; Rode, A.K.O.; Gravesen, E.; Olgaard, K.; Brunak, S.; et al. Impaired Vitamin D Signaling in T Cells From a Family With Hereditary Vitamin D Resistant Rickets. Front. Immunol. 2021, 12, 684015. [Google Scholar] [CrossRef]
  68. Yu, S.; Bruce, D.; Froicu, M.; Weaver, V.; Cantorna, M.T. Failure of T cell homing, reduced CD4/CD8alphaalpha intraepithelial lymphocytes, and inflammation in the gut of vitamin D receptor KO mice. Proc. Natl. Acad. Sci. USA 2008, 105, 20834–20839. [Google Scholar] [CrossRef]
  69. Yu, S.; Cantorna, M.T. The vitamin D receptor is required for iNKT cell development. Proc. Natl. Acad. Sci. USA 2008, 105, 5207–5212. [Google Scholar] [CrossRef]
  70. Kongsbak, M.; von Essen, M.R.; Boding, L.; Levring, T.B.; Schjerling, P.; Lauritsen, J.P.; Woetmann, A.; Odum, N.; Bonefeld, C.M.; Geisler, C. Vitamin D up-regulates the vitamin D receptor by protecting it from proteasomal degradation in human CD4+ T cells. PLoS ONE 2014, 9, e96695. [Google Scholar] [CrossRef]
  71. von Essen, M.R.; Kongsbak, M.; Schjerling, P.; Olgaard, K.; Odum, N.; Geisler, C. Vitamin D controls T cell antigen receptor signaling and activation of human T cells. Nat. Immunol. 2010, 11, 344–349. [Google Scholar] [CrossRef]
  72. Daryabor, G.; Gholijani, N.; Kahmini, F.R. A review of the critical role of vitamin D axis on the immune system. Exp. Mol. Pathol. 2023, 132–133, 104866. [Google Scholar] [CrossRef]
  73. 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]
  74. Lopez, D.V.; Al-Jaberi, F.A.H.; Damas, N.D.; Weinert, B.T.; Pus, U.; Torres-Rusillo, S.; Woetmann, A.; Ødum, N.; Bonefeld, C.M.; Kongsbak-Wismann, M.; et al. Vitamin D Inhibits IL-22 Production Through a Repressive Vitamin D Response Element in the il22 Promoter. Front. Immunol. 2021, 12, 715059. [Google Scholar] [CrossRef] [PubMed]
  75. Boonstra, A.; Barrat, F.J.; Crain, C.; Heath, V.L.; Savelkoul, H.F.; O’Garra, A. 1alpha,25-Dihydroxyvitamin d3 has a direct effect on naive CD4+ T cells to enhance the development of Th2 cells. J. Immunol. 2001, 167, 4974–4980. [Google Scholar] [CrossRef] [PubMed]
  76. Barrat, F.J.; Cua, D.J.; Boonstra, A.; Richards, D.F.; Crain, C.; Savelkoul, H.F.; de Waal-Malefyt, R.; Coffman, R.L.; Hawrylowicz, C.M.; O’Garra, A. In vitro generation of interleukin 10-producing regulatory CD4+ T cells is induced by immunosuppressive drugs and inhibited by T helper type 1 (Th1)- and Th2-inducing cytokines. J. Exp. Med. 2002, 195, 603–616. [Google Scholar] [CrossRef] [PubMed]
  77. Toews, G.B.; Bergstresser, P.R.; Streilein, J.W. Epidermal Langerhans cell density determines whether contact hypersensitivity or unresponsiveness follows skin painting with DNFB. J. Immunol. 1980, 124, 445–453. [Google Scholar] [CrossRef] [PubMed]
  78. De Fabo, E.C.; Noonan, F.P. Mechanism of immune suppression by ultraviolet irradiation in vivo. I. Evidence for the existence of a unique photoreceptor in skin and its role in photoimmunology. J. Exp. Med. 1983, 158, 84–98. [Google Scholar] [CrossRef] [PubMed]
  79. Elmets, C.A.; Bergstresser, P.R.; Tigelaar, R.E.; Wood, P.J.; Streilein, J.W. Analysis of the mechanism of unresponsiveness produced by haptens painted on skin exposed to low dose ultraviolet radiation. J. Exp. Med. 1983, 158, 781–794. [Google Scholar] [CrossRef]
  80. Robert, C.; Kupper, T.S. Inflammatory skin diseases, T cells, and immune surveillance. N. Engl. J. Med. 1999, 341, 1817–1828. [Google Scholar] [CrossRef]
  81. Schwarz, T. The dark and the sunny sides of UVR-induced immunosuppression: Photoimmunology revisited. J. Investig. Dermatol. 2010, 130, 49–54. [Google Scholar] [CrossRef]
  82. Kang, K.; Hammerberg, C.; Meunier, L.; Cooper, K.D. CD11b+ macrophages that infiltrate human epidermis after in vivo ultraviolet exposure potently produce IL-10 and represent the major secretory source of epidermal IL-10 protein. J. Immunol. 1994, 153, 5256–5264. [Google Scholar] [CrossRef]
  83. Hart, P.H.; Grimbaldeston, M.A.; Swift, G.J.; Jaksic, A.; Noonan, F.P.; Finlay-Jones, J.J. Dermal mast cells determine susceptibility to ultraviolet B-induced systemic suppression of contact hypersensitivity responses in mice. J. Exp. Med. 1998, 187, 2045–2053. [Google Scholar] [CrossRef] [PubMed]
  84. Vieyra-Garcia, P.A.; Wolf, P. From Early Immunomodulatory Triggers to Immunosuppressive Outcome: Therapeutic Implications of the Complex Interplay Between the Wavebands of Sunlight and the Skin. Front. Med. 2018, 5, 232. [Google Scholar] [CrossRef] [PubMed]
  85. Juzeniene, A.; Grigalavicius, M.; Juraleviciute, M.; Grant, W.B. Phototherapy and vitamin D. Clin. Dermatol. 2016, 34, 548–555. [Google Scholar] [CrossRef] [PubMed]
  86. Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef] [PubMed]
  87. El Bedewi, A.; El Anany, G.; El Mofty, M. Role of Synchrotron infra red microspectroscopy in studying epidermotropism of cutaneous T-cell lymphoma. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 1047–1050. [Google Scholar] [CrossRef] [PubMed]
  88. Sivanand, A.; Surmanowicz, P.; Alhusayen, R.; Hull, P.; Litvinov, I.V.; Zhou, Y.; Gniadecki, R. Immunotherapy for Cutaneous T-Cell Lymphoma: Current Landscape and Future Developments. J. Cutan. Med. Surg. 2019, 23, 537–544. [Google Scholar] [CrossRef] [PubMed]
  89. Assaf, C.; Waser, N.; Bagot, M.; He, M.; Li, T.; Dalal, M.; Gavini, F.; Trinchese, F.; Zomas, A.; Little, M.; et al. Contemporary Treatment Patterns and Response in Relapsed/Refractory Cutaneous T-Cell Lymphoma (CTCL) across Five European Countries. Cancers 2021, 14, 145. [Google Scholar] [CrossRef]
  90. Dai, J.; Duvic, M. Cutaneous T-Cell Lymphoma: Current and Emerging Therapies. Oncology 2023, 37, 55–62. [Google Scholar] [CrossRef]
  91. Weiner, D.M.; Durgin, J.S.; Wysocka, M.; Rook, A.H. The immunopathogenesis and immunotherapy of cutaneous T cell lymphoma: Current and future approaches. J. Am. Acad. Dermatol. 2021, 84, 597–604. [Google Scholar] [CrossRef]
  92. Dummer, R.; Vermeer, M.H.; Scarisbrick, J.J.; Kim, Y.H.; Stonesifer, C.; Tensen, C.P.; Geskin, L.J.; Quaglino, P.; Ramelyte, E. Cutaneous T cell lymphoma. Nat. Rev. Dis. Primers 2021, 7, 61. [Google Scholar] [CrossRef] [PubMed]
  93. Ghazawi, F.M.; Alghazawi, N.; Le, M.; Netchiporouk, E.; Glassman, S.J.; Sasseville, D.; Litvinov, I.V. Environmental and Other Extrinsic Risk Factors Contributing to the Pathogenesis of Cutaneous T Cell Lymphoma (CTCL). Front. Oncol. 2019, 9, 300. [Google Scholar] [CrossRef] [PubMed]
  94. Gordon, E.R.; Adeuyan, O.; Schreidah, C.M.; Chen, C.; Trager, M.H.; Lapolla, B.A.; Fahmy, L.M.; Weng, C.; Geskin, L.J. Clusters, crop dusters, and myth busters: A scoping review of environmental exposures and cutaneous T-cell lymphoma. Ital. J. Dermatol. Venerol. 2023, 158, 467–482. [Google Scholar] [CrossRef] [PubMed]
  95. Ghazawi, F.M.; Netchiporouk, E.; Rahme, E.; Tsang, M.; Moreau, L.; Glassman, S.; Provost, N.; Gilbert, M.; Jean, S.E.; Roshdy, O.; et al. Distribution and Clustering of Cutaneous T-Cell Lymphoma (CTCL) Cases in Canada During 1992 to 2010. J. Cutan. Med. Surg. 2018, 22, 154–165. [Google Scholar] [CrossRef]
  96. Ghazawi, F.M.; Netchiporouk, E.; Rahme, E.; Tsang, M.; Moreau, L.; Glassman, S.; Provost, N.; Gilbert, M.; Jean, S.E.; Pehr, K.; et al. Comprehensive analysis of cutaneous T-cell lymphoma (CTCL) incidence and mortality in Canada reveals changing trends and geographic clustering for this malignancy. Cancer 2017, 123, 3550–3567. [Google Scholar] [CrossRef] [PubMed]
  97. Litvinov, I.V.; Tetzlaff, M.T.; Thibault, P.; Gangar, P.; Moreau, L.; Watters, A.K.; Netchiporouk, E.; Pehr, K.; Prieto, V.G.; Rahme, E.; et al. Gene expression analysis in Cutaneous T-Cell Lymphomas (CTCL) highlights disease heterogeneity and potential diagnostic and prognostic indicators. Oncoimmunology 2017, 6, e1306618. [Google Scholar] [CrossRef] [PubMed]
  98. Choi, J.; Goh, G.; Walradt, T.; Hong, B.S.; Bunick, C.G.; Chen, K.; Bjornson, R.D.; Maman, Y.; Wang, T.; Tordoff, J.; et al. Genomic landscape of cutaneous T cell lymphoma. Nat. Genet. 2015, 47, 1011–1019. [Google Scholar] [CrossRef]
  99. da Silva Almeida, A.C.; Abate, F.; Khiabanian, H.; Martinez-Escala, E.; Guitart, J.; Tensen, C.P.; Vermeer, M.H.; Rabadan, R.; Ferrando, A.; Palomero, T. The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome. Nat. Genet. 2015, 47, 1465–1470. [Google Scholar] [CrossRef]
  100. Buus, T.B.; Willerslev-Olsen, A.; Fredholm, S.; Blümel, E.; Nastasi, C.; Gluud, M.; Hu, T.; Lindahl, L.M.; Iversen, L.; Fogh, H.; et al. Single-cell heterogeneity in Sézary syndrome. Blood Adv. 2018, 2, 2115–2126. [Google Scholar] [CrossRef]
  101. Jiang, T.T.; Cao, S.; Kruglov, O.; Virmani, A.; Geskin, L.J.; Falo, L.D., Jr.; Akilov, O.E. Deciphering Tumor Cell Evolution in Cutaneous T-Cell Lymphomas: Distinct Differentiation Trajectories in Mycosis Fungoides and Sézary Syndrome. J. Investig. Dermatol. 2023. [Google Scholar] [CrossRef]
  102. Rindler, K.; Bauer, W.M.; Jonak, C.; Wielscher, M.; Shaw, L.E.; Rojahn, T.B.; Thaler, F.M.; Porkert, S.; Simonitsch-Klupp, I.; Weninger, W.; et al. Single-Cell RNA Sequencing Reveals Tissue Compartment-Specific Plasticity of Mycosis Fungoides Tumor Cells. Front. Immunol. 2021, 12, 666935. [Google Scholar] [CrossRef]
  103. Gaydosik, A.M.; Tabib, T.; Geskin, L.J.; Bayan, C.A.; Conway, J.F.; Lafyatis, R.; Fuschiotti, P. Single-Cell Lymphocyte Heterogeneity in Advanced Cutaneous T-cell Lymphoma Skin Tumors. Clin. Cancer Res. 2019, 25, 4443–4454. [Google Scholar] [CrossRef] [PubMed]
  104. Hamrouni, A.; Fogh, H.; Zak, Z.; Ødum, N.; Gniadecki, R. Clonotypic Diversity of the T-cell Receptor Corroborates the Immature Precursor Origin of Cutaneous T-cell Lymphoma. Clin. Cancer Res. 2019, 25, 3104–3114. [Google Scholar] [CrossRef] [PubMed]
  105. Borcherding, N.; Voigt, A.P.; Liu, V.; Link, B.K.; Zhang, W.; Jabbari, A. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin. Cancer Res. 2019, 25, 2996–3005. [Google Scholar] [CrossRef] [PubMed]
  106. DeStefano, C.B.; Desale, S.; Fernandez, S.J.; Shenoy, A.G. The impact of environmental ultraviolet exposure on the clinical course of mycosis fungoides. J. Am. Acad. Dermatol. 2019, 81, 1074–1077. [Google Scholar] [CrossRef] [PubMed]
  107. Linser, K.; Harnack, K. Heliotherapy of mycosis fungoides. Arch. Klin. Exp. Dermatol. 1962, 215, 181–201. [Google Scholar] [CrossRef]
  108. Tippel, H.; Engst, R. Mycosis fungoides. Results of helioclimate therapy in high mountains (Davos, 1,560). Hautarzt 1986, 37, 450–453. [Google Scholar] [PubMed]
  109. Hodak, E.; Pavlovsky, L. Phototherapy of Mycosis Fungoides. Dermatol. Clin. 2015, 33, 697–702. [Google Scholar] [CrossRef]
  110. Morales-Suárez-Varela, M.M.; Olsen, J.; Johansen, P.; Kaerlev, L.; Guénel, P.; Arveux, P.; Wingren, G.; Hardell, L.; Ahrens, W.; Stang, A.; et al. Occupational sun exposure and mycosis fungoides: A European multicenter case-control study. J. Occup. Environ. Med. 2006, 48, 390–393. [Google Scholar] [CrossRef]
  111. Song, X.; Chang, S.; Seminario-Vidal, L.; de Mingo Pulido, A.; Tordesillas, L.; Song, X.; Reed, R.A.; Harkins, A.; Whiddon, S.; Nguyen, J.V.; et al. Genomic and Single-Cell Landscape Reveals Novel Drivers and Therapeutic Vulnerabilities of Transformed Cutaneous T-cell Lymphoma. Cancer Discov. 2022, 12, 1294–1313. [Google Scholar] [CrossRef]
  112. Jones, C.L.; Degasperi, A.; Grandi, V.; Amarante, T.D.; Mitchell, T.J.; Nik-Zainal, S.; Whittaker, S.J. Spectrum of mutational signatures in T-cell lymphoma reveals a key role for UV radiation in cutaneous T-cell lymphoma. Sci. Rep. 2021, 11, 3962. [Google Scholar] [CrossRef]
  113. Gniadecki, R.; O’Keefe, S.; Hennessey, D.; Iyer, A. Is Cutaneous T-Cell Lymphoma Caused by Ultraviolet Radiation? A Comparison of UV Mutational Signatures in Malignant Melanoma and Mycosis Fungoides. Cells 2023, 12, 1616. [Google Scholar] [CrossRef]
  114. Volden, G. A study of the photosensitive factor in relation to skin lesions of mycosis fungoides patients. Dermatologica 1980, 161, 89–92. [Google Scholar] [CrossRef] [PubMed]
  115. Allen, P.B.; Switchenko, J.; Ayers, A.; Kim, E.; Lechowicz, M.J. Risk of bacteremia in patients with cutaneous T-cell lymphoma (CTCL). Leuk. Lymphoma 2020, 61, 2652–2658. [Google Scholar] [CrossRef]
  116. Axelrod, P.I.; Lorber, B.; Vonderheid, E.C. Infections complicating mycosis fungoides and Sézary syndrome. JAMA 1992, 267, 1354–1358. [Google Scholar] [CrossRef] [PubMed]
  117. Bonin, S.; Tothova, S.M.; Barbazza, R.; Brunetti, D.; Stanta, G.; Trevisan, G. Evidence of multiple infectious agents in mycosis fungoides lesions. Exp. Mol. Pathol. 2010, 89, 46–50. [Google Scholar] [CrossRef]
  118. Dobos, G.; Assaf, C. Transcriptomic changes during stage progression of mycosis fungoides: From translational analyses to their potential clinical implications. Br. J. Dermatol. 2022, 186, 387–388. [Google Scholar] [CrossRef] [PubMed]
  119. Mirvish, E.D.; Pomerantz, R.G.; Geskin, L.J. Infectious agents in cutaneous T-cell lymphoma. J. Am. Acad. Dermatol. 2011, 64, 423–431. [Google Scholar] [CrossRef]
  120. Willerslev-Olsen, A.; Krejsgaard, T.; Lindahl, L.M.; Bonefeld, C.M.; Wasik, M.A.; Koralov, S.B.; Geisler, C.; Kilian, M.; Iversen, L.; Woetmann, A.; et al. Bacterial toxins fuel disease progression in cutaneous T-cell lymphoma. Toxins 2013, 5, 1402–1421. [Google Scholar] [CrossRef]
  121. Liu, X.; Sun, J.; Gao, Y.; Liu, F.; Pan, H.; Tu, P.; Wang, Y. Characteristics of Staphylococcus aureus Colonization in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2024, 144, 188–191. [Google Scholar] [CrossRef]
  122. Guenova, E.; Ødum, N. Old Sins Cast Long Shadows: News on Staphylococcus aureus in Cutaneous T Cell Lymphoma. J. Investig. Dermatol. 2024, 144, 8–10. [Google Scholar] [CrossRef]
  123. Odum, N.; Lindahl, L.M.; Wod, M.; Krejsgaard, T.; Skytthe, A.; Woetmann, A.; Iversen, L.; Christensen, K. Investigating heredity in cutaneous T-cell lymphoma in a unique cohort of Danish twins. Blood Cancer J. 2017, 7, e517. [Google Scholar] [CrossRef]
  124. Gluud, M.; Pallesen, E.M.H.; Buus, T.B.; Gjerdrum, L.M.R.; Lindahl, L.M.; Kamstrup, M.R.; Bzorek, M.; Danielsen, M.; Bech, R.; Monteiro, M.N.; et al. Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signaling in cutaneous T-cell lymphoma. Blood 2023, 141, 180–193. [Google Scholar] [CrossRef] [PubMed]
  125. Jackow, C.M.; Cather, J.C.; Hearne, V.; Asano, A.T.; Musser, J.M.; Duvic, M. Association of erythrodermic cutaneous T-cell lymphoma, superantigen-positive Staphylococcus aureus, and oligoclonal T-cell receptor V beta gene expansion. Blood 1997, 89, 32–40. [Google Scholar] [CrossRef]
  126. Nguyen, V.; Huggins, R.H.; Lertsburapa, T.; Bauer, K.; Rademaker, A.; Gerami, P.; Guitart, J. Cutaneous T-cell lymphoma and Staphylococcus aureus colonization. J. Am. Acad. Dermatol. 2008, 59, 949–952. [Google Scholar] [CrossRef]
  127. Kadin, M.E.; Hamilton, R.G.; Vonderheid, E.C. Evidence linking atopy and staphylococcal superantigens to the pathogenesis of lymphomatoid papulosis, a recurrent CD30+ cutaneous lymphoproliferative disorder. PLoS ONE 2020, 15, e0228751. [Google Scholar] [CrossRef] [PubMed]
  128. Vadivel, C.K.; Willerslev-Olsen, A.; Namini, M.R.J.; Zeng, Z.; Yan, L.; Danielsen, M.; Gluud, M.; Pallesen, E.M.H.; Wojewoda, K.; Osmancevic, A.; et al. Staphylococcus aureus induce drug resistance in cancer T cells in Sézary Syndrome. Blood 2024. [Google Scholar] [CrossRef]
  129. Talpur, R.; Bassett, R.; Duvic, M. Prevalence and treatment of Staphylococcus aureus colonization in patients with mycosis fungoides and Sézary syndrome. Br. J. Dermatol. 2008, 159, 105–112. [Google Scholar] [CrossRef]
  130. Lindahl, L.M.; Willerslev-Olsen, A.; Gjerdrum, L.M.R.; Nielsen, P.R.; Blümel, E.; Rittig, A.H.; Celis, P.; Herpers, B.; Becker, J.C.; Stausbøl-Grøn, B.; et al. Antibiotics inhibit tumor and disease activity in cutaneous T-cell lymphoma. Blood 2019, 134, 1072–1083. [Google Scholar] [CrossRef]
  131. Lindahl, L.M.; Iversen, L.; Ødum, N.; Kilian, M. Staphylococcus aureus and Antibiotics in Cutaneous T-Cell Lymphoma. Dermatology 2022, 238, 551–553. [Google Scholar] [CrossRef]
  132. Emge, D.A.; Bassett, R.L.; Duvic, M.; Huen, A.O. Methicillin-resistant Staphylococcus aureus (MRSA) is an important pathogen in erythrodermic cutaneous T-cell lymphoma (CTCL) patients. Arch. Dermatol. Res. 2020, 312, 283–288. [Google Scholar] [CrossRef]
  133. Lewis, D.J.; Holder, B.B.; Duvic, M. The “Duvic regimen” for erythrodermic flares secondary to Staphylococcus aureus in mycosis fungoides and Sézary syndrome. Int. J. Dermatol. 2018, 57, 123–124. [Google Scholar] [CrossRef] [PubMed]
  134. Pallesen, E.M.H.; Gluud, M.; Vadivel, C.K.; Buus, T.B.; de Rooij, B.; Zeng, Z.; Ahmad, S.; Willerslev-Olsen, A.; Röhrig, C.; Kamstrup, M.R.; et al. Endolysin Inhibits Skin Colonization by Patient-Derived Staphylococcus Aureus and Malignant T-Cell Activation in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2023, 143, 1757–1768. [Google Scholar] [CrossRef] [PubMed]
  135. Suga, H.; Sugaya, M.; Miyagaki, T.; Ohmatsu, H.; Kawaguchi, M.; Takahashi, N.; Fujita, H.; Asano, Y.; Tada, Y.; Kadono, T.; et al. Skin barrier dysfunction and low antimicrobial peptide expression in cutaneous T-cell lymphoma. Clin. Cancer Res. 2014, 20, 4339–4348. [Google Scholar] [CrossRef] [PubMed]
  136. Wolk, K.; Mitsui, H.; Witte, K.; Gellrich, S.; Gulati, N.; Humme, D.; Witte, E.; Gonsior, M.; Beyer, M.; Kadin, M.E.; et al. Deficient cutaneous antibacterial competence in cutaneous T-cell lymphomas: Role of Th2-mediated biased Th17 function. Clin. Cancer Res. 2014, 20, 5507–5516. [Google Scholar] [CrossRef] [PubMed]
  137. Nakajima, R.; Miyagaki, T.; Kamijo, H.; Oka, T.; Shishido-Takahashi, N.; Suga, H.; Sugaya, M.; Sato, S. Decreased progranulin expression in Mycosis fungoides: A possible association with the high frequency of skin infections. Eur. J. Dermatol. 2018, 28, 790–794. [Google Scholar] [CrossRef] [PubMed]
  138. Sawada, Y.; Nakamura, M.; Kabashima-Kubo, R.; Shimauchi, T.; Kobayashi, M.; Tokura, Y. Defective epidermal innate immunity and resultant superficial dermatophytosis in adult T-cell leukemia/lymphoma. Clin. Cancer Res. 2012, 18, 3772–3779. [Google Scholar] [CrossRef] [PubMed]
  139. Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef]
  140. Drake, M.T.; Maurer, M.J.; Link, B.K.; Habermann, T.M.; Ansell, S.M.; Micallef, I.N.; Kelly, J.L.; Macon, W.R.; Nowakowski, G.S.; Inwards, D.J.; et al. Vitamin D insufficiency and prognosis in non-Hodgkin’s lymphoma. J. Clin. Oncol. 2010, 28, 4191–4198. [Google Scholar] [CrossRef]
  141. Kelly, J.L.; Drake, M.T.; Fredericksen, Z.S.; Asmann, Y.W.; Liebow, M.; Shanafelt, T.D.; Feldman, A.L.; Ansell, S.M.; Macon, W.R.; Herr, M.M.; et al. Early life sun exposure, vitamin D-related gene variants, and risk of non-Hodgkin lymphoma. Cancer Causes Control 2012, 23, 1017–1029. [Google Scholar] [CrossRef]
  142. Kricker, A.; Armstrong, B.K.; Hughes, A.M.; Goumas, C.; Smedby, K.E.; Zheng, T.; Spinelli, J.J.; De Sanjosé, S.; Hartge, P.; Melbye, M.; et al. Personal sun exposure and risk of non Hodgkin lymphoma: A pooled analysis from the Interlymph Consortium. Int. J. Cancer 2008, 122, 144–154. [Google Scholar] [CrossRef]
  143. Yu, Z.; Wolf, P. How It Works: The Immunology Underlying Phototherapy. Dermatol. Clin. 2020, 38, 37–53. [Google Scholar] [CrossRef]
  144. Vieyra-Garcia, P.A.; Wolf, P. A deep dive into UV-based phototherapy: Mechanisms of action and emerging molecular targets in inflammation and cancer. Pharmacol. Ther. 2021, 222, 107784. [Google Scholar] [CrossRef]
  145. Ma, Y.; Trump, D.L.; Johnson, C.S. Vitamin D in combination cancer treatment. J. Cancer 2010, 1, 101–107. [Google Scholar] [CrossRef]
  146. Rasheed, H.; Hegazy, R.A.; Gawdat, H.I.; Mehaney, D.A.; Kamel, M.M.; Fawzy, M.M.; Nooh, M.M.; Darwish, H.A. Serum Vitamin D and Vitamin D Receptor Gene Polymorphism in Mycosis Fungoides Patients: A Case Control Study. PLoS ONE 2016, 11, e0158014. [Google Scholar] [CrossRef] [PubMed]
  147. Incel Uysal, P.; Alli, N.; Hayran, Y.; Candar, T. Mycosis Fungoides and Vitamin D Status: Analyses of Serum 25-Hydroxyvitamin D Levels and Single Nucleotide Polymorphisms in the Vitamin D Receptor Gene. Acta Dermatovenerol. Croat. 2018, 26, 8–14. [Google Scholar] [PubMed]
  148. Velissari, A.; Lakiotaki, E.; Nikolaou, V.; Argyropoulos, K.V.; Stratigos, A.; Daikos, G.; Konstantopoulos, K.; Siakantaris, M. Genetic polymorphisms in immunity related genes and the vitamin D receptor gene and risk of cutaneous T-cell lymphoma in Greek population. J. Eur. Acad. Dermatol. Venereol. 2021, 35, e805–e807. [Google Scholar] [CrossRef] [PubMed]
  149. Kaul, S.; Belzberg, M.; Hughes, J.M.; Mahadevan, V.; Khanna, R.; Bakhshi, P.R.; Hong, M.S.; Williams, K.A.; Grossberg, A.L.; Kwatra, S.G.; et al. Comorbidities in Mycosis Fungoides and Racial Differences in Co-Existent Lymphomatoid Papulosis: A Cross-Sectional Study of 580 Patients in an Urban Tertiary Care Center. Medicines 2019, 7, 1. [Google Scholar] [CrossRef] [PubMed]
  150. Mrotzek, C.; Felcht, M.; Sommer, A.; Schrader, A.; Klemke, C.D.; Herling, M.; Schlaak, M.; Fabri, M. Vitamin D controls apoptosis and proliferation of cutaneous T-cell lymphoma cells. Exp. Dermatol. 2015, 24, 798–800. [Google Scholar] [CrossRef]
  151. Kim, E.J.; Hess, S.; Richardson, S.K.; Newton, S.; Showe, L.C.; Benoit, B.M.; Ubriani, R.; Vittorio, C.C.; Junkins-Hopkins, J.M.; Wysocka, M.; et al. Immunopathogenesis and therapy of cutaneous T cell lymphoma. J. Clin. Investig. 2005, 115, 798–812. [Google Scholar] [CrossRef]
  152. Ødum, N. Deregulated signalling and inflammation in cutaneous T-cell lymphoma. Br. J. Dermatol. 2020, 182, 16–17. [Google Scholar] [CrossRef]
  153. Matsuda, Y.; Ikeda, S.; Abe, F.; Takahashi, Y.; Kitadate, A.; Takahashi, N.; Wakui, H.; Tagawa, H. Downregulation of miR-26 promotes invasion and metastasis via targeting interleukin-22 in cutaneous T-cell lymphoma. Cancer Sci. 2022, 113, 1208–1219. [Google Scholar] [CrossRef]
  154. Wang, L.; DeMarco, S.S.; Chen, J.; Phillips, C.M.; Bridges, L.C. Retinoids Bias Integrin Expression and Function in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2015, 135, 2102–2108. [Google Scholar] [CrossRef]
  155. Krejsgaard, T.; Lindahl, L.M.; Mongan, N.P.; Wasik, M.A.; Litvinov, I.V.; Iversen, L.; Langhoff, E.; Woetmann, A.; Odum, N. Malignant inflammation in cutaneous T-cell lymphoma-a hostile takeover. Semin. Immunopathol. 2017, 39, 269–282. [Google Scholar] [CrossRef]
  156. Liu, P.T.; Stenger, S.; Li, H.; Wenzel, L.; Tan, B.H.; Krutzik, S.R.; Ochoa, M.T.; Schauber, J.; Wu, K.; Meinken, C.; et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 2006, 311, 1770–1773. [Google Scholar] [CrossRef] [PubMed]
  157. Zasloff, M. Inducing endogenous antimicrobial peptides to battle infections. Proc. Natl. Acad. Sci. USA 2006, 103, 8913–8914. [Google Scholar] [CrossRef] [PubMed]
  158. Gunville, C.F.; Mourani, P.M.; Ginde, A.A. The role of vitamin D in prevention and treatment of infection. Inflamm. Allergy Drug Targets 2013, 12, 239–245. [Google Scholar] [CrossRef]
  159. Gaydosik, A.M.; Queen, D.S.; Trager, M.H.; Akilov, O.E.; Geskin, L.J.; Fuschiotti, P. Genome-wide transcriptome analysis of the STAT6-regulated genes in advanced-stage cutaneous T-cell lymphoma. Blood 2020, 136, 1748–1759. [Google Scholar] [CrossRef]
  160. Adorini, L.; Penna, G. Dendritic cell tolerogenicity: A key mechanism in immunomodulation by vitamin D receptor agonists. Hum. Immunol. 2009, 70, 345–352. [Google Scholar] [CrossRef] [PubMed]
  161. Li, W.; Liu, Z.; Tang, R.; Ouyang, S.; Li, S.; Wu, J. Vitamin D inhibits palmitate-induced macrophage pro-inflammatory cytokine production by targeting the MAPK pathway. Immunol. Lett. 2018, 202, 23–30. [Google Scholar] [CrossRef] [PubMed]
  162. Dimitrov, V.; Bouttier, M.; Boukhaled, G.; Salehi-Tabar, R.; Avramescu, R.G.; Memari, B.; Hasaj, B.; Lukacs, G.L.; Krawczyk, C.M.; White, J.H. Hormonal vitamin D up-regulates tissue-specific PD-L1 and PD-L2 surface glycoprotein expression in humans but not mice. J. Biol. Chem. 2017, 292, 20657–20668. [Google Scholar] [CrossRef] [PubMed]
  163. Lovato, P.; Norsgaard, H.; Tokura, Y.; Ropke, M.A. Calcipotriol and betamethasone dipropionate exert additive inhibitory effects on the cytokine expression of inflammatory dendritic cell-Th17 cell axis in psoriasis. J. Dermatol. Sci. 2016, 81, 153–164. [Google Scholar] [CrossRef] [PubMed]
  164. Thien, R.; Baier, K.; Pietschmann, P.; Peterlik, M.; Willheim, M. Interactions of 1 alpha,25-dihydroxyvitamin D3 with IL-12 and IL-4 on cytokine expression of human T lymphocytes. J. Allergy Clin. Immunol. 2005, 116, 683–689. [Google Scholar] [CrossRef] [PubMed]
  165. Bendix, M.; Greisen, S.; Dige, A.; Hvas, C.L.; Bak, N.; Jørgensen, S.P.; Dahlerup, J.F.; Deleuran, B.; Agnholt, J. Vitamin D increases programmed death receptor-1 expression in Crohn’s disease. Oncotarget 2017, 8, 24177–24186. [Google Scholar] [CrossRef]
  166. Koren, R.; Liberman, U.A.; Maron, L.; Novogrodsky, A.; Ravid, A. 1,25-Dihydroxyvitamin D3 acts directly on human lymphocytes and interferes with the cellular response to interleukin-2. Immunopharmacology 1989, 18, 187–194. [Google Scholar] [CrossRef]
  167. Scott-Mackie, P.; Hickish, T.; Mortimer, P.; Sloane, J.; Cunningham, D. Calcipotriol and regression in T-cell lymphoma of skin. Lancet 1993, 342, 172. [Google Scholar] [CrossRef]
  168. French, L.E.; Ramelet, A.A.; Saurat, J.H. Remission of cutaneous T-cell lymphoma with combined calcitriol and acitretin. Lancet 1994, 344, 686–687. [Google Scholar] [CrossRef]
  169. Bagot, M. Treatment of cutaneous T-cell lymphoma by retinoids and calcitriol. Lancet 1995, 346, 376–377. [Google Scholar] [PubMed]
  170. Thomsen, K. Cutaneous T-cell lymphoma and calcitriol and isotretinoin treatment. Lancet 1995, 345, 1583. [Google Scholar] [CrossRef] [PubMed]
  171. Yu, Z.; Vieyra-Garcia, P.; Benezeder, T.; Crouch, J.D.; Kim, I.R.; O’Malley, J.T.; Devlin, P.M.; Gehad, A.; Zhan, Q.; Gudjonsson, J.E.; et al. Phototherapy Restores Deficient Type I IFN Production and Enhances Antitumor Responses in Mycosis Fungoides. J. Investig. Dermatol. 2023, 144, 621–632. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Simplified overview of the vitamin D/VDR signaling pathway. PM, plasma membrane; VDR, vitamin D receptor; RXR, retinoid X receptor; VDRE, vitamin D response element.
Figure 1. Simplified overview of the vitamin D/VDR signaling pathway. PM, plasma membrane; VDR, vitamin D receptor; RXR, retinoid X receptor; VDRE, vitamin D response element.
Cells 13 00503 g001
Figure 2. The many anti- and pro-cancer roles of vitamin D in CTCL. IL, interleukin; Th2, T helper 2 cell; DC, dendritic cell; PD-L1, programmed death ligand-1; Treg, regulatory T cell.
Figure 2. The many anti- and pro-cancer roles of vitamin D in CTCL. IL, interleukin; Th2, T helper 2 cell; DC, dendritic cell; PD-L1, programmed death ligand-1; Treg, regulatory T cell.
Cells 13 00503 g002
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

Ødum, A.-W.F.; Geisler, C. Vitamin D in Cutaneous T-Cell Lymphoma. Cells 2024, 13, 503. https://doi.org/10.3390/cells13060503

AMA Style

Ødum A-WF, Geisler C. Vitamin D in Cutaneous T-Cell Lymphoma. Cells. 2024; 13(6):503. https://doi.org/10.3390/cells13060503

Chicago/Turabian Style

Ødum, August-Witte Feentved, and Carsten Geisler. 2024. "Vitamin D in Cutaneous T-Cell Lymphoma" Cells 13, no. 6: 503. https://doi.org/10.3390/cells13060503

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