Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Seasonal Variation in Vitamin D3 Levels Is Paralleled by Changes in the Peripheral Blood Human T Cell Compartment

  • Ai-Leng Khoo ,

    louisaileng@gmail.com

    Affiliations Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands

  • Hans J. P. M. Koenen,

    Affiliations Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands

  • Louis Y. A. Chai,

    Affiliations Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands, Department of Medicine, National University Health System, Singapore, Singapore

  • Fred C. G. J. Sweep,

    Affiliation Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands

  • Mihai G. Netea,

    Affiliations Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands

  • André J. A. M. van der Ven,

    Affiliations Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands

  • Irma Joosten

    Affiliations Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands, Nijmegen Institute for Infection, Inflammation and Immunity, Nijmegen, The Netherlands

Abstract

It is well-recognized that vitamin D3 has immune-modulatory properties and that the variation in ultraviolet (UV) exposure affects vitamin D3 status. Here, we investigated if and to what extent seasonality of vitamin D3 levels are associated with changes in T cell numbers and phenotypes. Every three months during the course of the entire year, human PBMC and whole blood from 15 healthy subjects were sampled and analyzed using flow cytometry. We observed that elevated serum 25(OH)D3 and 1,25(OH)2D3 levels in summer were associated with a higher number of peripheral CD4+ and CD8+ T cells. In addition, an increase in naïve CD4+CD45RA+ T cells with a reciprocal drop in memory CD4+CD45RO+ T cells was observed. The increase in CD4+CD45RA+ T cell count was a result of heightened proliferative capacity rather than recent thymic emigration of T cells. The percentage of Treg dropped in summer, but not the absolute Treg numbers. Notably, in the Treg population, the levels of forkhead box protein 3 (Foxp3) expression were increased in summer. Skin, gut and lymphoid tissue homing potential was increased during summer as well, exemplified by increased CCR4, CCR6, CLA, CCR9 and CCR7 levels. Also, in summer, CD4+ and CD8+ T cells revealed a reduced capacity to produce pro-inflammatory cytokines. In conclusion, seasonal variation in vitamin D3 status in vivo throughout the year is associated with changes in the human peripheral T cell compartment and may as such explain some of the seasonal variation in immune status which has been observed previously. Given that the current observations are limited to healthy adult males, larger population-based studies would be useful to validate these findings.

Introduction

Vitamin D3 is traditionally associated with bone homeostasis and calcium metabolism. The extra-renal synthesis of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] by macrophages and other immune cells has re-invented the role of vitamin D3. In recent years, research efforts were also focused on understanding the immunemodulatory properties of vitamin D3. 1,25-dihydroxyvitamin D3 has been shown to influence the growth and differentiation of both the innate and acquired immune cells, as well as their functions such as cytokine production [1][3]. As such, there has been much interest to identify its therapeutic potential in autoimmune or inflammatory diseases.

Sources of vitamin D3 include dietary uptake (primarily fatty fish and cod liver oil) as well as cutaneous biosynthesis from UVB exposure causing 7-dehydrocholestrol to form previtamin D3 in the skin. Vitamin D3 is subsequently hydroxylated into 25-hydroxyvitamin D3 [25(OH)D3] by 25-hydroxylase in the liver. 25-hydroxyvitamin D3 is further hydroxylated by 1α-hydroxylase in the kidney into the biologically active metabolite, 1,25(OH)2D3 [4]. The main source of vitamin D3 derives from UVB-induced vitamin D3 production, accounting for 80–90% of circulating vitamin D3 [5].

The seasonal variation in vitamin D3 status in temperate and cold climates with reduced sunlight exposure during certain periods of the year is thought to be responsible for the high prevalence of vitamin D3 insufficiency among populations residing at higher latitudes [6]. Low wintertime vitamin D3 levels have been found partly accountable for the seasonal peak in influenza and URTI occurrence [7][9]. Moreover, reduced sun exposure and vitamin D3 status have been identified as risk factors for the development of autoimmune diseases. Epidemiological studies have implicated seasonality of birth as well as geographical variation in UV radiation and serum vitamin D3 levels as contributing factors to the prevalence of multiple sclerosis and insulin-dependent diabetes mellitus [10][15].

T cells are known targets for 1,25(OH)2D3 since they express vitamin D receptor [16], [17]. Upon T cell activation, the expression of vitamin D receptor is up-regulated, suggesting an important functional role for vitamin D3 in adaptive immunity. Both human in vitro and animal models revealed that vitamin D3 can suppress pro-inflammatory T helper (Th)1 and Th17 cytokine responses [18], [19], while enhancing the production of interleukin (IL)-4, IL-5 and IL-10, thereby promoting a Th2 and regulatory T cell (Treg) phenotype [20], [21]. Indeed, accumulating evidence supports the notion that vitamin D3 could favorably influence the course of certain autoimmune pathology by increasing the number of Treg [13], [15]. In addition, chemokine receptors expression is a determining factor in migration and localization of T lymphocytes during physiological and inflammatory responses [22], [23]. 1,25(OH)2D3 has been demonstrated to affect the homing capacity of the peripheral CD4+ T cell population in vitro and in an animal model [24], [25].

Taken together, the involvement of 1,25(OH)2D3 in the dynamics of T cell compartment warrants further investigation. Previously, we have found a down-regulation of Toll-like receptor (TLR)4-mediated proinflammatory cytokines production in association with an elevated vitamin D3 status in summer [26]. However, our current knowledge on the immunomodulatory role of vitamin D3 conveys limited information on how the adaptive immune response of healthy individuals varies in response to physiological changes in vitamin D3 status in vivo during the different seasons of the year. Intrigued by the strong epidemiological association between vitamin D3 deficiency and autoimmunity, and the proposed effects of 1,25(OH)2D3 on Treg, we investigated whether there is a seasonal variation in the composition of the peripheral T cell pool and the circulating Treg. A potential modification in these parameters may provide a better understanding on how sun exposure and vitamin D3 can act as candidate risk-modifying factors in certain autoimmune disorders.

Materials and Methods

Study subjects

Fifteen healthy male volunteers (median 36 years old, range 28–60; mean BMI 22.8 kg/m2, range 20.5–26.2) were recruited and followed up for one year. Body mass index (BMI) has been shown to be inversely related to vitamin D3 levels [27]. We have eliminated this confounder from our study since none of the 15 volunteers was obese (BMI>30 kg/m2). Venous blood was drawn from the subjects every three months, at the end of four consecutive seasons in 2009; February in winter, May in spring, August in summer and November in autumn. On the rare occasions that a participant reported on being unwell, the experiment would be postponed until one week post-recovery.

Ethics Statement

The study was approved by the Ethical Committee on Human Experimentation of the Radboud University Nijmegen. A written consent was obtained from all participants in the study.

Flowcytometry

Cells were phenotypically analyzed by five-color flow cytometry (Coulter Cytomics FC 500, Beckman Coulter, Fullerton, USA) using Coulter Epics Expo 32 software. PBMC as well as whole blood (after red cell lysis) were used for flow cytometric analysis. Peripheral blood mononuclear cells (PBMC) were isolated by density centrifugation on Ficoll-Hypaque (Pharmacia Biotech, Uppsala, Sweden). Cells were washed with PBS with 0.2% bovine serum albumin (BSA) before being labeled with fluorochrome-conjugated antibodies (mAb). After incubation for 20 minutes at room temperature, in the dark, cells were washed twice to remove unbound antibodies and analyzed. For cell surface staining, the following mAb were used: CD127 PC5- or PC7-labeled (RDR5; eBioscience, Uithoorn, The Netherlands), CD25-PE (M-A251), CD25-APC (2A3) CD45RA-FITC (HI100), CCR4-PC7 (1G1), CCR6-PE (11A9), CLA-FITC (HECA-452) (all from BD Biosciences, Breda, The Netherlands), CD3-ECD (UCHT1), CD4 ECD- or PC7-labeled (SFCI12T4D11), CD4-PC5 (13B8.2), CD8-ECD (SFCI21Thy2D3), CD8-PC5 (B9.11), CD27-PC5 (1A4CD27), CD45RA-ECD (2H4LDH11LDB9) CD45RO-ECD (UCHL1) (all from Beckman Coulter, Mijdrecht The Netherlands), CCR7-FITC (150503), CCR9-PE (112509) (both from R&D Systems, Minneapolis, USA), CD27-FITC (M-T271), CD45-PE (T29/33), CD45RA-PE (4KB5) (both from Dako, Glostrup, Denmark) and CD31 Alexa Fluor® 488 (WM59) (BioLegend, San Diego, USA). Appropriate isotype control mAbs were used for gate settings. The live gate was set based on the forward angle light scatter (FSCs) and the side angle light scatter (SSCs), and Annexin-V/PI staining.

For intracellular staining of FoxP3 and Ki-67, cells were fixed and permeabilized using Fix and Perm reagent (eBioscience) according to the manufacturer's recommendations. The following mAb were used for staining: anti-FoxP3 FITC- or PE- labeled (FCH101; eBioscience), anti-Ki-67-FITC (B56, BD Biosciences).

Intracellular staining of cytokines was performed after 4 hours stimulation with PMA (12.5 ng/ml) and ionomycin (500 ng/ml) in the presence of Brefeldin A (5 µg/ml; Sigma, Zwijndrecht, The Netherlands). Cells were fixed and permeabilized using Fix and Perm reagent (eBioscience) according to the manufacturer's recommendations. The following mAb were used for staining: anti-IFNγ-PC7 (4S.B3), anti-IL-17-Alexa Fluor® 647 (eBIO64DEC17) (both from eBioscience), and anti-IL-2-PE (MQ1-17H12) (BD Bioscience).

Vitamin D3 measurement

Serum 25(OH)D3 was determined using high-performance liquid chromatography (HPLC) with ultraviolet (UV) detection, after prior extraction on small SepPak columns as previously described [28]. Tritiated 25(OH)D3, collected from the HPLC system during passage of the UV peak, was used to correct for procedural losses. Serum 1,25(OH)2D3 was measured using a radioreceptor assay (RRA) with prior extraction and chromatographic pre-purification with correction for recovery as previously described [29]. For 25(OH)D3, the within run precision was 2.6% at 69 nmol/l and between run precision was 6.2% at 69 nmol/l. For 1,25(OH)2D3, the within run precision was 10.6% at 115 pmol/l and between run precision was 17.2% at 69 nmol/l.

Statistical analysis

Results were pooled and analyzed using SPSS 16.0 statistical software. Data given as means+SEM and the Analysis of Variance (ANOVA) was performed to assess overall variation. Where the ANOVA indicated a significant difference (p<0.05), the Friedman test using Graphpad PRISM software (Graphpad Prism Inc., version 4, CA, USA) was used to compare differences between groups (unless otherwise stated). The level of significance was set at p<0.05.

Results

Serum 25(OH)D3 and 1,25(OH)2D3 levels are increased during summer

First, we determined serum concentrations of both 25(OH)D3 and 1,25(OH)2D3 in 15 healthy volunteers (median 36 years old, range 28–60) through winter (December to February), spring (March to May), summer (June to August) and autumn (September to November). The median concentration of 25(OH)D3 varied between the four seasons and was doubled from 43 nmol/l in winter to 89 nmol/l in summer (Figure 1A). Also, the median serum concentration of 1,25(OH)2D3 raised significantly from 219 pmol/l in winter to 237 pmol/l in summer (Figure 1B). These observed trends paralleled the amount of sunlight in the study region. Likewise, there is considerable seasonal difference in ultraviolet B (UVB) radiation in the study region [30]. The total duration of daylight in a month prior to vitamin D3 measurement were 103 hours and 240 hours in winter and summer respectively, which worked out to an average daily duration of 3.3 hours in winter and 7.7 hours in summer (Figure 1C).

thumbnail
Figure 1. Seasonal variation in serum vitamin D3 levels and the amount of daylight.

Median serum A) 25(OH)D3 and B) 1,25(OH)2D3 concentrations of 15 healthy volunteers during each of the four seasons. C) Duration of daylight in the study region in a month prior to serum vitamin D3 concentration assay (source: the Royal Netherlands Meteorological Institute). * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g001

Seasonal variation in peripheral blood T cell subset numbers associated with vitamin D3 levels

Next, we investigated whether seasonal variation in vitamin D3 status was associated with changes in the peripheral T cell pool, by performing flowcytometric analysis on blood samples obtained during the different seasons of the year (Figure S1). In spring and summer months when serum vitamin D3 levels were elevated, the percentage as well as the absolute CD4+ T cell counts were significantly raised as compared to winter (Figure 2A). For CD8+ T cells, this effect was less outspoken (Figure 2B).

thumbnail
Figure 2. Peripheral T cell (subset) numbers throughout the four seasons.

A) Percentage (of live gate) and absolute numbers of CD4+ T cells. B) Percentage (of live gate) and absolute numbers of CD8+ T cells, over time. C) Percentage (within CD4+ T cells) and absolute counts, of CD4+CD45RA+ T cells. D) Percentage (within CD4+ T cells) and absolute counts, of CD4+CD45RO+ T cells. E) Percentage and absolute counts of Ki-67-expressing CD4+CD45RA+ T cells. Whole blood samples obtained from 15 healthy volunteers during each season were analyzed for the respective markers using flow cytometry. Ki-67 analysis was performed on PBMC. Data show results of viable cells from 15 healthy donors. * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g002

The composition and size of the naïve and memory T cell pools are regulated by cytokines and T cell receptor (TCR) signalling from contact with major histocompatibility complex (MHC). Naïve T cells predominately express CD45RA and memory T cell express CD45RO. Interestingly, during spring and summer, we observed a relative increase in the percentage of CD4+CD45RA+ T cells (Figure 2C), with a corresponding drop in CD4+CD45RO+ T cell percentage (Figure 2D). Also, absolute CD4+CD45RA+ T cell counts were increased in spring and summer months, while the number of CD4+CD45RO+ T cells was not significantly changed. To investigate whether the increase in peripheral CD4+CD45RA+ T cells as observed in spring and summer could be attributed to recent thymic emigration or a higher proliferative capacity; we stained cells with Ki-67 and CD31. Ki-67 is a nuclear protein associated with cellular proliferation, while CD31 has been used as a marker for recent thymic emigrants [31]. In spring and summer, an increased Ki-67-expressing population was found within the CD4+CD45RA+ T cells (Figure 2E). On the other hand, there were no significant differences in both the frequency of CD4+CD45RA+ T cell expressing CD31 as well as their level of expression between winter and summer (data not shown).

The increase in vitamin D3 levels found in summer, as compared to winter was paralleled by a reduction in the percentage of CD25hiCD127 Treg within the CD4+ T cell population (Figure 3A), however the absolute Treg numbers were not associated with the variation in vitamin D3 levels. Of note, the level of expression (mean fluorescence intensity, MFI) of Foxp3 by the peripheral regulatory T cell population was increased in summer (Figure 3B).

thumbnail
Figure 3. Seasonal variation in numbers and Foxp3 expression of Treg during the four seasons.

A) Percentage (within CD4+ T cells) and absolute numbers of CD4+CD25hiCD127 Treg and B) level of Foxp3 expression (mean fluorescence intensity; MFI). Whole blood and PBMC isolated from 15 healthy volunteers during each season were analyzed for the respective markers using flowcytometry. Data show results from 15 healthy donors. * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g003

Seasonal variation in homing potential of peripheral blood CD4+ T cells

Peripheral T cell trafficking is regulated by specific chemokine receptors which are selectively expressed by the various T cells subsets. As 1,25(OH)2D3 has been demonstrated to affect the homing capacity of the peripheral CD4+ T cell population in vitro and in vivo, we wondered if we could detect seasonal variation in homing receptors expression. We looked at the expression of homing markers on CD4+ T cells, as well as more specifically on the Treg population, and included chemokine receptors associated with migration to the skin (CCR4, CCR6 and CLA), gut (CCR9) and lymphoid tissues (CCR7).

In summer, an increased skin homing potential of CD4+ T cells was observed compared to winter, given that the percentage of CD4+ T cells expressing CCR4 and CCR6 (Figure 4A,B) was significantly increased together with elevated expression levels (MFI) of CCR4, CCR6 and CLA (Figure 4A–C). Also, the percentage of CD4+ T cells expressing the gut homing marker CCR9 was increased in summer, as well as the level of expression (Figure 4D). Similar observations were seen in the expression level of the chemokine receptor associated with lymphoid tissue homing, CCR7 (Figure 4E).

thumbnail
Figure 4. Skin, lymphoid tissue and gut homing receptor expression on CD4+ T cells.

Percentage and level of expression (MFI) of A) CCR4, B) CCR6, C) CLA, D) CCR7 and E) CCR9 by CD4+ T cells during the different seasons of the year. Whole blood from 15 healthy volunteers during each season was analyzed for the respective markers using flow cytometry. Data show results from 15 healthy donors. * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g004

The skin homing potential of the regulatory T cell subset mirrored that of the whole peripheral CD4+ T cell population. Notably, in especially in summer Treg displayed a heightened skin homing potential as seen by a significantly increased frequency of CCR4-expressing Treg (Figure 5A), and higher expression levels of CCR4, CCR6 and CLA (Figure 5A–C), when compared to winter. The level of expression (MFI) of chemokine receptors involved in gut homing, CCR9 (Figure 5D) and lymphoid tissue homing, CCR7 (Figure 5E) were also increased in summer.

thumbnail
Figure 5. Skin, lymphoid tissue and gut homing receptor expression on CD4+CD25hiCD127 regulatory T cells.

Percentage of Treg (within CD4+ T cells) and their level of expression (MFI) of A) CCR4, B) CCR6, C) CLA, D) CCR7 and E) CCR9 during the four seasons of the year. Whole blood from 15 healthy volunteers during each season was analyzed for the respective markers using flow cytometry. Data show results from 15 healthy donors. * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g005

Reduced proinflammatory cytokine production by peripheral blood T cells in summer

Intrigued by the increased CD4+ T cell numbers in spring and summer, we also looked at functional characteristics of the cells by examining the cytokine-producing capacity of CD4+ T cells using intracellular cytokine staining for interferon (IFN)γ, IL-2 and IL-17. There was no significant effect on the percentage of IFNy-producing CD4+ T cells (Figure 6A), but the level of expression was lowered in summer (p<0.05). The percentages of IL-2 and IL-17- secreting CD4+ T cells were reduced in summer (Figure 6B and 6C), with unchanged levels of production on a per cell basis.

thumbnail
Figure 6. Seasonal variation in cytokine profile of CD4+ and CD8+ T cells.

Percentage and the level of production on a per cell basis (MFI) of A) IFNγ, B) IL-2 and C) IL-17 by CD4+ T cells; and of D) IFNγ and E) IL-2 by CD8+ T cells analyzed by flow cytometry. PBMC isolated from 10 healthy volunteers and intracellular staining for cytokines was performed after the cells were stimulated with PMA plus ionomycin in the presence of brefeldin A. CD4+ T cells were defined as CD3+CD8. Data show results from 10 healthy donors. * p<0.05 as compared to winter.

https://doi.org/10.1371/journal.pone.0029250.g006

Also for CD8+ T cells we found lowered levels of IFNy production from spring to autumn (Figure 6D). The percentage of CD8+ T cells producing IL-2 was significantly reduced from spring to autumn (Figure 6E); expression levels were increased during spring.

Discussion

There is growing evidence that vitamin D3 plays a pivotal role in infections and autoimmune diseases. Whilst UV-induced vitamin D3 production serves as the main source of vitamin D3 in the body [5], it is not apparent whether seasonal variation in vitamin D3 can impact T cell immunity. We show for the first time that physiological elevation in vitamin D3 concentrations during summer is paralleled by changes in the peripheral T cell composition, with a notable shift in the naïve and memory CD4+ T cell balance as a consequence of increased proliferation of naïve CD4+CD45RA+ T cells.

By virtue of its stability and long half-life, 25(OH)D3 is the vitamin D metabolite that best reflects the vitamin D3 status [32]. Here, we found a significant difference between winter (December to February) and summer (June to August) 25(OH)D3 levels. Serum 1,25(OH)2D3 concentrations were also higher in summer as compared to winter. In our cohort of 15 subjects residing at 52°N from the Equator, this variation correlated with the amount of sunlight and ultraviolet B radiation received in the study region. Vitamin D3 insufficiency at high latitudes has been implicated in the prevalence of autoimmune diseases such as multiple sclerosis and insulin-dependent diabetes [33], [34]. Therefore, we investigated whether the peripheral T cell compartment might vary with physiological changes in vitamin D3 status throughout the year.

We found higher percentages of peripheral CD4+ and CD8+ T cells concomitant with a heightened vitamin D3 status during summer. Of note, we observed a higher proportion of CD4+CD45RA+ naïve T cells in the spring/summer months with a corresponding drop in the percentage, but not in the absolute number, of CD4+CD45RO+ memory T cells. When investigated further, the expansion of CD4+CD45RA+ naïve T cells resulted from an increased proliferative capacity as seen by a higher absolute cell count and an increased population expressing the proliferative marker, Ki-67. One of the key targets of 1,25(OH)2D3 are the CD4+ T cells. In vitro, 1,25(OH)2D3 inhibits T cell proliferation [35], [36]. Though few studies examined the differential effects on naïve and memory T cells, the inhibitory effect has been found to be more pronounced in the memory T cell compartment [37].

1,25(OH)2D3 exerts a marked inhibitory effect on cells of the adaptive immune system and it has been consistently described that 1,25(OH)2D3 inhibits cytokines such as IFNγ [21], [38] and IL-17, as well as IL-2 [19], [39], both under in vitro conditions and in animal models. Our data reveal that in healthy adult males residing at 52°N from the Equator, the percentages of IL-17- and IL-2-producing CD4+ T cells were down-regulated in summer and the IFNy expression levels in both CD4+ and CD8+ T cells were also reduced.

Regulatory T cells are characterized by a constitutively high expression of the transcription factor, Foxp3. We observed that, although the percentage of peripheral Treg was lower in summer as compared to winter, there was no correlation between absolute numbers of Treg and vitamin D3 levels. This is in concert with findings of Smolders et al, who failed to detect a correlation between Treg numbers and serum 25(OH)D3 levels in patients with multiple sclerosis [40]. Of note, they did find that higher 25(OH)D3 levels were associated with improved suppressive function. This fits our data on increased expression of Foxp3 in the Treg during summer. Morales-Tirado et al reported that in vitro, 1,25(OH)2D3 enhanced Treg function by increasing the expression of Foxp3 and that this was shown to be associated with modulation of cell cycle progression by vitamin D3 [41].

T cell migration is determined by the presence of specific selectins, chemokine receptors and integrins. Homing receptors are selectively expressed and regulated in different T cell subsets [23], [42]. Our results are suggestive of a vitamin-D3 associated up-regulation of skin, gut- and lymphoid tissue- homing expression on CD4+ T cells, including Treg. Although not previously described in the context of physiological variation, 1,25(OH)2D3 has been reported to influence certain skin homing markers in human T cells. In vitro, it has been shown that addition of 1,25(OH)2D3 resulted in induction of CCR10, inhibition of CLA, but not CCR4 and CCR6 expression [38], [43]. In our study, we found that during summer an increased frequency of CCR4-expressing cells as well as an increased level of expression (mean fluorescence intensity; MFI) of CCR4, CCR6 and CLA. These data suggest that in summer CD4+ T cells, including Treg, are better equipped to migrate to the skin. Also, we observed higher levels of CCR9 and thus heightened potential to migrate to the gut. Previously, 1,25(OH)2D3 was described not to affect gut-homing markers [25]. However, it should be appreciated that the physiological up-regulation of vitamin D3 levels by UV light through the skin is likely to yield distinct effects from those obtained through supraphysiological doses employed in these in vitro studies.

In the present study, we assessed a homogenous study population (healthy, adult males of normal BMI) to establish if and how the human peripheral T cell compartment varies with the season. Unique to previous in vitro and in vivo studies examining the role of 1,25(OH)2D3 on T cells, our current results suggest that physiological variation in serum vitamin D3 levels throughout the four seasons might influence CD4+ and CD8+ T cell homeostasis and homing behavior. Given that serum 25(OH)D3 levels can be affected by various factors, our observations warrant future validation in a larger and more diverse population cohort to identify any possible differences in adaptive immune responses among the extreme of ages and different genders. Nevertheless, our data provide insight on previous epidemiological findings regarding the prevalence of certain autoimmune diseases and infections, which have been attributed to seasonal variation in sun exposure and serum 25(OH)D3 levels [10][12],[44]. Although not as extensively reported as vitamin D3 status, certain hormones and corticosteroids such as catecholamine and aldosterone seem to vary with seasons as well [45], [46]. It would be of interest to find out if these factors are associated with changes in immunological characteristics of T cell.

In conclusion, we have demonstrated for the first time the existence of variations in adaptive immunity throughout the four seasons of the year in association with physiological changes in serum 25(OH)D3 levels in vivo. These novel findings further our understanding on the seasonal variability between vitamin D3 and human peripheral T cell composition, and support the basis for conducting larger population-based studies to investigate the benefits of vitamin D3 supplementation in temperate regions during winter.

Supporting Information

Figure S1.

Gate setting for CD4+ and CD8+ T cells gated on CD45+ cells; and CD4+CD45RA+ T cells, CD4+CD45RO+ T cells and CD4+CD25hiCD127 regulatory T cells gated on CD4+ T cells. Dotplots show surface staining for markers performed on whole blood.

https://doi.org/10.1371/journal.pone.0029250.s001

(EPS)

Acknowledgments

The authors are grateful to André Brandt (Department of Laboratory Medicine, RUNMC) for his technical assistance.

Author Contributions

Conceived and designed the experiments: A-LK HJPMK MGN AJAMvdV IJ. Performed the experiments: A-LK LYAC. Analyzed the data: A-LK HJPMK MGN AJAMvdV IJ. Contributed reagents/materials/analysis tools: FCGJS. Wrote the paper: A-LK HJPMK MGN AJAMvdV IJ.

References

  1. 1. Mora JR, Iwata M, von Andrian UH (2008) Vitamin effects on the immune system: vitamins A and D take centre stage. Nat Rev Immunol 8: 685–698.
  2. 2. Bikle D (2009) Nonclassic actions of vitamin D. J Clin Endocrinol Metab 94: 26–34.
  3. 3. Adams JS, Hewison M (2008) Unexpected actions of vitamin D: new perspectives on the regulation of innate and adaptive immunity. Nat Clin Pract Endocrinol Metab 4: 80–90.
  4. 4. Lips P (2006) Vitamin D physiology. Prog Biophys Mol Biol 92: 4–8.
  5. 5. Webb AR, Holick MF (1988) The role of sunlight in the cutaneous production of vitamin D3. Annu Rev Nutr 8: 375–399.
  6. 6. Lips P (2010) Worldwide status of vitamin D nutrition. J Steroid Biochem Mol Biol.
  7. 7. Cannell JJ, Vieth R, Umhau JC, Holick MF, Grant WB (2006) Epidemic influenza and vitamin D. Epidemiol Infect 134: 1129–1140.
  8. 8. Ginde AA, Mansbach JM, Camargo CA Jr (2009) Association between serum 25-hydroxyvitamin D level and upper respiratory tract infection in the Third National Health and Nutrition Examination Survey. Arch Intern Med 169: 384–390.
  9. 9. Sabetta JR, DePetrillo P, Cipriani RJ, Smardin J, Burns LA (2010) Serum 25-hydroxyvitamin d and the incidence of acute viral respiratory tract infections in healthy adults. PLoS One 5: e11088.
  10. 10. Fernandes de Abreu DA, Babron MC, Rebeix I, Fontenille C, Yaouanq J (2009) Season of birth and not vitamin D receptor promoter polymorphisms is a risk factor for multiple sclerosis. Mult Scler 15: 1146–1152.
  11. 11. Moltchanova EV, Schreier N, Lammi N, Karvonen M (2009) Seasonal variation of diagnosis of Type 1 diabetes mellitus in children worldwide. Diabet Med 26: 673–678.
  12. 12. Willer CJ, Dyment DA, Sadovnick AD, Rothwell PM, Murray TJ (2005) Timing of birth and risk of multiple sclerosis: population based study. BMJ 330: 120.
  13. 13. Correale J, Ysrraelit MC, Gaitan MI (2009) Immunomodulatory effects of Vitamin D in multiple sclerosis. Brain 132: 1146–1160.
  14. 14. Simpson S Jr, Blizzard L, Otahal P, van dM I, Taylor B (2011) Latitude is significantly associated with the prevalence of multiple sclerosis: a meta-analysis. J Neurol Neurosurg Psychiatry.
  15. 15. Zold E, Barta Z, Bodolay E (2011) Vitamin D deficiency and connective tissue disease. Vitam Horm 86: 261–286.
  16. 16. Veldman CM, Cantorna MT, DeLuca HF (2000) Expression of 1,25-dihydroxyvitamin D(3) receptor in the immune system. Arch Biochem Biophys 374: 334–338.
  17. 17. Mahon BD, Wittke A, Weaver V, Cantorna MT (2003) The targets of vitamin D depend on the differentiation and activation status of CD4 positive T cells. J Cell Biochem 89: 922–932.
  18. 18. Lemire JM, Archer DC, Beck L, Spiegelberg HL (1995) Immunosuppressive actions of 1,25-dihydroxyvitamin D3: preferential inhibition of Th1 functions. J Nutr 125: 1704S–1708S.
  19. 19. Tang J, Zhou R, Luger D, Zhu W, Silver PB, Grajewski RS (2009) Calcitriol suppresses antiretinal autoimmunity through inhibitory effects on the Th17 effector response. J Immunol 182: 4624–4632.
  20. 20. Boonstra A, Barrat FJ, Crain C, Heath VL, Savelkoul HF, O'Garra A (2001) 1alpha,25-Dihydroxyvitamin d3 has a direct effect on naive CD4(+) T cells to enhance the development of Th2 cells. J Immunol 167: 4974–4980.
  21. 21. Imazeki I, Matsuzaki J, Tsuji K, Nishimura T (2006) Immunomodulating effect of vitamin D3 derivatives on type-1 cellular immunity. Biomed Res 27: 1–9.
  22. 22. Marelli-Berg FM, Cannella L, Dazzi F, Mirenda V (2008) The highway code of T cell trafficking. J Pathol 214: 179–189.
  23. 23. Bromley SK, Mempel TR, Luster AD (2008) Orchestrating the orchestrators: chemokines in control of T cell traffic. Nat Immunol 9: 970–980.
  24. 24. Topilski I, Flaishon L, Naveh Y, Harmelin A, Levo Y (2004) The anti-inflammatory effects of 1,25-dihydroxyvitamin D3 on Th2 cells in vivo are due in part to the control of integrin-mediated T lymphocyte homing. Eur J Immunol 34: 1068–1076.
  25. 25. Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A (2007) DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol 8: 285–293.
  26. 26. Khoo AL, Chai LY, Koenen HJ, Sweep FC, Joosten I, Netea MG, et al. (2011) Regulation of cytokine responses by seasonality of vitamin D status in healthy individuals. Clin Exp Immunol 164: 72–79.
  27. 27. Moan J, Lagunova Z, Lindberg FA, Porojnicu AC (2009) Seasonal variation of 1,25-dihydroxyvitamin D and its association with body mass index and age. J Steroid Biochem Mol Biol 113: 217–221.
  28. 28. Van Den Bout-Van Den Beukel CJ, Fievez L, Michels M, Sweep FC, Hermus AR, et al. (2008) Vitamin D deficiency among HIV type 1-infected individuals in the Netherlands: effects of antiretroviral therapy. AIDS Res Hum Retroviruses 24: 1375–1382.
  29. 29. van Hoof HJ, Swinkels LM, van Stevenhagen JJ, van den BH, Ross HA (1993) Advantages of paper chromatography as a preparative step in the assay of 1,25-dihydroxyvitamin D. J Chromatogr 621: 33–39.
  30. 30. Anonymous (2011) Seasonal variation of UV index in the Netherlands. http://www.rivm.nl/Onderwerpen/Onderwerpen/U/UV_ozonlaag_en_klimaat/Zonkracht/Seizoensvariatie. Date accessed: 11/11/2011.
  31. 31. Kimmig S, Przybylski GK, Schmidt CA, Laurisch K, Mowes B, Radbruch A, Thiel A (2002) Two subsets of naive T helper cells with distinct T cell receptor excision circle content in human adult peripheral blood. J Exp Med 195: 789–794.
  32. 32. Hollis BW (1996) Assessment of vitamin D nutritional and hormonal status: what to measure and how to do it. Calcif Tissue Int 58: 4–5.
  33. 33. Alonso A, Hernan MA (2008) Temporal trends in the incidence of multiple sclerosis: a systematic review. Neurology 71: 129–135.
  34. 34. Ponsonby AL, McMichael A, van dM I (2002) Ultraviolet radiation and autoimmune disease: insights from epidemiological research. Toxicology 181–182: 71–78.
  35. 35. Lemire JM, Adams JS, Sakai R, Jordan SC (1984) 1 alpha,25-dihydroxyvitamin D3 suppresses proliferation and immunoglobulin production by normal human peripheral blood mononuclear cells. J Clin Invest 74: 657–661.
  36. 36. Rigby WF, Stacy T, Fanger MW (1984) Inhibition of T lymphocyte mitogenesis by 1,25-dihydroxyvitamin D3 (calcitriol). J Clin Invest 74: 1451–1455.
  37. 37. Muller K, Bendtzen K (1992) Inhibition of human T lymphocyte proliferation and cytokine production by 1,25-dihydroxyvitamin D3. Differential effects on CD45RA+ and CD45R0+ cells. Autoimmunity 14: 37–43.
  38. 38. Baeke F, Korf H, Overbergh L, van EE, Verstuyf A, Gysemans C, Mathieu C (2010) Human T lymphocytes are direct targets of 1,25-dihydroxyvitamin D3 in the immune system. J Steroid Biochem Mol Biol 121: 221–227.
  39. 39. Jeffery LE, Burke F, Mura M, Zheng Y, Qureshi OS, Hewison M, et al. (2009) 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 183: 5458–5467.
  40. 40. Smolders J, Thewissen M, Peelen E, Menheere P, Tervaert JW, Damoiseaux (2009) Vitamin D status is positively correlated with regulatory T cell function in patients with multiple sclerosis. PLoS One 4: e6635.
  41. 41. Morales-Tirado V, Wichlan DG, Leimig TE, Street SE, Kasow KA, Riberdy JM (2011) 1alpha,25-dihydroxyvitamin D3 (vitamin D3) catalyzes suppressive activity on human natural regulatory T cells, uniquely modulates cell cycle progression, and augments FOXP3. Clin Immunol 138: 212–221.
  42. 42. Mora JR, von Andrian UH (2006) T-cell homing specificity and plasticity: new concepts and future challenges. Trends Immunol 27: 235–243.
  43. 43. Yamanaka K, Dimitroff CJ, Fuhlbrigge RC, Kakeda M, Kurokawa I, et al. (2008) Vitamins A and D are potent inhibitors of cutaneous lymphocyte-associated antigen expression. J Allergy Clin Immunol 121: 148–157.
  44. 44. Simpson S Jr, Taylor B, Blizzard L, Ponsonby AL, Pittas F (2010) Higher 25-hydroxyvitamin D is associated with lower relapse risk in multiple sclerosis. Ann Neurol 68: 193–203.
  45. 45. Radke KJ, Izzo JL Jr (2010) Seasonal variation in haemodynamics and blood pressure-regulating hormones. J Hum Hypertens 24: 410–416.
  46. 46. Van Cauter EW, Virasoro E, Leclercq R, Copinschi G (1981) Seasonal, circadian and episodic variations of human immunoreactive beta-MSH, ACTH and cortisol. Int J Pept Protein Res 17: 3–13.