Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

Contents lists available at ScienceDirect

Journal of Steroid Biochemistry and Molecular Biology


journal homepage: www.elsevier.com/locate/jsbmb

Vitamin D-modulated dendritic cells delay lethal graft-versus-host disease T


through induction of regulatory T cells
An-Sofie Vanherwegen, Dana Paulina Cook, Gabriela Bomfim Ferreira, Conny Gysemans1,

Chantal Mathieu ,1
Clinical and Experimental Endocrinology, KU Leuven, Belgium

A R T I C LE I N FO A B S T R A C T

Keywords: Graft-versus-host disease (GVHD) is the most lethal complication after allogeneic bone marrow transplantation
Vitamin D (allo-BMT). Current approaches to prevent GVHD rely on donor lymphocyte/T cell depletion or general im-
GVHD munosuppression, leading to opportunistic infections and cancer relapse. Tolerogenic dendritic cells can induce
Tolerogenic dendritic cells regulatory T cells (Tregs) with the ability to suppress inflammation and prevent transplant rejection, making
Regulatory T cells
them an attractive cellular therapy to control GVHD. Active vitamin D (1α,25-dihydroxyvitamin D3;
1α,25(OH)2D3) promotes the generation of tolerogenic dendritic cells (1,25D3-DCs). This study aimed to de-
termine the ability of ex vivo generated 1,25D3-DCs to trigger the expansion of Tregs that are able to control
lethal xenogeneic GVHD in humanized NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice. We demonstrate that 1,25D3-
DCs express lower levels of HLA-DR and costimulatory molecules, such as CD80 and CD86, and produce higher
levels of IL-10 and TNF-α and lower amounts of IL-12, compared to vehicle-treated DCs. Moreover, these cells
express increased levels of various co-inhibitory molecules such as PD-L1 and ILT-3 and the glycoprotein CD52
that is known to suppress T cell activation. Consequently, 1,25D3-DCs are poor stimulators of alloantigen-primed
T cells, but foster the generation of antigen-specific suppressive Tregs. When adoptively transferred in huma-
nized NSG mice, these 1,25D3-DC-induced Tregs delayed GVHD caused by the co-transferred autologous human
peripheral blood mononuclear cells (PBMCs). These results indicate that 1,25D3-DC-induced Tregs can inhibit
xenogeneic GVHD and maintain their immunomodulatory function under conditions of inflammation.

1. Introduction interferes with the differentiation and maturation processes [4], locking
DCs in a tolerogenic state [5]. DCs generated in the presence of
Allogeneic bone marrow transplantation (allo-BMT) has been 1α,25(OH)2D3 (1,25D3-DCs) maintain CD14 expression, express little
exploited as a curative treatment for hematological malignant and non- CD1a, show reduced expression of antigenpresenting (e.g. HLA-ABC and
malignant diseases since the 70 s [1]. The alloreactivity of these cells -DR) and costimulatory molecules (e.g. CD80, CD86) and diminished
against human leukocyte antigen (HLA) mismatches and minor histo- secretion of stimulatory cytokines (e.g. interleukin (IL)-12, IL-23) that
compatibility antigens can trigger life-threatening graft-versus-host are known to regulate the polarization of Teff cells [6–8]. Moreover, the
disease (GVHD) [2]. Recent studies suggest that an imbalance in ef- expression of inhibitory molecules, including programmed death-ligand
fector (Teff) and regulatory T cell (Treg) reconstitution after allo-BMT (PD-L) 1, PD-L2, and immunoglobulin-like transcript (ILT)-3 and -4, is
can amplify alloreactivity and promote GVHD. upregulated and the production of regulatory cytokines like IL-10,
Vitamin D3 is a fat-soluble hormone that can be acquired from food transforming growth factor (TGF)-β and tumor necrosis factor (TNF)-α
or be synthesized in the skin upon ultraviolet-B radiation. In addition to is enhanced [6,9,10]. As a result, 1,25D3-DCs influence T cell behavior
its classic role in calcium/phosphate and bone homeostasis, 1α,25-di- inducing hyporesponsiveness and shifting T cell polarization from T
hydroxyvitamin D3 (1α,25(OH)2D3), which is the active metabolite of helper (Th)1- and Th17-mediated inflammatory responses towards Treg
vitamin D3, has been identified as a potent natural modulator of innate responses [8–10]. In addition to these effects, 1α,25(OH)2D3 has a
and adaptive immunity [3]. In dendritic cells (DCs), generated from profound impact on the migratory capacity of DCs through upregula-
bone marrow precursors or peripheral blood monocytes, 1α,25(OH)2D3 tion of the chemokine receptor CXCR3, guiding cells to inflamed


Corresponding author at: Clinical and Experimental Endocrinology, KU Leuven, Campus Gasthuisberg O&N1, Herestraat 49 box 902, 3000, Leuven, Belgium.
E-mail address: chantal.mathieu@uzleuven.be (C. Mathieu).
1
Shared senior author.

https://doi.org/10.1016/j.jsbmb.2018.12.013
Received 1 October 2018; Received in revised form 17 December 2018; Accepted 31 December 2018
Available online 31 December 2018
0960-0760/ © 2019 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

tissues. and TNF-α secreted in the DC culture supernatant and similarly the
In rodents, adoptively transferred tolerogenic DCs can efficiently secretion of IFN-γ and IL-10 in the DC-T cell co-culture.
suppress lethal acute GVHD by the in vivo generation of inducible Tregs
(iTregs) [11–14]. However, high numbers of these cells are needed to 2.4. Measurement of indoleamine-2,3-dioxygenase (IDO)
reduce the incidence and severity of GVHD and so far, complete pro-
tection from GVHD has never been achieved. In addition, a major fear is Secretion levels of IDO in the DC culture supernatant was de-
the reversal of the ex vivo induced tolerogenic phenotype when exposed termined by the RayBio® Human IDO ELISA Kit (RayBiotech, Norcross,
to the in vivo pro-inflammatory environment of the host, resulting in GA, USA).
immunogenic DCs that could induce Teff cells, rather than Tregs. Thus,
intervention strategies that generate stable tolerogenic DCs that induce 2.5. In vitro allogenic T cell proliferation and flow cytometry analysis
Tregs with superior suppressive ability would be advantageous to
control GVHD as well as prevent transplant rejection or treat auto- CD3+ T cells were isolated from PBMCs of healthy donors using the
immunity. Although rodents continue to be important in vivo models for Pan T cell Kit II (Miltenyi Biotec) according to manufacturer’s guide-
studying the human immune system, we currently recognize the po- lines. These purified CD3+ T cells were labelled with 1 μM carboxy-
tential limitations of generalizing data from mice to humans. The model fluorescein succinimidyl ester (CFSE; Thermo Fisher Scientific/
of NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice allows the study of an eBioscience) and subsequently co-cultured with DCs in a 1:10 DC:T cell
extreme form of GVHD, namely a xenogeneic GVHD, when human ratio for 4 days. T cell proliferation was determined in CD4+ and CD8+
immune cells are infused. T cell subsets by flow cytometry (antibodies from Thermo Fisher
In the current study, we explored whether 1,25D3-DCs can induce Scientific/eBioscience). Doublets were excluded based on signal height
suppressive Tregs that are able to prevent lethal xenogeneic GVHD in and widths. Dead cells were stained by Zombie Yellow Fixable Viability
NSG mice infused with human peripheral blood mononuclear cells Kit (BioLegend) and excluded from analysis. Samples were acquired on
(PBMCs). a Gallios™ Flow Cytometer (Beckman Coulter) and analyzed with
FlowJo software (TreeStar).
2. Materials and methods
2.6. Induction of antigen-specific Tregs and suppression assay
2.1. Ex vivo generation of human monocyte-derived 1,25D3-DCs
T cell lines were induced as described previously [10]. In brief,
Human monocytes were freshly isolated and differentiated into DCs CD4+ T cells were isolated from PBMCs using the Human CD4+ T cell
as previously described [6], with the approval of the ethics committee Isolation Kit (Miltenyi Biotec), according to manufacturer’s specifica-
(S-number 52679) of the University hospital of Leuven (UZ Leuven, tions. 1 × 106 purified CD4+ T cells were co-cultured with vehicle-
Leuven, Belgium). In brief, PBMCs were isolated by Lymphoprep gra- treated and 1,25D3-DCs from an haplo-identical donor at a 1:10 DC:T
dient centrifugation (Alere Technologies AS, Oslo, Norway) from blood cell ratio in IMDM (Thermo Fisher Scientific/Gibco) with 10% human
from healthy donors. Monocytes were purified from PBMCs by positive serum (Biowest, Nuaillé, France). After 5 days, T cells were recovered
selection using the CD14 Microbeads Kit (Miltenyi Biotec, Bergisch and rested for 2 days in the presence of 10 ng/ml IL-7 and 5 ng/ml IL-15
Gladbach, Germany) according to manufacturer’s protocol. Isolated (both from Genscript, Nanjing, China). On day 7 after primary stimu-
monocytes were cultured in differentiation medium containing 500 IU/ lation, the CD4+ T cells were restimulated under the same conditions. T
ml human recombinant IL-4 (Gentaur, Kampenhout, Belgium) and 800 cells stimulated with 1,25D3-DCs are referred to as Treg lines, while
IU/ml human recombinant GM-CSF (Gentaur). Medium and cytokines those stimulated with vehicle-treated DCs are referred to as Teff lines.
were refreshed on day 3. On day 6, maturation was induced in the To assess the suppressive capacity of the generated T cell lines, CD4+
presence of 800 IU/ml GM-CSF (Gentaur), 100 ng/ml LPS (Sigma-Al- responder T cells (donor A) were labelled with 1 μM CFSE (Thermo
drich, St. Louis, MO, USA) and 1,000 IU/ml human recombinant IFN-γ Fisher Scientific/eBioscience) and cultured in the presence of activated
(Roche, Basel, Switzerland). To obtain 1,25D3-DCs, 10−8 M stimulator DCs (donor B) at a 1:10 DC:T cell ratio in 96-well round
1α,25(OH)2D3 (Sigma-Aldrich) was added at day 0 and day 3. Similar bottom plates coated with 0.1 μg/ml anti-human CD3 monoclonal an-
dilution of ethanol was used as vehicle control. tibody (clone OKT-3; Exbio Antibodies, Vestec, Czech Rebublic). Tregs
(donor A) were added at a 1:1 ratio to responder T cells (donor A). After
2.2. Flow cytometry analysis of surface markers on human DCs 4 days, the proliferation of the responder T cells was analyzed on a
Gallios™ Flow Cytometer (Beckman Coulter). Prior to acquisition,
Mature vehicle-treated and 1,25D3-DCs were analyzed for their CountBright Absolute Counting Beads (Thermo Fisher Scientific/In-
surface marker expression using flow cytometry. DCs were pretreated vitrogen) were added and an equal amount of beads was obtained to
with Fc block (anti-CD16/CD32) and subsequently labelled with the facilitate comparison between samples. To asses IFN-γ and IL-10 cyto-
following conjugated antibodies: CD1a, CCR7, CXCR3 (from BioLegend, kine production by the induced T cell lines, T cells were stimulated with
San Diego, CA, USA), CD14, HLA-DR, PD-L1, ILT-3 (from Thermo Fisher LPS-activated control DCs (donor A) for 24 h after which culture su-
Scientific/ eBioscience, San Diego, CA, USA), CD80, CD86 (from BD pernatant was collected.
Biosciences, San Jose, CA, USA), CD52 (Bio-Rad, Hercules, CA, USA).
Doublets were excluded based on signal height and widths. Dead cells 2.7. Xenogeneic graft-versus-host disease model
were stained by Zombie Yellow/Aqua Fixable Viability Kit (BioLegend)
according to manufacturer’s guidelines and excluded from analysis. NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice were purchased from
Samples were acquired on a Gallios™ Flow Cytometer (Beckman The Jackson Laboratory (Bar Harbor, ME, USA) and inbred in the an-
Coulter) and analyzed with the FlowJo software (TreeStar, Ashland, imal facility of KU Leuven (Leuven, Belgium). Housing of all mice oc-
OR, USA). curred under semi-barrier conditions and animals were fed sterile food
and water ad libitum. Handling of mice and experimental procedures
2.3. Cytokine production in DC cultures and DC:T cell co-cultures were performed in accordance to guidelines of the institutional animal
care and use committee of the KU Leuven (project number ECD 221/
The V-plex Proinflammatory Panel 1 Human kit from MesoScale 2013). Xenogeneic GVHD was induced as described previously [15]. In
Discovery (Rockville, MD, USA) was used according to the manu- brief, NSG mice received total body irradiation with 1.5 Gy one day
facturer’s instructions for the quantitative detection of IL-12p70, IL-10 before intravenous injection of purified single-donor human PBMCs

104
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

(3 × 106). Simultaneously, PBS or autologous DCs (0.3 × 106) were phenotype and cytokine secretion resulted in an altered T cell stimu-
injected intraperitoneally. Mice were treated with antibiotic drinking latory capacity, DCs were co-cultured with T cells. Unstimulated T cells
water (Baytril, Bayer) after xenogeneic PBMC transplantation to pre- have a low basal proliferation rate, which can be boosted by anti-CD3/
vent unspecific infections. Transplanted mice were assessed twice CD28 stimulation (Fig. 3A). Addition of mature vehicle-treated DCs
weekly for weight loss and clinical scoring of GVHD symptoms. A cu- stimulated the proliferative capacity of both CD4+ and CD8+ allo-
mulative global disease score was established for five clinical para- geneic T cells with a respective 4- and 5-fold increase, which was re-
meters on a scale from 0 to 8: weight loss (2, if > 10%; 6, if > 20%), duced to at least half in the condition with 1,25D3-DCs. Analysis of
anemia (1, white ears; 2, white ears and tail), posture (1, kyphosis), fur cytokines secreted into the co-culture supernatant showed that T cells
(1, hair loss) and activity (1, stationary > 50% of the time). Mice were that were co-cultured with 1,25D3-DCs produced lower amounts of IFN-
euthanized when reaching a global score ≥6, with the date of death γ (Fig. 3B) a typical Th1 cytokine, and high amounts of the tolerogenic
recorded as the maximum score of 8 in accordance with institutional cytokine IL-10 compared to mature vehicle-treated DCs (Fig. 3C). Of
animal ethics guidelines. Mice were bled every 2 weeks after transfer in note, with the used measuring technique, it is impossible to determine
order to determine the presence of human cells by flow cytometry. whether the IL-10 cytokine levels in the co-culture supernatant are
Blood cells were labelled with antibodies against CD3, CD4, FoxP3 produced by DCs or by T cells. These data show that 1,25D3-DCs are
(Thermo Fisher Scientific/eBioscience) and CD25 (Miltenyi Biotec) . potent antigen-presenting cells that can suppress the activation of al-
Doublets were excluded based on signal height and widths. Dead cells logeneic Teff responses.
were stained by Zombie Aqua Fixable Viability Kit (BioLegend) ac-
cording to manufacturer’s guidelines and excluded from analysis. 3.3. 1α,25(OH)2D3-modulated dendritic cells induce suppressive antigen-
Samples were acquired on a Canto II Flow Cytometer (BD Biosciences) specific Tregs and delay GVHD
and analyzed with the FlowJo software (TreeStar, Ashland, OR, USA).
Next, 1,25D3-DC-induced Treg lines were generated and assessed for
2.8. Statistics their functional capacity. Tregs, primed in the presence of 1,25D3-DCs,
potently suppressed the proliferation of responder T cells to allogeneic
The normality of all datasets was tested. When data was normally mature DCs compared to Teff cells, primed with mature vehicle-treated
distributed, either unpaired Student’s t test or ANOVA with Sidak’s DCs, as evidenced by decreased proliferation and division indexes
post-test for multiple comparison was used. When data was not nor- (Fig. 3D).
mally distributed, statistical significance was determined using the Finally, we investigated whether 1,25D3-DCs were able to mediate
Mann-Whitney U test or the Kruskal-Wallis test with subsequent Dunn’s immune suppression when transferred in vivo. We used a xenogeneic
post-test. Differences in survival rates were assessed using the Mantel- GVHD model induced by transferring human PBMCs into NSG mice that
Cox Log-rank test. All statistical analyses were performed using were co-injected with PBS or autologous, mature vehicle-treated DCs or
GraphPad Prism software (GraphPad Software, La Jolla, CA, USA). 1,25D3-DCs (Fig. 4A). All recipients of human PBMCs developed GVHD
Significance was defined as *p < 0.05; **p < 0.01; ***p < 0.001; (Fig. 4B) and died between day 16 and day 72 following transplanta-
**** p < 0.0001. tion. In mice receiving only human PBMCs, clinical signs of GVHD in-
cluding reduced weight, anemia and hair loss were evident from day 16
3. Results onwards (Fig. 4B). No significant delay in disease occurrence or mor-
tality was observed when control DCs were co-transferred with human
3.1. 1α,25(OH)2D3 treatment induces stable tolerogenic DCs capable of PBMCs (Fig. 4B, C). A single injection of co-transferred 1,25D3-DCs
homing to inflammatory sites markedly attenuated GVHD onset to 34 days on average (ranged from
day 28 until day 40), with reduced GVHD disease manifestations at a
We first studied the cellular phenotype and function of 1,25D3-DCs time when most PBS or vehicle DC-treated mice had died (Fig. 4C).
from healthy donors after an inflammatory challenge (LPS and IFN-γ). Mice were bled every 2 weeks after transfer to determine the presence
We demonstrate that even after exposure to inflammation, 1,25D3-DCs of human cells and the in vivo induction of CD4+CD25+FoxP3+ Tregs
displayed a stable tolerogenic phenotype with reduced surface expres- by flow cytometry. Interestingly, at the first time point of 2 weeks post-
sion of the antigen-presenting molecules HLA-DR and CD1a and the engraftment, a significant higher percentage of human FoxP3-expres-
costimulatory molecules CD80 and CD86 (Fig. 1). The expression of the sing Tregs were present in the mice that received 1,25D3-DCs compared
monocytic marker CD14 was retained in 1,25D3-DCs exposed to in- to vehicle-treated DCs or only PBMCs, while the total CD4+CD25+
flammation. The inhibitory molecules PD-L1 and ILT-3 as well as the population remained equal over the different groups and over time
newly identified tolerogenic marker CD52 [16] were increased 1.3-, (Fig. 4D). However, 4 and 8 weeks post-engraftment this effect was no
7.5- and 6.4-fold, respectively, at the cell surface after 1α,25(OH)2D3 longer detectable. This observation indicates that 1,25D3-DCs are able
treatment, a general feature of tolerogenic DCs [17]. Moreover, surface to induce CD4+CD25+FoxP3+ Tregs in an in vivo setting, but that this
expression of CXCR3, responsible for homing to inflammatory sites, was is a transient phenomenon.
upregulated in 1,25D3-DCs, while the chemokine receptor CCR7 was
similarly expressed as in control cells, still allowing these cells to mi- 4. Discussion
grate to the lymphoid tissues (Fig. 1). Also, the cytokine pattern of DCs
was altered upon 1α,25(OH)2D3 exposure with decreased IL-12p70 and The active metabolite of vitamin D3, 1α,25(OH)2D3, potently
elevated IL-10 and TNF-α secretion levels (Fig. 2). Additionally, 1,25D3- modulates the behavior of several immune cells. In DCs, 1α,25(OH)2D3
DCs produced higher amounts of the indoleamine-2,3-dioxygenase induces a tolerogenic profile. As DCs play a central role in regulating
(IDO) enzyme, although this difference was not statistically significant the delicate balance between immunogenicity and tolerance, 1,25D3-
(Fig. 2). Taken together, the inhibition of full maturation, coupled with DCs possess a great potential for therapeutic applications in auto-
decreased IL-12p70 and enhanced IL-10 and TNF-α production, hints at immune diseases and transplantation settings. In the current manu-
a superior immunoregulatory capacity of 1,25D3-DCs. script, we confirmed previous studies showing the 1α,25(OH)2D3-
mediated downregulation of antigen-presentation, costimulation and
3.2. 1α,25(OH)2D3-modulated dendritic cells induce allogeneic T cell pro-inflammatory cytokine production. Moreover, an increased surface
hyporesponsiveness expression of inhibitory molecules and tolerogenic cytokine secretion
was observed. 1,25D3-DCs maintained a specific migratory signature
To determine whether these 1α,25(OH)2D3-induced changes in DC upon 1α,25(OH)2D3 modulation, allowing them to migrate to the

105
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

Fig. 1. The tolerogenic phenotype of 1α,25(OH)2D3-modulated human dendritic cells. Surface marker analysis of mature vehicle-treated (CTR; black) and 1,25D3-
DCs (1,25D3; grey). The expression level of phenotypical markers HLA-DR, CD1a, CD80, CD86, CD14, PD-L1, ILT-3, CD52, CXCR3, and CCR7 was determined by flow
cytometry (n = 6–12). Bar graphs represent mean fluorescence intensity (MFI) ± SEM. Each dot represents a different DC donor. * p < 0.05, ** p < 0.01, ***
p < 0.001, **** p < 0.0001.

lymph nodes and the site of inflammation [6,10,18–22]. Interestingly, regulation in more detail on a genome-wide scale. For example, the
these changes remained stable upon inflammatory challenge in vitro CD14 gene expression has been found to be early upregulated by
[10,23], which is crucial for in vivo applications where 1,25D3-DCs will 1α,25(OH)2D3 treatment [25,26]. Moreover, the promoter region of
have to exert their functions in a highly inflammatory environment. The CD14 comprises several VDR binding sites, confirming that CD14 is
elevated expression of the new tolerogenic DC biomarker CD52 [16] indeed a direct target of active vitamin D3 [26]. Over the past few years,
was validated as well. 1α,25(OH)2D3 has been shown to rapidly regulate several immune
Many microarray analyses have been performed on several cell genes in monocytes and DCs (e.g. CD274, CD40, CD80, HLA-DR)
types, including immune cell subsets, to identify direct transcriptional [16,25–27]. With regard to the physiological actions of active vitamin
targets of 1α,25(OH)2D3. However, only early time points are useful to D, not only the primary 1α,25(OH)2D3 targets are important and sec-
distinguish primary and secondary target genes as the full 1,25(OH)2D3- ondary targets should be taken into account as well. In this regard,
mediated transcriptional process is believed to take 2–3 h [24]. In ad- 1α,25(OH)2D3 induces other transcription factors that could regulate
dition, primary 1α,25(OH)2D3 targets are expected to have at least one the expression of secondary target genes [28]. Other mechanism of
vitamin D responsive element (VDRE) in their promoter region. indirect regulation by 1α,25(OH)2D3 include protein-protein interac-
Therefore, establishment of the chromatin immunoprecipitation se- tions with other transcription factors, such as NF-κB in the case of IL-12
quencing (ChIP-seq) technique allowed the study of transcriptional [29], hereby preventing association with its genomic binding sites or

Fig. 2. 1α,25(OH)2D3-modulated human den-


dritic cells secrete low levels of stimulatory
cytokines and high levels of tolerogenic cyto-
kines and markers. Secretion levels of the cy-
tokines IL-12p70, IL-10 and TNF-α into the DC
culture supernatant were measured by MSD
multiplex (n = 6–11). Bar graphs represent
mean cytokine levels ± SEM. Production of
the enzyme IDO into DC culture supernatant
was determined by ELISA (n = 6). Bar graphs
represent mean IDO protein level ± SEM.
Each dot represents a different DC donor. *
p < 0.05, ** p < 0.01, *** p < 0.001, ****
p < 0.0001.

106
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

Fig. 3. 1α,25(OH)2D3-modulated human dendritic cells suppress allogeneic T cell activation and induce suppressive Tregs. (A) Mature CTR- and 1,25D3-DCs were
harvested and co-cultured with CD3+ T cells. After 4 days, proliferation of CD4+ (left) and CD8+ (right) T cells was assessed through CFSE dilution by flow
cytometry. Unstimulated (T alone) and anti-CD3/CD28-stimulated (aCD3/aCD28) T cells were included as negative and positive controls, respectively. Bar graphs
represent percentage of proliferation relative to CTR condition ± SEM (n = 3–5). Secretion of IFN-γ (B) and IL-10 (C) into the supernatant was measured after 4
days of co-culture by MSD multiplex (n = 4). (D) Mature CTR- and 1,25D3-DCs were co-cultured with CD4+ T cells to generate Teff or Treg lines, respectively.
Suppressive capacity of Teff and Treg lines determined in a suppression assay with autologous responder T cells in a 1:1 T cell line: T responder ratio. Plots show CFSE
dilutions in CD4+ cells of Teff lines primed with mature CTR- (left) or Treg lines primed with mature 1,25D3-DCs (right). Representative figure for two independent
experiments. Proliferation index and division index expressed as percent proliferation relative to the crowding control (n = 2). Bar graphs represent mean ± SEM.
Each dot represents a different DC donor. Data are pooled from at least two independent experiments. * p < 0.05, ** p < 0.01.

the induction of epigenetic changes [30]. Future ChIP-seq experiments TR1 cells was solely based on the observation that the iTregs produced
in human 1,25D3-DCs will provide more insights in genomic VDR high amounts of IL-10 and was not confirmed by surface expression
binding sites and allow better discrimination between primary and analysis of TR1-specific parameters, such as CD49b and LAG3 [35]. It
secondary targets. remains to be elucidated which Treg subsets are exactly being induced
As a result of these aforementioned phenotypic changes, 1,25D3- by 1,25D3-DCs in vitro and in vivo. Moreover, the exact mechanisms by
DCs are poor T cell stimulators and simultaneously drive the induction which 1,25D3-DCs induce antigen-specific Tregs to restore peripheral
of antigen-specific Tregs [10,31]. Our current data indeed showed tolerance or immune homeostasis are still not fully understood. A
strongly reduced T cell proliferation and the generation of functional crucial role for PD-L1, upregulated upon 1α,25(OH)2D3 modulation of
Tregs in vitro and in vivo. Interestingly, repetitive priming of CD4+ T the DCs, has been reported as PD-L1 blockade resulted in Th1-like cells
cells with 1,25D3-DCs has been shown to result in the de novo induction without suppressive capacity [10]. It is thought that increased PD-L1
of IL-10-producing Tregs, and not only by the expansion of already triggers IL-10 production by the DCs, thereby driving Treg differentia-
existing CD4+CD25+ Tregs [10,32]. The exact nature of these induced tion [36]. Moreover, 1,25D3-DCs produced elevated amounts of soluble
Tregs (iTregs) remains controversial. 1,25D3-DCs can drive the differ- TNF after maturation, but mTNF was shown to be essential for the in-
entiation of IL-10-producing type 1 regulatory (TR1) cells and duction of suppressive T cells through the mTNF-TNF receptor II sig-
CD4+CD25+FoxP3+ Tregs [10,33,34]. The characterization of these naling pathway [20]. Tolerogenic DCs expressing IDO promote immune

107
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

Fig. 4. 1α,25(OH)2D3-modulated dendritic


cells suppress xenogeneic GVHD. (A)
Overview of the experimental set-up. NSG
mice were irradiated with 1,5 Gy (day -1).
On day 0, they received 3 × 106 human
PBMCs (intraveneously, i.v.) simulta-
neously with 0.3 × 106 mature DCs (CTR-
or 1,25D3-, as indicated) or PBS (in-
traperitoneally, i.p.). (B) Disease onset, thus
when mean disease score ≥ 1, and (C)
mean GVHD scores based on weight loss,
anemia, hair loss, posture and reduced mo-
bility were monitored twice per week. (D)
Mice were bled every 2 weeks (if still alive)
and the presence of human Tregs was de-
termined by flow cytometry. Percentage of
human CD4+CD25+ (top) and
CD4 CD25 FoxP3+ (bottom) T cells 2
+ +

weeks, 4 weeks and 8 weeks post-engraft-


ment. Graphs represent mean ± SEM
(n = 4–6). * p < 0.05.

tolerance since tryptophan depletion reduces T cell proliferation, while required to induce FoxP3+ Tregs, it is dispensable for Treg induction by
kynurenine pushes Treg differentiation via the aryl hydrocarbon re- 1,25D3-DCs [37]. Antigen-specific iTregs are believed to suppress dia-
ceptor (AhR). In addition, low tryptophan levels increase ILT-3 and ILT- betogenic Th1 cells via linked suppression and infectious tolerance
4 expression on human DCs [33]. Although ILT-3 expression by DCs is [38].

108
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

In the present report, we showed that 1,25D3-DCs secrete IL-10 and 46 (2017) 1061–1094.
were fully active in vitro and in vivo. The xenogeneic GVHD model in [4] R.M. Steinman, Some interfaces of dendritic cell biology, Apmis 111 (2003)
675–697.
NSG mice provides the unique opportunity to test the functionality of [5] L. Adorini, G. Penna, Dendritic cell tolerogenicity: a key mechanism in im-
human tolerogenic DCs in an in vivo situation. NSG mice are a model for munomodulation by vitamin D receptor agonists, Hum. Immunol. 70 (2009)
immunodeficiency as these animals lack functional T cells, B cells and 345–352.
[6] G.B. Ferreira, et al., Differential protein pathways in 1,25-dihydroxyvitamin D 3
NK cells. Moreover, NSG mice do not express the IL-2Rγ resulting in and dexamethasone modulated tolerogenic human dendritic cells, J. Proteome Res.
defective cytokine signaling pathways [39]. In this mouse model, 11 (2012) 941–971.
human PBMC transfer will cause GVHD due to the immune reaction of [7] G.B. Ferreira, et al., 1,25-Dihydroxyvitamin D3 alters murine dendritic cell beha-
viour in vitro and in vivo, Diabetes Metab. Res. Rev. 27 (2011) 933–941.
human T cells against murine tissues. The severity of the induced GVHD [8] A.G.S. Van Halteren, et al., Redirection of human autoreactive T-cells upon inter-
depends on the dose of PBMCs infused. Interestingly, infusion of en- action with dendritic cells modulated by TX527, an analog of 1,25 dihydrox-
riched Tregs have been shown to delay the development of xenogeneic yvitamin D3, Diabetes 51 (2002) 2119–2125.
[9] G.B. Ferreira, et al., 1,25-Dihydroxyvitamin D3 promotes tolerogenic dendritic cells
GVHD in NSG mice [40]. In the current study, the co-transfer of 1,25D3-
with functional migratory properties in NOD Mice, J. Immunol. 192 (2014).
DCs alongside human PBMCs could significantly delay xenogeneic [10] W.W.J. Unger, S. Laban, F.S. Kleijwegt, A.R. Van Der Slik, B.O. Roep, Induction of
GVHD, suggesting the induction of Tregs in this model and demon- Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone:
strating the stability of the tolerogenic profile in the highly pro-in- Differential role for PD-L1, Eur. J. Immunol. 39 (2009) 3147–3159.
[11] P. Reddy, et al., Histone deacetylase inhibition modulates indoleamine 2,3-dioxy-
flammatory milieu in GVHD. Although we did not completely prevent genase–dependent DC functions and regulates experimental graft-versus-host dis-
GVHD, it would be interesting to study whether higher DC doses or ease in mice, J. Clin. Invest. 118 (2008) 2562–2573.
repetitive dosing need to be explored. Our results indeed showed that [12] W. Reichardt, et al., Impact of mammalian target of rapamycin inhibition on lym-
phoid homing and tolerogenic function of nanoparticle-labeled dendritic cells fol-
the induction of Tregs in vivo is a transient phenomenon that probably lowing allogeneic hematopoietic cell transplantation, J. Immunol. 181 (2008)
requires multiple doses of 1,25D3-DCs to obtain a stable Treg popula- 4770–4779.
tion. Moreover, we believe that 1,25D3-DCs are a safer and more spe- [13] K. Sato, N. Yamashita, N. Yamashita, M. Baba, T. Matsuyama, Regulatory dendritic
cells protect mice from murine acute graft-versus-host disease and leukemia re-
cific way to interfere with GVHD in the allo-BMT setting compared to lapse, Immunity 18 (2003) 367–379.
general immunosuppression with pharmacologic inhibitors. This ob- [14] A. Chorny, E. Gonzalez-Rey, A. Fernandez-Martin, D. Ganea, M. Delgado,
servation is also of great importance for the current application of Vasoactive intestinal peptide induces regulatory dendritic cells that prevent acute
graft-versus-host disease while maintaining the graft-versus-tumor response, Blood
1,25D3-DCs in several autoimmune disorders, like type 1 diabetes 107 (2006) 3787–3794.
(Trialregister.nl; NTR5542) and multiple sclerosis (MS) (Clinical- [15] J. Cuende, et al., Monoclonal antibodies against GARP/TGF-β1 complexes inhibit
trial.gov; NCT02903537 and NCT02618902). In these circumstances, the immunosuppressive activity of human regulatory T cells in vivo, Sci. Transl.
Med. 7 (2015) 284ra56.
monocytes from autoimmune patients are isolated, treated ex vivo with
[16] T. Nikolic, et al., Differential transcriptome of tolerogenic versus inflammatory
high doses of 1α,25(OH)2D3, and loaded with disease-relevant antigens dendritic cells points to modulated T1D genetic risk and enriched immune reg-
(e.g. proinsulin in type 1 diabetes or myelin peptides in MS). Once the in ulation, Genes Immun. 18 (2017) 176–183.
vitro induction of the tolerogenic profile of these antigen-specific [17] J.S. Manavalan, et al., High expression of ILT3 and ILT4 is a general feature of
tolerogenic dendritic cells, Transpl. Immunol. 11 (2003) 245–258.
1,25D3-DCs is confirmed, the cells are transferred back into the patients [18] G. Penna, L. Adorini, 1, 25-Dihydroxyvitamin D3 Inhibits Differentiation,
in order to induce Tregs specific for the antigens used. Our data indicate Maturation, Activation, and Survival of Dendritic Cells Leading to Impaired
that it is highly unlikely that these cells would revert to an im- Alloreactive T Cell Activation, J. Immunol. 164 (2000) 2405–2411.
[19] L. Piemonti, et al., Vitamin D3 affects differentiation, maturation, and function of
munogenic phenotype and aggravate disease upon transfer into a pro- human monocyte-derived dendritic cells, J. Immunol. 164 (2000) 4443–4451.
inflammatory environment in vivo [20] F.S. Kleijwegt, et al., Critical role for TNF in the induction of human antigen-specific
In conclusion, we verified the potential of 1α,25(OH)2D3 to induce regulatory T cells by tolerogenic dendritic cells, J. Immunol. 185 (2010)
1412–1418.
tolerogenic DCs, that remain stable when challenged with pro-in- [21] G. Ferreira, et al., Vitamin D3 induces tolerance in human dendritic cells by acti-
flammatory signals both in vitro and in vivo, strengthening the potential vation of intracellular metabolic pathways, Cell Rep. 10 (2015) 711–725.
use of these 1,25D3-DCs for tolerance induction in autoimmune diseases [22] G.B. Ferreira, et al., Proteome analysis demonstrates profound alterations in human
dendritic cell nature by TX527, an analogue of vitamin D, Proteomics 9 (2009)
and open therapeutic routes for the treatment of GVHD.
3752–3764.
[23] M. Naranjo-Gomez, et al., Comparative study of clinical grade human tolerogenic
Competing interests dendritic cells, J. Transl. Med. 9 (2011) 89.
[24] C. Carlberg, M.J. Campbell, Vitamin D receptor signaling mechanisms: integrated
actions of a well-defined transcription factor, Steroids 78 (2013) 127–136.
There are no conflicts of interest to be disclosed. [25] S. Heikkinen, et al., Nuclear hormone 1α,25-dihydroxyvitamin D3 elicits a genome-
wide shift in the locations of VDR chromatin occupancy, Nucleic Acids Res. 39
Acknowledgements (2011) 9181–9193.
[26] L. Széles, et al., 1,25-Dihydroxyvitamin D3 is an autonomous regulator of the
transcriptional changes leading to a tolerogenic dendritic cell phenotype, J.
We would like to thank Frea Coun and Dries Swinnen for their Immunol. 182 (2009) 2074–2083.
technical assistance. We thank the staff at the VIB-KU Leuven FACS [27] A. Neme, V. Nurminen, S. Seuter, C. Carlberg, The vitamin D-dependent tran-
scriptome of human monocytes, J. Steroid Biochem. Mol. Biol. 164 (2016) 180–187.
Core Facility for assistance with flow cytometry and the staff of the [28] V. Nurminen, et al., The transcriptional regulator BCL6 participates in the sec-
laboratory of Experimental Radiotherapy for assistance with the irra- ondary gene regulatory response to vitamin D, Biochim. Biophys. Acta - Gene Regul.
diations. This work was supported by grants from the Flemish Research Mech. 1849 (2015) 300–308.
[29] D. D’Ambrosio, et al., Inhibition of IL-12 production by 1,25-dihydroxyvitamin D3.
Foundation (FWO Vlaanderen) through an aspirant mandate to AS.V. Involvement of NF-kappaB downregulation in transcriptional repression of the p40
(11Y5915N) and D.P.C. (11Y6716N), FWO G.0672.14N, FWO Krediet gene, J. Clin. Invest. 101 (1998) 252–262.
aan Navorser 1.5140.15N, the Boehringer Ingelheim Fund and the KU [30] V. Nurminen, A. Neme, S. Seuter, C. Carlberg, The impact of the vitamin D-
modulated epigenome on VDR target gene regulation, Biochim. Biophys. Acta -
Leuven (GOA2014/10).
Gene Regul. Mech. 1861 (2018) 697–705.
[31] S.P. Cobbold, E. Adams, K.F. Nolan, F.S. Regateiro, H. Waldmann, Connecting the
References mechanisms of T-cell regulation: dendritic cells as the missing link, Immunol. Rev.
236 (2010) 203–218.
[32] G. Penna, et al., Expression of the inhibitory receptor ILT3 on dendritic cells is
[1] G.L. Phillips, Allogeneic hematopoietic stem cell transplantation (HSCT) for high- dispensable for induction of CD4+Foxp3+ regulatory T cells by 1,25-dihydrox-
risk acute myeloid leukemia (AML)/myelodysplastic syndrome (MDS): How can we yvitamin D3, Blood 106 (2005) 3490–3497.
improve outcomes in the near future? Leuk. Res. 36 (2012) 1490–1495. [33] R. Kushwah, J. Hu, Role of dendritic cells in the induction of regulatory T cells, Cell
[2] J.L.M. Ferrara, P. Reddy, Pathophysiology of graft-versus-host disease, Semin. Biosci. 1 (2011) 20.
Hematol. 43 (2006) 3–10. [34] D.X. Beringer, et al., T cell receptor reversed polarity recognition of a self-antigen
[3] A.S. Vanherwegen, C. Gysemans, C. Mathieu, Regulation of immune function by major histocompatibility complex, Nat. Immunol. 16 (2015) 1153–1161.
vitamin d and its use in diseases of immunity, Endocrinol. Metab. Clin. North Am. [35] N. Gagliani, et al., Coexpression of CD49b and LAG-3 identifies human and mouse T

109
A.-S. Vanherwegen et al. Journal of Steroid Biochemistry and Molecular Biology 188 (2019) 103–110

regulatory type 1 cells, Nat. Med. 19 (2013) 739–746. Proinflammatory Dendritic Cells via Induced Autoreactive Regulatory T Cells, J.
[36] H. Kuipers, et al., Contribution of the PD-1 ligands/PD-1 signaling pathway to Immunol. 187 (2011) 6357–6364.
dendritic cell-mediated CD4+ T cell activation, Eur. J. Immunol. 36 (2006) [39] L.D. Shultz, M.A. Brehm, J.V. Garcia-Martinez, D.L. Greiner, Humanized mice for
2472–2482. immune system investigation: progress, promise and challenges, Nat. Rev.
[37] G. Penna, et al., 1,25-Dihydroxyvitamin D3 selectively modulates tolerogenic Immunol. 12 (2012) 786–798.
properties in myeloid but not plasmacytoid dendritic cells, J. Immunol. 178 (2007) [40] M. Hannon, et al., Infusion of clinical-grade enriched regulatory T cells delays ex-
145–153. perimental xenogeneic graft-versus-host disease, Transfusion 54 (2013).
[38] F.S. Kleijwegt, et al., Transfer of Regulatory Properties from Tolerogenic to

110

You might also like