Original Article
Print
Original Article
Live but not heat-killed Staphylococcus epidermidis elicit an anti-inflammatory phenotype in human embryonic Langerhans cells
expand article infoYi Pan§, Mathias Hochgerner|, Natalie Bordag, Markus Absenger-Novak, Herbert Strobl, Thomas Bieber§, Peter Wolf
‡ Medical University of Graz, Graz, Austria
§ Christine Kühne-Center for Allergy Research and Education, Medicine Campus, Davos, Switzerland
| Fudan University, Shanghai, China
Open Access

Abstract

Background: In atopic dermatitis (AD), the skin is colonized by high levels of Staphylococcus aureus (SA), while levels of the commensal Staphylococcus epidermidis (SE) are reduced. Levels of SA correlate with the severity of the disease, while transplantation of SE into the skin can ameliorate AD.

Objectives: It is hypothesized that the underlying mechanism is supported by Langerhans cells (LCs) which sense a shift in the microbiome and then either continue to perpetuate the disease (with SA) or to act as anti-inflammatory agents, ameliorating the inflammation (with SE). However, many different experimental protocols generating very different results have made the literature on this topic complex and hard to understand.

Methods: In this study, we specifically tested human, embryonic stem cell-derived LCs and measured their reactions to SA and SE. We directly compared co-cultures exposed to living SA and SE and to heat-killed bacteria. We analyzed the gene expression of the LCs with qPCR and flow cytometry, the secreted cytokines via ELISA, and their downstream effect on T cells.

Results: Our data show that exposure to living SA causes LCs to mature; in turn, this activates a T-cell response of cytokine secretions resembling the inflammatory phenotype observed in AD patients. Meanwhile, SE-primed LCs do not activate T cells, but instead act as anti-inflammatory agents by secreting high levels of IL-10. Importantly, this difference can only be observed in living bacteria. LCs react to heat-killed SE as pro-inflammatory agents, as they do to SA (albeit slightly weaker).

Conclusions: Using living bacteria is absolutely crucial when performing experiments to reflect the in vivo situation. Our results expand on the work of others and contribute to the ongoing investigation of interactions of the skin and its microbiome.

Why was the study undertaken?

Numerous studies have observed that SE not only correlates with less severe AD, but can ameliorate the disease. This study was undertaken to further investigate the immunological mechanisms related to this effect.

What does this study add?

Our data shows that SE modifies T-cell reactions via LCs. Importantly, only living SE has this anti-inflammatory effect; heat-killed SE instead has a pro-inflammatory effect similar to that of S. aureus.

What are the implications of this study for the understanding of skin physiology and pathology and/or disease management?

The skin microbiome plays a key role in AD, but at the heart of AD pathophysiology are T cells. We present further evidence that the missing link between the microbiome and the T cells is in fact the LCs.

List of abbreviations: AD: Atopic dermatitis; APC: Antigen-presenting cell; DC: Dendritic cell; LC: Langerhans cell; mo-LC: Monocyte-derived Langerhans cell; LN: Lymph nodes; PBMCs: Peripheral blood mononuclear cells; SA: Staphylococcus aureus; SE: Staphylococcus epidermidis.

Key words:

Langerhans cell, atopic dermatitis, skin microbiome, immunology, Staphylococcus aureus, Staphylococcus epidermidis

Introduction

Langerhans cells are myeloid immune cells that display dendritic morphology and form a tight network in the epidermis [1, 2]. They exhibit characteristics of both macrophages and dendritic cells: LCs are embryonically derived and self-renewing [3]. They sample antigens and migrate to the skin-draining lymph nodes (LN). As professional antigen-presenting cells (APCs), LCs can present antigens via both MHC-1 to CD8+ T cells and via MHC-II to CD4+ T cells. LCs are also excellent cross-presenters [4, 5]. LCs are key players in skin immunity, and especially in the defence against viral infections. However, in their un-activated (immature) state, LCs act anti-inflammatory agents [6, 7]. In fact, even in the steady state, LCs constantly migrate towards the LN, carrying self-antigens or antigens from non-pathogenic microbes [8]. Thus, LCs act to maintain immunological tolerance and prevent pathological immune reactions [9].

LC dendrites extend through tight junctions, also allowing them to take up antigens on the surface of the skin, which have not penetrated into the body [10]. Thus, LCs are an important sensor of the skin microbiome.

Healthy human skin is colonized by a wide variety of microbes. The skin and the microbiome constantly interact, influencing each other [11, 12]. While much progress in this field has been made in recent decades, the exact mechanisms remain incompletely understood.

The microbiome is of special interest in chronic skin diseases. Many skin diseases are associated with changes in the microbiome. One such disease is atopic dermatitis [13]. AD is the most common chronic inflammatory skin disease worldwide. It is strongly associated with microbial dysbiosis and most importantly with an overgrowth of Staphylococcus aureus (SA) and a reduction of Staphylococcus epidermidis (SE) [14]. Higher levels of SA correlate with more severe disease forms, while higher levels of SE correlate with milder forms [15]. Transplanting a healthy microbiome, especially with SE, to patients has been shown to ameliorate AD [16, 17]. However, the exact mechanism by which the microbiome influences AD, and especially how SE can ameliorate the symptoms, remains incompletely understood.

LCs are key cells in AD; while their exact function remains incompletely understood, studies have shown that LCs in AD patients are highly activated and pro-inflammatory [18, 19].

Currently, scientists hypothesize that LCs in the epidermis sense SE, react by inducing anti-inflammatory programs and then shape the downstream immune reaction accordingly. Thus, not only is the inflammation perpetuated by SA in AD, but the important anti-inflammatory signal from SE is also lost [20].

Much research has already been performed in this field. However, information in the literature is far from consistent. Some studies have used human cells, while others have used mouse cells. While the two seem very similar, murine and human skin are actually quite different [21]. Furthermore, mouse skin contains a population of Langerin-expressing dermal dendritic cells (DCs), which are not present in human skin and not equivalent to LCs [22]. Conversely, human skin in AD is infiltrated by inflammatory dendritic epidermal cells (IDEC), which are not found in mice [23]. Both of these cell types have at times been confused with real LCs. Additionally, human Langerin reacts to specific cell wall components of SA, which mouse Langerin does not [24].

Many studies use bone marrow-derived, LC-like DCs or work with monocyte-derived LCs (mo-LCs), which are both quite different from real, embryonic LCs. For example, mo-LCs but not real LCs have been shown to be drivers of AD [25], while the deletion of real LCs exacerbates skin inflammations [6].

On the other hand, some studies have used living bacteria (or the cell culture supernatant from these), while others have used killed bacteria (produced with different protocols) or only bacterial components. Importantly, one study has demonstrated that the method of killing bacteria strongly changes their antigenicity and the reaction of immune cells to them [26]. Thus, such experiments may not reflect the in vivo situation very well.

In this study, we compared the reactions of human embryonically derived LCs to SA and to SE. We compared the effects of exposing both heat-killed and living bacteria to LC and analyzed LC gene expression, marker expression and secreted cytokines. The study results help to clarify some of the confusion in the literature.

Materials and methods

Isolation of CD34+ cord blood stem cells

Cord blood was collected after healthy full/term deliveries, and peripheral blood mononuclear cells (PBMCs) were isolated by performing density gradient centrifugation with Ficoll® Paque Premium media. Afterwards, CD34+ stem cells were isolated by applying magnetic sorting with a Miltenyi Biotech CD34+ micro bead kit according to the manufacturer’s instructions. The purity of each batch of enriched cells was confirmed with flow cytometry. Only isolates with a purity of > 95% were used for experiments. CD34+ stem cells were stored in liquid nitrogen until use. Informed consent forms were obtained from all patients as required by the Declaration of Helsinki. Ethics approval was obtained from the ethics committee of the University of Bonn and the Medical University of Graz Institutional Review Board (26-520).

In vitro generation of Langerhans cells

Thawed CD34+ hematopoietic stem cells were differentiated into LCs over 8 days in cell culture. We used RMPI 1640 with GlutaMAXTM, 50 µM 2-mercaptoethanol, 10% FBS and 1% penicillin-streptomycin as a differentiation medium with GM-CSF (300 IU/ml), mIgE (1 µg/ml), FLT3L (10 ng/ml), SCF (10 ng/ml), TGF-β (0.5 ng/ml) and TNF-α (20 U/ml). After 8 days of differentiation, LCs were collected and enriched with a Miltenyi Biotech CD207+ micro bead kit according to the manufacturer’s instructions. The purity of the generated CD207+ cells was confirmed with flow cytometry. In further experiments, only isolates with a purity of > 90% were used.

Flow cytometry

Antibodies used for flow cytometry are listed in Table 1. Intracellular staining was performed with a BD Cytofix/Cytoperm Kit (Beckton Dickinson, Germany) according to the manufacturer’s protocol. Experiments performed in Bonn used 7-AAD (Biolegend, USA) as viability stain and were performed on a FACSCantoTM flow cytometer. Experiments performed in Graz used Viability™ 405/520 Fixable Dye (Miltenyi Biotec, Germany) and were performed on a CytoFlex LX (Beckman Coulter, Germany). Data was analyzed with FACSDiva™ (Beckton Dickinson, Germany) or FlowJo v10 (FlowJo, USA).

Table 1.

Antibodies analyzed using flow cytometry.

Antibody Source Clone Identifier #
Biotin anti-human CD281 (TLR1) Antibody Biolegend TLR1.136 334504
Brilliant Violet 605™ Mouse IgG1 Biolegend MOPC-21 400161
CCR5 APC Miltenyi Biotec REA245 130-123-057
CCR6 PE Miltenyi Biotec REA190 130-120-458
CCR7 PE Miltenyi Biotec REA546 130-119-583
CD11b Percpvio700 Miltenyi Biotec REA713 130-110-557
CD11c Viobright515 Miltenyi Biotec REA618 130-127-207
CD14 APC Miltenyi Biotec TÜK4 130-113-705
CD14 Percpvio700 Miltenyi Biotec REA599 130-110-523
CD1a RD1 Beckman Coulter SCFI19Thy1A8 6603185
CD1a VioBlue Miltenyi Biotec REA736 130-111-875
CD1b PEvio770 Miltenyi Biotec SN13 130-101-578
CD1c PEvio615 Miltenyi Biotec REA618 130-126-883
CD206 Percpvio700 Miltenyi Biotec DCN228 130-104-129
CD207 Beckman Coulter DCGM4 PN IM3449
CD207 APC-Vio770 Miltenyi Biotec REA770 130-112-214
CD209 Percpvio700 Miltenyi Biotec REA617 130-109-593
CD324 PEvio615 Miltenyi Biotec REA811 130-125-732
CD34 PE Becton Dickinson 581 (RUO) 555822
CD36 PE Miltenyi Biotec REA760 130-110-740
CD40 PEvio770 Miltenyi Biotec REA733 130-110-948
CD80 PE Miltenyi Biotec REA661 130-123-253
CD83 Santa Cruz Biotechnology HB15a sc-19677
CD83 APC Miltenyi Biotec REA714 130-110-504
CD86 Viobright515 Miltenyi Biotec REA968 130-116-165
CX3CR1 APC Miltenyi Biotec REA385 130-122-912
CXCR1 PEvio770 Miltenyi Biotec REA958 130-115-881
FceR1a eBioscience AER-37 16-5899-025
FceRIa FITC Miltenyi Biotec REA758 130-110-726
Gam FITC Jackson ImmunoResearch Polyclonal 115-095-003
HLA-DR PEvio615 Miltenyi Biotec REA805 130-111-797
IgG1 PE Becton Dickinson X40 340761
IgG1 RD1 Beckman Coulter 2T8-2F5 6602884
IgG2a APC Miltenyi Biotec S43.10 130-113-831
IgG2b Sigma-Aldrich MOPC-141 M5534
mouse IgG1κ Percpvio700 Miltenyi Biotec IS5-21F5 130-113-776
mouse IgG1κ PEvio770 Miltenyi Biotec IS5-21F5 130-113-764
Recombinant human IgG1 APC Miltenyi Biotec REA293 130-113-446
Recombinant human IgG1 APC-Vio770 Miltenyi Biotec REA293 130-113-447
Recombinant human IgG1 FITC Miltenyi Biotec REA293 130-113-449
Recombinant human IgG1 PE Miltenyi Biotec REA293 130-113-450
Recombinant human IgG1 Percpvio700 Miltenyi Biotec REA293 130-113-453
Recombinant human IgG1 PEvio615 Miltenyi Biotec REA293 130-113-451
Recombinant human IgG1 PEvio770 Miltenyi Biotec REA293 130-113-452
Recombinant human IgG1 VioBlue Miltenyi Biotec REA293 130-113-454
Recombinant human IgG1 Viobright515 Miltenyi Biotec REA293 130-113-457
TLR2 Imgenex 1030A5.138 MAB0066
TLR2 PEvio615 Miltenyi Biotec REA109 130-107-462
TLR6 PE-Vio770 Miltenyi Biotec REA382 130-106-590
Trop1 FITC Miltenyi Biotec REA764 130-110-998
Trop2 APC Miltenyi Biotec REA916 130-115-056

Isolation of RNA

RNA was isolated from harvested cells by using the TRIzol® reagent and following the manufacturer’s protocol. The RNA concentration and purity were measured with a NanoDrop™ spectrophotometer (Thermo Fisher Scientific, MA, USA). The RNA was then transcribed to cDNA by using the SuperScript™ reverse transcriptase.

qRT-PCR

qRT-PCR was performed with the SYBR® Green Supermix with ROXTM according to the manufacturer’s instructions. The primers used are listed in Table 2.

Table 2.

Primers used to perform qRT-PCR.

Gene Forward sequence Reverse sequence
ACTB AGCGCGGCTACAGCTTCA TCCTTAATGTCACGCACGATTT
CCL13 GTGCCTGCTGCTCATGACA TGCATCTGGCTGAGCAAGTC
CCL17 GAAGACGTGGTACCAGACATCTGA CCCTGCACAGTTACAAAAACGA
CCL22 CGGCGCCAACATGGAA CAGACGGTAACGGACGTAATCA
CCR6 CCATTCTGGGCAGTGAGTCA GCACGTGGCATTGCTGAA
CCR7 GCTGCGTCAACCCTTTCTTG AAGAGATCGTTGCGGAACTTG
CD83 GCCTCGAAAACCATCACATGA GGTGGCCATGGAGAAGCA
Elf.1 TGCCCCAGTCACCCATGT ACCCGGTGAGTCTGCATATT
FceRIa GGCAGCTGGACTATGAGTCTGA CTTCTCACGCGGAGCTTTTATT
FceRIg GATGCCATCCTGTTTCTGTATGG CACTTGGATCTTCAGTCGACAGTAG
HMGB1 GATCCTAAGAAGCCGAGAGGC CTTATGCTCCTCCCGACAAGT
HMGB2 CCCGGACTCTTCCGTCAATT TCTTCCATCTCTCCGAACACTTC
IL-13Ra1 AAGCGCAATTCCACACTCTACA TGCACCTGCGACGATGACT
IL-2Rg TGTCTAAGGGACTGGCTGAGAGT TGACGAGGCAGAGTCGTTCA
IL-4Ra CCTGGAGCAACCCGTATCC CAAATGTTGACTGCATAGGTGAGAT
Jak1 CGAGATCCCCTTGAAAGACAAG TGCACCGGCTTTCATAGAATC
Jak2 TGCTCCAGAATCACTGACAGAGA ACCACTCCAAAGCTCCAAACA
Jak3 TGCCATCAACAAGCTCAAGACT TGCCATCAACAAGCTCAAGACT
MY88 TCACTGTCTGCGACTACACCAA GGCAAGGCGAGTCCAGAAC
PU.1 GGAGAGCCATAGCGACCATT GGAGCTCCGTGAAGTTGTTC
SP1 GGACTACCTGGAGTGATGCCTAA CCCATCAACGGTCTGGAACT
TLR1 TGTGCTGCCAATTGCTCATT TTTTCCCCATAAGTCTCTCCTAAGAC
TLR2 CCAAGGAAGAATCCTCCAATCA GCTGCCCTTGCAGATACCA
TLR4 CCTCGGCGGCAACTTCATAA AGAGCGGATCTGGTTGTACTG
TLR6 GGGACTCAGCATGGTAGAAGGTA CTCCTGTTACTCTGCAAGCTTTCA
Tyk2 TTCTCTCTGCGTCGCTGTTG CCGCATGATGATGAGATTGG
YY.1 GTTCAGGGATAACTCGGCCA TTCTGCACAGACGTGGACTC

Live imaging

LCs were stained with 5 μM CellTracker™ Blue CMHC Dye in clear, serum-free media. Live SA from 1 ml stock (OD600 = 0.5) was stained with 5 μg/ml SA polyclonal antibody and 5 μg/ml goat anti-rabbit IgG secondary antibody Alexa Fluor 546. Live SE from 1 ml stock (OD600 = 0.5) was stained with 5 μg/ml SE monoclonal antibody and 5 μg/ml goat anti-mouse IgG secondary antibody Alexa Fluor 488. Then, 100 μl of stained live bacteria suspension were added to 5 ml of LCs. Cells were imaged with a Nikon Eclipse Ti2-E inverted microscope, taking one image every 10 min. NIS-Elements C software was used for the image analysis. The antibodies used for live imaging are listed in Table 3.

Table 3.

Antibodies detected with live imaging.

Antibody Source Clone Identifier #
Goat anti-Mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 Invitrogen Polyclonal A-11001
Goat anti-Rabbit IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 546 Invitrogen Polyclonal A-11035
Staphylococcus aureus Polyclonal Antibody Invitrogen Polyclonal PA1-7246
Staphylococcus epidermidis Monoclonal Antibody Invitrogen 17-5 MA1-35788

Bacterial culture

SA (DSM 20372) and SE (DSM 1798) cultures were purchased from the Leibniz Institute DSMZ (German Collection of Microorganisms and Cell Cultures GmbH) in Braunschweig Science Campus, Braunschweig-Süd, Germany. bacteria were grown on tryptic soy agar, and colonies were then inoculated into tryptic soy broth (TSB). bacteria were grown to an OD600 of 0.6, then harvested. For experiments with inactivated bacteria, these were fixed in formalin (1.5%, 1 h, RT) and then heat-inactivated (80 °C, 10 min). For experiments with live bacteria, the overnight culture was used to inoculate fresh RT TSB at a dilution of 1:200. bacteria used for infections were harvested at an OD600 of 0.5. Serial dilutions of the inoculum were plated onto tryptic soy agar to quantify the titre (viable bacteria).

Cytokine ELISA

Cell culture supernatants were collected from the experiments and sterile filtered. Cytokines were measured with LEGENDplex™ cytokine bead assays according to the manufacturer’s instructions. The following kits were used: LEGENDplex™ Human Proinflammatory Chemokine Panel 1 (13-plex), LEGENDplex™ Human Inflammation Panel 1 (13-plex) and LEGENDplex™ Human Th Cytokine Panel (12-plex).

Isolation of CD4+ T cells

Peripheral blood was collected from healthy volunteers. PBMCs were isolated with density gradient centrifugation as for cord blood. CD4+ T cells were isolated by performing magnetic sorting with a Miltenyi Biotech CD4+ micro bead kit (130-096-533) according to the manufacturer’s instructions. The purity of the enriched cells was assessed with flow cytometry. In further experiments, only isolations with a cell purity of ≥ 95% were used. Isolated CD4+ T cells were stored in liquid nitrogen until use.

LC – T cell co-culture

Cord blood-derived LCs were primed with live SA or live SE cultures for 1 hour, then harvested, thoroughly washed and counted. 1×105 viable LCs were co-cultured with 5×105 T cells (LC:TC = 1:5) in 96-well round bottom tissue culture plates in RMPI 1640 10% FBS, 5% penicillin-streptomycin. Cells were harvested after five days and analyzed with flow cytometry. The cell culture medium was analyzed by performing cytokine ELISA. All experiments were performed in duplicate.

Statistical analysis

Gene expression (qRT-PCR) and cytokine measurements (ELISA) were visualized and analyzed with R [27] (v4.4.0, packages stringr, colorspace, RColorBrewer, dplyr, tidyr, readxl, pheatmap, ggplot2, ggpubs, ggbeeswarm, dendsort, nlme, emmeans, rstatix) and Tibco Spotfire (v14.4.0). Normality and scedasticity was tested with a Kolmogorov-Smirnov test with stats::ks.test() and a Bartlett test with stats::bartlett.test(), additionally applying the Benjamini-Hochberg (BH) multiple test adjustment method with stats::p.adjust(). The data distribution improved sufficiently after performing a log10 transformation, and only transformed data were used for the subsequent statistical analysis. A hierarchical clustering analysis was performed centred and scaled to unit variance base::scale() over both the parameters and the samples. Dendrograms were calculated by performing standard clustering stats::hclust(dist()) (Lance-Williams dissimilarity update with complete linkage) and sorted dendsort::dendsort() according to the average distance between the subtrees at every merging point. Heatmaps were plotted using pheatmap::pheatmap(). For the univariate analysis, linear models were fitted by applying the generalized least squares method [28] with nlme::gls() on log10-transformed data with BH multiple test adjustment.

All other statistical analyses were performed using GraphPad Prism V8 software (GraphPad Software). Differences between the two groups were calculated with a two-tailed Student’s t-test. Multiple groups were compared by applying a one-way analysis of variance (ANOVA), corrected with Tukey’s multiple comparison test.

Results

Heat-killed SA and SE both lead to LC activation

In mice and humans, different types of cells have been called “LCs” or “LC-like cells”. However, there are significant differences between real LCs (embryonically derived and self-renewing), mo-LCs (monocyte-derived) and mucosal LCs (bone marrow-derived). In our study, we generated LCs from human cord-blood-derived CD34+ embryonic stem cells. We used a protocol previously developed by our group to generate LCs matching the phenotype found in the skin of AD patients [29]. Our LCs are TLR2+, FCεRI+ and CD83-, i.e. immature cells. The LC quality and phenotypes were controlled for each batch with flow cytometry (Suppl. material 1: fig. S1).

First, we exposed our LCs to heat-killed SA and SE overnight and analyzed LC activation with qPCR and flow cytometry (Fig. 1A). As expected, exposure to SA led to LC maturation, as indicated by the upregulation of CD83 and CCR7 and downregulation of CCR6, TLR2 and FCεRIα (Fig. 1B, C). Exposure to SE led to the same LC maturation; while slight quantitative differences were observed (e.g. SA exposure leads to the higher expression of CCR6 and lower expression of CD83), the overall trend is very much the same.

Next, we analyzed the expression of key transcription factors, receptors and cytokines with qPCR (Fig. 1D, violin plots in Suppl. material 1: fig. S2). Again, exposure to both SA and SE led to the initiation of the relevant maturation-associated genetic programs: TRLs were downregulated as well as the transcription factors PU1 and YY1, while JAK1 and JAK3 were upregulated. For all observed genes, the only significant difference was a slightly higher expression of IL-4Rα in SA-treated as compared to SE-treated LCs. All other analyzed genes showed no difference between these two groups. Thus, both heat-killed SA and SE activate LCs in the same way.

Figure 1. 

Exposure to heat-killed SA and SE causes LCs to activate and mature: A) CD34+ stem cell-derived LCs were exposed for 24 h to heat-killed SA or SE; LCs were harvested and analyzed with qPCR; B) violin plots depict the distribution of cytokine cDNA levels. Each dot represents an individual biological replicate (n = 6). Significant differences between groups were determined by linear mixed-effects models with generalized least squares, followed by the application of a Benjamini-Hochberg correction for multiple comparisons. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001; C) flow cytometry; D) expression levels were normalized to actin. One-way ANOVA, dots represent biological replicates, n = 7–9.

Heat-killed SA and SE both promote the secretion of pro-inflammatory cytokines

Next, we collected the supernatant of our cell cultures and analyzed the secreted cytokines by performing multiplex ELISA (Fig. 2A, B). While the gene expression and expression of surface markers hardly differed between the SA and SE groups, significant differences were observed in the secreted cytokines. Exposure to both SA and SE promoted the secretion of pro-inflammatory cytokines in LCs while this secretion was not observed in the untreated control. However, regarding almost all analyzed cytokines, SA-treated LCs (SA-LCs) secreted significantly higher amounts than SE-treated LCs (SE-LCs). Specifically, SA-treated LCs secreted more CCL11, CCL5, CXCL1, IP-10, TNFα, IL-1β, IL-12p70, IFN-α2, CXCL11, CCL2, CCL4, IL-10, CXCL9, CCL20, CXCL5 and IL-18. SE-LCs also secreted all observed cytokines, but most secreted less of these than SA-LCs. Thus, while SA induces the stronger secretion of cytokines, in principle, exposure of LCs to both SA and SE triggers the same type of reaction. The differences seen are only quantitative.

Figure 2. 

Exposure to heat-killed SA and SE causes LCs to secrete pro-inflammatory cytokines. LCs were stimulated with heat-killed SA or SE for 24 h; then cell culture supernatants were collected and secreted cytokines were analyzed via multiplex ELISA: A) heat-map and B) violin plots depict the distribution of secreted cytokine levels. Each dot represents an individual biological replicate (n = 6). Significant differences between groups were determined by linear mixed-effects models with generalized least squares, followed by the application of a Benjamini-Hochberg correction for multiple comparisons. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001, n = 6.

LCs react very differently to living SA than to SE

In vivo observations correlate high levels of SA with increased inflammation and high levels of SE with reduced inflammation. Contrary to this observation, our first set of experiments showed that LCs react the same way to SA and SE. However, these experiments were performed with heat-killed cells.

In order to better reflect the in vivo situation, we next co-cultured LCs with living SA or SE. We observed the LCs by performing live cell imaging for 4 h. Additionally, LCs were collected after 1 h for flow cytometry analysis.

LCs co-cultured with SA displayed fast, mesenchymal migration. They were also observed to actively engulf SA. LCs co-cultured with SE displayed slow, amoeboid movement and did not engulf bacteria (Fig. 3A, B). Co-culturing LCs with SA also results in rapid cell death, while exposure of LCs to SE only slightly decreases the LC viability at later timepoints (Fig. 3C). This is probably due to SA’s ability to survive phagocytosis and to kill phagocytes from within [30].

SA-LCs were strongly activated, as shown by the upregulation of HLA-DR, CD80, CD86 and CCR7. Interestingly, SE-LCs did not display an upregulation of any of these markers. LCs definitely reacted to exposure to SE, as evidenced by the upregulation of CD1b and CCR5 (also upregulated in SA to comparable levels). However, these cells did not mature (Fig. 3D and Suppl. material 1: fig. S3). Thus, live bacteria affect LCs very differently than heat-killed bacteria.

Figure 3. 

Exposure to live SA activates LCs, while exposure to live SE does not. LCs were co-cultured with live SA or SE: A) live microscopy of LCs co-cultured with SA (red); B) live microscopy of LCs co-cultured with live SE (green); C) viability of LCs co-cultured with live bacteria; D) flow cytometry analysis of LC activation after 1 h. One-way ANOVA, dots represent biological replicates, n = 4.

Live SA induces LCs to produce a very different secretome than live SE

We next collected the supernatants from the co-cultures of LCs and living bacteria and analyzed the cytokines secreted into this supernatant after 1 h, 2 h and 3 h of exposure, respectively, with multiplex ELISA. Already after 1 h of co-culture, SA-LCs and SE-LCs show highly distinct cytokine profiles. These differences are only stronger at later timepoints (Fig. 4A, B).

Specifically, SA-LCs express more IFN-α2, IL-18, IL-1β, IP-10 and (at early timepoints only) TNFα and CCL5. In contrast, SE-LCs express (at later timepoints) more IL-10, IL-6, IL-8, CCL2 and CCL4. Cytokines that were identified but where amounts did not significantly change are shown in Suppl. material 1: fig. S4A.

Thus, LCs secrete cytokines upon exposure to both SA and SE, but the secretory profiles of these LCs are very different. Of note, LC death in the SA culture impacts the secretory profile. The observations of early (1 h) increases in cytokine levels in these cultures may be partly due to their release upon LC death. However, in AD patients where SA infiltrates the epidermis and releases exotoxins there, LC death may also be expected.

Figure 4. 

LCs co-cultured with live SA or SE show different cytokine profiles. LCs were co-cultured with living SA or SE for 1, 2 or 3 h, then supernatant was collected and analyzed with multiplex ELISA. A) heatmap and B) violin plots of secreted cytokines. One-way ANOVA, dots represent biological replicates, n = 4.

LCs primed with live SA induce a T-cell response, but LCs primed with SE do not

Next, we investigated the downstream effect of SA-/SE-LC co-culturing. We exposed LCs to live SA or SE for 1 h, then isolated and washed the LCs. We then co-cultured them together with T cells for five days. The flow cytometry analyses show that exposure to SA-primed LCs induced the activation and proliferation of T cells. T cells co-cultured with SE-primed LCs did not proliferate. They did however still upregulate CD25, indicating that some form of activation occurred (Fig. 5A, B).

The multiplex ELISA of the cell culture supernatant shows that SA-primed cultures contained secretions of high levels of the interleukins IL-5, IL-6, IL-9 and IL-22. SE-primed cultures show no significant upregulation of these cytokines compared to un-primed controls. SE-priming of LCs, however, induced very high secretions of IL-10, which was not found in SA-primed cultures (Fig. 5C). Cytokines that were identified but where amounts did not significantly change are shown in Suppl. material 1: fig. S4B.

Of note, patients suffering from AD display increased serum levels of IL-5 and IL-6 [31]. T cells producing IL-22 are key players in the pathophysiology of AD, directly causing epidermal hyperproliferation [32]. Thus, LCs primed with SA display a cytokine profile similar to that found in LCs of AD-patients, while LCs primed with SE do not.

Figure 5. 

SA-primed LCs cause T-cell proliferation and secretion of cytokines. LCs were exposed to SA or SE for 1 h, then washed and co-cultured with T cells for 5 d. A, B) flow cytometry measurements of activation and proliferation of T cells; C) multiplex ELISA measurements of secreted cytokines. One-way ANOVA, dots represent biological replicates, n = 4.

Discussion

Due to their position upstream of T cells, LCs may be important therapeutic targets for the treatment of many skin diseases. Especially AD is a highly heterogenous disease, manifesting Th1, Th2, Th17 or Th22 biases. SA is associated with the severity of AD, but different sub-strains of SA are additionally associated with more severe or specific sub-forms of the disease. Conversely, transplanting SE onto the skin of patients suffering from AD has been shown to ameliorate their symptoms [16, 17].

Understanding the underlying mechanisms of this effect would allow dermatologists to control the disease more efficiently and offer more effective treatments. Targeting the LCs may offer an effective treatment strategy as an alternative to live-bacteria transplantation; however, this topic is complex and still incompletely understood.

In this study, we show that embryonically derived human LCs react very differently to SA than to SE. Specifically, exposure to SA leads to the activation and maturation of LCs and, further downstream, to the initiation of an inflammatory T-cell response.

In contrast, exposure to SE does not induce LC maturation, but definitely has an effect beyond merely not being pro-inflammatory. Our study findings demonstrate that SE-primed LCs alter their receptor expression and cytokine secretion patterns and influence T cells differently. However, the secretome of these LCs differs from that of SA-primed LCs. T cells co-cultured with SE-primed LCs show activation, but not proliferation. Importantly, SE-LC-primed T cells secrete high levels of IL-10. These findings indicate that SE-LCs may push T cells towards an anti-inflammatory/tolerogenic phenotype, but more research on T cells is needed to clarify this point.

Importantly, these results are only true for LCs co-cultured with living bacteria. While we could detect some differences in LC response when they were exposed to heat-killed SA and SE, these differences were only quantitative. In essence, the LCs show the same type of reaction to heat-killed SA as to SE. Therefore, heat-killing the bacteria seems to destroy whatever anti-inflammatory properties SE may have.

Our results match observations made in clinical settings and also reported in the literature. Laborel-Prenéron et al. showed that the secretome of live SA induces maturation of mo-DCs, but exposure to SE induces an anti-inflammatory phenotype [20]. Strunk et al. previously reported major differences in innate immune reactions to killed and live bacteria [26]; however, most studies on this topic use either mo-LCs or not further described “DCs”.

LCs express a wide variety of pattern-recognition-receptors (PRR), but their PRR expression is very different from that of conventional DCs or macrophages. For this reason, LCs may react differently when exposed to the same antigen [33]. In our study, we used embryonically derived LCs, such as those that have been described to have an anti-inflammatory function in AD. Finally, we have added to the literature on this subject by analysing the expression of a wide range of transcription factors, receptors, ligands and secreted cytokines by LCs.

We hope that our work can help guide further research on this topic. While our findings match those previously obtained in studies on other types of APCs, we again stress the huge impact of heat-killing the bacteria. This denatures the proteins, prohibits the bacteria from producing secreted factors, and exposes molecules which would otherwise be hidden inside the bacteria from the host’s immune system. Thus, heat-killing of bacteria not only destroys antigens, but may also produce/expose neo-antigens which would otherwise not exist in the in vivo situation.

One possible alternative to heat killing may be inactivation with radiation. In a direct comparison, several strains of bacteria elicited significantly stronger immune reactions when heat-killed than when radiation-inactivated [34]. We recommend conducting further experiments to investigate interactions between the microbiome and the immune system to take this into account and to carefully consider older literature in this light.

Acknowledgements

Parts of the laboratory work of this research were conducted by Yi Pan when she was a PhD student at the University Hospital of Bonn, Bonn, Germany. We sincerely thank Christina Passegger for her excellent technical assistance.

Additional information

Conflict of interest

The authors have declared that no competing interests exist.

Ethical statement

This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of the University Bonn (Nr. 204/09) and the Medical University of Graz Institutional Review Board (26-520).

The authors declared that no clinical trials were used in the present study.

The authors declared that no experiments on humans or human tissues were performed for the present study.

The authors declared that no informed consent was obtained from the humans, donors or donors’ representatives participating in the study.

The authors declared that no experiments on animals were performed for the present study.

The authors declared that no commercially available immortalised human and animal cell lines were used in the present study.

Funding

PW was supported by the Austrian Science Fund (FWF - W1241). YP was supported by the China Scholarship Council and received research support from the company Shanghai Biocelline Enterprise Co. Ltd, China. The project was supported by the Christine Kühne Center for Allergy Research and Education, Davos, Switzerland.

Author contributions

YP: Conceptualization; Funding acquisition; Material preparation; Planning and performing the experiments; Data collection and analysis; Writing original draft. MH: Planning and performing the experiments; Data analysis; Writing and editing. NB: Data analysis; Software; Visualization. MA: Live imaging. HS: Collect and prepare human stem cells. TB: Conceptualization; Funding acquisition; Supervision; Review and editing. PW: Conceptualization; Funding acquisition; Supervision; Review and editing.

Author ORCIDs

Yi Pan https://orcid.org/0000-0001-6723-1370

Mathias Hochgerner https://orcid.org/0000-0003-4987-4393

Natalie Bordag https://orcid.org/0000-0002-9505-6271

Herbert Strobl https://orcid.org/0000-0002-8070-4977

Thomas Bieber https://orcid.org/0000-0002-8800-3817

Peter Wolf https://orcid.org/0000-0001-7777-9444

Data availability

All of the data that support the findings of this study are available in the main text or Supplementary Information.

References

  • 1. Schuler G, Steinman RM. Murine epidermal Langerhans cells mature into potent immunostimulatory dendritic cells in vitro. J Exp Med. 1985;161(3):526–46. https://doi.org/10.1084/jem.161.3.526
  • 2. Stingl G, Wolff-Schreiner EC, Pichler WJ, Gschnait F, Knapp W, Wolff K. Epidermal Langerhans cells bear Fc and C3 receptors. Nature. 1977;268(5617):245–6. https://doi.org/10.1038/268245a0
  • 3. Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med. 2012;209(6):1167–81. https://doi.org/10.1084/jem.20120340
  • 5. Fehres CM, Duinkerken S, Bruijns SCM, Kalay H, van Vliet SJ, Ambrosini M, et al. Langerin-mediated internalization of a modified peptide routes antigens to early endosomes and enhances cross-presentation by human Langerhans cells. Cell Mol Immunol. 2017;14(4):360–70. https://doi.org/10.1038/cmi.2015.87
  • 6. Bobr A, Olvera-Gomez I, Igyarto BZ, Haley KM, Hogquist KA, Kaplan DH. Acute ablation of Langerhans cells enhances skin immune responses. J Immunol. 2010;185(8):4724–8. https://doi.org/10.4049/jimmunol.1001802
  • 7. Gomez de Agüero M, Vocanson M, Hacini-Rachinel F, Taillardet M, Sparwasser T, Kissenpfennig A, et al. Langerhans cells protect from allergic contact dermatitis in mice by tolerizing CD8+ T cells and activating Foxp3+ regulatory T cells. J Clin Invest. 2012;122(5):1700–11. https://doi.org/10.1172/JCI59725
  • 8. Chorro L, Sarde A, Li M, Woollard KJ, Chambon P, Malissen B, et al. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J Exp Med. 2009;206(13):3089–100. https://doi.org/10.1084/jem.20091586
  • 9. Kitashima DY, Kobayashi T, Woodring T, Idouchi K, Doebel T, Voisin B, et al. Langerhans cells prevent autoimmunity via expansion of keratinocyte antigen-specific regulatory T cells. EBioMedicine. 2018;27:293–303. https://doi.org/10.1016/j.ebiom.2017.12.022
  • 10. Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med. 2009;206(13):2937–46. https://doi.org/10.1084/jem.20091527
  • 11. Chen H, Zhao Q, Zhong Q, Duan C, Krutmann J, Wang J, et al. Skin Microbiome, Metabolome and Skin Phenome, from the Perspectives of Skin as an Ecosystem. Phenomics. 2022;2(6):363–82. https://doi.org/10.1007/s43657-022-00073-y
  • 12. Meisel JS, Sfyroera G, Bartow-McKenney C, Gimblet C, Bugayev J, Horwinski J, et al. Commensal microbiota modulate gene expression in the skin. Microbiome. 2018;6(1):20. https://doi.org/10.1186/s40168-018-0404-9
  • 13. Hrestak D, Matijašić M, Čipčić Paljetak H, Ledić Drvar D, Ljubojević Hadžavdić S, Perić M. Skin Microbiota in Atopic Dermatitis. Int J Mol Sci. 2022;23(7). https://doi.org/10.3390/ijms23073503
  • 15. Simpson EL, Villarreal M, Jepson B, Rafaels N, David G, Hanifin J, et al. Patients with Atopic Dermatitis Colonized with Staphylococcus aureus Have a Distinct Phenotype and Endotype. J Invest Dermatol. 2018;138(10):2224–33. https://doi.org/10.1016/j.jid.2018.03.1517
  • 16. Liu X, Qin Y, Dong L, Han Z, Liu T, Tang Y, et al. Living symbiotic bacteria-involved skin dressing to combat indigenous pathogens for microbiome-based biotherapy toward atopic dermatitis. Bioact Mater. 2023;21:253–66. https://doi.org/10.1016/j.bioactmat.2022.08.019
  • 17. Myles IA, Williams KW, Reckhow JD, Jammeh ML, Pincus NB, Sastalla I, et al. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight. 2016;1(10). https://doi.org/10.1172/jci.insight.86955
  • 18. Pan Y, Hochgerner M, Cichoń MA, Benezeder T, Bieber T, Wolf P. Langerhans cells: Central players in the pathophysiology of atopic dermatitis. J Eur Acad Dermatol Venereol. 2024. https://doi.org/10.1111/jdv.20291
  • 19. Schuller E, Teichmann B, Haberstok J, Moderer M, Bieber T, Wollenberg A. In situ expression of the costimulatory molecules CD80 and CD86 on Langerhans cells and inflammatory dendritic epidermal cells (IDEC) in atopic dermatitis. Arch Dermatol Res. 2001;293(9):448–54. https://doi.org/10.1007/s004030100263
  • 20. Laborel-Préneron E, Bianchi P, Boralevi F, Lehours P, Fraysse F, Morice-Picard F, et al. Effects of the Staphylococcus aureus and Staphylococcus epidermidis Secretomes Isolated from the Skin Microbiota of Atopic Children on CD4+ T Cell Activation. PLoS One. 2015;10(10):e0141067. https://doi.org/10.1371/journal.pone.0141067
  • 23. Wollenberg A, Kraft S, Hanau D, Bieber T. Immunomorphological and ultrastructural characterization of Langerhans cells and a novel, inflammatory dendritic epidermal cell (IDEC) population in lesional skin of atopic eczema. J Invest Dermatol. 1996;106(3):446–53. https://doi.org/10.1111/1523-1747.ep12343596
  • 24. van Dalen R, De La Cruz Diaz JS, Rumpret M, Fuchsberger FF, van Teijlingen NH, Hanske J, et al. Langerhans cells sense Staphylococcus aureus wall teichoic acid through langerin to induce inflammatory responses. mBio. 2019;10(3). https://doi.org/10.1128/mBio.00330-19
  • 25. Xiao C, Zhu Z, Zhang C, Gao J, Luo Y, Fang H, et al. A population of dermal Langerin+ dendritic cells promote the inflammation in mouse model of atopic dermatitis. Front Immunol. 2022;13:981819. https://doi.org/10.3389/fimmu.2022.981819
  • 26. Strunk T, Richmond P, Prosser A, Simmer K, Levy O, Burgner D, et al. Method of bacterial killing differentially affects the human innate immune response to Staphylococcus epidermidis. Innate Immun. 2011;17(6):508–16. https://doi.org/10.1177/1753425910379840
  • 27. R Core Team. R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing. 2024. Available from: https://www.R-project.org/
  • 29. Novak N, Valenta R, Bohle B, Laffer S, Haberstok J, Kraft S, et al. FcεRI engagement of Langerhans cell-like dendritic cells and inflammatory dendritic epidermal cell-like dendritic cells induces chemotactic signals and different T-cell phenotypes in vitro. J Allergy Clin Immunol. 2004;113(5):949–57. https://doi.org/10.1016/j.jaci.2004.02.005
  • 30. Huitema L, Phillips T, Alexeev V, Tomic-Canic M, Pastar I, Igoucheva O. Intracellular escape strategies of Staphylococcus aureus in persistent cutaneous infections. Exp Dermatol. 2021;30(10):1428–39. https://doi.org/10.1111/exd.14235
  • 31. Krupka-Olek M, Bożek A, Kawczyk-Krupka A. The Immunological and Allergen Profiles of Patients with Atopic Dermatitis or Psoriasis. Medicina (Kaunas). 2022;58(3). https://doi.org/10.3390/medicina58030367
  • 32. Nograles KE, Zaba LC, Shemer A, Fuentes-Duculan J, Cardinale I, Kikuchi T, et al. IL-22–producing “T22” T cells account for upregulated IL-22 in atopic dermatitis despite reduced IL-17–producing TH17 T cells. J Allergy Clin Immunol. 2009;123(6). https://doi.org/10.1016/j.jaci.2009.03.041
  • 33. Flacher V, Bouschbacher M, Verronèse E, Massacrier C, Sisirak V, Berthier-Vergnes O, et al. Human Langerhans cells express a specific TLR profile and differentially respond to viruses and Gram-positive bacteria. J Immunol. 2006;177(11):7959–67. https://doi.org/10.4049/jimmunol.177.11.7959
  • 34. Wong C, Ustunol Z. Mode of inactivation of probiotic bacteria affects interleukin 6 and interleukin 8 production in human intestinal epithelial-like Caco-2 cells. J Food Prot. 2006;69(9):2285–8. https://doi.org/10.4315/0362-028X-69.9.2285

1Yi Pan and Mathias Hochgerner contributed equally to this work.

Supplementary material

Supplementary material 1 

Supplementary figures

Yi Pan, Mathias Hochgerner, Natalie Bordag, Markus Absenger-Novak, Herbert Strobl, Thomas Bieber, Peter Wolf

Data type: pdf

Explanation note: figure S1. Generation of LCs from human cord blood CD34+ stem cells. A) CD34+ stem cells were isolated from human cord blood from healthy deliveries and differentiated to LCs as shown above. B) LCs were enriched and qualitycontrolled via flow cytometry. figure S2. Heat-killed S.A. and S.E. both cause LCs to activate and mature. LCs were exposed to S.A. or S.E. for 24h, then gene expression was analyzed via qPCR. Violin plots depict the distribution of cytokine cDNA levels, normalized to actin and log10-transformed. Each dot represents an individual biological replicate (n=6). Significant differences between groups were determined by linear mixed-effects models with generalized least squares, followed by Benjamini-Hochberg correction for multiple comparisons. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001. figure S3. Living SA leads to LC activation, live SE does not. LCs were co-cultured with living SA or SE; Flow cytometry analysis of LC activation after 1h. Grey: Isotype control. Blue: unstimulated LCs. Green: LCs stimulated with S.e. Red: LCs stimulated with S.a. Representative plots from one experiment of two; n=4 per group. figure S4. Tested, but non-significant cytokines (cytokine ELISA from cell culture supernatants). A) LCs were co-cultured with living SA or SE for 1, 2 or 3h (Ad Figure 4B). B) LCs were exposed to SA or SE for 1h, then washed and co-cultured with T-cells for 5d. Then, supernatants were collected and analysed (Ad Figure 5C).

This dataset is made available under the Open Database License (http://opendatacommons.org/licenses/odbl/1.0/). The Open Database License (ODbL) is a license agreement intended to allow users to freely share, modify, and use this Dataset while maintaining this same freedom for others, provided that the original source and author(s) are credited.
Download file (2.29 MB)
login to comment