homeostatic nf-κb signaling in steady-state …...immunity article homeostatic nf-kb signaling in...

28
Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance Graphical Abstract Highlights d A NF-kB-regulated gene network in steady-state migratory NLT-DCs d Spontaneous autoimmunity from targeted deletion of IKKb in DCs d IKKb is required for steady-state migratory NLT-DC accumulation d IKKb in DCs regulates tolerance to endogenous tissue antigens Authors Myriam Baratin, Chloe Foray, ..., Marc Dalod, Toby Lawrence Correspondence [email protected] In Brief Baratin et al. uncover a new role for NF-kB in tolerance. They show that NF-kB regulates migratory dendritic cells (DCs), which are required to maintain tolerance to tissue antigens. When they block NF- kB activation in DCs, these cells no longer migrate to draining lymph nodes, leading to impaired tolerance to tissue antigens. Baratin et al., 2015, Immunity 42, 627–639 April 21, 2015 ª2015 Elsevier Inc. http://dx.doi.org/10.1016/j.immuni.2015.03.003

Upload: others

Post on 31-Aug-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Article

Homeostatic NF-kB Signal

ing in Steady-StateMigratory Dendritic Cells Regulates ImmuneHomeostasis and Tolerance

Graphical Abstract

Highlights

d A NF-kB-regulated gene network in steady-state migratory

NLT-DCs

d Spontaneous autoimmunity from targeted deletion of IKKb in

DCs

d IKKb is required for steady-state migratory NLT-DC

accumulation

d IKKb in DCs regulates tolerance to endogenous tissue

antigens

Baratin et al., 2015, Immunity 42, 627–639April 21, 2015 ª2015 Elsevier Inc.http://dx.doi.org/10.1016/j.immuni.2015.03.003

Authors

Myriam Baratin, Chloe Foray, ...,

Marc Dalod, Toby Lawrence

[email protected]

In Brief

Baratin et al. uncover a new role for NF-kB

in tolerance. They show that NF-kB

regulates migratory dendritic cells (DCs),

which are required to maintain tolerance

to tissue antigens. When they block NF-

kB activation in DCs, these cells no longer

migrate to draining lymph nodes, leading

to impaired tolerance to tissue antigens.

Page 2: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Immunity

Article

Homeostatic NF-kB Signalingin Steady-State Migratory Dendritic CellsRegulates Immune Homeostasis and ToleranceMyriam Baratin,1,2,3 Chloe Foray,1,2,3 Olivier Demaria,1,2,3,6 Mohamed Habbeddine,1,2,3 Emeline Pollet,1,2,3

Julien Maurizio,1,2,3 Christophe Verthuy,1,2,3 Suzel Davanture,1,2,3 Hiroaki Azukizawa,4 Adriana Flores-Langarica,5

Marc Dalod,1,2,3 and Toby Lawrence1,2,3,*1Centre d’Immunologie de Marseille-Luminy (CIML), Aix-Marseille Universite, UM2, 13288 Marseille, France2Institut National de la Sante et de la Recherche Medicale (INSERM), U1104, 13288 Marseille, France3Centre National de la Recherche Scientifique (CNRS), UMR7280, 13288 Marseille, France4Osaka University, Osaka 565-0871, Japan5IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Edgbaston,Birmingham B15 2TT, UK6Present address: Immuno-Dermatologie, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland

*Correspondence: [email protected]

http://dx.doi.org/10.1016/j.immuni.2015.03.003

SUMMARY

Migratory non-lymphoid tissue dendritic cells (NLT-DCs) transport antigens to lymph nodes (LNs) andare required for protective immune responses in thecontext of inflammation and to promote tolerance toself-antigens in steady-state. However, themolecularmechanisms that elicit steady-state NLT-DCmatura-tion and migration are unknown. By comparing thetranscriptomeofNLT-DCs in the skinwith theirmigra-tory counterparts in draining LNs, we have identifieda novel NF-kB-regulated gene network specific tomigratory DCs. We show that targeted deletion ofIKKb in DCs, a major activator of NF-kB, preventsNLT-DC accumulation in LNs and compromises reg-ulatory T cell conversion in vivo. This was associatedwith impaired tolerance and autoimmunity. NF-kB isgenerally considered the prototypical pro-inflamma-tory transcription factor, but this study describes arole for NF-kB signaling in DCs for immune homeo-stasis and tolerance that could have implications inautoimmune diseases and immunity.

INTRODUCTION

Our skin and mucosal tissues, such as lung and intestine, are

continually exposed to foreign antigens and microorganisms in

our diet and the environment. Maintenance of tissue homeosta-

sis is dependent upon the immune system’s ability to remain

tolerant of these antigens and commensal microflora while re-

taining the ability to mount the appropriate immune responses

to pathogenic microorganisms upon infection or injury. Defects

in this delicate balance can lead to the development of chronic

tissue inflammation and autoimmune diseases.

Dendritic cells (DCs) are key regulators of both protective im-

mune responses and tolerance to self-antigens (Steinman et al.,

2003). In steady state, two main groups of DCs can be distin-

guished according to their location and migratory capacity.

Resident lymphoid-tissue (LT)-DCs include plasmacytoid DCs

(pDCs) and CD8a+ and CD11b+ conventional DCs (cDCs).

Non-lymphoid tissue (NLT)-DCs, found in the parenchyma of tis-

sues, also include CD8a-type and CD11b+ cDC subsets. How-

ever, in steady-state a fraction of NLT-DCs migrate through the

lymphatics to draining lymph nodes (LNs), where they are classi-

fied as migratory NLT-DCs (migDCs). NLT-DCs in mucosal tis-

sues and skin regulate both immune tolerance and the response

to invading pathogens by virtue of their ability tomigrate to drain-

ing LNs and transport antigens. In steady-state, this function

promotes the expansion of inducible regulatory T (iTreg) cells,

required for tolerance to self-antigens (Azukizawa et al., 2011;

Guilliams et al., 2010). However, in the context of inflammation,

these cells can transport self or foreign antigens and promote

pathogenic or protective T cell responses, respectively (Bedoui

et al., 2009; Henri et al., 2010).

The molecular mechanisms that regulate NLT-DC migration

in steady state are poorly understood. Accumulation of migDCs

in draining LNs is dependent on expression of the chemokine

receptor CCR7 and its ligands CCL21 and CCL19, which are

highly expressed in lymphoid tissues and present all along

the lymphatic endothelium (Ohl et al., 2004). It has been

proposed that NLT-DCs undergo a ‘‘limited’’ maturation pro-

gram that results in upregulated expression of CCR7 and

subsequent migration in steady-state (Lutz and Schuler,

2002). The functional maturation of DCs has been widely

studied in the context of microbial stimuli or pro-inflammatory

cytokines, which promote morphological, phenotypic, and

functional changes required for the effective priming and acti-

vation of naive T cells. Migratory NLT-DCs undergo similar

morphological and phenotypic changes, including increased

expression of MHC class II and CD40. But in contrast with

DC maturation during inflammation, the steady-state matura-

tion of NLT-DCs is associated with induction of tolerance rather

than T cell priming and activation. However, the signaling path-

ways that regulate steady-state NLT-DC maturation remain an

enigma.

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 627

Page 3: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Comparing the transcriptional program of resident NLT-DCs

to their migratory LN counterparts, we have identified a

novel NF-kB-regulated gene network specific to steady-state

migratory DCs that includes genes previously shown to regulate

migration and Treg cell conversion. Although the NF-kB pathway

is thought to play a prominent role in DC activation in the context

of microbial stimuli and pro-inflammatory cytokines (Dev et al.,

2011), the role of NF-kB in homeostatic DC maturation had not

been addressed. We therefore generated mice with a targeted

deletion of IkB kinase b (IKKb) in the DC lineage—a critical regu-

lator of NF-kB activation. We found that IKKb deletion in DCs

results in dis-regulated immune homeostasis and spontaneous

autoimmunity. Moreover our studies show that homeostatic

NF-kB signaling is required for the steady-state accumulation

of migratory DCs in draining LNs and maintenance of immune

tolerance to tissue antigens. This represents a new and unex-

pected role for NF-kB in immune homeostasis and tolerance

that could have important implications in host defense and

autoimmune diseases.

RESULTS

Steady-State Migratory NLT-DCs Activate aNF-kB-Regulated Gene NetworkMigratory NLT-DCs (migDCs) are critical for both T cell priming in

the context of inflammation and tolerance to innocuous tissue

antigens in steady-state. But the molecular pathways that regu-

late NLT-DC maturation and migration are unknown. Skin

contains two major DC populations, epidermal Langerhans

cells (LCs) and dermal DCs (dDCs), the latter of which can be

divided into three subsets (CD11bloCD207�, CD11bloCD207+,

and CD11bhiCD207�) that represent more than 60% of all

dDCs (Henri et al., 2010). Migratory counterparts of all four pop-

ulations are found in the cutaneous LNs (CLNs) draining the skin.

To investigate the genes and signaling pathways that are

specifically associated with steady-state migDCs from the

skin, we examined the global gene expression profiles of

skin-resident CD11bhiCD207�CD64� dDCs and their migratory

counterparts in the CLNs from published microarray data

(Tamoutounour et al., 2013). We found several hundred genes

that were differentially expressed between migDCs in the

CLNs and their resident counterparts in the dermis (Figures

1A and S1). Ingenuity Pathway Analysis (IPA) revealed that

this set of genes was significantly enriched for annotations

pertaining to ‘‘DC maturation’’ and for targets of the ‘‘NF-kB

complex’’ (Figure 1A). Moreover, by using distant regulatory

elements (DiRE) analysis, we found that NF-kB was the tran-

scription factor most significantly associated with the migDC-

specific gene signature (Figure 1B). The over-representation

of NF-kB binding motifs in these genes was also confirmed

by Clover (cis-element overrepresentation) analysis with a p

value of 10�3 (Figure 1C). The vast majority of these genes

were validated as bona fide NF-kB target genes in DCs by pre-

vious ChIP-seq analysis of RelA binding sites during TLR-

induced DC maturation (Figure 1D; Garber et al., 2012). To

further analyze the commonalities between the strong NF-kB

signature in DCs during TLR-induced maturation and the

NF-kB-associated gene signature in steady-state migDCs, we

compared the fold changes in expression of all validated

628 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

RELA-target genes from TLR-induced DC maturation (Garber

et al., 2012) and predicted NF-kB targets induced in steady-

state migDCs, compared to their tissue-resident counterparts.

This comparison revealed a strong association between TLR-

induced DC maturation and high expression of canonical NF-

kB target genes, such as Il1b, Il1a, Il6, Il12a, Il12b, and Tnfsf1a

(Figure 1E). In contrast, these genes were not highly expressed,

or even repressed, in steady-state migDCs. Reciprocally, many

of the predicted NF-kB target genes that were induced specif-

ically in migDCs (Fscn1, Il15ra, Aldh1a2, and Il4i1) were not

regulated during LPS-induced DC maturation, and several

genes were in fact strongly repressed in LPS-activated DCs

(Sox4, Idh1). This indicates a NF-kB-regulated gene network

in migDCs that is likely to have unique upstream regulators.

These analyses suggest that NF-kB activation might control

the steady-state maturation of NLT-DCs through regulation of

a novel gene network that is distinct from its function in DCmatu-

ration during inflammation or infection.

Targeted Deletion of IKKb in DCs Leads to SpontaneousAutoimmunityTo examine the role of NF-kB signaling in NLT-DCs, we gener-

ated mice with a targeted deletion of IKKb (Ikbkb) in the DC line-

age. IkbkbF/F mice were crossed with Tg(Itgax-Cre) mice, which

express Cre recombinase from the CD11c (Itgax) promoter (Ca-

ton et al., 2007). Homozygous IkbkbDItgax/DItgax (IkbkbDItgax) mice

were born in normal Mendelian ratios and we confirmed efficient

deletion of IKKb expression specifically in DCs by analysis of

both mRNA and protein expression (Figure S2). From adulthood,

IkbkbDItgax mice presented with frequent LN adenopathy and

splenomegaly (Figures 2A and 3A). HE staining and immuno-

histochemical (IHC) analysis of spleens from IkbkbDItgax mice

showed a dramatic increase in B cell follicle germinal centers

(GCs), marked by PNA staining, compared to littermate controls

(Figure 2B), reflecting an increase in antigen-induced B cell pro-

liferation. In addition, there was a striking increase in the infiltra-

tion of DCs into the T cell zone of B cell follicles in IkbkbDItgax mice

(Figure 2C), suggesting increased DC-mediated T cell activation.

Polyclonal B cell activation was associated with the presence

of anti-nuclear antibodies (ANA) in the serum of IkbkbDItgax

mice (Figure 2D), a common indicator of autoimmune disease.

Another frequent manifestation of autoimmune disease is the

deposition of immune complexes in glomeruli of the kidney,

which subsequently trigger inflammation and glomerular

nephritis. IHC analysis also revealed glomerular antibody depo-

sition (IgG) in the kidneys of IkbkbDItgax mice, whichwas absent in

littermate controls (Figure 2C). Collectively, these data suggest

that inhibition of steady-state NF-kB signaling in DCs results in

spontaneous autoimmunity.

IKKb in DCs Regulates Immune HomeostasisTo further characterize the autoimmune phenotype in IkbkbDItgax

mice, we performed flow cytometry analysis on LN and spleen

cell suspensions. This revealed a significant increase in propor-

tions of neutrophils (PMNs), Ly6Chi ‘‘inflammatory’’ monocytes,

and B cells compared to IkbkbF/F control mice (Figure 3B). In

contrast, the proportions of CD8+ and CD4+ T cells were signif-

icantly reduced in LNs (Figure 3B). Resident LT-DCs play an

important role in maintaining immune homeostasis (Birnberg

Page 4: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A B

C D

E

Figure 1. A NF-kB-Regulated Gene Network in Steady-State Migratory DCs

(A) Heat map showing the expression pattern of upregulated genes in migDCs from CLNs compared to dermal DCs (dDCs). Relative expression values are

represented for the genes of the ‘‘DC maturation’’ pathway (black) and the upstream regulator ‘‘NF-kB complex’’ (black bold), based on IPA. Genes that are

common to both analyses are in bold red.

(B) Diagram showing the top 10 transcription factor motifs enriched in migDC-upregulated genes. The occurrence represents percent of genes with a conserved

binding site for the TRANSFAC transcription factors. The importance is the product between the occurrence and the weight assigned to each transcription factor.

(C) The consensus NF-kB binding motif (MA0061.1) was used for Clover analysis that revealed a significant enrichment in genes upregulated in migDCs; p value

and score are indicated.

(D) Venn diagram showing the overlap between the migDC-upregulated genes with RelA target genes identified in LPS-stimulated DCs by ChIP-seq analyses

(Garber et al., 2012).

(E) Scatter plot comparing fold changes (log2) in the expression of bona fide NF-kB target genes (based on RELA binding) and putative NF-kB target genes in

steady-state migDCs, as shown in (D). LPS-stimulated DCs (y axis) and steady-state migDCs (x axis). Examples of canonical pro-inflammatory NF-kB target

genes upregulated in LPS-stimulated DCs and newly identified NF-kB target genes specifically regulated in migDCs are highlighted by red circles.

et al., 2008), but we found no significant differences in numbers

of LT-DCs in LNs and spleen from IkbkbDItgax mice (Figure 3C).

The proportions of CD11b+ and CD8+ LT-DC subsets was also

not altered in IkbkbDItgax mice, but there was a decrease in the

frequency of CD4+ DCs among the CD11b+ subset and a recip-

rocal increase in CD4�CD11b+ DCs (Figure S3). These data

showed that IKKb signaling was not required LT-DC develop-

ment or survival.

CD11c is expressed throughout the DC lineage, including

monocyte-derived DCs (moDCs), but also by somemacrophage

populations (Miller et al., 2012). To assess the role of IKKb

more specifically in cDCs, we compared IkbkbDItgax mice with

IkbkbDLyz2 mice, where IKKb is deleted in lysozyme M (Lyz2)-

expressing myeloid cells, including macrophages, PMNs, and

moDCs, but is not highly expressed in cDCs (Figure S2). Although

IkbkbDLyz2 mice also showed some degree of splenomegaly,

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 629

Page 5: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A B

C D

Figure 2. IKKb Deficiency in DCs Leads to Spontaneous Autoimmunity

(A) Spleen and CLNs of IkbkbF/F and IkbkbDItgax mice. Abbreviations are as follows: BLN, brachial LN; ILN, inguinal LN; AxLN, axillary LN; ALN, auricular LN.

(B) Spleen sections were analyzed by HE and PNA histochemistry, GC development was scored in n = 4 mice.

(C) Spleen sections were analyzed for Ter119, CD11c, and CD3 staining by IHC (left), kidney sections were analyzed by HE (middle), and IgG deposition by IHC

(right); scale bars represent 100 mm.

(D) Anti-nuclear autoantibodies (ANA) were measured in serum from IkbkbF/F and IkbkbDItgax mice. Statistical analysis was performed with a Fischer exact test.

compared to littermate controls, they did not present with LN

adenopathy or increased LN cellularity (Figure 3A). Furthermore,

flow cytometry analysis did not reveal any significant changes in

the proportions of B cells or T cells in LN and spleen from

IkbkbDLyz2 mice in contrast to IkbkbDItgax mice (Figure 3B). There

was a slight increase in the proportions of PMNs and Ly6C+

monocytes in LN and spleen from IkbkbDLyz2 mice (Figure 3B).

These data indicate that IKKb signaling in cDCs is not required

for LT-DC development or survival in steady-state but has an

important role in maintaining immune homeostasis.

IKKb in DCs Regulates Treg Cell Accumulation in CLNsDCs contribute to the maintenance of immune tolerance by

regulating the accumulation of Treg cells. There are two major

populations of Treg cells in mice: natural Treg (nTreg) cells pro-

duced in the thymus and iTreg cells generated in peripheral

lymphoid organs. Thymus-dependent nTreg cells provide cen-

tral tolerance to self-antigens expressed by thymic medullary

630 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

epithelial cells (mTECs), and there might also be a role for

thymic DCs in nTreg cell selection through the cross-presen-

tation of mTEC-associated antigens. In the periphery, naive

T cells can be converted to iTreg cells by DC-mediated antigen

presentation (Steinman et al., 2003). To assess the role of IKKb

in DCs for Treg cell accumulation, we measured the number of

CD25+FOXP3+CD4+ Treg cells in thymus, spleen, and CLNs of

IkbkbDItgax and littermate IkbkbF/F control mice. There was a sig-

nificant reduction in the proportion of Treg cells in CLNs from

IkbkbDItgax mice compared to littermate controls (Figure 3D),

but the proportions of Treg cells in the spleen and thymus

were not affected (Figure 3D). However, the frequency of

L-selectin (CD62L)-expressing Treg cells was significantly

reduced in spleen, whereas NRP1+ (neuropillin) thymus-depen-

dent nTreg cells were unaffected (Figure S3). This suggests that

IKKb in DCs contributes to iTreg cell conversion and activation

in the periphery and particularly in CLNs that acquire antigens

from the skin.

Page 6: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A B C

D

Figure 3. Targeted Deletion of IKKb in DCs Disrupts Immune Homeostasis

Flow cytometry analysis of spleen and CLNs from IkbkbF/F, IkbkbDItgax, and IkbkbDLyz2 mice.

(A–C) Total cell number (A), proportions of immune cells represented as the ratio between IkbkbF/F and IkbkbDItgaxmice (B), and percentage of resident LT-DCs (C).

(D) Proportions of Foxp3+CD4+ cells in thymus, spleen, and CLN.

Data are shown as mean ± SEM; graphs represent pooled data from at least three independent experiments. Statistical analysis was performed with Mann-

Whitney test; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

IKKb Is Required for Steady-State NLT-DCAccumulation in the CLNsThe generation of iTreg cells in CLNs is dependent on migratory

NLT-DCs (migDCs) from the dermis that transport antigens to the

CLNs in steady-state (Azukizawa et al., 2011; Guilliams et al.,

2010). These migDCs can be distinguished from resident LT-

DCs on the basis of their MHCIIhiCD11cintCCR7+ phenotype.

We measured the accumulation of migDCs in the CLNs from

IkbkbDItgax mice and littermate controls by flow cytometry.

IkbkbDItgax mice showed a dramatic reduction in accumulation

of migDCs, whereas the proportion of resident LT-DCs was un-

affected (Figures 4A and 3C). The specificity of the analysis for

migDCs was confirmed with CLNs from CCR7-deficient mice

(Ccr7�/�), which lack migDCs in draining LNs (Figure 4A; Ohl

et al., 2004). We also performed IHC analysis on LN sections

from IkbkbDItgax mice; this analysis confirmed the lack of accu-

mulation of CD207+ skin-derived DCs in the T cell zone of the

LN (Figure 4B). These data clearly showed that IKKb expression

in DCs is required for the steady-state accumulation of migDCs

in the CLNs.

To evaluate the role of IKKb in NLT-DC development and

homeostasis, we analyzed the distribution of NLT-DCs in the

epidermis (LCs) and dermis (dDCs). IHC analysis showed no

change in LC number or distribution in epidermis from IkbkbDItgax

mice (Figure 4B). FACS analysis of dermal cell suspensions also

showed no significant differences in the frequency of dDCs in

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 631

Page 7: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A B

C D

E

Figure 4. IKKb Is Required for Accumulation of migDCs in Steady-State

(A) Flow cytometry analysis of CLNs from IkbkbF/F, IkbkbDItgax, and Ccr7�/� mice. Resident LT-DCs and migDCs were distinguished by MHCII and CD11c

staining, representative FACS plots are shown, and the graph represents pooled data from three independent experiments. Statistical analysis was performed

with Mann-Whitney test; ***p < 0.001.

(B) Sections of intact CLNs were stained for CD207, LYVE1, B220, and CD11c and analyzed by confocal microscopy. Representative images are shown; scale

bars indicate 200 mM.

(C) Ear epidermal sheets were stained for MHCII and CD3 and epidermal DCs counted from at least three fields (n = 6). Lower panels: Dermal cell suspensions

were prepared and stained forMHCII, CD45.2, andCD3. Plots represent CD45.2 gated cells, the total number of DCs/ear from three independent experiments are

represented in the graph.

(D) BrdU was administered i.p. daily for 7 consecutive days. On day 3 and 7, mice were sacrificed, dermal and epidermal cell suspensions from the ears were

prepared, cellswere stained asdescribed in (C), andBrdU+ incorporationbyDCswasdetermined.Graphs represent twopooled independent experiments of n=3.

(E) Tg(Itgax-Bcl2) mice were crossed with IkbkbDItgax mice to generate mice overexpressing BCL2 in DCs (Tg(Bcl2)) and proportions of resDCs and migDCs in

CLNs were analyzed as described in (A); data are shown asmean ± SEM of n = 5 and are representative of at least 2 independent experiments. Statistical analysis

was performed with Mann-Whitney test; ***p < 0.001.

IkbkbDItgax mice (Figure 4B). Moreover, the different dDC subsets

were present in similar proportions as littermate control mice

(Figure S4). These data showed that IKKb was not required for

632 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

the development or accumulation of NLT-DCs in the skin. Previ-

ous studies have estimated the half-life of NLT-DCs in the dermis

to be between 3 and 5 days, being constantly replenished from

Page 8: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

circulating common dendritic cell progenitors (CDPs) that

develop in the bone marrow (Poulin et al., 2007). On the other

hand, epidermal LCs are maintained by self-renewal in the tissue

with little contribution from blood-borne progenitors (Poulin

et al., 2007). To assess the role of IKKb in NLT-DC turnover,

we used in vivo BrdU labeling. Epidermal LCs and dermal DCs

from IkbkbDItgax mice showed no difference in BrdU incorpora-

tion compared with littermate controls during a 7 day labeling

period (Figure 4C), indicating that IKKb deletion does not affect

the proliferation or turnover of skin DCs in steady-state.

The experiments described above clearly showed that IKKb

deletion in DCs did not affect the homeostasis of NLT-DCs in

the skin, yet there was a dramatic absence of migDCs in

CLNs. This presented the possibility that IKKb might regulate

migDC survival in the draining LN after maturation and migration

from the skin. To address this question, we crossed IkbkbDItgax

mice to transgenic mice that expressed the anti-apoptotic pro-

tein Bcl2 specifically in the DC lineage (Tg(Itgax-Bcl2)) (Gautier

et al., 2009). The proportions of DCs in the dermis were not

significantly altered in IkbkbF/F or IkbkbDItgax mice expressing

the Bcl2 transgene (Figure 4E). However, although there was a

significant increase in the accumulation of migDCs in CLNs

from IkbkbF/F mice expressing the Bcl2 transgene, this did not

rescue the defect inmigDC accumulation in IkbkbDItgax mice (Fig-

ure 4E). These studies demonstrate that the accumulation of

migDCs in steady state is limited by apoptotic cell death; how-

ever, rescuing cell survival is not sufficient to restore migDC

accumulation in the absence of IKKb signaling, suggesting that

IKKb activation in steady-state migDCs drives the maturation

andmigration of NLT-DCs from the skin and not their persistence

in the CLNs.

IKKb Regulates Spontaneous Maturation of NLT-DCs inthe SkinSteady-state NLT-DC maturation is associated with the upre-

gulation of major histocompatibility complex II (MHCII) and

co-stimulatory molecules such as CD40 on migratory DCs. Cul-

ture of intact skin explants ex vivo results in spontaneous DC

maturation and the acquisition of migratory properties that

can be measured by DC emigration into the culture medium.

The number of DCs that emigrated from skin explants of

IkbkbDItgax mice was significantly reduced compared to litter-

mate controls (Figure 5A). Emigrated DCs from IkbkbDItgax

mice showed no difference in Annexin V staining, suggesting

that the reduced emigration was not due to effects on cell sur-

vival (Figure 5B). Culture of intact skin explants also results

in accumulation of DCs along dermal lymphatic vessels in

a CCR7-dependent manner, forming so-called dermal cord

structures (Ohl et al., 2004; Weinlich et al., 1998); however, ex-

plants from IkbkbDItgax mice showed no defect in dermal cord

formation compared to skin from littermate control mice (Fig-

ure 5C). In contrast, those structures were absent from the

dermis of skin explants from Ccr7�/� mice (Figure 5D), as pre-

viously described by Ohl et al. (2004), suggesting that IKKb

is not required for CCR7-mediated sensing of chemokines ex-

pressed by lymphatic endothelium. This is supported by the

increased accumulation of LT-DCs in the T cell zones of splenic

B cell follicles in IkbkbDItgax mice (Figure 2C), which is also

CCR7 dependent (Ohl et al., 2004).

To further examine the role of IKKb in spontaneous matura-

tion of skin DCs, we isolated epidermal and dermal DCs from

IkbkbDItgax mice and littermate controls and measured their

phenotype ex vivo. The isolation and culture of skin DCs at

37�C, without exogenous stimulation, was sufficient to induce

the upregulation of typical markers of maturation including sur-

face expression of MHCII, CD40, and CCR7, as well as intracel-

lular expression of the actin bundling protein FASCIN (Fscn1)—

which has been shown to be required for migration of mature

DCs (Yamakita et al., 2011) and to be a putative NF-kB-regulated

gene in steady-state migDCs (Figure 1A). IKKb-deficient DCs

showed no difference in upregulation of MHCII or survival upon

culture ex vivo (Figure 5E) but showed reduced expression

of CD40, CCR7, and FASCIN compared to wild-type cells (Fig-

ure 5E), demonstrating that IKKb contributes to the intrinsic

spontaneous maturation of isolated skin DCs. Furthermore,

addition of specific small molecule inhibitor of IKKb (Zhang

et al., 2014) blocked the spontaneous maturation of isolated

dermal DCs in this context (Figure S5).

Finally, to confirm the IKKb-dependent expression of putative

NF-kB target genes upregulated in steady-state migDCs, we

performed quantitative PCR analysis of mRNA expression in iso-

lated dermal DCs before and after spontaneous maturation. We

confirmed the IKKb-dependent induction of several predicted

NF-kB target genes during spontaneous maturation of dermal

DCs, including Fscn1 and Aldh1a2, but not the canonical pro-

inflammatory target gene Tnfsf1a, which was in fact downregu-

lated during spontaneous maturation (Figure 5F).

These studies demonstrate that IKKb activation regulates the

spontaneous maturation of skin DCs ex vivo and the upregula-

tion of genes specifically associated with steady-state migDCs,

including genes required for migration and Treg cell conversion.

Steady-State NLT-DC Migration Does Not Depend onCanonical NF-kB StimuliThesignaling pathways that regulate steady-stateNLT-DCmatu-

ration are unknown. The canonical NF-kBpathway ismostwidely

studied in the context of TNF-a, IL-1b, and TLR signaling (Dev

et al., 2011), so todetermine the role of thesepathways in homeo-

static NLT-DC migration, we analyzed CLNs from Myd88�/�

Trif�/�mice, deficient in all TLR-signaling pathways and signaling

through the IL-1 receptor, and frommice deficient in both TNF-a

receptors (Tnfsfr1�/�Tnfsfr2�/�). We found no defects in the

accumulation of migDCs in either of these strains (Figure S6).

This is in keepingwith our analysis of the transcriptional signature

of steady-state migDCs, which indicates that the NF-kB-regu-

lated gene network in migDCs is distinct from the network of

NF-kB-regulated genes during TLR-induced DCmaturation (Fig-

ure 1E). In addition, Ingenuity Pathway Analysis (IPA) with the

genes differentially expressed by migDCs compared to their tis-

sue-resident counterparts showed a negative enrichment for

known upstream regulators of pro-inflammatory NF-kB signaling

(Figure S1), indicating that the ‘‘canonical’’ NF-kB pathway is in

fact downregulated in these cells.

The alternative NF-kB pathway, leading to activation of RELB

p52 heterodimers, has also been suggested to play an important

role in DC maturation (Azukizawa et al., 2011; Shih et al., 2012).

The alternative pathway is normally dependent on IKKa activa-

tion and not IKKb, but a recent study showed that IKKb can

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 633

Page 9: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A B C D

E

F

Figure 5. IKKb Regulates Spontaneous Skin NLT-DC Maturation

(A and B) Ear skin flaps from IkbkbF/F and IkbkbDItgax mice were floated on tissue culture medium for 72 hr and DC emigration was analyzed by flow cytometry; (A)

represents total emigrated DCs per ear and (B) the percentage of Annexin V+ cells among emigrated DCs.

(legend continued on next page)

634 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

Page 10: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

regulate activation of RELB in DCs after lymphotoxin b (LTb)

stimulation (Shih et al., 2012). We found no defect in accumula-

tion of migDCs in the CLN of IKKaAA knock-in mice (Mancino

et al., 2013), which express an inactive mutant of IKKa (Fig-

ure S6). Furthermore, blockade of LTbR signaling through the

administration of LTbR-Ig fusion protein had no impact on

migDC accumulation in CLNs (Figure S6). These data indicate

that the alternative NF-kB pathway and LTbR signaling through

either IKKa or IKKb is not required for the steady-state migration

of NLT-DCs in the skin.

IKKb Regulates Dermal NLT-DC Migration and AntigenTrafficking to CLNsMigratory dermal NLT-DCs transport antigens from the skin

to the CLNs for subsequent presentation to naive T cells.

To confirm the role of IKKb in NLT-DC migration to the CLNs,

we applied FITC epicutaneously to the ear skin of IkbkbF/F and

IkbkbDItgax mice andmeasured the accumulation of FITC-labeled

migDCs in the CLNs. Previous studies have established that all

FITC+ DCs in the CLNs via this procedure represent migratory

dDCs or LCs (Ohl et al., 2004). As expected, we observed a

profound defect in accumulation of FITC+ DCs in the CLNs

from IkbkbDItgaxmice compared to littermate controls (Figure 6A).

Because FITC acts as a hapten, forming adducts with endoge-

nous proteins, these data suggest that IKKb deficiency in NLT-

DCs would result in impaired trafficking of antigens from the

skin to theCLNs. To test the role of IKKb inNLT-DC-mediated an-

tigen presentation and T cell priming in the CLNs, we applied the

TLR7 agonist R848, as an adjuvant, in combination with MHC I-

restricted ovalbumin peptide (OVAp) to the ear skin of IkbkbDItgax

and littermate control mice; we then measured the expansion of

OVAp-specificCD8+ T cells in theCLNs by FACSby usingOVAp-

specific tetramers. There was a significant reduction in antigen-

specific CD8+ T cell expansion in CLNs from IkbkbDItgax mice

compared to littermate controls (Figure 6B), demonstrating that

impaired NLT-DC migration in IkbkbDItgax mice prevented the

transport and presentation of an exogenous antigen applied to

the skin. In addition, the significant increase in NLT-DCmigration

promoted by R848 application was impaired in IkbkbDItgax mice

(Figure 6C), suggesting that IKKb is required for NLT-DC migra-

tion in both steady-state and inflammatory conditions.

T cell priming upon sub-cutaneous (s.c.) immunization with the

commonly used adjuvant CFA is dependent on recruitment

of monocyte-derived DCs (moDCs) into draining LNs through

high endothelial venules (HEVs) (Nakano et al., 2009)—as

opposed to migration of NLT-DCs through lymphatics. We did

(C and D) Ear skin flaps were floated on tissue culture medium for 72 hr befo

Quantitative analysis was performed with ImageJ software and data are plotted

represent 100 mm. Graphs represent pooled data from at least three independ

**p < 0.01, ***p < 0.001.

(E) Epidermal and dermal cell suspensions were prepared and cultured overnight

Fascin expression by flow cytometry. The first graphs represent the ratio of viable

and Fascin expression by epidermal and dermal DCs are shown at 4�C and aft

because they were uniformly expressed by DCs. CCR7 and Fascin expression are

at 37�C. Data are shown as mean ± SEM and are representative of three indepen

*p < 0.05.

(F) Dermal cell suspensions were prepared as described in (E); DCs were sorted

expression of specific genes in dermal DCs at 4�C and 37�C is indicated norm

replicates from pooled cells of n = 3 mice and are representative of two indepen

not observe any defect in the accumulation of OVAp-specific

CD8+ T cells in the CLNs of IkbkbDItgax mice after s.c. immuniza-

tion with CFA and OVA, and moDC accumulation in CLNs was

not affected in IkbkbDItgax mice (data not shown). These data

suggest that IKKb is not required for the recruitment of moDCs

or subsequent T cell priming in inflammatory conditions, but spe-

cifically regulates the maturation and migration of NLT-DCs.

IKKb in DCs Promotes Tolerance to an EndogenousTissue AntigenIn steady state, migratory NLT-DCs are required for conversion

of naive CD4+ T cells to FOXP3+ Treg (iTreg) cells and tolerance

to self-antigens (Azukizawa et al., 2011; Guilliams et al., 2010).

IkbkbDItgax mice showed a profound absence of steady-state

migDCs and significantly reduced accumulation of FOXP3+

Treg cells (Figures 4A and 3D). The data described above

show that IKKb signaling in NLT-DCs is required for the transport

and presentation of exogenous antigens from the skin in the

draining LNs. To evaluate the role of IKKb in skin NLT-DCs for

tolerance to an endogenous self-antigen, we used K5-mOVA

transgenic mice (Tg(K5-mOVA)), which express membrane-

bound OVA in epidermal keratinocytes (Azukizawa et al.,

2003). Previous studies with K5-mOVA mice have shown that

migratory NLT-DCs are required for the transport and presenta-

tion of OVA antigens from the skin to naive T cells in the CLNs. In

the case of CD8a-type migDCs, this was required for cross-pre-

sentation of OVA-peptide to autoreactive CD8+ T cells and their

subsequent deletion (Waithman et al., 2007). However, in the

case of CD11b+ migDCs, this was required for the conversion

of OVA-specific naive CD4+ T cells to iTreg cells (Azukizawa

et al., 2011; Guilliams et al., 2010). To test the role of IKKb in

this process, we generated K5-mOVA mice with a targeted

deletion of IKKb in DCs (Tg(K5-mOVA) IkbkbDItgax). First, we

confirmed the absence of migDCs in CLNs from Tg(K5-mOVA)

IkbkbDItgax mice compared to Tg(K5-mOVA) IkbkbF/F littermate

controls (Figure 7A). This was associated with a significant

reduction in the accumulation of CD25+FOXP3+ Treg cells in

the CLNs of Tg(K5-mOVA) IkbkbDItgax mice (Figure 7A). To test

the role of IKKb in migDCs for deletion of autoreactive CD8+

T cells, we adoptively transferred purified OVA-specific CD8+

T cells (OT-I) to cohorts of Tg(K5-mOVA) IkbkbDItgax and Tg(K5-

mOVA) IkbkbF/F mice and monitored the deletion of OT-I cells

in the blood and CLNs by FACS, using specific tetramers. All

control mice deleted adoptively transferred OT-I cells within

2 weeks (Figure 7B); however, OT-I cells expanded in a signifi-

cant proportion of mice with IKKb deletion in DCs (Figure 7B),

re dermis was separated from epidermis and stained for MHCII and LYVE1.

as proportion of MHCII+ cells co-localized to Lyve1+ lymphatics. Scale bars

ent experiments. Statistical analysis was performed with Mann-Whitney test;

at 37�C. DCs were counted and stained for viability, MHCII, CD40, CCR7, and

DCs upon culture at 37�C compared to cells kept at 4�C. MHCII, CD40, CCR7,

er culture at 37�C. MHCII and CD40 expression are represented as MFI only,

expressed as proportion of positive cells among DCs andMFI of positive cells

dent experiments. Statistical analysis was performed with Mann-Whitney test;

and mRNA expression analyzed by quantitative RT-PCR (qRT-PCR). Relative

alized to cyclophillin (CPH) expression. Data represent mean ± SEM of three

dent experiments.

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 635

Page 11: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

A

B

C

Figure 7. IKKb in DCs Regulates Tolerance to an Endogenous Cell-

Associated Antigen in the Skin

(A) The proportions of migDCs and Foxp3+ Treg cells in CLNs from Tg(K5-

mOVA)/IkbkbDItgax mice and Tg(K5-mOVA)/IkbkbF/F littermates was evaluated

by flow cytometry. Graphs represent pooled data from three independent

experiments.

(B) Purified OVA-specific CD8 T cells (OT-I) were transferred to Tg(K5-mOVA)/

IkbkbDItgax and Tg(K5-mOVA)/IkbkbF/F mice and the proportion of activated

OT-I cells measured in peripheral blood after 11 days by flow cytometry using

tetramers. Data from four independent experiments are represented on the

graph. Each point corresponds to one mouse.

(C) Purified naive CD25�CD4+OT-II cells were adoptively transferred to Tg(K5-

mOVA)/IkbkbDItgax and Tg(K5-mOVA)/IkbkbF/F mice, and after 13 days the

proportion of CD25+FOXP3+ OT-II cells among CD4+ cells in the CLN was

evaluated by flow cytometry.

Data are representative of three independent experiments. Statistical analyses

in (A) and (C) were performed with Mann-Whitney test; **p < 0.01, ***p < 0.001,

****p < 0.0001. Statistical analysis in (B) was performed by Fischer exact test;

*p < 0.05.

A

B

C

Figure 6. IKKb Is Required for Dermal NLT-DC Migration and

Antigen Trafficking to CLNs

(A) FITC was applied epicutaneously to the ear skin of IkbkbF/F and IkbkbDItgax

mice, and 3 days later the accumulation of FITC-labeled DCs in draining ALNs

was measured by flow cytometry. Representative data showing the percent-

age and the number of FITC+ DCs are plotted from three independent ex-

periments.

(B) Mice were immunized by topical application of R848 on the ear in combi-

nation with MHC I-restricted ovalbumin peptide (OVAp). After 7 days, the

expansion of OVAp-specific CD8+ T cells was measured in the ALN by cy-

tometry using tetramers. Graphs represent pooled data from three indepen-

dent experiments and show the percentage and the number of OVAp-specific

CD8+ T cells in ALNs.

(C) R848was applied to the ears and accumulation ofmigDCs in draining ALNs

after 2 days was evaluated by flow cytometry. Representative data showing

the percentage and the number of migDCs from three independent experi-

ments are shown.

Statistical analysis was performed with Mann-Whitney test; **p < 0.01, ***p <

0.001, ****p < 0.0001.

indicating impaired deletional tolerance. In fact, the expansion of

adoptively transferred autoreactive CD8+ T cells in Tg(K5-mOVA)

IkbkbDItgax mice frequently led to inflammatory lesions in the skin

as a result of the activation of autoreactive CD8+ T cells (data not

shown).

To evaluate the role of IKKb-dependent migDCs in the conver-

sion of naive OVA-specific CD4+ T cells to iTreg cells, we adop-

tively transferred purified naive CD25�CD4+ OT-II cells to

cohorts of Tg(K5-mOVA) IkbkbDItgax and Tg(K5-mOVA) IkbkbF/F

mice and measured the accumulation of CD25+FOXP3+ OT-II

636 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

cells in theCLNs. In these experiments, Tg(K5-mOVA) IkbkbDItgax

mice showed a profound impairment in accumulation of

OT-II iTreg cells compared to littermate controls (Figure 7C),

demonstrating that IKKb-dependent migDCs were required for

OVA-specific iTreg cell conversion in CLNs.

Collectively, these data demonstrate that IKKb-dependent

accumulation of migDCs in the CLNs in steady state plays

an important contribution to tolerance toward endogenously

expressed cell-associated antigens by regulating both iTreg

cell conversion, through MHCII-mediated presentation to naive

CD4+ T cells, and deletional tolerance, through cross-presenta-

tion of cell-associated antigens to CD8+ T cells.

Page 12: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

DISCUSSION

The homeostatic maturation of NLT-DCs and their subsequent

migration from the tissue parenchyma to the draining LNs is crit-

ical tomaintain tolerance to innocuous tissue antigens. Triggering

of pattern recognition receptors (PRRs), such as TLRs, on DCs is

well known to induce a process of functional maturation that is

required for the priming of naive T cells. Similar changes are asso-

ciated with steady-state migDCs compared to their tissue-resi-

dent counterparts, but results in tolerogenic activity rather than

T cell priming. However, the signaling pathways that regulate

the maturation and accumulation of migDCs in steady state

have remained an enigma. Here we have shown that NF-kB

signaling is required for thesteady-statematurationandmigration

of NLT-DCs and we demonstrate that IKKb deletion in DCs was

sufficient to prevent the steady-state accumulation of migDCs

from the skin. Deletion of IKKb did not affect DC development

or survival but was critically required formaturation andmigration

of NLT-DCs in the skin. Furthermore, the IKKb-dependent accu-

mulation of migDCs played an important role in tolerance toward

endogenously expressed cell-associated antigens by regulating

both iTregcell conversion, throughMHCII-mediatedpresentation

tonaiveCD4+Tcells, anddeletional tolerance, throughcross-pre-

sentation of cell-associated antigens to CD8+ T cells.

Our analysis of the specific gene expression signature in

steady-state migDCs reveals a strong enrichment for NF-kB-

regulated genes. However, canonical NF-kB target genes that

are highly expressed upon TLR-induced DC maturation were

not upregulated or were even repressed, indicating a novel NF-

kB-regulated gene network in migDCs that is likely to depend

on unique upstream regulators. In keeping with this hypothesis,

we showed that mice deficient in TLR, IL-1b, or TNF-a signaling

had no defects in accumulation ofmigDCs in steady state. This is

in accordance with a previous study that showed no alterations

in migDC accumulation or maturation in germ-free mice and

mice deficient in TLR signaling (Wilson et al., 2008). The NF-

kB-regulated gene network in steady-state migDCs from the

skin included genes previously linked with DC migration

(Fscn1) and Treg cell conversion (Aldh1a2), both functions that

were impaired in vivo upon deletion of IKKb in DCs, as well as

several genes with no known function in DCs.

One possible explanation for the distinct networks of NF-kB-

regulated genes during steady-state (tolerogenic) and Toll-like

receptor (TLR)-induced (immunogenic) DC maturation might be

the context-specific activation or expression of transcriptional

co-regulators. For example, the transcription factor IRF4 was

recently shown to be required for development of CD11b+

NLT-DCs in both skin and intestine and the steady-state accu-

mulation of migDCs in CLNs and MLNs, respectively (Persson

et al., 2013; Schlitzer et al., 2013). IRF4 and NF-kB are known

to interact in B cells and co-operatively regulate specific genes.

We have also found significant enrichment for IRF binding sites in

genes upregulated in steady-state migDCs (Dalod et al., 2014),

suggesting that IRF4 and NF-kB might co-operatively regulate

steady-state NLT-DC maturation.

The homeostatic signals that drive NLT-DC maturation and

the accumulation of migratory DCs are completely unknown. In

fact, it isn’t even clear whether this is an intrinsically stochastic

process or one driven by instructive signals in the tissue micro-

environment. Factors that have been suggested to play a role

in NLT-DC homeostasis and maturation include CSF2 (GM-

CSF) and TGF-b. CSF2 was shown to promote the survival of

NLT-DCs and thus affect the accumulation of migDCs in draining

LNs (Greter et al., 2012), but we found no role for IKKb in NLT-DC

survival, and furthermore, blocking CSF2 failed to prevent the

spontaneousmaturation of dermal DCs ex vivo, which was abro-

gated by inhibition of IKKb (Figure S5). TGF-b has been linked

with tolerogenic functions in DCs (Travis et al., 2007) and plays

a critical role in the homeostasis of LCs in the epidermis (Kel

et al., 2010). However, we also found that TGF-b neutralization

did not affect spontaneous maturation of dermal DCs ex vivo

in contrast to IKKb inhibition (Figure S5).

Several previous studies have suggested that WNT-b-catenin

signaling in DCs is important for tolerogenic function (Jiang et al.,

2007; Manicassamy et al., 2010). Targeted deletion of b-catenin

in DCs led to impaired Treg cell conversion by DCs in the intes-

tine and disruption of immune homeostasis (Manicassamy et al.,

2010). However, these authors focused on NLT-DCs in the intes-

tine lamina propria and did not analyze migDCs from the mesen-

teric LNs. Our analysis shows that WNT-b-catenin target genes

are in fact downregulated in migDCs from the CLNs compared

to their dermal counterparts (Figure S1), indicating that this

pathway is not upregulated during steady-state DC maturation

in the skin. This might reflect tissue specificity in pathways

regulating NLT-DC maturation, or alternatively, WNT-b-catenin

signaling might affect DC function independently of NLT-DC

maturation and migration. Another link between WNT-b-catenin

signaling and tolerogenic DC maturation was suggested by

Jiang et al. (2007), when these authors showed that disruption

of homotypic E-cadherin interactions between clusters of

bone-marrow-derived DCs (BMDCs) in culture led to b-cate-

nin-mediated maturation, including upregulation of CCR7,

CD40, and CD86. This maturation was associated with tolero-

genic function rather than T cell priming. We generated

IKKb-deficient BMDCs but found no impact on cluster-disrup-

tion-induced maturation, including upregulation of CCR7,

CD40, and CD86 (data not shown). Furthermore, a number of

the genes induced by BMDCs in this context were in fact down-

regulated in migDCs from the skin compared to dermal DCs

(Figure S1), as were other WNT-b-catenin pathway genes.

In summary, we have uncovered a new role for NF-kB

signaling in the accumulation of steady-state migDCs and

maintenance of immune tolerance in the skin. Although NF-kB

is generally considered as a major pro-inflammatory transcrip-

tion factor, these data show an important role for NF-kB

signaling in immune homeostasis and tolerance that could be

significant in the context of autoimmune diseases and can-

cer—where the molecular mechanisms regulating tolerance to

tissue antigens are therapeutic targets. Our analysis of the

global gene expression profile of NLT-DCs and their migratory

counterparts suggests that a novel NF-kB-regulated gene

network is associated with the steady-state maturation of

NLT-DCs and their tolerogenic function; however, the homeo-

static signals that trigger IKKb-NF-kB activation in this context

are yet to be revealed. Future characterization of the upstream

regulators and key nodes in this network might reveal new mo-

lecular targets to specifically modulate migDCs and promote

immunity or tolerance.

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 637

Page 13: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

EXPERIMENTAL PROCEDURES

Detailed experimental procedures are described in the Supplemental Informa-

tion section.

Mice

K5-mOVA, OT-I, OT-II, Ccr7�/�, and Tg(Itgax-hBcl2) mice have been previ-

ously described (Azukizawa et al., 2003; Hogquist et al., 1994; Barnden

et al., 1998; Forster et al., 1999; Gautier et al., 2009).

Flow Cytometry

Anti-CD11c (HL3), anti-CD8a (53-6.7), anti-MHC II (M5/114), anti-CD45.2

(104), anti-CCR7 (4B12), anti-CD11b (M1/70), anti-CD24 (M1/69), anti-Foxp3

(FJK-16 s), and anti-CD103 (M290) antibodies were all purchased from either

BDeBioscience or Biolegend. Anti-Fascin (EP5902) antibody was purchased

from Abcam. H-2Kb-OVA257-264 PE tetramers were purchased from

Beckman Coulter. Analysis was performed with FACSCanto system (BD)

and Flowjo software (Tree Star).

Microarray Analysis

Microarray analyses were performed as reported previously (Yamakita et al.,

2011). The genes differentially expressed between steady-state dDCs and

migDCs were identified with the Bioconductor Limma statistical package,

with cutoff values of 0.05 for the false discovery rate and of 2 for the fold

change. A pathway analysis was then performed on the 1,095 genes upregu-

lated in migDCs via the Ingenuity Pathway Analysis facility. Distant regulatory

elements analysis was performed with default parameters and a motif search

with the Clover software as previously reported (Baranek et al., 2012); p value

threshold and score were fixed respectively at 0.05 and 4.

Skin Explants

Ears were split into ventral and dorsal parts and floated split side down on 1ml

medium containing RPMI 10% FCS and 50 mM b-mercaptoethanol.

Isolation of Skin DCs

Epidermis was separated from the dermis and single-cell suspensions were

prepared and cultured for 18 hr at 37�C prior to analysis by flow cytometry.

For inhibitor experiments, single-cell suspensions of skin DCs were prepared

from epidermis and treated with or without various inhibitors for 18 hr at 4�Cor 37�C: IKKb inhibitor BI605906 (Zhang et al., 2014), 10 and 50 mM; JAK3

inhibitor CP690550 (Tocris), 1 mM; anti-mouse TGF-b1 neutralizing Ab

(Uyttenhove et al., 2011), 10 mg/ml; anti-mouse CSF2 neutralizing Ab

(eBioscience), 5 mg/ml.

Adoptive T Cell Transfer

OT-I and OT-II cells were purified from pooled LNs and spleen by negative

selection via a T cell isolation kit (Invitrogen); CD25� OT-II cells were subse-

quently sorted by flow cytometry using anti-CD4 and anti-CD25 antibodies.

BrdU Labeling In Vivo

Mice were injected i.p. with 1.5 mg of BrdU (Sigma Aldrich) and their drinking

water was supplemented with 0.8 mg/ml BrdU and 2% glucose. BrdU staining

was performed according to the manufacturer’s recommendations (BrdU

labeling Flow kit, BD).

FITC Painting

FITC (0.5 mg/ml) was dissolved in acetone and dibutyl phthalate 1:1 and 30 ml

was applied on the ear of anesthetized mice.

Topical Immunization

50 mg of H-2Kb-OVA257-264 peptide was mixed in 50 mg of imiquimod cream

(Aldara; 3M Pharceuticals) and applied to ears of anesthetized animals.

Histopathology

Hematoxylin and eosin staining was perfomed on paraffin-embedded spleen

and kidney tissue. Immunohistochemistry was performed on frozen tissue

sections with the indicated antibodies. GC development was evaluated via

the following scoring: 1, weak; 2, moderate; 3, marked; 4, high. Serum was

638 Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc.

analyzed for autoantibody production by a standard autoantibody test with

Hep-2 cells fixed on slides (Biomedical Diagnosis).

Statistical Analysis

p values were calculated with either the Mann-Whitney test or the Fisher test

when appropriate with Graph pad prism software.

SUPPLEMENTAL INFORMATION

Supplemental Information includes six figures and Supplemental Experimental

Procedures and can be found with this article online at http://dx.doi.org/10.

1016/j.immuni.2015.03.003.

AUTHOR CONTRIBUTIONS

M.B. and C.F. performed most of the experiments with technical support from

S.D. and C.V. O.D., M.H., and A.F.-L. contributed important data and proto-

cols. J.M., E.P., andM.D. performed bioinformatics analysis. M.B. and T.L. de-

signed the experiments and wrote the manuscript.

ACKNOWLEDGMENTS

We thank Boris Reizis (Columbia University) for supplying Tg(Itgax-Cre) mice,

Jorge Caamano (University of Birmingham) for Tnfrsf1�/�Tnfrsf2�/� mice,

Yang-Xin Fu (University of Chicago) for LTBR-Ig, Philippe Lesnik (UPMC) for

Tg(Itgax-hBcl2) mice, and Michael Karin (University of California, San Diego)

for IkbkbF/F mice. We also thank our colleagues Bernard Malissen, Sandrine

Henri, andMarc Bajenoff for helpful advice and for sharing unpublished obser-

vations. These studies were supported by grants to T.L. from L’Agence Natio-

nale de la Recherche (ANR) (ANR-09-MIEN-029-01, ANR-10-BLAN-1302-01)

and European Research Council (FP7/2007–2013 Grant agreement number

260753), and institutional funding from INSERM, CNRS, and Aix-Marseille-

Universite. E.P. and M.D. received financial support from Sanofi during the

course of these studies.

Received: March 27, 2014

Revised: December 23, 2014

Accepted: February 10, 2015

Published: April 7, 2015

REFERENCES

Azukizawa, H., Kosaka, H., Sano, S., Heath, W.R., Takahashi, I., Gao, X.H.,

Sumikawa, Y., Okabe, M., Yoshikawa, K., and Itami, S. (2003). Induction of

T-cell-mediated skin disease specific for antigen transgenically expressed in

keratinocytes. Eur. J. Immunol. 33, 1879–1888.

Azukizawa, H., Dohler, A., Kanazawa, N., Nayak, A., Lipp, M., Malissen, B.,

Autenrieth, I., Katayama, I., Riemann, M., Weih, F., et al. (2011). Steady state

migratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction

of antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells. Eur. J. Immunol.

41, 1420–1434.

Baranek, T., Manh, T.P., Alexandre, Y., Maqbool, M.A., Cabeza, J.Z.,

Tomasello, E., Crozat, K., Bessou, G., Zucchini, N., Robbins, S.H., et al.

(2012). Differential responses of immune cells to type I interferon contribute

to host resistance to viral infection. Cell Host Microbe 12, 571–584.

Barnden, M.J., Allison, J., Heath, W.R., and Carbone, F.R. (1998).

Defective TCR expression in transgenic mice constructed using cDNA-based

alpha- and beta-chain genes under the control of heterologous regulatory

elements. Immunol. Cell Biol. 76, 34–40.

Bedoui, S., Whitney, P.G., Waithman, J., Eidsmo, L., Wakim, L., Caminschi, I.,

Allan, R.S., Wojtasiak, M., Shortman, K., Carbone, F.R., et al. (2009). Cross-

presentation of viral and self antigens by skin-derived CD103+ dendritic cells.

Nat. Immunol. 10, 488–495.

Birnberg, T., Bar-On, L., Sapoznikov, A., Caton, M.L., Cervantes-Barragan, L.,

Makia, D., Krauthgamer, R., Brenner, O., Ludewig, B., Brockschnieder, D.,

et al. (2008). Lack of conventional dendritic cells is compatible with normal

Page 14: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

development and T cell homeostasis, but causes myeloid proliferative syn-

drome. Immunity 29, 986–997.

Caton, M.L., Smith-Raska, M.R., and Reizis, B. (2007). Notch-RBP-J signaling

controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Med.

204, 1653–1664.

Dalod, M., Chelbi, R., Malissen, B., and Lawrence, T. (2014). Dendritic cell

maturation: functional specialization through signaling specificity and tran-

scriptional programming. EMBO J. 33, 1104–1116.

Dev, A., Iyer, S., Razani, B., and Cheng, G. (2011). NF-kB and innate immunity.

Curr. Top. Microbiol. Immunol. 349, 115–143.

Forster, R., Schubel, A., Breitfeld, D., Kremmer, E., Renner-Muller, I., Wolf, E.,

and Lipp, M. (1999). CCR7 coordinates the primary immune response by

establishing functional microenvironments in secondary lymphoid organs.

Cell 99, 23–33.

Garber, M., Yosef, N., Goren, A., Raychowdhury, R., Thielke, A., Guttman, M.,

Robinson, J., Minie, B., Chevrier, N., Itzhaki, Z., et al. (2012). A high-throughput

chromatin immunoprecipitation approach reveals principles of dynamic gene

regulation in mammals. Mol. Cell 47, 810–822.

Gautier, E.L., Huby, T., Saint-Charles, F., Ouzilleau, B., Pirault, J., Deswaerte,

V., Ginhoux, F., Miller, E.R., Witztum, J.L., Chapman, M.J., and Lesnik, P.

(2009). Conventional dendritic cells at the crossroads between immunity and

cholesterol homeostasis in atherosclerosis. Circulation 119, 2367–2375.

Greter, M., Helft, J., Chow, A., Hashimoto, D., Mortha, A., Agudo-Cantero, J.,

Bogunovic, M., Gautier, E.L., Miller, J., Leboeuf, M., et al. (2012). GM-CSF

controls nonlymphoid tissue dendritic cell homeostasis but is dispensable

for the differentiation of inflammatory dendritic cells. Immunity 36, 1031–1046.

Guilliams, M., Crozat, K., Henri, S., Tamoutounour, S., Grenot, P., Devilard, E.,

de Bovis, B., Alexopoulou, L., Dalod, M., and Malissen, B. (2010). Skin-drain-

ing lymph nodes contain dermis-derived CD103(-) dendritic cells that constitu-

tively produce retinoic acid and induce Foxp3(+) regulatory T cells. Blood 115,

1958–1968.

Henri, S., Poulin, L.F., Tamoutounour, S., Ardouin, L., Guilliams, M., de Bovis,

B., Devilard, E., Viret, C., Azukizawa, H., Kissenpfennig, A., and Malissen, B.

(2010). CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-

derived antigens irrespective of the presence of Langerhans cells. J. Exp.

Med. 207, 189–206.

Hogquist, K.A., Jameson, S.C., Heath, W.R., Howard, J.L., Bevan, M.J., and

Carbone, F.R. (1994). T cell receptor antagonist peptides induce positive

selection. Cell 76, 17–27.

Jiang, A., Bloom, O., Ono, S., Cui, W., Unternaehrer, J., Jiang, S., Whitney,

J.A., Connolly, J., Banchereau, J., and Mellman, I. (2007). Disruption of E-cad-

herin-mediated adhesion induces a functionally distinct pathway of dendritic

cell maturation. Immunity 27, 610–624.

Kel, J.M., Girard-Madoux, M.J., Reizis, B., and Clausen, B.E. (2010). TGF-beta

is required to maintain the pool of immature Langerhans cells in the epidermis.

J. Immunol. 185, 3248–3255.

Lutz, M.B., and Schuler, G. (2002). Immature, semi-mature and fully

mature dendritic cells: which signals induce tolerance or immunity? Trends

Immunol. 23, 445–449.

Mancino, A., Habbeddine, M., Johnson, E., Luron, L., Bebien, M., Memet, S.,

Fong, C., Bajenoff, M., Wu, X., Karin, M., et al. (2013). I kappa B kinase alpha

(IKKa) activity is required for functional maturation of dendritic cells and

acquired immunity to infection. EMBO J. 32, 816–828.

Manicassamy, S., Reizis, B., Ravindran, R., Nakaya, H., Salazar-Gonzalez,

R.M., Wang, Y.C., and Pulendran, B. (2010). Activation of beta-catenin in den-

dritic cells regulates immunity versus tolerance in the intestine. Science 329,

849–853.

Miller, J.C., Brown, B.D., Shay, T., Gautier, E.L., Jojic, V., Cohain, A., Pandey,

G., Leboeuf, M., Elpek, K.G., Helft, J., et al.; Immunological Genome

Consortium (2012). Deciphering the transcriptional network of the dendritic

cell lineage. Nat. Immunol. 13, 888–899.

Nakano, H., Lin, K.L., Yanagita, M., Charbonneau, C., Cook, D.N., Kakiuchi, T.,

and Gunn, M.D. (2009). Blood-derived inflammatory dendritic cells in lymph

nodes stimulate acute T helper type 1 immune responses. Nat. Immunol. 10,

394–402.

Ohl, L., Mohaupt, M., Czeloth, N., Hintzen, G., Kiafard, Z., Zwirner, J.,

Blankenstein, T., Henning, G., and Forster, R. (2004). CCR7 governs skin den-

dritic cell migration under inflammatory and steady-state conditions. Immunity

21, 279–288.

Persson, E.K., Uronen-Hansson, H., Semmrich, M., Rivollier, A., Hagerbrand,

K., Marsal, J., Gudjonsson, S., Hakansson, U., Reizis, B., Kotarsky, K., and

Agace, W.W. (2013). IRF4 transcription-factor-dependent CD103(+)CD11b(+)

dendritic cells drive mucosal T helper 17 cell differentiation. Immunity 38,

958–969.

Poulin, L.F., Henri, S., de Bovis, B., Devilard, E., Kissenpfennig, A., and

Malissen, B. (2007). The dermis contains langerin+ dendritic cells that develop

and function independently of epidermal Langerhans cells. J. Exp. Med. 204,

3119–3131.

Schlitzer, A., McGovern, N., Teo, P., Zelante, T., Atarashi, K., Low, D., Ho,

A.W., See, P., Shin, A., Wasan, P.S., et al. (2013). IRF4 transcription factor-

dependent CD11b+ dendritic cells in human and mouse control mucosal

IL-17 cytokine responses. Immunity 38, 970–983.

Shih, V.F., Davis-Turak, J., Macal, M., Huang, J.Q., Ponomarenko, J., Kearns,

J.D., Yu, T., Fagerlund, R., Asagiri, M., Zuniga, E.I., and Hoffmann, A. (2012).

Control of RelB during dendritic cell activation integrates canonical and nonca-

nonical NF-kB pathways. Nat. Immunol. 13, 1162–1170.

Steinman, R.M., Hawiger, D., and Nussenzweig, M.C. (2003). Tolerogenic den-

dritic cells. Annu. Rev. Immunol. 21, 685–711.

Tamoutounour, S., Guilliams, M., Montanana Sanchis, F., Liu, H., Terhorst, D.,

Malosse, C., Pollet, E., Ardouin, L., Luche, H., Sanchez, C., et al. (2013).

Origins and functional specialization of macrophages and of conventional

and monocyte-derived dendritic cells in mouse skin. Immunity 39, 925–938.

Travis, M.A., Reizis, B., Melton, A.C., Masteller, E., Tang, Q., Proctor, J.M.,

Wang, Y., Bernstein, X., Huang, X., Reichardt, L.F., et al. (2007). Loss of integ-

rin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice.

Nature 449, 361–365.

Uyttenhove, C., Marillier, R.G., Tacchini-Cottier, F., Charmoy, M., Caspi, R.R.,

Damsker, J.M., Goriely, S., Su, D., Van Damme, J., Struyf, S., et al. (2011).

Amine-reactive OVA multimers for auto-vaccination against cytokines

and other mediators: perspectives illustrated for GCP-2 in L. major infection.

J. Leukoc. Biol. 89, 1001–1007.

Waithman, J., Allan, R.S., Kosaka, H., Azukizawa, H., Shortman, K., Lutz, M.B.,

Heath, W.R., Carbone, F.R., and Belz, G.T. (2007). Skin-derived dendritic cells

can mediate deletional tolerance of class I-restricted self-reactive T cells.

J. Immunol. 179, 4535–4541.

Weinlich, G., Heine, M., Stossel, H., Zanella, M., Stoitzner, P., Ortner, U.,

Smolle, J., Koch, F., Sepp, N.T., Schuler, G., and Romani, N. (1998). Entry

into afferent lymphatics and maturation in situ of migrating murine cutaneous

dendritic cells. J. Invest. Dermatol. 110, 441–448.

Wilson, N.S., Young, L.J., Kupresanin, F., Naik, S.H., Vremec, D., Heath, W.R.,

Akira, S., Shortman, K., Boyle, J., Maraskovsky, E., et al. (2008). Normal propor-

tion and expression of maturation markers in migratory dendritic cells in the

absenceofgermsorToll-like receptorsignaling. Immunol.CellBiol.86, 200–205.

Yamakita, Y., Matsumura, F., Lipscomb, M.W., Chou, P.C., Werlen, G.,

Burkhardt, J.K., and Yamashiro, S. (2011). Fascin1 promotes cell migration

of mature dendritic cells. J. Immunol. 186, 2850–2859.

Zhang, J., Clark, K., Lawrence, T., Peggie, M.W., and Cohen, P. (2014). An un-

expected twist to the activation of IKKb: TAK1 primes IKKb for activation by

autophosphorylation. Biochem. J. 461, 531–537.

Immunity 42, 627–639, April 21, 2015 ª2015 Elsevier Inc. 639

Page 15: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Immunity

Supplemental Information

Homeostatic NF-B Signaling

in Steady-State Migratory Dendritic Cells

Regulates Immune Homeostasis and Tolerance

Myriam Baratin, Chloe Foray, Olivier Demaria, Mohamed Habbeddine, Emeline Pollet,

Julien Maurizio, Christophe Verthuy, Suzel Davanture, Adriana Flores-Langarica, Marc

Dalod, and Toby Lawrence

Page 16: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

1    

Supplementary information

Figure S1. Related to Fig.1; Geneset enrichment analysis (GSEA) of differentially expressed genes between migDC in CLN and dermal DC. (A) Gene set enrichment analyses (GSEA) for the pairwise comparison of CD64-

CD11b+ migratory DC from CLN (migDC) compared to dermal CD64- CD11b+ DC

(dDC) using several publically available DC gene signatures and gene sets compiled

from genes modulated by Wnt/β-catenin signaling in DC (extracted from Jiang et al.,

2007; GENESET_UP_BETA_CATENIN and GENESET_REDUCED_MELLMAN).

The number of genes in each signature is indicated in brackets. (B) Relative

expression values for direct Wnt-βcatenin target genes are shown in dDC compared

to migDC (GENESET_UP_BETA_CATENIN), genes extracted from Jiang et al., 2007

(GENESET_REDUCED_MELLMAN) are indicated in bold. (C) Ingenuity pathway

analysis (IPA) of upstream regulators associated with the migDC specific gene set,

selected terms associated with known activators of IKKβ-NF-κB pathway are shown.

Figure S2. Related to Fig.2; Specificity of IKKβ deletion in Tg(Itgax-Cre) mice.

(A) Tg(Itgax-Cre) and Lyz2Cre/+ mice were crossed with IkbkbF/F mice carrying a RFP

reporter gene (Rosa26LSL-RFP); DC subsets (migDC, cDC and moDC), T cells and B

cells were subsequently analysed by flow cytometry. Representative histograms are

shown and the proportions of RFP positive cells indicated. (B-C) DC, T cells and B

cells were sorted by flow cytometry from spleens of IkbkbF/F and IkbkbΔItgax mice; (B)

protein extracts were prepared for Western blot analysis of IKKβ expression, (C) total

RNA was isolated from cells in parallel for analysis of Ikbkb mRNA expression by

quantitative RT-PCR (qPCR), expression was normalized to Cph (cyclophillin)

mRNA.

Figure S3. Related to Fig.3; Further analysis of spleen DC subsets and Treg phenotype in IkbkbF/F and IkbkbΔItgax mice. (A) Spleen cell suspensions from IkbkbF/F and IkbkbΔItgax mice were prepared and

stained with anti-B220, anti-MHC II, anti-CD8, anti-CD11b and anti-CD4 to analyze

different DC subsets; after gating out dead cells and excluding B cells, DC were

gated as MHCIIhi CD11chi and proportions of CD11b+ and CD8+ cells were

Page 17: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

2    

determined; CD11b+ cells were further separated as CD4 expression. Data are

shown as mean ± SEM, graphs represent pooled data from at least 2 independent

experiments. Statistical analysis was performed with Mann Whitney test; ***p<0.001.

(B) Spleen and (C) CLN cell suspensions were prepared and stained with anti-CD4,

anti-CD25, anti-Foxp3, anti-Nrp1 and anti-CD62l to analyze different Treg

populations in IkbkbF/F and IkbkbΔItgax mice. Data are shown as mean ± SEM, graphs

represent pooled data from at least 2 independent experiments. Statistical analysis

was performed with Mann Whitney test; **p<0.01, ****p<0.0001.

 

Figure S4. Related to Fig.4; Distribution of dermal DC subsets after IKKβ

deletion in DC. Dermal cell suspensions were prepared and stained with anti-CD45.2, anti-MHC II,

anti-CD3, anti-CD11b, anti-CD24, and anti-CD103 to analyze the different DC

subsets. CD45.2+, MHC II+, CD3- gated cells are represented on the plots. Four

populations (#1 to 4) were determined according to their expression of CD11b and

CD24. The percentage of CD103+ cells included in population #2 was also

determined. Graphs represent the percentage of each population and are

representative of 2 independent experiments. Figure S5. Related to Fig.5; Spontaneous maturation of dermal DC ex vivo. Single cell suspensions of skin DC were prepared from epidermis and treated with or

without various inhibitors overnight at 4oC or 37oC (IKKβ inhibitor BI605906, 10 and

50µM; JAK3 inhibitor CP690550 1µM; anti-mouse TGFβ1 neutralizing ab 10µg/ml;

anti-mouse CSF2 neutralizing ab 5µg/ml). DC maturation was measured by

expression of CD40, CCR7 and Fascin by flow cytometry, gating on Sytox-negative

MHCIIhi cells. Data are shown as mean ± SEM of biological replicates and are

representative of 3 independent experiments. Statistical analysis was performed by

ANOVA with Bonferroni correction; ****p<0.0001.

Figure S6. Related to Fig.4; TNFα, IL-1β, TLR and IKKα signaling are not

required for steady-state migration of NLT-DC in the skin. Flow cytometry analysis of CLN was performed on C57BL/6 littermate control mice,

Myd88/Trif-/- mice, Tnfsfr1/Tnfsfr2-/- mice, IKKαAA/AA mice and LTβR-Ig treated mice.

Page 18: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

3  

After gating out dead cells, B cells, PMN and monocytes, migDC were identified by

anti-MHC II and anti-CD11c staining. Plots represent the percentage of migDC from

n=3 mice of each group (mean ± SEM), and are representative of at least two

independent experiments.

Page 19: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

4    

Experimental procedures Mice. Mice were housed under specific pathogen free conditions and handled in

accordance with French and European directives. All animal experimentation was

approved by Direction Départementale des Services Vétérinaires des Bouches du

Rhône. K5-mOVA, OT-I, OT-II, Ccr7-/- and Tg(Itgax-hBcl2) mice have been

previously described respectively by (Azukizawa et al., 2003), (Hogquist et al., 1994),

(Barnden et al., 1998), (Forster et al., 1999) and (Gautier et al., 2009). OT-II mice

were crossed with CD45.1 C57BL/6 mice.

Antibodies. Anti-CD11c (HL3), anti-CD8α (53-6.7), anti-MHC II (M5/114), anti-

CD45.2 (104), anti-CCR7 (4B12), anti-CD11b (M1/70), anti-CD24 (M1/69), anti-

Foxp3 (FJK-16s) and anti-CD103 (M290) antibodies were all purchased from either

BDeBioscience or Biolegend. Anti-Fascin (EP5902) antibody was purchased from

Abcam. H-2Kb-OVA257-264 PE tetramers were purchased from Beckman Coulter.

Flow cytometry. Cells were first incubated at 4°C for 10 min with the 2.4.G2

antibody to block Fc receptors and then stained with the adequate antibodies during

30 min at 4°C. Dead cells were gated out using sytox blue dye. For fascin staining,

cells were first fixed with PFA 2% and permeabilized by incubation with 90%

methanol for 30 min. Foxp3 expression in CD4+ T cells was evaluated using the

Foxp3 staining kit (eBioscience) following the manufacturer's protocol. Samples were

run with a FACSCanto system (BD). Analysis were performed using Flowjo software

(Tree Star, Inc).

Microarray Analysis. Microarray analyses were performed as reported previously

(Yamakita et al., 2011). The genes differentially expressed between steady state

dDC and mig-DC were identified using the Bioconductor Limma statistical package,

with a cutoff value of 0.05 for the false discovery rate and of 2 for the fold change. A

pathway analysis was then performed on the 1.095 genes up-regulated in mig-DC

using the Ingenuity Pathway Analysis facility. This gene list was also inspected for

enrichment for transcription factor binding sites using different methods, including a

Distant Regulatory Elements analysis using default parameters, and a motif search

using the Clover (Cis-eLement OVER representation) software as previously reported

Page 20: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

5    

(Baranek et al. Cell Host Microbe. 2012) with the p-value threshold and the score

fixed respectively at 0.05 and 4.

Skin explant culture. Ears were split into ventral and dorsal parts and floated split

side down on 1ml medium containing RPMI 10% FCS and 50µM β-mercaptoethanol.

Medium was replaced daily. After 3 days of culture, emigrated cells in the culture

media were collected and stained to evaluate DC number, viability and phenotype. In

parallel experiments, dermis was separated from the epidermis and stained as

described in the following section.

IHC on skin explants. Ear were split into dorsal and ventral halves and floated split

side down for 2h on 20mM EDTA in PBS at 37°C. The epidermis was separated from

the dermis with forceps and both were washed in PBS and fixed in ice cold

ANTIGENFIX (DIAPATH) 20min at 4°C. After rehydration in PBS and

permeabilisation in Perm Wash buffer (BD), sheets were stained with anti-MHC II and

anti-CD3. Staining were performed in Perm Wash buffer (BD). Sheets were mounted

with Fluoromount-G (Southern Biotech) and were analysed with the Carl Zeiss LSM

780 confocal microscope. Pictures were analysed using Image J software.

Isolation of skin DC. Ears were split into dorsal and ventral halves and floated split

side down for 1h on dispase (1mg/ml) in PBS at 37°C. The epidermis was separated

from the dermis with forceps and each sheet was minced with a razor blade before

2h incubation in RPMI, HEPES, 2%FCS, penicillin/streptomycin and collagenase A

(1mg/ml) at 37°C under agitation. Cell suspensions were filtered through a 70µm

mesh and washed in culture medium. Cells were cultured for 18h at 37°C in 96well

plates prior to analysis by flow cytometry. For inhibitor experiments; single cell

suspensions of skin DC were prepared from epidermis and treated with or without

various inhibitors for 18h at 4oC or 37oC (IKKβ inhibitor BI605906 (Zhang et al.,

2014), 10 and 50µM; JAK3 inhibitor CP690550 (Tocris) 1µM; anti-mouse TGFβ1

neutralising ab (Uyttenhove et al., 2011) 10µg/ml; anti-mouse CSF2 neutralising ab

(eBioscience) 5µg/ml)

Adoptive T cell transfer. OT-I and OT-II cells were purified from pooled LN and

spleen by negative selection using a T cell isolation kit (Invitrogen). CD25- OT-II cells

were further sorted by FACS using anti-CD4 and anti-CD25 antibodies. Purity was

determined by staining with anti-Vα2 and anti-CD8 or CD4 antibodies. For CFSE

Page 21: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

6    

labelling, purified cells were suspended in PBS and labelled with 2.5 µM CFSE

(Invitrogen) for 10 min at 37°C. 1 to 2x106 cells were injected i.v. into the recipient

mice. Nine or 13 days after injection of OT-I or OT-II cells respectively, single cell

suspensions from CLN were prepared and stained with H-2Kb-OVA257-264 PE

tetramers to detect OT-I cells or with anti-CD45.1 antibody to detect OT-II cells.

Brdu labelling in vivo. Mice were injected i.p. with 1,5 mg of BrdU (sigma Aldrich) to

ensure its immediate availability and their drinking water was supplemented for up to

7 days with 0,8 mg/ml of BrdU and 2% glucose and changed daily. DC from

epidermis and dermis were isolated and stained for surface markers and

permeabilised for BrdU staining according to the manufacturer recommendations

(BrdU labelling Flow kit, BD).

FITC painting. One µl of FITC (0.5 mg/ml) was dissolved in 50µl acetone and 50µl

dibutyl phthalate. 30 µl were epicutaneously applied on the ear of anesthetized mice.

After 48h, single cell suspensions from auricular LN were prepared and stained for

flow cytometry.

Topical immunization. 50µg of H-2Kb-OVA257-264 peptide were included in 50mg

of imiquimod ointment (Aldara; 3M Pharceuticals). This was applied to the shaved

ears of anesthesized animals. 7 days later, single cell suspensions from CLN were

prepared and stained with H-2Kb-OVA257-264 PE tetramers to detect CD8 specific T

cells.

Histopathology and IHC. The spleen and kidney were processed, stained with

hematoxylin an eosin and analysed by the CIML pathology core. For

immunochemistry, spleen and kidney were snap-frozen and 5µm sections were

stained with indicated antibodies. GC development was evaluated using the following

scoring: 1, weak; 2, moderate; 3, marked; 4, high.

Bone marrow derived dendritic cells. Bone marrow cells from IkbkbF/F Tg(Ubc-

Cre/ERT2) or IkbkbF/F control mice were cultured in 6-well plates for 8 days in RPMI

containing 10% heat-inactivated FBS, 2mM L-glutamine, 100 U/ml

penicillin/streptomycin and 20 ng/ml recombinant murine CSF2 (Peprotech). At days

2 and 4, half of the medium was aspirated and replaced by new medium taking care

not to disrupt develping cell clusters. On day 4, 5µM 4-OHT was added to each well

and 4 days later cell clusters were collected and replated in 24-well plates for 24h

Page 22: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

 

7    

with or without disruption of cell clusters by repeated pipetting. Cells were analyzed

by flow cytometry at days 8 and 9.

Autoantibody detection in the serum. Serum was tested for autoantibody

production by a standard autoantibody test with Hep-2 cells fixed on slides

(Biomedical Diagnosis). Sera were diluted 1:160, and a donkey anti-mouse IgG

secondary antibody (Invitrogen) was used to identify autoreactive sera.

Statistical analysis. P values were calculated using either the Mann Whitney test or

the Fisher test when appropriate using Graph pad prism software.

References.  Azukizawa, H., Kosaka, H., Sano, S., Heath, W.R., Takahashi, I., Gao, X.H., Sumikawa, Y., Okabe, M., Yoshikawa, K., and Itami, S. (2003). Induction of T-cell-mediated skin disease specific for antigen transgenically expressed in keratinocytes. European Journal of Immunology 33, 1879-1888. Barnden, M.J., Allison, J., Heath, W.R., and Carbone, F.R. (1998). Defective TCR expression in transgenic mice constructed using cDNA-based alpha- and beta-chain genes under the control of heterologous regulatory elements. Immunology and Cell Biology 76, 34-40. Forster, R., Schubel, A., Breitfeld, D., Kremmer, E., Renner-Muller, I., Wolf, E., and Lipp, M. (1999). CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23-33. Gautier, E.L., Huby, T., Saint-Charles, F., Ouzilleau, B., Pirault, J., Deswaerte, V., Ginhoux, F., Miller, E.R., Witztum, J.L., Chapman, M.J., and Lesnik, P. (2009). Conventional dendritic cells at the crossroads between immunity and cholesterol homeostasis in atherosclerosis. Circulation 119, 2367-2375. Hogquist, K.A., Jameson, S.C., Heath, W.R., Howard, J.L., Bevan, M.J., and Carbone, F.R. (1994). T-Cell Receptor Antagonist Peptides Induce Positive Selection. Cell 76, 17-27. Jiang, A., Bloom, O., Ono, S., Cui, W., Unternaehrer, J., Jiang, S., Whitney, J.A., Connolly, J., Banchereau, J., and Mellman, I. (2007). Disruption of E-cadherin-mediated adhesion induces a functionally distinct pathway of dendritic cell maturation. Immunity 27, 610-624. Uyttenhove, C., Marillier, R.G., Tacchini-Cottier, F., Charmoy, M., Caspi, R.R., Damsker, J.M., Goriely, S., Su, D., Van Damme, J., Struyf, S., et al. (2011). Amine-reactive OVA multimers for auto-vaccination against cytokines and other mediators: perspectives illustrated for GCP-2 in L. major infection. Journal of leukocyte biology 89, 1001-1007. Yamakita, Y., Matsumura, F., Lipscomb, M.W., Chou, P.C., Werlen, G., Burkhardt, J.K., and Yamashiro, S. (2011). Fascin1 promotes cell migration of mature dendritic cells. Journal of immunology 186, 2850-2859. Zhang, J., Clark, K., Lawrence, T., Peggie, M.W., and Cohen, P. (2014). An unexpected twist to the activation of IKKbeta: TAK1 primes IKKbeta for activation by autophosphorylation. The Biochemical journal 461, 531-537.  

Page 23: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Fst

Cd44Cdx1

MitfLgr5Cldn1Gja1Ccnd1Fgf18Fosl1MycVegfaFzd7Krt1MycbpTcf4Wisp1Hnf1aNeurog1Axin2En2Ephb2Ephb3Fgf4Ovol1PpardSp5Gbx2Lef1JunId2Mmp7Neurod1Ctla4Pitx2Tnfrsf19Itgb1NrcamTVcan

-3.2 2,30

Relative expression value (log2 , normalized to total mean)

Legend :

dDC Dermis versus

migDC CLN CCNB2 (267)

LENAOUR_DENDRITIC_CELL_MATURATION_DN (138)

LINDSTEDT_DENDRITIC_CELL_MATURATION_D (80)

DC_MAT_CORE_DOWN (58)

LINDSTEDT_DENDRITIC_CELL_MATURATION_A (68)

KEGG_TOLL_LIKE_RECEPTOR_SIGNALING_PATHWAY (102)

GENESET_UP_BETA_CATENIN (37)

GENESET_REDUCED_MELLMAN (10)

MIGRATION (315)

PRO-APOPTOTIC (71)

LINDSTEDT_DENDRITIC_CELL_MATURATION_B (54)

LINDSTEDT_DENDRITIC_CELL_MATURATION_C (66)

DC_MAT_CORE_UP (72)

KEGG_JAK_STAT_SIGNALING_PATHWAY (158)

REACTOME_INTRINSIC_PATHWAY_FOR_APOPTOSIS (36)

Legend : Circle area = NES 3.3 2.2 1.6 1.0

Color intensity = FDR

<0.0025 <0.025 <0.13 NS (>0.25)

Enriched in Population A

Enriched in Population B

B A migDC CLN

dDC Dermis

Figure S1

Upstream Regulator Log Ratio Molecule Type Predicted Activation StateActivation z-score p-value of overlap

lipopolysaccharide chemical drug Inhibited -2,529 2,19E-70TNF -1,763 cytokine Inhibited -2,187 9,64E-51IL1B -5,071 cytokine Inhibited -4,300 1,03E-37TLR4 -1,040 transmembrane receptor Inhibited -2,948 5,62E-20TLR3 transmembrane receptor Inhibited -2,422 7,46E-18MYD88 other Inhibited -3,257 2,91E-15© 2000-2013 Ingenuity Systems, Inc. All rights reserved.

C

9  

Page 24: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Figure S2

10  

Page 25: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Figure S3

11  

Page 26: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Figure S4

12  

Page 27: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Figure S5

13  

Page 28: Homeostatic NF-κB Signaling in Steady-State …...Immunity Article Homeostatic NF-kB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance

Figure S6

14