Activation of Transcription Factor 4 in Dendritic Cells Controls Th1-Th17 Responses and Autoimmune Neuroinflammation

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Activation of Transcription Factor 4 in

Dendritic Cells Controls Th1/Th17 Responses


and Autoimmune Neuroinflammation
This information is current as Indumathi Manoharan, Daniel Swafford, Arulkumaran
of November 14, 2021. Shanmugam, Nikhil Patel, Puttur D. Prasad, Muthusamy
Thangaraju and Santhakumar Manicassamy

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
J Immunol 2021; 207:1428-1436; Prepublished online 4
August 2021;
doi: 10.4049/jimmunol.2100010
http://www.jimmunol.org/content/207/5/1428

Supplementary http://www.jimmunol.org/content/suppl/2021/08/04/jimmunol.210001
Material 0.DCSupplemental
References This article cites 62 articles, 20 of which you can access for free at:
http://www.jimmunol.org/content/207/5/1428.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2021 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Activation of Transcription Factor 4 in Dendritic Cells Controls


Th1/Th17 Responses and Autoimmune Neuroinflammation

Indumathi Manoharan,*,† Daniel Swafford,† Arulkumaran Shanmugam,† Nikhil Patel,‡


Puttur D. Prasad,* Muthusamy Thangaraju,* and Santhakumar Manicassamy*,†,§
Dendritic cells (DCs) are professional APCs that play a crucial role in initiating robust immune responses against invading
pathogens while inducing regulatory responses to the body's tissues and commensal microorganisms. A breakdown of DC-
mediated immunological tolerance leads to chronic inflammation and autoimmune disorders. However, cell-intrinsic molecular

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
regulators that are critical for programming DCs to a regulatory state rather than to an inflammatory state are not known. In this
study, we show that the activation of the TCF4 transcription factor in DCs is critical for controlling the magnitude of
inflammatory responses and limiting neuroinflammation. DC-specific deletion of TCF4 in mice increased Th1/Th17 responses and
exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of TCF4 in DCs led to heightened
activation of p38 MAPK and increased levels of proinflammatory cytokines IL-6, IL-23, IL-1b, TNF-a, and IL-12p40. Consistent
with these findings, pharmacological blocking of p38 MAPK activation delayed experimental autoimmune encephalomyelitis onset
and diminished CNS pathology in TCF4DDC mice. Thus, manipulation of the TCF4 pathway in DCs could provide novel
opportunities for regulating chronic inflammation and represents a potential therapeutic approach to control autoimmune
neuroinflammation. The Journal of Immunology, 2021, 207: 14281436.

M
ultiple sclerosis (MS) is a chronic autoimmune inflam- activation (3, 4, 5). Thus, DCs play a key role in bridging innate
matory condition that leads to multifocal demyelination and adaptive immunity. Although DCs are present in low numbers
in the white matter of the human CNS. Using experi- in the CNS under homeostatic conditions, their numbers increase
mental autoimmune encephalomyelitis (EAE), a mouse model for drastically during autoimmune inflammation, infection, or trauma
MS, studies have shown that dendritic cells (DCs) play a critical (18). However, cell-intrinsic molecular regulators that are critical for
role in the initiation and development of CNS pathology (13). DCs programming DCs to a regulatory state rather than to an inflamma-
are a specialized subset of APCs that form a critical link between tory state are not known. Thus, understanding these events may pre-
innate and adaptive immune cells (4, 5). Accumulating evidence sent promising new targets for therapeutic intervention of various
suggests that DCs play a pivotal role in instigating inflammation as autoimmune and chronic inflammatory conditions.
well as suppressing inflammation and restoring immune homeostasis Aberrant activation of the wingless/integrated (Wnt) signaling
(4, 5). Recent studies have shown that ablation of DCs in mice pathway occurs in several inflammatory diseases, including neurode-
breaks self-tolerance of CD41 T cells and results in spontaneous generative and neuroinflammatory diseases (1921). Many studies
fatal autoimmunity (6, 7). Likewise, depletion of DCs in mice have documented that Wnt ligands are highly expressed in several
resulted in stronger inflammatory responses with exacerbated EAE chronic inflammatory diseases and autoimmune diseases (2224).
(7, 8). Furthermore, depletion of specific DC subsets in lymphoid or The T cell factor/lymphoid enhancerbinding factor (TCF/LEF)
nonlymphoid tissues during the acute or relapse phase of EAE family of transcription factors are known to be critical for embryo-
results in stronger inflammatory responses and exacerbates the dis- genesis and in the development of hematopoietic stem cells and
ease (912). Besides, other studies have shown that these cells lose immune cells (25). The TCF/LEF family comprises four NFs,
their regulatory properties, resulting in uncontrolled chronic inflam- namely TCF1, LEF1, TCF3, and TCF4 (also designated as TCF7,
mation (1315). DCs contribute to CNS pathology through differen- LEF1, TCF7L1, and TCF7L2) (25). They act as one of the main
tiation and activation of naive CD41 T cells to myelin-specific Th1 downstream mediators of the Wnt/b-catenin signaling pathway (25,
and Th17 cells (16, 17). Conversely, emerging evidence suggests 26), and DCs highly express the TCF4 isoform (27, 28). However,
that DCs are also critical in resolving inflammation and limiting the functional and biological significance of TCF4 in DCs in regu-
immune-mediated pathology in EAE by producing immune regula- lating ongoing inflammation and establishing immune homeostasis
tory factors and driving regulatory T cell (Treg) differentiation and is poorly understood.

*Department of Biochemistry and Molecular Biology, Medical College of Augusta University, 1120 15th Street, CN 4153, Augusta, GA 30912. E-mail address:
Georgia, Augusta University, Augusta, GA; †Georgia Cancer Center, Medical Col- smanicassamy@augusta.edu
lege of Georgia, Augusta University, Augusta, GA; ‡Department of Pathology, Medical
College of Georgia, Augusta University, Augusta, GA; and xDepartment of Medicine, The online version of this article contains supplemental material.
Medical College of Georgia, Augusta University, Augusta, GA Abbreviations used in this article: DC, dendritic cell; DLN, draining lymph
ORCIDs: 0000-0001-9335-0082 (N.P.); 0000-0002-8455-0186 (P.D.P.); 0000-0002- node; EAE, experimental autoimmune encephalomyelitis; ICS, intracellular
2203-3917 (S.M.). cytokine staining; LRP5/6, low density lipoprotein receptor-related protein 5 and
Received for publication January 7, 2021. Accepted for publication June 28, 2021. 6; MF, macrophage; MHC II, MHC class II; MOG, myelin oligodendrocyte
glycoprotein; MS, multiple sclerosis; pDC, plasmacytoid DC; pi, post-
This work was supported by the National Institute of Diabetes and Digestive and immunization; TCF/LEF, T cell factor/lymphoid enhancerbinding factor;
Kidney Diseases Awards (DK097271 and DK123360) and Augusta University Awards Tr1, type 1 Treg; Treg, regulatory T cell; Wnt, wingless/integrated; WT,
(IGPB0003 and ESA00041) (to S.M.). wild-type.
Address correspondence and reprint requests to Dr. Santhakumar Manicassamy,
Department of Biochemistry and Molecular Biology, Medical College of Georgia, Copyright © 2021 by The American Association of Immunologists, Inc. 0022-1767/21/$37.50

www.jimmunol.org/cgi/doi/10.4049/jimmunol.2100010
The Journal of Immunology 1429

In the current study, we report that during the induction and Flow cytometry
effector phase of EAE, TCF4 activation in DCs plays a key role in Single-cell suspensions from CNS leukocytes, DLNs, and spleen were sus-
regulating the magnitude of inflammatory responses and limiting pended in PBS containing 5% FBS. After incubation for 15 min at 4 C with
collateral damage to the host. Accordingly, our data demonstrate the blocking Ab 2.4G2 (anti-FcgRIII/I), the cells were stained at 4 C for 30
that the DC-specific deletion of TCF4 in mice results in severe EAE min with the appropriately labeled Abs. Samples were then washed two
times in PBS containing 5% FBS. The samples were either immediately ana-
pathology. This was because of heightened activation of p38 MAPK
lyzed at this point or fixed in PBS containing 2% paraformaldehyde and
and increased expression of proinflammatory cytokines by DCs stored at 4 C. Intracellular staining for phospho-p38 MAPK-PE and GFP-
lacking TCF4, resulting in increased Th1 and Th17 cell polarization. FITC was performed using rabbit mAb or with appropriate isotype control in
In contrast, pharmacological blocking of p38 MAPK activation in TBS containing 1% BSA. For intracellular cytokine staining (ICS), single-
TCF4-deficient DCs markedly reduced the expression of proinflam- cell suspensions from the DLNs or CNS were ex vivo stimulated with PMA/
matory cytokines and diminished EAE severity in TCF4DDC mice. ionomycin and brefeldin A/monensin for 6 h at 37 C. The cells were then
stained for CD4 and CD8 followed by intracellular staining of IFN-g, IL-
These results reveal a novel mechanism by which the TCF4 in DCs
17A, TNF-a, and IL-10. To measure Foxp31 Tregs or phospho-p38 MAPK
controls chronic inflammation and limits immune-mediated pathol- or GFP expression, the corresponding Abs were added after permeabilization
ogy in EAE. Thus, manipulating TCF4 activation in DCs may repre- and fixation of cells. Flow cytometric analyses were performed using a
sent a convenient therapeutic approach to improve the outcome of FACS LSRII system (BD Biosciences), and the data were analyzed using

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
MS and other inflammatory diseases. FlowJo software (Ashland, OR).

Tetramer staining
Materials and Methods MOG3849-IAb tetramers were kindly provided by the National Institutes of
Mice Health Tetramer Core Facility at Emory University. CNS-infiltrating leuko-
cytes were resuspended in PBS containing 5% FBS followed by tetramer
C57BL/6, CD11c-cre (29), TCF/LEF-reporter mice (30), and 2D2 myelin staining for 1 h at 37 C as described in a previous study (34, 36). After 1 h,
oligodendrocyte glycoprotein (MOG)specific TCR transgenic mice (31) cells were stained with anti-CD4 (APC) and anti-CD45 (Alexa Fluor 700)
were originally obtained from The Jackson Laboratory and bred on site.
for 30 min on ice. Cells were washed three times, followed by data acquisi-
TCF4 floxed mice (32) were kindly provided by Dr. Melinda L Angus-Hill
tion on the flow cytometer. A nonspecific peptide tetramer was used as a
(University of Utah) and were crossbred to CD11c-cre mice to generate
negative control. The percentage of tetramer-PEpositive cells was deter-
mice lacking TCF4 in DCs (TCF4DDC) (27). The successful cre-mediated
deletion was confirmed by PCR and protein expression analyses. All mice mined in CD4-positive populations based on negative control staining.
were housed under specific pathogen-free conditions in the Laboratory Ani-
2D2 CD41 T cell adoptive transfer
mal Services of Augusta University. Animal care protocols were approved
by the Institutional Animal Care and Use Committee of Augusta University. TCF4FL/FL and TCF4DDC recipient mice were reconstituted with 2.5  106
2D2 TCR transgenic CD41 T cells followed by EAE immunization, as
Reagents and Abs described in our previous study (34). Five days later, DLNs were removed,
MOG3555 peptide (MEVGWYRSPFSRVVHLYRNGK) was purchased and after RBC lysis, in vitro recall responses were assayed by restimulating
from AnaSpec. Abs against mouse CD4 (GK1.5), CD8a (53-6.7), CD45 (30- DLN cells (2  106/ml) for 6 h with PMA/ionomycin in the presence of bre-
F11), Foxp3 (FJK-16s), IL-10 (JES5-16E3), TNF-a (MP6-XT22), CD11c feldin A/monensin for intracellular cytokine detection.
(N418), CD90.1 (HIS51), IL-17 (TC11-18H10), and IFN-g (XMG1.2) were
purchased from eBioscience and BioLegend. GFP, phospho-p38 MAPK, In vitro culture of murine DCs and T cells
p38 MAPK, and TCF4 Abs were obtained from Cell Signaling Technology. In vitro 2D2 CD41 T cell differentiation was performed as previously
p38 MAPK inhibitor (SB203580) was purchased from Calbiochem. described (33, 37). In brief, purified DLNs and splenic CD11c1 DCs (pooled
EAE induction together) (106 cells/ml) from immunized mice on day 5 were cultured together
with naive 2D2 CD41CD62L1 T cells (105) in 200 ml RPMI-1640 complete
EAE induction experiments were performed as described in our previous medium in 96-well round-bottom plates in the presence of the MOG3555 pep-
studies (33, 34). EAE was induced by s.c. immunization in the hind flanks tide (1 mg/ml). In some experiments, DCs purified from immunized mice
on day 0 using 100 mg of the MOG3555 peptide emulsified in CFA contain-
treated with p38 inhibitor or vehicle were cultured with naive 2D2 T cells.
ing 2.5 mg/ml heat-inactivated Mycobacterium tuberculosis (Difco Laborato-
After 96 h, cells were restimulated with PMA/ionomycin for 6 h in the pres-
ries). Mice also received 250 ng of pertussis toxin (List Biological
ence of brefeldin A/monensin for intracellular cytokine detection.
Laboratories) i.p. on days 0 and 2 postimmunization (pi). Disease severity
was assessed on different days pi, according to the following scale: 0, no dis-
Quantitative real-time PCR
ease; 1, flaccid tail; 2, hind limb weakness; 3, hind limb paralysis; 4, fore-
limb weakness; and 5, moribund. In some experiments, EAE-induced Total RNA was isolated from purified DLNs and spleen (pooled together) or
TCF4FL/FL and TCF4DDC were treated with either p38 MAPK inhibitor (5 CNS-infiltrating CD11c1 DCs using the QIAGEN RNeasy Mini Kit, accord-
mg/kg) or vehicle by i.p. injection every 2 d starting on day 3 pi. ing to the manufacturer's protocol (QIAGEN). cDNA was generated using
the superscript First-Strand Synthesis System for RT-PCR and random hex-
CNS leukocyte isolation amer primers (Invitrogen), according to the manufacturer's protocol. cDNA
Mice were euthanized with CO2 and perfused through the left ventricle with was used as a template for quantitative real-time PCR using SYBR Green
PBS. The brain and spinal cord were removed from each animal and dis- Master Mix (Bio-Rad Laboratories) and gene-specific primers, as described
sected into small fragments followed by digestion with collagenase type 4 (1 in our previous studies (33, 34). Gene expression across samples was nor-
mg/ml) in complete DMEM plus 2% FBS for 30 min at 37 C. Leukocytes malized relative to the housekeeping gene GAPDH.
were isolated using 40% Percoll (Sigma-Aldrich) and then were stained for
CD41 T cells expressing Foxp3 and different intracellular cytokines. Histopathology
1 Spinal cords from EAE-induced mice were removed after perfusion and
CD11c cell isolation
1 fixed using 10% formalin in PBS. Fixed spinal cords were embedded in par-
CD11c DCs were purified from the CNS or draining lymph nodes (DLNs) affin and sectioned, after which the sections were stained with H&E to study
and spleen (pooled together) as previously described (33). In brief, the spleen
leukocyte infiltration and pathology as well as Luxol fast blue stain to dem-
or brain was cut into small fragments and then digested with collagenase
onstrate CNS demyelination (34, 37).
type 4 (1 mg ml1) in complete DMEM plus 2% FBS for 30 min at 37 C.
Cells were washed twice and enriched for CD11c1 DCs with the CD11c- Statistical analysis
specific microbeads from Miltenyi Biotec. The resulting purity of CD11c1
DCs was 95%. Enriched CD11c1 DCs (106 cells/ml) from immunized Statistical analyses were conducted using Prism (GraphPad). Mean clinical
mice were cultured ex vivo for 24 h. The supernatants were collected for scores were analyzed using the MannWhitney nonparametric t test. The sta-
cytokine analysis by ELISA, whereas cells were collected for gene expres- tistical significance of differences in the means ± SD of cytokines released
sion analysis by RT-PCR (34, 35). by cells of various groups was calculated with the Student t test (one-tailed).
1430 TCF4 DEFICIENCY IN DCs AGGRAVATES THE PATHOGENESIS OF EAE

Results in DC subsets in TCF4DDC mice (Supplemental Fig. 1E). However,


Deletion of TCF4 in DCs exacerbates EAE pathology TCF4 mRNA levels were comparable in splenic MFs isolated from
WTFL/FL and TCF4DDC mice, demonstrating efficient target gene
To investigate the selective role of TCF4 in DC function during auto-
deletion in DCs (Supplemental Fig. 1E). Next, we investigated
immune neuroinflammation, we first assessed whether TCF4 is acti-
vated in DCs during autoimmune neuroinflammation using the TCF/ whether TCF4 deficiency affects DC numbers during the steady
LEF-GFP reporter mice (30). In this reporter mice, GFP reporter gene state. We did not observe any significant differences in the fre-
is under the control of six copies of a TCF/Lef response element (30). quency and total number of splenic CD11c1MHC II1 DCs between
We immunized TCF-reporter mice with the MOG3555 peptide plus WTFL/FL and TCF4DDC mice (Supplemental Fig. 1F). Similarly, the
CFA and assessed GFP expression in DCs isolated from the spleen dur- frequencies of DC subsets in the spleen were comparable between
ing the preclinical stage of EAE phase (day 5 pi) and disease phase WTFL/FL and TCF4DDC mice (Supplemental Fig. 1G), suggesting
(day 14 pi). We observed a marked increase in GFP expression in that TCF4 deletion does not affect the development and differentia-
splenic DC of TCF/LEF-GFP reporter mice pi during the induction tion of these DC subsets.
and disease phase of EAE (Fig. 1A). Likewise, CNS-infiltrating DCs Next, we analyzed EAE disease progression and pathology in the
from the reporter mice showed increased GFP expression during the WTFL/FL and TCF4DDC mice. WTFL/FL mice immunized with the
clinical stage of EAE (Fig. 1A). Next, we analyzed GFP expression in MOG3555 peptide plus CFA showed onset of neurologic impairment

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
DC subsets, macrophages (MFs), and T cells. We observed increased occurring around day 14. In contrast, TCF4DDC mice showed an earlier
GFP expression in CD81 DCs (CD451MHC class II [MHC II]1 onset of neurologic impairment and developed a more severe form of
CD11c1CD11bCD8a1), CD11b1 DCs (CD451MHC II1CD11c1 EAE (Fig. 1B). The histopathological analysis of CNS showed a
CD11b1CD8a), plasmacytoid DCs (pDCs) (CD451CD11c1B2201 marked increase in leukocyte infiltration and intense demyelination in
PDCA-11), and MFs (CD451MHC II1CD11cCD11b1CD641F4/ TCF4DDC mice compared with WTFL/FL mice (Fig. 1C, 1D). In line
801) (Supplemental Fig. 1A). Of note, we do not detect any reporter with these observations, flow cytometric analysis confirmed a signifi-
gene expression in CD41 and CD81 T cells during the preclinical cant increase in the total number of CD451 cells in the CNS of
stage of EAE phase (day 5 pi) (Supplemental Fig. 1A). We asked TCF4DDC mice. Further characterization of CNS-infiltrating leukocytes
whether increased GFP expression in DCs is due to changes in the in TCF4DDC mice showed more DCs (CD45hiMHC IIhiCD11c1),
TCF4 expression during EAE. Enriched splenic and CNS DCs from MFs (CD45hiMHC IIhiCD11cCD11b1CD641), monocytes (CD
the EAE mice showed similar TCF4 mRNA expression levels under 45hiMHC IICD11cCD11b1Ly6ChiLy6Glow), neutrophils (CD45hi
steady-state conditions as well as during the induction and effector MHC IICD11cCD11b1Ly6ClowLy6Ghi), and CD41 T cells
phase of the disease (Supplemental Fig. 1B). These results suggest that (Fig. 1E). Collectively, these data suggest that DC-specific expression
TCF4 is activated in DCs in EAE. of TCF4 is critical for limiting autoimmune CNS pathology.
To determine the selective role of TCF4 in DC function in EAE,
TCF4DDC mice display increased infiltration of Th1 and Th17 cells
we deleted TCF4 specifically in DCs (TCF4DDC) by crossing TCF4
in the CNS during EAE
floxed (WTFL/FL) mice to CD11c-cre mice. Subsequently, we con-
firmed the efficient deletion of TCF4 by quantitating the mRNA and CNS-infiltrating CD41 effector T cells contribute to the pathogene-
protein expression levels of TCF4 in the splenic DCs isolated from sis of EAE (16, 17, 38), whereas the CD41 Tregs play a key role in
the WTFL/FL and TCF4DDC mice. As expected, we noted a marked limiting the disease severity and associated tissue injury. Thus, we
decrease in the TCF4 mRNA and protein levels in TCF4-deficient next investigated if increased EAE severity observed in TCF4DDC
DCs compared with the wild-type (WT) DCs (Supplemental Fig. mice was due to effector CD41 T cell subsets. There was a signifi-
1C, 1D). Moreover, TCF4 mRNA levels were markedly decreased cant increase in the frequency of the MOG3849 tetramerspecific

FIGURE 1. Loss of TCF4 in DCs exacerbates EAE. TCF/LEF-GFP reporter mice were immunized with 100 mg of the MOG3555 peptide in CFA on day 0.
Mice also received 250 ng of pertussis toxin on days 0 and 2 pi. (A) Representative histogram of GFP expression by DCs isolated from the DLNs, spleen (day 5
and 14 pi), and CNS (day 14 pi) of EAE-induced TCF/LEF reporter mice and analyzed by ICS. (BE) WTFL/FL and TCF4DDC mice were immunized with 100
mg of the MOG3555 peptide in CFA on day 0. Mice also received 250 ng of pertussis toxin on days 0 and 2 pi. The EAE disease progression was monitored at
various days pi. (B) Mean clinical EAE score in WTFL/FL and TCF4DDC mice. (C and D) H&E and Luxol fast blue staining of spinal cords of WTFL/FL and
TCF4DDC mice to reveal the degree of demyelination and inflammation on day 16 pi. Scale bars (blue), 200 mm. (E) Total number of leukocytes (CD451), DCs
(CD45hiMHC IIhiCD11c1), MFs (CD45hiMHC IIhiCD11cCD11b1CD641), monocytes (CD45hiMHC IICD11cCD11b1Ly6ChiLy6GLow), neutrophils
(CD45hiMHC IICD11cCD11b1Ly6ClowLy6Ghi), and CD41 T cells from the CNS of WTFL/FL and TCF4DDC mice (day 16 pi) analyzed by flow cytometry.
Data are representative of two experiments (n = 45 mice per experiment). Error bars show mean values ± SEM. *p < 0.01, **p < 0.001, ***p < 0.0001.
The Journal of Immunology 1431

CD41 T cells in the CNS of TCF4DDC mice when compared with critical for limiting inflammatory cytokine expression while inducing
WT control mice (Fig. 2A, 2B). ICS of CD41 T cells showed a sig- IL-10 during the induction and effector phase of EAE.
nificant increase in the frequencies of IFN-g1, IL-171, and TNF- TCF4 activation in DCs limits Th1 and Th17 differentiation
a1 CD41 T cells in the CNS of TCF4DDC mice (Fig. 2C, 2D). In
contrast, we observed a marked decrease in the frequency of IL- As DCs dictate the fate of naive CD41 T cells through differential pro-
101CD41 regulatory (type 1 Treg [Tr1]) cells in the CNS of duction of pro- and anti-inflammatory cytokines (39), we further con-
TCF4DDC mice compared with WT control mice, whereas the fre- sidered the functional relevance of DC-specific TCF4-mediated
signaling in naive CD41 T cell differentiation. First, we performed an
quency of Foxp31CD41 T cells was comparable in both groups
ex vivo CD41 T cell differentiation assay by coculturing naive MOG-
(Fig. 2C, 2D). Consistent with these observations, following ex vivo
specific 2D2 T cells with DCs isolated from the DLNs and spleen of
MOG stimulation, leukocytes isolated from the CNS of TCF4DDC
WTFL/FL and TCFDDC mice challenged with the MOG3555 peptide
mice produced markedly higher levels of IFN-g, IL-17A, and TNF-
plus CFA. T cells cultured with TCF4-deficient DCs contained signifi-
a and lower levels of IL-10 compared with the leukocytes isolated
cantly higher frequencies of Th1 and Th17 cells compared with T cells
from the CNS of WTFL/FL mice (Fig. 2E). Collectively, these data
cultured with WT DCs (Fig. 4A, 4B). Further, analysis of the culture
suggest that DC-specific expression of TCF4 is critical for limiting supernatant showed that T cells cultured with TCF4-deficient DCs

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
autoimmune CNS pathology, indicating a possible regulatory role produced markedly higher levels of IFN-g and IL-17A than that of T
for TCF4 in DCs during ongoing neuroinflammation. cells cultured with WT DCs (Fig. 4C). Therefore, deletion of TCF4 in
TCF4 programs DCs to induce IL-10 and limit proinflammatory DCs augments Th1 and Th17 cell differentiation.
cytokine expression To extend these observations in vivo, we adoptively transferred
naive CD41CD25 T cells from 2D2 mice into WTFL/FL and TCFDDC
These results prompted us to examine whether TCF4 regulates DC
mice and then challenged these mice with the MOG3555 peptide plus
function through the expression of inflammatory and anti-inflamma-
CFA. There was a significant increase in the number of CD412D21 T
tory cytokines during the preclinical stage and clinical stage of
cells in the DLNs of TCF4DDC mice when compared with WT mice
EAE. We observed significantly increased mRNA levels for IL-6,
on day 5 postchallenge (Fig. 4D). In line with these observations, intra-
TNF-a, IL-1b, IL-23p19, and IL-12p40 and a decreased level of cellular cytokine analysis showed a significant increase in donor T cell
IL-10 in DCs isolated from the DLNs and spleen of TCF4DDC mice differentiation toward Th1 and Th17 cells in TCF4DDC mice compared
compared with WT control mice (Fig. 3A). In line with this obser- with the WT mice (Fig. 4E). Consistent with these observations, donor
vation, TCF4-deficient DCs produced markedly higher levels of IL- T cells isolated from the DLNs and spleen of TCF4DDC mice produced
6, IL-1b, TNF-a, IL-23, and IL-12p40 and reduced levels of IL-10 markedly higher levels of IFN-g and IL-17A compared with the donor
compared with WT DCs (Fig. 3B). In addition to the effector T T cells isolated from the WT mice in response to ex vivo MOG3355
cells, DCs infiltrating the CNS contributes to the pathogenesis by peptide stimulation (Fig. 4F). Therefore, TCF4 signaling in DCs limits
reactivating the primed T cells and functions as effector cells to Ag-specific Th1 and Th17 cell differentiation and expansion in vivo.
cause CNS lesions. Thus, we examined the expression of inflamma- Next, we asked whether increased inflammatory Th1 and Th17
tory and anti-inflammatory cytokines in CNS-infiltrating DCs. As cell differentiation observed in TCF4DDC mice is due to altered DC
shown in Fig. 3C, there was a significant increase in mRNA levels maturation and activation. Characterization of DLN and splenic
of IL-6, IL-1b, TNF-a, IL-23p19, and IL-12p40 but decreased lev- DCs from TCF4DDC mice showed no significant difference in the
els of IL-10 in TCF4-deficient DCs compared with WT control DCs expression of MHC II, CD40, CD80, and CD86 as compared with
infiltrating CNS. Thus, our data demonstrates that in DCs, TCF4 is control WT mice on day 5 pi (Fig. 4G). Collectively, our results

FIGURE 2. TCF4 deletion in DCs leads to increased frequencies of Th1 and Th17 cells in the CNS during EAE. (A) Representative FACS plot and
(B) frequencies of the MOG3849 tetramerspecific CD41 T cells isolated from CNS of WTFL/FL and TCF4DDC mice on day 16 pi. (C) Representative FACS
plots and (D) frequencies of IFN-g1, IL-17A1, TNF-a1, IL-101 (after PMA and ionomycin stimulation), and Foxp31CD41 T cells isolated from the CNS
of WTFL/FL and TCF4DDC EAE mice on day 16 pi. (E) Cytokine concentrations in supernatants obtained after culture of CNS-infiltrating leukocytes stimu-
lated ex vivo with the MOG3555 peptide for 48 h. Data are representative of two experiments (n = 45 mice per experiment). Error bars show mean values ±
SEM. **p < 0.001, ***p < 0.0001.
1432 TCF4 DEFICIENCY IN DCs AGGRAVATES THE PATHOGENESIS OF EAE

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
FIGURE 3. TCF4 regulates the expression of proinflammatory and anti-inflammatory cytokines in DCs during EAE. (A and C) Quantitative real-time PCR analy-
sis of Il1b, Tnfa , Il6, Il23p19, Il12p40, and Il10 mRNA expression in CD11c1 DCs isolated from the DLNs and spleen or CNS of WTFL/FL and TCF4DDC EAE mice
on day 5 and day 14 pi. (B) Cytokine secretion by CD11c1 DCs isolated from DLNs and spleen of WTFL/FL and TCF4DDC EAE mice (day 5 or 14 pi) after ex vivo cul-
ture for 48 h. Data are representative of two experiments (n > 3 per experiment). Error bars show mean values ± SEM. *p < 0.01, **p < 0.001, ***p < 0.0001.

indicate that TCF4 activation in DCs regulates the differentiation of EAE and other inflammatory diseases (40, 41). Thus, we assessed
naive CD41 T cells into Th1 and Th17 cells through the regulation the activation status of p38a MAPK in DCs in TCF4DDC and WT
of DC-specific cytokine production. mice during the induction phase and disease phase of EAE. DCs
isolated from the DLNs and spleen of TCF4DDC mice showed a
TCF4 limits the expression of inflammatory factors and Th1/Th17
marked increase in the phosphorylated (active) form of p38a
cell differentiation by regulating p38 MAPK activation in DCs MAPK compared with DCs isolated from WT control mice both
The p38a MAPK signaling pathway is critical for the expression of during the induction and effector phase of EAE (Fig. 5A, 5B). Like-
inflammatory factors and plays a key role in the pathogenesis of wise, the activity of p38a MAPK was markedly elevated in CNS-

FIGURE 4. TCF4 activation in DCs limits Th1 and Th17 cell differentiation. (AC) CD11c1 DCs were isolated from DLNs and spleen on day 5 from
WTFL/FL and TCF4DDC mice immunized with the MOG3555 peptide plus CFA (MOG) or control mice without immunization (none) and cultured ex vivo with
naive CD41CD62L1 T cells from 2D2 mice. After 5 d, cultured 2D2 cells were stimulated with anti-CD3/CD28 Ab for 48 or 6 h (in the presence of brefeldin
A and monensin). (A) Representative FACS plots and (B) frequencies for IFN-g and IL-17Aproducing CD41 2D2 T cells are shown. (C) Cytokine concentra-
tions in the culture supernatants obtained from differentiated 2D2 cells. (D and E) Naive CD41CD62L1 T cells from 2D2 mice were adoptively transferred into
WTFL/FL and TCF4DDC mice followed by immunization with the MOG3555 peptide plus CFA (MOG) or control mice without immunization (none). Five days
after the challenge, DLNs cells were stimulated in vitro for 6 h with CD3/CD28 in the presence of brefeldin A and monensin followed by ICS. (D) Total number
of CD41 2D2 cells and (E) cumulative frequencies for IFN-g and IL-17Aproducing CD41 2D2 cells are shown. (F) Cytokine concentrations in supernatants
were obtained after the culture of in vivo differentiated 2D2 cells from DLNs and spleen of WTFL/FL and TCF4DDC mice and stimulated ex vivo with the
MOG3555 peptide for 48 h. (G) CD11c1 DCs from the DLNs and spleen of WTFL/FL and TCF4DDC mice on day 5 postEAE induction were analyzed for acti-
vation and maturation. The representative histogram shows the expression of MHC II, CD40, CD80, and CD86 expression on CD11c DCs. Data are representa-
tive of two experiments (n = 45 mice per experiment). Error bars show mean values ± SEM. ***p < 0.0001.
The Journal of Immunology 1433

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
FIGURE 5. TCF4 regulates the expression of proinflammatory cytokines and Th1/Th17 cell differentiation through p38 MAPK activity. (A) Representative
histogram of phosphorylated p38 MAPK (p-p38 MAPK) in CD11c1 DCs isolated from the spleen (days 5 and 14 pi) and CNS (day 14 pi) of WTFL/FL
and TCF4DDC EAE mice. (B) Immunoblot analysis of expression of native (p38) and phosphorylated p38 MAPK (phospho-p38 MAPK) in CD11c1 DCs iso-
lated from the spleen of WTFL/FL and TCF4DDC EAE mice (day 5 pi). (CE) CD11c1 DCs were isolated from the spleen on day 5 from TCF4DDC mice
immunized with the MOG3555 peptide plus CFA (MOG) and treated daily with either SB203580 or vehicle. (C) Cytokine concentrations in supernatants
were obtained after the culture of CD11c1 DCs isolated from the spleen from TCF4DDC. (D and E) DCs were isolated from TCF4DDC mice and cultured with
naive CD41CD62L1 T cells from 2D2 mice. After 5 d, cultured 2D2 cells were stimulated in vitro for 6 h with CD3/CD28 in the presence or absence of bre-
feldin A and monensin. (D) Frequencies for IFN-g and IL-17Aproducing CD41 2D2 T cells are shown. (E) Cytokine concentrations in the culture superna-
tants obtained from differentiated 2D2 cells stimulated with anti-CD3/CD28 Ab for 48 h. Data are representative of two experiments (n = 3 per experiment).
Error bars show mean values ± SEM. ***p < 0.0001.

infiltrating DCs isolated from TCF4DDC mice as compared with of pharmacologically blocking the p38 MAPK pathway in TCF4DDC
CNS DCs isolated from WT control mice (Fig. 5A). mice during ongoing neuroinflammation. Thus, we assessed whether
Based on these observations, we hypothesized that in EAE, TCF4 blocking this pathway could ameliorate EAE pathology in TCF4DDC
regulates the expression of proinflammatory cytokines by controlling mice. To study this, we immunized TCF4DDC mice with the
the p38a MAPK activation in DCs. Thus, we examined the effect MOG3355 peptide plus CFA (MOG) and treated with either
of blocking the p38a pathway in TCF4-deficient DCs on the SB203580 or vehicle i.p. every 2 d from day 3 pi. The treatment of
expression of inflammatory and anti-inflammatory cytokines. To test TCF4DDC mice with p38 inhibitor markedly delayed EAE onset and
this, we immunized TCF4DDC mice with the MOG3355 peptide reduced EAE severity compared with vehicle-treated control mice
plus CFA (MOG) and treated with either SB203580 or vehicle . As (Fig. 6A). Consistent with disease severity score, histopathological
shown in Fig. 5C, DCs isolated from TCF4DDC mice with analysis of the CNS showed reduced inflammation as marked by
SB203580 produced markedly lower levels of IL-6, TNF-a, IL-23, reduced infiltration of leukocytes and diminished demyelination in
and IL-12p40 compared with DCs isolated from mice treated with the p38 inhibitortreated group compared with mice in the vehicle-
vehicle. However, p38 inhibitor treatment had no effect on IL-1b treated group (Fig. 6B, 6C). Besides, flow cytometric analysis con-
and IL-10 production by TCF4-deficient DCs (Fig. 5C). These firmed a marked decrease in the total number of leukocytes as well
results indicate that TCF4 limits the expression of proinflammatory as decreased number of DCs, MFs, monocytes, neutrophils, and
cytokines by regulating the activity of p38a MAPK. CD41 T cells in the CNS of TCF4DDC mice treated with SB203580
Next, we asked whether blocking p38a MAPK activation in TCF4- compared with the vehicle-treated group (Fig. 6D). Further charac-
deficient DCs affects Th1/Th17 cell differentiation. For this, we per- terization of CNS-infiltrating CD41 T cells showed a significant
formed ex vivo 2D2 T cell differentiation assay by coculturing naive reduction in the frequencies of the MOG3849 tetramerspecific
2D2 T cells with DCs isolated from TCF4DDC mice immunized with the CD41 T cells, Th1, and Th17 cells upon SB203580 treatment
MOG3555 peptide plus CFA (MOG) and treated with either SB203580 (Fig. 6EG) . Consistent with these observations, leukocytes isolated
or vehicle. T cells cultured with TCF4-deficient DCs obtained from from the CNS of TCF4DDC mice treated with the p38 inhibitor pro-
mice treated with vehicle contained higher frequencies of Th1/Th17 duced markedly lower levels of IFN-g and IL-17A compared with
cells compared with T cells cultured with TCF4-deficient DCs obtained the leukocytes isolated from the CNS of vehicle-treated mice in
from mice treated with SB203580 (Fig. 5D). Further, analysis of the cul- response to ex vivo stimulation with the MOG3355 peptide
ture supernatant showed that SB203580 treatment of TCF4-deficient (Fig. 6H). Collectively, these results indicate that TCF4 activation in
DCs markedly reduced IFN-g and IL-17A produced by T cells (Fig. DCs controls CNS inflammation and EAE disease severity by regu-
5E). Based on these results, we conclude that TCF4 activation in DCs lating the p38 MAPK pathway.
limits Th1/Th17 differentiation by regulating the activity of p38a.
Pharmacological blocking of p38 MAPK activation limits EAE Discussion
pathology in TCF4DDC mice The current study defines an essential role for the TCF4 transcrip-
Our data have thus far indicated that p38 MAPK is downstream of tion factor in DCs in controlling CNS inflammation and EAE dis-
the TCF4 pathway in DCs. So, we explored the possible implication ease severity. Accordingly, DC-specific deletion of TCF4 led to an
1434 TCF4 DEFICIENCY IN DCs AGGRAVATES THE PATHOGENESIS OF EAE

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
FIGURE 6. Pharmacological inhibition of p38 MAPK activation diminished EAE pathology in TCF4DDC mice. The progression of EAE disease course in
TCF4DDC mice treated with vehicle alone or SB203580. (A) Mean clinical EAE score in vehicle- (open circle) and SB203580- (closed circle) treated mice.
(B and C) H&E and luxol fast blue (LFB) staining of spinal cords of TCF4DDC mice to reveal the degree of demyelination and inflammation on day 16 pi.
Scale bars (blue), 200 mm. (D) Total number of leukocytes (CD451), DCs (CD45hiMHC IIhiCD11c1), MFs (CD45hiMHC IIhiCD11cCD11b1CD641),
monocytes (CD45hiMHC IICD11cCD11b1Ly6ChiLy6GLow), neutrophils (CD45hiMHC IICD11cCD11b1Ly6ClowLy6Ghi), and CD41 T cells in the
CNS of TCF4DDC EAE mice (day 16 pi) analyzed by flow cytometry. (E) Frequencies of the MOG3849 tetramerspecific CD41 T cells isolated from CNS
of TCF4DDC EAE mice treated with vehicle alone or SB203580 (day 16 pi). (F) Representative FACS plots and (G) frequencies of IFN-g1 and IL-17A1
CD41 T cells isolated from the CNS of TCF4DDC EAE mice on day 16 pi. (H) Cytokine concentrations in supernatants were obtained after culture of CNS-
infiltrating leukocytes stimulated ex vivo with the MOG3555 peptide for 72 h. Data are representative of two experiments (n = 45 mice per experiment).
Error bars show mean values ± SEM. **p < 0.001, ***p < 0.0001.

increased expression of IL-6, TNF-a, IL-1b, IL-23, and IL-12p40 shows that TCF4 is one of the key downstream mediators of the LRP5/
with diminished production of IL-10 during the induction and effec- 6b-catenin signaling in DCs. As seen in LRP5/6DDC and b-catDDC
tor phase of EAE. Consequently, the absence of TCF4 signaling in mice (34), DC-specific deletion of TCF4 in mice increased Th1/Th17
DCs suppressed Treg responses yet promoted Th1 and Th17 cell responses and exacerbated EAE pathology. Furthermore, b-catenin tar-
differentiation. Accordingly, mice lacking TCF4 in DCs exhibited a get genes such as Axin1, Axin 2, and WISP1 are markedly decreased
very severe EAE pathology characterized by intense demyelination in DCs isolated from TCF4DDC mice during the induction and effector
in CNS with increased effector CD41 T cell infiltration in the CNS. phase of the disease (Supplemental Fig. 1H). Many of the LRP5/6- and
Mechanistically, TCF4 controls the expression of proinflammatory b-catenincontrolled cytokines, such as IL-12, IL-6, IL-23, and
cytokines and neuroinflammation by regulating p38 activity in DCs. TNF-a, are elevated in TCF4DDC mice. These proinflammatory cyto-
Finally, pharmacological blocking of p38 MAPK activation in kines are also elevated in MS (5254) and are essential to induce EAE
TCF4-deficient DCs markedly reduced the expression of proinflam- (55, 56). These observations strongly suggest that TCF4 is a key down-
matory cytokines and delayed EAE onset with diminished neuropa- stream mediator of the LRP5/6b-catenin signaling in DCs. In mice
thology in TCF4DDC mice. These data indicate an important role for and humans, multiple subsets of DCs exist in both lymphoid (CD8a1
TCF4 in DCs in controlling CNS inflammation during EAE and DCs, pDCs, and CD11b1 DCs) and nonlymphoid tissues (CD1031
represent a logical target to control chronic inflammatory conditions DCs, pDCs, and CD11b1 DCs)2, 3. Depletion of DCs in mice resulted
such as MS. in stronger inflammatory responses with exacerbated EAE7, 8. Further-
Aberrant activation of the Wnt signaling pathway occurs in sev- more, depletion of specific DC subsets in lymphoid or nonlymphoid
eral inflammatory diseases, including neurodegenerative and neuro- tissues during the acute or relapsing phase of EAE results in stronger
inflammatory diseases (19, 20, 42). Many studies have documented inflammatory responses and exacerbates the disease9-12. The present
that Wnt ligands are highly expressed in several chronic inflamma- study shows that the TCF4 pathway is active in all three major DC sub-
tory diseases and autoimmune diseases (2224). The coreceptors, sets (CD8a1 DCs, CD11b1 DCs, and pDCs). Deletion of TCF4 does
low density lipoprotein receptor-related proteins 5 and 6 (LRP5/6), not dramatically alter the frequencies and number of CD8a1 DCs,
and coactivator b-catenin are critical for mediating canonical Wnt CD11b1 DCs, and pDCs. However, further studies are warranted to
signaling in APCs (21, 26, 42). In murine models of intestinal understand the role of TCF4 in these subsets in regulating CNS inflam-
inflammation and colitis-associated colon cancer, recent studies have mation. Although not analyzed in the current study, it is possible that
shown that the canonical Wnt pathway in DCs induce T regulatory TCF4 might modulate EAE by regulating DC migration.
response and suppress intestinal inflammation and inflammation- A fine balance between Tregs versus pathological Th1/Th17 cells
associated colon cancer. Our previous studies have shown that in underlies disease progression in many inflammatory diseases,
conditional knockout mice that specifically lack LRP5/6 (LRP5/ including EAE. Past studies have shown that LRP5/6b-catenin sig-
6DDC) or b-catenin (b-catDDC) in DCs are more susceptible to naling in APCs limits chronic inflammation through the induction of
inflammatory diseases (28, 43). These mice develop more severe Tregs while limiting the differentiation of pathological Th1/Th17
EAE (34). b-Catenin interacts with several transcription factors, cells (28, 34, 43). Accumulating evidence suggests that Th1/Th17
including TCFs, PPARg (44, 45), Foxo (4648), VDR (46, 49), cells play an important role in the pathogenesis of EAE and MS
IRF3 (50), and IRF8 (51). Yet, downstream mediators of LRP5/ (5759). In contrast, Tregs such as Foxp31 Tregs and IL-
6b-catenin signaling in DCs are not known. The present study 10producing Tr1 cells play a pivotal role in neuroinflammation
The Journal of Immunology 1435

and EAE disease severity (59). Our studies show that TCF4-defi- of naive CD41 T cells to Th1 and Th17 cells. Thus, manipulation of
cient DCs are potent in inducing MOG-specific Th1/Th17 cell dif- the TCF4 pathway in DCs could provide novel opportunities for regu-
ferentiation, at least in part because of increased production of IL-6, lating chronic inflammation and represents a potential therapeutic
IL-23, and IL-12. Indeed, conditional deletion of TCF4 in DCs approach to control autoimmune neuroinflammation.
exacerbated EAE severity because of a marked increase in Th1/
Th17 cells and a decrease in IL-10producing Tr1 cells in the CNS.
A recent study on intestinal inflammation has shown that the b-cate- Acknowledgments
nin directly regulates IL-10 production through TCF4 in intestinal We thank Dr. Melinda L Angus-Hill (University of Utah) for kindly provid-
DCs and MFs (28). Furthermore, b-catenin/TCF4 signaling regu- ing TCF4 floxed mice. We also thank Jeanene Pihkala for technical help
lates the expression of inflammatory factors through the autocrine with FACS sorting and analysis and Janice Randall for expert technical
effects of IL-10 on colonic APCs (28). Although not analyzed in assistance with the mice used in this study.
the current study, it is possible that TCF4-mediated signals in DCs
might modulate EAE by inducing Tr1 cell differentiation and regu-
lating the suppressive function of Foxp31 Tregs. Disclosures
The authors have no financial conflicts of interest.
Innate immune receptors, including TLR-mediated signaling in

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
DCs, play a critical role in the initiation of EAE. These receptors on
DCs sense various danger signals and induce the activation of sev- References
eral signaling networks with the secretion of cytokines that drive the 1. Terry, R. L., I. Ifergan, and S. D. Miller. 2016. Experimental autoimmune
differentiation of naive CD41 and CD81 T cells to pathological encephalomyelitis in mice. Methods Mol. Biol. 1304: 145160.
effector T cells or Tregs (39). Activation of most TLRs on DCs 2. Ganguly, D., S. Haak, V. Sisirak, and B. Reizis. 2013. The role of dendritic cells
in autoimmunity. Nat. Rev. Immunol. 13: 566577.
induces secretion of key proinflammatory cytokines that promote 3. Steinman, R. M., D. Hawiger, and M. C. Nussenzweig. 2003. Tolerogenic den-
Th1 or Th17 cell differentiation (5, 39). M. tuberculosis, an adjuvant dritic cells. Annu. Rev. Immunol. 21: 685711.
used for the induction of EAE, activates several TLRs on APCs. 4. Manicassamy, S., and B. Pulendran. 2011. Dendritic cell control of tolerogenic
responses. Immunol. Rev. 241: 206227.
p38 MAPK is one of the key downstream mediators of TLR signal- 5. Pulendran, B., H. Tang, and S. Manicassamy. 2010. Programming dendritic cells
ing and is critical for the production of proinflammatory cytokines to induce T(H)2 and tolerogenic responses. Nat. Immunol. 11: 647655.
upon TLR activation (60). The p38a MAPK signaling pathway 6. Ohnmacht, C., A. Pullner, S. B. King, I. Drexler, S. Meier, T. Brocker, and D.
Voehringer. 2009. Constitutive ablation of dendritic cells breaks self-tolerance of
plays a key role in the pathogenesis of EAE and other inflammatory CD4 T cells and results in spontaneous fatal autoimmunity. J. Exp. Med. 206:
diseases (40, 61). Moreover, p38 expression and activation in DCs 549559.
are critical for the expression of the proinflammatory factors that 7. Yogev, N., F. Frommer, D. Lukas, K. Kautz-Neu, K. Karram, D. Ielo, E. von Ste-
but, H. C. Probst, M. van den Broek, D. Riethmacher, et al. 2012. Dendritic cells
drive Th1/Th17 cell differentiation (62). Indeed, DC-specific dele- ameliorate autoimmunity in the CNS by controlling the homeostasis of PD-1
tion of p38a in mice increased the expression of the proinflamma- receptor(1) regulatory T cells. Immunity. 37: 264275.
tory cytokines and Th1/Th17 responses during EAE (62). More 8. Deshpande, P., I. L. King, and B. M. Segal. 2007. Cutting edge: CNS CD11c1
cells from mice with encephalomyelitis polarize Th17 cells and support
importantly, pharmacological inhibitors of p38 markedly reduced CD251CD41 T cell-mediated immunosuppression, suggesting dual roles in the
Th1/Th17 cell differentiation and ameliorated EAE in mice (40). disease process. J. Immunol. 178: 66956699.
Our data indicate that TCF4 in DCs limits the expression of proin- 9. Li, H., G. X. Zhang, Y. Chen, H. Xu, D. C. Fitzgerald, Z. Zhao, and A. Rostami.
2008. CD11c1CD11b1 dendritic cells play an important role in intravenous tol-
flammatory cytokines and Th1/Th17 differentiation by regulating erance and the suppression of experimental autoimmune encephalomyelitis.
the activity of p38a. Accordingly, loss of TCF4 in DCs led to J. Immunol. 181: 24832493.
heightened activation of p38 MAPK and increased levels of proin- 10. Bailey-Bucktrout, S. L., S. C. Caulkins, G. Goings, J. A. Fischer, A. Dzionek,
and S. D. Miller. 2008. Cutting edge: central nervous system plasmacytoid den-
flammatory cytokines IL-6, IL-23, IL-1b, TNFa, and IL-12. Consis- dritic cells regulate the severity of relapsing experimental autoimmune encepha-
tent with these observations, pharmacological blocking of p38 lomyelitis. J. Immunol. 180: 64576461.
MAPK activation in TCF4-deficient DCs markedly reduced the 11. Wang, L., Z. Li, B. Ciric, F. Safavi, G. X. Zhang, and A. Rostami. 2016. Selec-
tive depletion of CD11c1 CD11b1 dendritic cells partially abrogates tolerogenic
expression of proinflammatory cytokines and decreased its ability to effects of intravenous MOG in murine EAE. Eur. J. Immunol. 46: 24542466.
drive Th1/Th17 cell differentiation. Besides, the treatment of 12. King, I. L., M. A. Kroenke, and B. M. Segal. 12010. GM-CSF-dependent,
TCF4DDC mice with p38 inhibitor markedly delayed EAE onset and CD1031 dermal dendritic cells play a critical role in Th effector cell differentia-
tion after subcutaneous immunization. J. Exp. Med. 207: 953961.
reduced EAE severity. Although not analyzed in the current study, 13. Isaksson, M., B. Ardesj€o, L. R€onnblom, O. K€ampe, H. Lassmann, M. L. Eloranta,
further studies are warranted to understand whether SB203580 treat- and A. Lobell. 2009. Plasmacytoid DC promote priming of autoimmune Th17
ment affects the Ag-presenting capacity or the migratory properties cells and EAE. Eur. J. Immunol. 39: 29252935.
14. Duraes, F. V., C. Lippens, K. Steinbach, J. Dubrot, D. Brighouse, N. Bendriss-
of DCs in TCF4DDC mice. Although our data indicate an important Vermare, S. Issazadeh-Navikas, D. Merkler, and S. Hugues. 2016. pDC therapy
role for TCF4 in modulating p38 activity in DCs during EAE, it is induces recovery from EAE by recruiting endogenous pDC to sites of CNS
not clear whether TCF4 regulates p38 activity directly or indirectly. inflammation. J. Autoimmun. 67: 818.
15. Bailey, S. L., P. A. Carpentier, E. J. McMahon, W. S. Begolka, and S. D. Miller.
A recent study has shown that the b-catenin/TCF4/IL-10 signaling 2006. Innate and adaptive immune responses of the central nervous system. Crit.
axis in intestinal APCs regulates IL-6, IL-12, and IL-23 through the Rev. Immunol. 26: 149188.
suppression of p38 (28). It is possible that TCF4 could regulate p38 16. Bettelli, E., M. Oukka, and V. K. Kuchroo. 2007. T(H)-17 cells in the circle of
immunity and autoimmunity. Nat. Immunol. 8: 345350.
activation indirectly through IL-10. In the current study, we also 17. Hirota, K., J. H. Duarte, M. Veldhoen, E. Hornsby, Y. Li, D. J. Cua, H. Ahlfors,
show that TCF4 is critical for IL-10 production by DCs during C. Wilhelm, M. Tolaini, U. Menzel, et al. 2011. Fate mapping of IL-17-producing
EAE. However, p38 inhibition in TCF4-deficient DCs failed to T cells in inflammatory responses. Nat. Immunol. 12: 255263.
18. Bailey, S. L., B. Schreiner, E. J. McMahon, and S. D. Miller. 2007. CNS myeloid
restore the IL-10 production, suggesting that TCF4 regulates IL-10 DCs presenting endogenous myelin peptides ‘preferentially’ polarize CD41
production independent of p38. T(H)-17 cells in relapsing EAE. Nat. Immunol. 8: 172180.
In summary, our study reveals an important role for TCF4 in DCs 19. Clevers, H., and R. Nusse. 2012. Wnt/b-catenin signaling and disease. Cell. 149:
11921205.
in exerting a protective effect on CNS inflammation during EAE by 20. Joiner, D. M., J. Ke, Z. Zhong, H. E. Xu, and B. O. Williams. 2013. LRP5 and
regulating the expression of key proinflammatory and anti-inflamma- LRP6 in development and disease. Trends Endocrinol. Metab. 24: 3139.
tory mediators. Furthermore, our study indicates a regulatory role for 21. Suryawanshi, A., R. K. Tadagavadi, D. Swafford, and S. Manicassamy. 2016.
Modulation of inflammatory responses by Wnt/b-catenin signaling in dendritic
the TCF4 during ongoing neuroinflammation, in which activation of cells: a novel immunotherapy target for autoimmunity and cancer. Front. Immu-
this transcription factor in DCs limits the uncontrolled differentiation nol. 7: 460.
1436 TCF4 DEFICIENCY IN DCs AGGRAVATES THE PATHOGENESIS OF EAE

22. Rabelo, F. S., L. M. da Mota, R. A. Lima, F. A. Lima, G. B. Barra, J. F. de Car- 41. Kumar, S., J. Boehm, and J. C. Lee. 2003. p38 MAP kinases: key signalling mol-
valho, and A. A. Amato. 2010. The Wnt signaling pathway and rheumatoid ecules as therapeutic targets for inflammatory diseases. Nat. Rev. Drug Discov. 2:
arthritis. Autoimmun. Rev. 9: 207210. 717726.
23. Hughes, K. R., F. Sablitzky, and Y. R. Mahida. 2011. Expression profiling of 42. Swafford, D., and S. Manicassamy. 2015. Wnt signaling in dendritic cells: its
Wnt family of genes in normal and inflammatory bowel disease primary human role in regulation of immunity and tolerance. Discov. Med. 19: 303310.
intestinal myofibroblasts and normal human colonic crypt epithelial cells. 43. Swafford, D., A. Shanmugam, P. Ranganathan, M. S. Hussein, P. A. Koni, P. D.
Inflamm. Bowel Dis. 17: 213220. Prasad, M. Thangaraju, and S. Manicassamy. 2018. Canonical Wnt signaling in
24. Gudjonsson, J. E., A. Johnston, S. W. Stoll, M. B. Riblett, X. Xing, J. J. Kochko- CD11c1 APCs regulates microbiota-induced inflammation and immune cell
dan, J. Ding, R. P. Nair, A. Aphale, J. J. Voorhees, and J. T. Elder. 2010. Evi- homeostasis in the colon. J. Immunol. 200: 32593268.
dence for altered Wnt signaling in psoriatic skin. J. Invest. Dermatol. 130: 44. Alastalo, T. P., M. Li, V. J. Perez, D. Pham, H. Sawada, J. K. Wang, M. Kosken-
18491859. vuo, L. Wang, B. A. Freeman, H. Y. Chang, and M. Rabinovitch. 2011. Disrup-
25. Staal, F. J., and H. C. Clevers. 2005. WNT signalling and haematopoiesis: a tion of PPARg/b-catenin-mediated regulation of apelin impairs BMP-induced
WNT-WNT situation. Nat. Rev. Immunol. 5: 2130. mouse and human pulmonary arterial EC survival. J. Clin. Invest. 121:
26. Suryawanshi, A., M. S. Hussein, P. D. Prasad, and S. Manicassamy. 2020. Wnt 37353746.
45. Liu, J., H. Wang, Y. Zuo, and S. R. Farmer. 2006. Functional interaction between
signaling cascade in dendritic cells and regulation of anti-tumor immunity. Front.
peroxisome proliferator-activated receptor gamma and beta-catenin. Mol. Cell.
Immunol. 11: 122.
Biol. 26: 58275837.
27. Hong, Y., I. Manoharan, A. Suryawanshi, T. Majumdar, M. L. Angus-Hill, P. A.
46. Hoogeboom, D., M. A. Essers, P. E. Polderman, E. Voets, L. M. Smits, and
Koni, B. Manicassamy, A. L. Mellor, D. H. Munn, and S. Manicassamy. 2015.
B. M. Burgering. 2008. Interaction of FOXO with beta-catenin inhibits beta-cate-
b-catenin promotes regulatory T-cell responses in tumors by inducing vitamin A nin/T cell factor activity. J. Biol. Chem. 283: 92249230.
metabolism in dendritic cells. Cancer Res. 75: 656665. 47. Yan, Y., and M. R. Lackner. 2012. FOXO3a and b-catenin co-localization: dou-

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
28. Swafford, D., A. Shanmugam, P. Ranganathan, I. Manoharan, M. S. Hussein, N. ble trouble in colon cancer? Nat. Med. 18: 854856.
Patel, H. Sifuentes, P. A. Koni, P. D. Prasad, M. Thangaraju, and S. Manicass- 48. Kamo, N., B. Ke, R. W. Busuttil, and J. W. Kupiec-Weglinski. 2013. PTEN-
amy. 2020. The Wnt-b-catenin-IL-10 signaling axis in intestinal APCs protects mediated Akt/b-catenin/Foxo1 signaling regulates innate immune responses in
mice from colitis-associated colon cancer in response to gut microbiota. J. Immu- mouse liver ischemia/reperfusion injury. Hepatology. 57: 289298.
nol. 205: 22652275. 49. Shah, S., M. N. Islam, S. Dakshanamurthy, I. Rizvi, M. Rao, R. Herrell, G.
29. Caton, M. L., M. R. Smith-Raska, and B. Reizis. 2007. Notch-RBP-J signaling Zinser, M. Valrance, A. Aranda, D. Moras, et al. 2006. The molecular basis of
controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Med. 204: vitamin D receptor and beta-catenin crossregulation. Mol. Cell 21: 799809.
16531664. 50. Yang, P., H. An, X. Liu, M. Wen, Y. Zheng, Y. Rui, and X. Cao. 2010. The cyto-
30. Ferrer-Vaquer, A., A. Piliszek, G. Tian, R. J. Aho, D. Dufort, and A. K. Hadjan- solic nucleic acid sensor LRRFIP1 mediates the production of type I interferon
tonakis. 2010. A sensitive and bright single-cell resolution live imaging reporter via a beta-catenin-dependent pathway. Nat. Immunol. 11: 487494.
of Wnt/ß-catenin signaling in the mouse. BMC Dev. Biol. 10: 121. 51. Scheller, M., J. Sch€onheit, K. Zimmermann, U. Leser, F. Rosenbauer, and A.
31. Bettelli, E., M. Pagany, H. L. Weiner, C. Linington, R. A. Sobel, and V. K. Kuch- Leutz. 2013. Cross talk between Wnt/b-catenin and Irf8 in leukemia progression
roo. 2003. Myelin oligodendrocyte glycoprotein-specific T cell receptor trans- and drug resistance. J. Exp. Med. 210: 22392256.
genic mice develop spontaneous autoimmune optic neuritis. J. Exp. Med. 197: 52. Martino, G., R. Furlan, E. Brambilla, A. Bergami, F. Ruffini, M. Gironi, P. L.
10731081. Poliani, L. M. Grimaldi, and G. Comi. 2000. Cytokines and immunity in multiple
32. Angus-Hill, M. L., K. M. Elbert, J. Hidalgo, and M. R. Capecchi. 2011. T-cell sclerosis: the dual signal hypothesis. J. Neuroimmunol. 109: 39.
factor 4 functions as a tumor suppressor whose disruption modulates colon cell 53. Bar-Or, A., E. M. Oliveira, D. E. Anderson, and D. A. Hafler. 1999. Molecular
proliferation and tumorigenesis. Proc. Natl. Acad. Sci. USA. 108: 49144919. pathogenesis of multiple sclerosis. J. Neuroimmunol. 100: 252259.
33. Manicassamy, S., R. Ravindran, J. Deng, H. Oluoch, T. L. Denning, S. P. Kasturi, 54. Uccelli, A., E. Pedemonte, E. Narciso, and G. Mancardi. 2003. Biological
K. M. Rosenthal, B. D. Evavold, and B. Pulendran. 2009. Toll-like receptor 2- markers of the inflammatory phase of multiple sclerosis. Neurol. Sci. 24(0, Suppl
dependent induction of vitamin A-metabolizing enzymes in dendritic cells pro- 5): S271S274.
motes T regulatory responses and inhibits autoimmunity. Nat. Med. 15: 401409. 55. Samoilova, E. B., J. L. Horton, B. Hilliard, T. S. Liu, and Y. Chen. 1998. IL-6-
34. Suryawanshi, A., I. Manoharan, Y. Hong, D. Swafford, T. Majumdar, M. M. deficient mice are resistant to experimental autoimmune encephalomyelitis: roles
Taketo, B. Manicassamy, P. A. Koni, M. Thangaraju, Z. Sun, et al. 2015. Canoni- of IL-6 in the activation and differentiation of autoreactive T cells. J. Immunol.
cal wnt signaling in dendritic cells regulates Th1/Th17 responses and suppresses 161: 64806486.
56. Schiffenbauer, J., W. J. Streit, E. Butfiloski, M. LaBow, C. Edwards III, and
autoimmune neuroinflammation. J. Immunol. 194: 32953304.
L. L. Moldawer. 2000. The induction of EAE is only partially dependent on TNF
35. Manoharan, I., A. Suryawanshi, Y. Hong, P. Ranganathan, A. Shanmugam, S.
receptor signaling but requires the IL-1 type I receptor. Clin. Immunol. 95:
Ahmad, D. Swafford, B. Manicassamy, G. Ramesh, P. A. Koni, et al. 2016.
117123.
Homeostatic PPARa signaling limits inflammatory responses to commensal
57. Segal, B. M. 2010. Th17 cells in autoimmune demyelinating disease. Semin.
microbiota in the intestine. J. Immunol. 196: 47394749. Immunopathol. 32: 7177.
36. Sabatino, J. J., Jr., J. Huang, C. Zhu, and B. D. Evavold. 2011. High prevalence 58. Cua, D. J., J. Sherlock, Y. Chen, C. A. Murphy, B. Joyce, B. Seymour, L. Lucian,
of low affinity peptide-MHC II tetramer-negative effectors during polyclonal W. To, S. Kwan, T. Churakova, et al. 2003. Interleukin-23 rather than interleu-
CD41 T cell responses. J. Exp. Med. 208: 8190. kin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature.
37. Manoharan, I., Y. Hong, A. Suryawanshi, M. L. Angus-Hill, Z. Sun, A. L. Mel- 421: 744748.
lor, D. H. Munn, and S. Manicassamy. 2014. TLR2-dependent activation of 59. Jones, A., and D. Hawiger. 2017. Peripherally induced regulatory T cells:
b-catenin pathway in dendritic cells induces regulatory responses and attenuates recruited protectors of the central nervous system against autoimmune neuroin-
autoimmune inflammation. J. Immunol. 193: 42034213. flammation. Front. Immunol. 8: 532.
38. Ji, Q., and J. Goverman. 2007. Experimental autoimmune encephalomyelitis 60. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat. Rev. Immunol.
mediated by CD81 T cells. Ann. N. Y. Acad. Sci. 1103: 157166. 4: 499511.
39. Manicassamy, S., and B. Pulendran. 2009. Modulation of adaptive immunity 61. Ashwell, J. D. 2006. The many paths to p38 mitogen-activated protein kinase
with Toll-like receptors. Semin. Immunol. 21: 185193. activation in the immune system. Nat. Rev. Immunol. 6: 532540.
40. Krementsov, D. N., T. M. Thornton, C. Teuscher, and M. Rincon. 2013. The 62. Huang, G., Y. Wang, P. Vogel, T. D. Kanneganti, K. Otsu, and H. Chi. 2012. Sig-
emerging role of p38 mitogen-activated protein kinase in multiple sclerosis and naling via the kinase p38a programs dendritic cells to drive TH17 differentiation
its models. Mol. Cell. Biol. 33: 37283734. and autoimmune inflammation. Nat. Immunol. 13: 152161.

You might also like