EXPERIMENTAL IMMUNOLOGY doi: 10.1111/sji.12334 ..................................................................................................................................................................

Inhibition of Interferon Regulatory Factor 4 Suppresses Th1 and Th17 Cell Differentiation and Ameliorates Experimental Autoimmune Encephalomyelitis C. Yang, D. He, C. Yin & J. Tan

Abstract Neurology Department, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China

Received 12 February 2015; Accepted in revised form 9 June 2015 Correspondence to: J. Tan, The Eastern Part of Hulan Road, Xinxiang 453003, Henan Province, China. E-mail: [email protected]

Multiple sclerosis (MS) is an autoimmune disease that is characterized by recurrent episodes of T-cell-mediated immune attack on central nervous system (CNS) myelin, leading to axon damage and progressive disability. Interferon regulatory factor 4 (IRF4) is expressed predominantly in the immune system and plays an important role in its development and function. Recent study demonstrated that IRF4 was critical for the generation of IL-17-producing Th17 cells. However, the effect of IRF4 on experimental autoimmune encephalomyelitis (EAE), an animal model of MS, needs to be further investigated. In our current study, inhibition of IRF4 with IRF4 siRNA (SiIRF4) decreases EAE scores and infiltration of Th1 and Th17 cells, but increases Treg infiltration. SiIRF4 inhibits Th1 and Th17 cell differentiation in vivo and in vitro. In our DC-T-cell coculture system, SiIRF4treated DCs resulted in significantly less IFN-c and IL-17 production from T cells. Next, we adoptively transfer CD11c+ DCs from SiIRF4-treated mice into recipient mice and found that these CD11c+ DCs ameliorated EAE. Furthermore, CD11c+ DCs from SiIRF4-treated naive mice exhibited significantly reduced expression of pro-inflammatory cytokines TNF-a, IL-1b, IL-6 and IL-12/IL-23 (p40), and a corresponding increase in anti-inflammatory IL-10 expression. In conclusion, inhibition of IRF4 suppresses Th1 and Th17 cell differentiation and ameliorates EAE, via a direct regulation of DCs.

Introduction Multiple sclerosis (MS) is an autoimmune disease that is characterized by recurrent episodes of T-cell-mediated immune attack on central nervous system (CNS) myelin, leading to axon damage and progressive disability [1]. Effector CD4+ T cells of Th1 and Th17 subsets are found in MS lesion and can mediate experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Expression of Th1 and Th17 cytokines, IFN-c and IL-17 is detected in MS lesions [2]. EAE can be induced by the adoptive transfer of CNS antigen-reactive Th1 and Th17 cells [3–5]. Neutralization of IL-17 by treatment with antiIL-17 antibodies has been demonstrated to reduce motor symptoms of EAE [6]. Interferon regulatory factors (IRFs), consisting of nine members, were originally identified as transcriptional regulators of type I IFNs. Interferon regulatory factor 4 (IRF4) is expressed predominantly in the immune system and plays an important role in its development and function [7, 8]. Recent study demonstrated that IRF4 was

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

critical for the generation of IL-17-producing Th17 cells [9]. Moreover, previous study by Magdalena Huber et al. [10] indicated that IRF4 was pivotal for differentiation of Tc17 cells in vitro and in vivo during CNS autoimmunity. However, the effect of IRF4 on EAE needs to be further investigated. In our current study, we demonstrate a crucial role for IRF4 in the development of EAE. Inhibition of IRF4 with IRF4 siRNA (SiIRF4) decreases EAE scores and infiltration of Th1 and Th17 cells, but increases Treg infiltration. SiIRF4 inhibits Th1 and Th17 cell differentiation in vivo and in vitro. In our DC-T-cell coculture system, SiIRF4treated DCs resulted in significantly less IFN-c and IL-17 production from T cells. Next, we adoptively transfer CD11c+ DCs from SiIRF4-treated mice into recipient mice and found that these CD11c+ DCs ameliorated EAE. Furthermore, CD11c+ DCs from SiIRF4-treated naive mice exhibited significantly reduced expression of pro-inflammatory cytokines TNF-a, IL-1b, IL-6 and IL-12/IL-23 (p40), and a corresponding increase in anti-inflammatory IL-10 expression.

345

346 Inhibition of IRF4 suppresses Th1 and Th17 C. Yang et al. ..................................................................................................................................................................

Materials and methods Animals. Male C57BL/6 mice, all aged 6–8 weeks, were purchase from Center of Xinxiang Medical University. Mice were maintained in a specific pathogen-free condition. All experimental procedures were examined and approved by the Institutional Animal Care and Use Committee at Xinxiang Medical University. Induction of EAE. Induction of EAE was conducted as described previously [11]. EAE was induced in C57BL/6 mice by immunization with 250 lg of MOG35-55 (myelin oligodendrocyte glycoprotein (MOG)). All peptides were dissolved in complete Freund’s adjuvant (CFA) containing 4 mg/ml of heat-killed mycobacterium tuberculosis H37Ra. At day 1 and 48 h after immunization, C57BL/ 6 mice were injected with 500 ng of pertussis toxin in PBS, intraperitoneally. Clinical assessment of EAE was performed after disease induction by the following criteria: 0, no disease; 1, tail paralysis; 2, hindlimb weakness or partial paralysis; 3, complete hindlimb paralysis; 4, forelimb and hindlimb paralysis; and 5, death. A total of 100 lg of IRF4 siRNA or control siRNA was administrated via caudal vein for every 2 days from day 5 to day 15 after immunization [12]. Sequences of siRNAs were as follows: control siRNA, 50 -UAAGGCUAUGAAGA GAUACUU-30 ; and IRF4 siRNA, 50 -GGACACAC CUAUGAUGUUAUU-30 . These siRNAs were obtained from Dharmacon. For passive EAE experiments, donor mice were immunized with MOG/CFA in the same fashion as when inducing EAE, but no pertussis toxin was administered. Spleens and draining lymph nodes were collected 10 days later, single-cell suspensions were prepared, and RBCs were lysed. Cells (6 9 106 cells per ml) were cultured in RPMI1640 medium with 40 lg/ml of MOG35–55 peptide and 10 ng/ml of recombinant mouse IL-12 (R&D Systems, Houston, USA). After 3 days of culture, cells were harvested and T cells were isolated by negative selection using Dynabeads (Invitrogen, Carlsbad, CA, USA). Recipient mice were irradiated sublethally (500 cGy) and received 5 9 106 cells intravenously [13]. Histopathology. Mice were humanely euthanatized, and spinal cord was immersed in 4% paraformaldehyde for 48 h at 4 °C and paraffin-embedded. Six-micrometre-thick longitudinal sections of the spinal cord were cut to include the majority of the length of the spinal cord (from cervical to lumbar regions), containing both grey and white matters. Prior to staining, sections were deparaffinized in xylene (2 9 5 min), hydrated in graded ethanol (2 9 5 min in 100%, 5 min in 85%, 5 min 70%) to distilled water and finally rinsed in PBS. Spinal cord sections were stained with haematoxylin/eosin (Sigma, St. Louis, MO, USA) and with Luxol fast blue (American Mastertech, CA, Houston, USA) following the manufacturer’s recommendations to assess cell infiltration and demyelination, respectively. Four to five different sections

per mouse were analysed under light microscopy. At least 10 randomly distributed 409 fields within the white matter of the spinal cord were captured for each section. To determine the demyelination, the white matter was outlined automatically using the wand tool. The areas covered by the Luxol fast blue stain were quantified using Image-Pro Plus software (Media Cybernetics, MD, Houston, USA). The demyelination was determined as follows: (Total white matter area-Luxol fast blue area)/Total white matter area 9 100%. Preparation and evaluation of CNS cells. Brains of mice, which were perfuse with cold PBS, were dissected and incubated in 2.5 mg/ml collagenase D for 30 min at 37 °C. Single-cell suspensions were prepared. Cells were washed in RPMI 1640 medium, and mononuclear cells were isolated using a discontinuous Percoll gradient [14]. Flow cytometry analysis. Isolated mononuclear cells from spleen, LN and CNS were cultured in 24-well plates in RPMI1640 medium supplemented with 10% FBS, 200 ng/ml phorbol myristate acetate (PMA; Sigma), 400 ng/ml ionomycin and brefeldin A (Sigma) for 4 h. The cells were harvested and stained with FITC-antihuman CD4 at 4 °C for 30 min. After washing with PBS, the cells were fixed, permeabilized and stained with APCanti-IL-17, APC-anti-IFN-c or APC-anti-Foxp3 (eBioscience, San Diego, CA, USA) at 4 °C for 30 min. The frequencies of Th17 and Th1 cells were analysed using a FACS cytometer equipped with CellQuest software (BD Pharmingen, New York, USA). Cytokine analysis. Spleens, LN cells and CNS mononuclear cells were harvested and pooled from EAE mice, and single-cell suspensions were prepared. Cells were cultured at 5 9 107 cells/well in 24-well U-bottom plates with 10 mg/ml MOG35-55 peptide in complete RPMI 1640 medium. For ELISA analysis, supernatants were harvested at 24 h of culture. The concentrations of indicated cytokines were measured by quantitative capture ELISA, according to the guidelines of the manufacturers. Adoptive transfer of DCs. DCs were isolated from the spleens of 8- to 10-week-old mice treated with SiIRF4 or control for 10 days. DCs were enriched by magnetic cell sorting using CD11c+ magnetic beads (Miltenyi, Berlin, Germany), resulting in a general purity of at least 95%. Recipient C57BL/6 mice were injected intravenously (i.v.) with 5 9 106 SiIRF4- or control-treated DCs and then immunized with MOG35–55 immediately [15]. Real-time PCR. Total RNA was extracted from cultured cells using Trizol (Invitrogen) and reverse-transcribed into cDNA using the PrimeScript RT reagent kit (Takara Biotechnology, Dalian, China) according to the manufacturer’s instructions. mRNA levels of target genes were quantified using SYBR Green Master Mix (Takara Biotechnology) with ABI PRISM 7900 Sequence Detector system (Applied Biosystems, Foster City, CA, USA). Each reaction was performed in duplicate, and changes in

Scandinavian Journal of Immunology, 2015, 82, 345–351

C. Yang et al. Inhibition of IRF4 suppresses Th1 and Th17 347 ..................................................................................................................................................................

Figure 1 SiIRF4 ameliorates EAE development. (A) Clinical scores for mice treated with SiIRF4 after the onset of EAE; (B) At day 25 after immunization, CNSinfiltrating CD4+ and CD8+ T cells were isolated, analysed by flow cytometry and absolute numbers were calculated; (C) Indicating the demyelination rate at day 25 after immunization; (D) MOG-primed T cells from control or SiIRF4-treated mice were transferred into sublethally irradiated mice. The mice were scored for clinical signs of EAE and the mean clinical score was calculated by averaging the scores of all mice. *P < 0.05. N = 6. The experiments were repeated three times in each animal.

A

B

C

D

relative gene expression normalized to 18sRNA levels were determined using the relative threshold cycle method. Western blotting. The protein levels were determined by Western blotting. Protein extracted from cells or tissue was separated on 10% SDS-polyacrylamide electrophoresis gels and transferred to nitrocellulose membranes (Pierce, Rockford, IL, USA). After being blocked with 5% nonfat milk in TBS for 3 h, the membranes were incubated with indicated primary antibodies (0.2 lg/ml) at 4 °C overnight, followed by incubation with HRP-conjugated secondary antibody (1:5000) for 3 h. b-Actin was used as a loading control for comparison between samples. Immunoreactive bands were quantified with densitometry using Image J software (NIH, Maryland, USA). Statistical analysis. All data were presented as means  SEM. ANOVA was used for statistical analysis with P < 0.05 being considered statistically significant. Data were analysed using Prism software (GraphPad Software, Inc., Beijing, China).

A

B

Results SiIRF4 treatment ameliorates EAE development

In this study, we used a mouse model of EAE to evaluate the therapeutic effect of IRF4 inhibition with SiIRF4. Results showed that SiIRF4 treatment decreased the EAE scores and enhanced clinical recovery from EAE (Fig. 1A). Analysis of CNS-infiltrating mononuclear showed that mice treated with control PBS developed prominent inflammatory infiltration, whereas SiIRF4 treatment

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

Figure 2 In vivo generation of Th17 cells and Th1 cells in EAE inhibited by SiIRF4. (A) Intracellular staining of IFN-c, IL-17 or Foxp3 in spleen cells, LN cells and CNS-infiltrating mononuclear cells. The frequencies of Th17 and Th1 cells were analysed using a FACS cytometer equipped with CellQuest software (BD Pharmingen); (B) Indicating the absolute number of IFN-c+, IL-17+ or Foxp3+ cells in CNS. *P < 0.05. N = 6.

348 Inhibition of IRF4 suppresses Th1 and Th17 C. Yang et al. .................................................................................................................................................................. A

B

C

showed decreased CD4+ and CD8+ leucocytes infiltration and significantly alleviated demyelination (Fig. 1B–C). Next, we transferred T cells from MOG-immunized control or SiIRF4-treated mice into wild-type recipients. Mice that received SiIRF4-treated T cells displayed lower EAE scores and better clinical recovery from EAE (Fig. 1D). SiIRF4 inhibits Th1 and Th17 cell differentiation in EAE

We examined the presence of IL-17 (Th17), interferon-c (IFN-c) (Th1) or Foxp3 (Treg)-producing CD4+ T cells during EAE in lymph nodes, spleen and the CNS. Mice were harvested on day 25 after immunization with MOG35-55. Intracellular cytokine staining showed that the frequencies of both Th1 and Th17 cells in the splenocytes, lymph nodes cells and CNS mononuclear cells were lower in SiIRF4-treated mice than that in controls (Fig. 2A). The frequencies of Foxp3+ Tregs in the splenocytes, lymph nodes cells and CNS mononuclear cells were much higher in SiIRF4-treated mice than that in controls (Fig. 2A). The absolute numbers of Th1 and Th17 cells in CNS were also markedly smaller in SiIRF4-treated mice than that in controls (Fig. 2B). And there was no significant difference in the number of Treg infiltration in CNS between the two groups (Fig. 2B). CD4+ T cells in control-treated mice underwent cell divisions in vitro with MOG35-55, whereas CD4+ T cells in SiIRF4-treated mice had a reduced proliferation (Fig. 3A). In parallel, CCK8 assays via splenocytes produced similar differences (Fig. 3B). Supernatants were harvested at 24 h of culture and analysed by

Figure 3 SiIRF4 affects inflammatory T cell development in vitro during EAE. (A) To investigate the effect of SiIRF4 on T cell proliferation in vitro, the splenocytes harvested from mice 25 days after EAE induction were labelled with carboxyfluorescein succinimidyl amino ester (CFSE). CFSE loss by CD4+ proliferating cells from the two groups was assayed by flow cytometry after restimulation with MOG35-55 (10 mg/ml) for 72 h. The average frequency of CD4+ proliferating cells is presented; (B) The harvested splenocytes was also restimulated with MOG35-55 (10 mg/ml) for 72 h, and added CCK-8 solution incubated at 37 °C for the final 4 h. Cell proliferation was evaluated by stimulation index and the average frequency is presented; (C) Indicates the production of IFN-c and IL-17A by splenocytes, LN cells and CNS-infiltrating mononuclear cells from control and SiIRF4treated EAE mice. *P < 0.05. N = 6.

ELISA. SiIRF4-treated mice showed decreased production of both IL-17A and IFN-c compared to their controls (Fig. 3C). SiIRF4 attenuates EAE via a direct effect on DC

Myeloid-derived suppressive cells (MDSCs) are a heterogeneous group of myeloid cells comprised of hematopoietic progenitor cells and precursors of macrophages, granulocytes and dendritic cells (DCs) [16]. Recently, the close relationship between IRF4 and DCs has been demonstrated [17, 18]. Therefore, we tested whether SiIRF4 regulates the expression of Th1- and Th17polarizing cytokine by DCs. For this, we cultured CD4+ T cells with CD11c+ DCs from control- and SiIRF4treated mice and analysed cytokine expression. In our DC-T-cell coculture system, SiIRF4-treated DCs resulted in significantly less IFN-c and IL-17 production from T cells (Fig. 4A). To investigate whether SiIRF4-induced myeloid DCs could modulate clinical responses in vivo, we isolated CD11c+ DCs from SiIRF4-treated mice, adoptively transferred them into recipient mice and then immunized with MOG35-55. These CD11c+ DCs ameliorated EAE development in recipient mice (Fig. 4B–C). SiIRF4 alters cytokine expression in DCs

DCs are a major source of different cytokines important for initiation and regulation of an immune response. Therefore, we analysed the cytokines expression by Q-PCR. As

Scandinavian Journal of Immunology, 2015, 82, 345–351

C. Yang et al. Inhibition of IRF4 suppresses Th1 and Th17 349 .................................................................................................................................................................. A

Figure 4 SiIRF4 attenuates EAE via a direct effect on DC. (A) CD4+ T cells isolated from C57BL/6 mice were cultured with CD11c+ DCs derived from control and SiIRF4-treated mice on day 25 after EAE induction. Supernatants from cultures were harvested 72 h after initiation of cultures and assayed by ELISA for IFN-c and IL-17; (B) CD11c+ cells from SiIRF4-treated donor mice attenuates EAE. 5 9 106 purified splenic CD11c+ cells from mice treated with SiIRF4 or control were injected i.v. into recipient C57BL/6 mice and then immunized with MOG35–55. (C) At day 25 after immunization, CNS-infiltrating CD4+ and CD8+ T cells were isolated and analysed by flow cytometry and absolute numbers were calculated. *P < 0.05. N = 6.

B

C

A

B

Figure 5 SiIRF4 alters cytokine expression in DCs. CD11c+ DCs were isolated from SiIRF4- or control-treated naive (unimmunized) mice, and the expression of TNF-a, IL-1b, IL-6, IL-12/23 (p40), IL-10 and CD80 was analysed by Q-PCR (A) and Western blotting (B). *P < 0.05. N = 6.

shown in Fig. 5, CD11c+ DCs from SiIRF4-treated naive (unimmunized) mice exhibited significantly reduced expression of pro-inflammatory cytokines TNF-a, IL-1b, IL-6 and IL-12/IL-23 (p40), and a corresponding increase in anti-inflammatory IL-10 expression. We also found that SiIRF4 treatment resulted in significant reduction in the expression of CD80 (Fig. 5).

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

Discussion Our current study demonstrates a crucial role for IRF4 in the development of EAE. Inhibition of IRF4 with SiIRF4 decreases EAE scores and infiltration of Th1 and Th17 cells, but increases Treg infiltration. SiIRF4 inhibits Th1 and Th17 cell differentiation in vivo and in vitro. In our

350 Inhibition of IRF4 suppresses Th1 and Th17 C. Yang et al. .................................................................................................................................................................. DC-T-cell coculture system, SiIRF4-treated DCs resulted in significantly less IFN-c and IL-17 production from T cells. Next, we adoptively transfer CD11c+ DCs from SiIRF4-treated mice into recipient mice and found that these CD11c+ DCs ameliorated EAE. Furthermore, CD11c+ DCs from SiIRF4-treated naive mice exhibited significantly reduced expression of pro-inflammatory cytokines TNF-a, IL-1b, IL-6 and IL-12/IL-23 (p40), and a corresponding increase in anti-inflammatory IL-10 expression. CD4+ Th cells play an important role in the adaptive immune system by activating and directing other immune cells. There are at least four distinct Th cell subsets (Th1, Th2, Th17 and regulatory T cells), which have been demonstrated to control immune response [19]. Both Th1 and Th17 cells have been shown in clinical and preclinical studies to be critically involved in the pathogenesis of autoimmune diseases including MS, rheumatoid arthritis and inflammatory bowel disease, whereas Th2 cells are implicated in asthma and allergy when aberrantly stimulated [20, 21]. IFN-c-producing Th1 and IL-17-producing Th17 cells are key proinflammatory mediators of cellular immunity that underlie crucial events during development of EAE. Both MS and EAE are thought to be initiated by myelin-reactive CD4+ T cells that produce IFN-c and IL-17 [22–24]. The Th1 lineage of cytokine can help Th17 cells invade the brain and spinal cord, thus triggering EAE [25, 26]. The high percentage of Th17 cells has an impact on the inflammation in the brain and the severity of disease [27]. Treg cells have been demonstrated to be capable of controlling CNS autoimmunity in several EAE models. Several studies described that the transfer of CD25+ Treg ameliorated EAE symptoms [28–30]. This study showed that the attenuation of EAE in SiIRF4-treated mice is associated with decreased expression of IL-17 and IFN-c and increased expression of Foxp3 in the peripheral lymphoid organs and CNS. The need for parenchymal APC is based on the fundamental immunological principle of reactivation for CD4+ T-cell effector function within the target tissue. A role for DC in directing T-cell transit from the perivascular space in postcapillary venules has been proposed [31, 32]. Recently, DCs have been demonstrated as a therapeutic target in MS and EAE [33]. CD11c+ DCs have been shown to be sufficient to initiate this autoimmune demyelinating disorder [28]. Furthermore, antigen presentation by myeloid DCs has been implicated in driving progression of relapsing EAE [34]. In our DC-T-cell coculture system, SiIRF4-treated DCs resulted in significantly less IFN-c and IL-17 production from T cells. Furthermore, we isolated CD11c+ DCs from SiIRF4-treated mice, adoptively transferred them into recipient mice and then immunized with MOG35-55. The results showed that these CD11c+ DCs ameliorated EAE in recipient mice. All of these indicated that the effect of SiIRF4 on Th1 and Th17 cell differentiation may be mediated via modulation of DCs.

Cytokines secreted by DCs are critical messengers that control T-cell activation and differentiation. CD11c+ DCs from SiIRF4-treated mice displayed significantly reduced production of the pro-inflammatory cytokines TNF-a, IL-1b, IL-6 and IL-12/IL-23 (p40), and a corresponding increase in anti-inflammatory IL-10 expression. These proinflammatory cytokines have been shown to promote CNS tissue damage in EAE [35, 36]. IL-1b is required for Th17 differentiation and in mice; it synergizes with IL-6 and IL-23 in induction of Th17 cells [37]. In addition to its potential direct toxicity, TNF-a perpetuates the inflammatory process through recruitment of cells [38]. IL-10 is an anti-inflammatory cytokine with paracrine and autocrine effects [39]. We conclude that SiIRF4 reduces the Tcell stimulatory capacity of DCs, which has been shown to be necessary for the survival of encephalitogenic T cells in the effector phase of the immune response [31]. In summary, our study provides evidence that SiIRF4 suppresses Th1 and Th17 cell differentiation and ameliorates EAE. Inhibition of IRF4 with SiIRF4 decreases EAE scores and leucocyte infiltration. SiIRF4 inhibits Th1 and Th17 cell differentiation in vivo and in vitro. And this effect was via a direct regulation of DCs. Although further investigations are needed to fully clarify the precise molecular and cellular mechanism involved in immunoregulation, IRF4 may be an effective target in attenuating MS.

References 1 Keegan BM, Noseworthy JH. Multiple sclerosis. Annu Rev Med 2002;53:285–302. 2 Steinman L. Nuanced roles of cytokines in three major human brain disorders. J Clin Invest 2008;118:3557–63. 3 Ando DG, Clayton J, Kono D, Urban JL, Sercarz EE. Encephalitogenic T cells in the B10.PL model of experimental allergic encephalomyelitis (EAE) are of the Th-1 lymphokine subtype. Cell Immunol 1989;124:132–43. 4 Jager A, Dardalhon V, Sobel RA, Bettelli E, Kuchroo VK. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J Immunol 2009;183:7169–77. 5 Domingues HS, Mues M, Lassmann H, Wekerle H, Krishnamoorthy G. Functional and pathogenic differences of Th1 and Th17 cells in experimental autoimmune encephalomyelitis. PLoS ONE 2010;5: e15531. 6 Knier B, Rothhammer V, Heink S et al. Neutralizing IL-17 protects the optic nerve from autoimmune pathology and prevents retinal nerve fiber layer atrophy during experimental autoimmune encephalomyelitis. J Autoimmun. 2015;56:34–44. 7 Lu R. Interferon regulatory factor 4 and 8 in B-cell development. Trends Immunol 2008;29:487–92. 8 Gabriele L, Ozato K. The role of the interferon regulatory factor (IRF) family in dendritic cell development and function. Cytokine Growth Factor Rev 2007;18:503–10. 9 Brustle A, Heink S, Huber M et al. The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol 2007;8:958–66. 10 Huber M, Heink S, Pagenstecher A et al. IL-17A secretion by CD8+ T cells supports Th17-mediated autoimmune encephalomyelitis. J Clin Invest 2013;123:247–60.

Scandinavian Journal of Immunology, 2015, 82, 345–351

C. Yang et al. Inhibition of IRF4 suppresses Th1 and Th17 351 .................................................................................................................................................................. 11 Zhang J, Cheng Y, Cui W, Li M, Li B, Guo L. MicroRNA-155 modulates Th1 and Th17 cell differentiation and is associated with multiple sclerosis and experimental autoimmune encephalomyelitis. J Neuroimmunol 2014;266:56–63. 12 Watanabe T, Asano N, Murray PJ et al. Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest 2008;118:545–59. 13 Yamamoto M, Suzuki K, Okuno T et al. Plexin-A4 negatively regulates T lymphocyte responses. Int Immunol 2008;20:413–20. 14 Fujihashi K, Taguchi T, Aicher WK et al. Immunoregulatory functions for murine intraepithelial lymphocytes: gamma/delta T cell receptor-positive (TCR+) T cells abrogate oral tolerance, while alpha/beta TCR+ T cells provide B cell help. J Exp Med 1992;175:695–707. 15 Schulze-Topphoff U, Shetty A, Varrin-Doyer M et al. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PLoS ONE 2012;7:e33797. 16 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162–74. 17 Akbari M, Honma K, Kimura D et al. IRF4 in dendritic cells inhibits IL-12 production and controls Th1 immune responses against Leishmania major. J Immunol 2014;192:2271–9. 18 Williams JW, Tjota MY, Clay BS et al. Transcription factor IRF4 drives dendritic cells to promote Th2 differentiation. Nat Commun 2013;4:2990. 19 Zhou L, Chong MM, Littman DR. Plasticity of CD4 + T cell lineage differentiation. Immunity 2009;30:646–55. 20 Hofstetter H, Gold R, Hartung HP. Th17 Cells in MS and experimental autoimmune encephalomyelitis. Int MS J 2009;16:12–8. 21 Qin X, Guo BT, Wan B et al. Regulation of Th1 and Th17 cell differentiation and amelioration of experimental autoimmune encephalomyelitis by natural product compound berberine. J Immunol 2010;185:1855–63. 22 Kroenke MA, Carlson TJ, Andjelkovic AV, Segal BM. IL-12- and IL23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. J Exp Med 2008;205:1535–41. 23 Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol 2007;8:345–50. 24 Yang Y, Weiner J, Liu Y et al. T-bet is essential for encephalitogenicity of both Th1 and Th17 cells. J Exp Med 2009;206:1549–64. 25 Lassmann H, van Horssen J. The molecular basis of neurodegeneration in multiple sclerosis. FEBS Lett 2011;585:3715–23. 26 Reboldi A, Coisne C, Baumjohann D et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

27

28

29

30

31

32

33

34

35

36

37

38

39

plexus is required for the initiation of EAE. Nat Immunol 2009;10:514–23. Stromnes IM, Cerretti LM, Liggitt D, Harris RA, Goverman JM. Differential regulation of central nervous system autoimmunity by T (H)1 and T(H)17 cells. Nat Med 2008;14:337–42. McGeachy MJ, Stephens LA, Anderton SM. Natural recovery and protection from autoimmune encephalomyelitis: contribution of CD4+CD25+ regulatory cells within the central nervous system. J Immunol 2005;175:3025–32. Mekala DJ, Alli RS, Geiger TL. IL-10-dependent infectious tolerance after the treatment of experimental allergic encephalomyelitis with redirected CD4 + CD25 + T lymphocytes. Proc Natl Acad Sci USA 2005;102:11817–22. Stephens LA, Malpass KH, Anderton SM. Curing CNS autoimmune disease with myelin-reactive Foxp3 + Treg. Eur J Immunol 2009;39:1108–17. Greter M, Heppner FL, Lemos MP et al. Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat Med 2005;11:328–34. Owens T, Bechmann I, Engelhardt B. Perivascular spaces and the two steps to neuroinflammation. J Neuropathol Exp Neurol 2008;67:1113– 21. Sagar D, Lamontagne A, Foss CA, Khan ZK, Pomper MG, Jain P. Dendritic cell CNS recruitment correlates with disease severity in EAE via CCL2 chemotaxis at the blood-brain barrier through paracellular transmigration and ERK activation. J Neuroinflammation 2012;9:245. Miller SD, McMahon EJ, Schreiner B, Bailey SL. Antigen presentation in the CNS by myeloid dendritic cells drives progression of relapsing experimental autoimmune encephalomyelitis. Ann N Y Acad Sci 2007;1103:179–91. Centonze D, Muzio L, Rossi S et al. Inflammation triggers synaptic alteration and degeneration in experimental autoimmune encephalomyelitis. J Neurosci 2009;29:3442–52. Rossi S, De Chiara V, Furlan R et al. Abnormal activity of the Na/Ca exchanger enhances glutamate transmission in experimental autoimmune encephalomyelitis. Brain Behav Immun 2010;24:1379–85. Chung Y, Chang SH, Martinez GJ et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity 2009;30:576–87. Skarica M, Wang T, McCadden E et al. Signal transduction inhibition of APCs diminishes th17 and Th1 responses in experimental autoimmune encephalomyelitis. J Immunol 2009;182:4192–9. Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol 2010;10:170–81.

Inhibition of Interferon Regulatory Factor 4 Suppresses Th1 and Th17 Cell Differentiation and Ameliorates Experimental Autoimmune Encephalomyelitis.

Multiple sclerosis (MS) is an autoimmune disease that is characterized by recurrent episodes of T-cell-mediated immune attack on central nervous syste...
679KB Sizes 2 Downloads 14 Views