Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jun 27.
Published in final edited form as: J Immunol. 2009 Jun 17;183(1):613–620. doi: 10.4049/jimmunol.0802258

IL-1R signaling within the CNS regulates CXCL12 expression at the blood-brain barrier and disease severity during experimental autoimmune encephalomyelitis

Erin E McCandless 1, Matthew Budde 2, Jason R Lees 3, Denise Dorsey 4, Eric Lyng 4, Robyn S Klein 1,4,5,6
PMCID: PMC2892701  NIHMSID: NIHMS212942  PMID: 19535637

Abstract

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by disruption of the blood-brain barrier (BBB). This breach in CNS immune privilege allows undeterred trafficking of myelin-specific lymphocytes into the CNS where they induce demyelination. Although the mechanism of BBB compromise is not known, the chemokine CXCL12 has been implicated as a molecular component of the BBB whose pattern of expression is specifically altered during MS and which correlates with disease severity. The inflammatory cytokine IL-1β has recently been shown to contribute not only to BBB permeability, but also to the development of IL-17-driven autoimmune responses. Using the rodent model of MS, experimental autoimmune encephalomyelitis (EAE), we demonstrate that IL-1β mediates pathologic relocation of CXCL12 during the induction phase of the disease, prior to the development of BBB disruption. We also show that CD4, CD8 and, surprisingly, γδ T cells are all sources of IL-1β. In addition, γδ T cells are also targets of this cytokine, contributing to IL-1β mediated production of IL-17. Finally, we show that the level of CNS IL-1R determines the clinical severity of EAE. These data suggest that T cell-derived IL-1β contributes to loss of immune privilege during CNS autoimmunity via pathologic alteration in the expression of CXCL12 at the BBB.

Keywords: central nervous system, autoimmunity, chemokine, blood-brain barrier

INTRODUCTION

Multiple sclerosis (MS) is believed to be an autoimmune disease in which myelin-specific CD4+ T cells gain excessive entry to the central nervous system (CNS) parenchyma and induce demyelinating disease (1, 2). The increased trafficking of mononuclear cells during MS exacerbations is accompanied by BBB disruption, as evidenced by the appearance of gadolinium (gd)-enhancing lesions on magnetic resonance (MR) imaging (3). However, serial radiologic studies of MS patients indicate that normal appearing white matter may exhibit altered water diffusion prior to the development of gd enhancement (4). Thus, alterations in BBB biology may occur prior to the onset of severe disruption. Understanding how changes in BBB function lead to loss of leukocyte restriction is essential for developing therapies that limit inflammation during MS.

CXCL12 is a secondary lymphoid chemokine that is constitutively expressed by the central nervous system (CNS) (5). Under normal conditions, CXCL12 is detected on the basolateral surface of the CNS endothelium where it serves to restrict the entry of infiltrating leukocytes (6-8). This polarized expression is altered in individuals with CNS autoimmune diseases with loss of perivascular CXCL12 expression and relocation of the chemokine to the luminal side of the microvasculature (7, 8). This pathologic pattern of CXCL12 expression is associated with enhanced activation of the CXCL12 receptor CXCR4 on infiltrating leukocytes and is unique to MS and its murine model, experimental autoimmune encephalomyelitis (EAE) (8). Thus, the altered expression of CXCL12 at the BBB could promote inappropriate leukocyte trafficking and contribute to disease pathogenesis.

Several molecules, including TNF-α, IL-1β, and CD40, can affect CXCL12 expression. In vitro, TNF-α and IL-1β increase CXCL12 in osteoblasts (9) and astrocytes (10), respectively, while IL-1β decreases CXCL12 expression in dermal fibroblasts (11). Studies have shown that, in vivo, CXCL12 expression is increased by CD40/CD40L in the joint synovium during arthritis (12) and by macrophage-produced IL-1β astrocytes during HIV encephalitis (13).

IL-1β is an important component of disease pathogenesis in MS (14). IL-1β is found in MS lesions (15) and individuals with a relatively high ratio of IL-1β to the naturally occurring receptor antagonist are genetically predisposed to MS (16). Studies have indicated that IL-1β induces BBB plasticity by inducing angiogenic transcription factors (17) and chronic expression of IL-1β leads to leukocyte accumulation in the otherwise homeostatic CNS (18). BBB endothelial cells (19) and astrocytes (20) express the IL-1 receptor (IL-1R) and recent studies have shown that the IL-1R deficient mouse is protected from EAE induced by active immunization with myelin peptide (21). Additionally, IL-1β is required for the proper development of Th17 cells in mice (21, 22) and humans (23). In this study, we utilized EAE to evaluate the kinetics of and identify cytokines and their cellular sources responsible for the pathologic relocation of CXCL12 at the BBB. We demonstrate that relocation of CXCL12 occurs at pre-clinical time-points in mice with EAE, prior to the development of spinal cord gd-enhancement as demonstrated by MRI and is mediated by IL-1β derived from IL-17-expressing γδT cells during the induction phase of disease. In addition, via generation of chimeric mice, we determined that CNS levels of IL-1R determine the severity of EAE.

Materials and Methods

Animals

Either C57BL/6 wild-type, IL-1R deficient, or TNFR1 deficient mice (Jackson Laboratories, Bar Harbor, ME) were used for all experiments. All mice were maintained under pathogen free conditions (Department of Comparative Medicine, Washington University, St. Louis, MO) and studies were performed in compliance with the guidelines of the Washington University School of Medicine Animal Safety Committee.

EAE induction

Active EAE was induced in 8- to 12-wk-old mice by s.c. immunization with a murine myelin oligodendroglial glycoprotein (MOG) peptide (MOGp35-55) (Sigma Genosys) in PBS emulsified 1:1 with IFA supplemented with 500μg/ml inactivated Mycobacterium tuberculosis for a total 50μg immunizing dose of MOGp35-55.Mice received 300ng of pertussis toxin (List Laboratories) i.v. at the time of immunization and 48 h later, as previously described (7). For adoptive transfer studies, MOG-specific cell lines, from C57BL/6 or IL-1R deficient C57BL/6 mice, were generated from animals immunized s.c. with 50 μg MOG35-55 peptide (Sigma-Aldrich) emulsified in IFA and supplemented with 500 μg/ml Mycobacterium tuberculosis. CD4+ cells were isolated from the spleen as previously described (24). Cells were then stimulated at a concentration of 106 cells/ml in the presence of 5 × 106 cells/ml of irradiated C57BL/6 splenocytes, 10 μg/ml MOG35-55 peptide, 10 U/ml IL-12, and 10 U/ml IL-2, in 10% FCS containing RPMI complete media to polarize cells to a Th1 phenotype. Cells were isolated 7 d later by Histopaque 1.077 (Sigma-Aldrich) and then restimulated in the absence of IL-12 under the other conditions outlined earlier in the section. Cells underwent two rounds of stimulation before transferred by retroorbital i.v. injection to 6–8-wk-old mice, which also received 300 ng pertussis toxin on the day of transfer and two days later. The grading scale used for scoring clinical manifestations of EAE in both models was the following: 1, tail weakness; 2, difficulty righting; 3, hind limb paralysis; 4, forelimb weakness or paralysis; 5, moribund or dead.

Administration of cytokines

Systemic administration of recombinant murine IL-1β (100 ng), TNFα (500 ng) and CD40L (500 ng) (R&D Systems) was accomplished via tail-vein injection of reconstituted protein in normal saline. Administration of TNF-α (10 ng/ml) directly into the spinal cord was also performed. For these experiments, isofluorane-anesthetized animals were immobilized in a steel harness and the entire length of spine exposed via incision and retraction of the skin. Individual injection sites were initiated at sections of the spinal cord separated by two vertebrae of space and carried out via spot reduction of the bone covering the target injection sites. TNF-α was injected at a volume of 1 μl per injection point using a calibrated microinjector. Individual mice received three injections at different sections of the spine, with interspersed sections of uninjected spinal cord acting as internal controls for unactivated tissue. Injections were made using microscopic needle-points produced by melting capillary tubes. Following injections, skin was sutured and animals monitored until awake and ambulating.

Antibodies

The following antibodies were used in this study. CXCL12 rabbit polyclonal antibody (Peprotech, Rocky Hills, NJ); IgG isotype control antibodies (Jackson ImmunoResearch); monoclonal rat anti-mouse-CD31 (PECAM) and CD11b (BD Pharmingen); CD3 (Dako Cytomation); IL-1β (R&D Systems); and fluorescently-conjugated antibodies against CD4, CD8, CD3, (BD Pharmingen), IL-17, and γδTCR (eBioscience).

Immunohistochemistry

Frozen sections were permeabilized and stained as described (7). For detection of CXCL12, additional blocking with image-iT Fx signal enhancer (Molecular Probes, Eugene, O.R.) solution was used according to the manufacturer's instructions. Primary antibodies were used at the following dilutions: anti-CXCL12 (1:20), anti-mPECAM-1 (2ug/mL), anti-CD11b (1:25), anti-CD3 (1:500 dilution) and anti-IL-1β (1:66 dilution). Primary antibodies were detected with secondary goat anti-rabbit and anti-rat IgG conjugated to Alexa 555 or Alexa 488 and donkey anti-goat IgG conjugated to Alexa 568 (Molecular Probes Inc.) for immunofluorescence and nuclei were counterstained with ToPro3. Control sections were incubated with isotype-matched IgG. Sections were analyzed using a Zeiss LSM 510 laser scanning confocal microscope and accompanying software (Zeiss).

Flow cytometry

Cells were isolated from the spinal cords of mice with EAE as previously described (7) and stained with fluorescently conjugated antibodies to CD4, CD8, CD3, IL-17, and γδTCR (see above) or with unconjugated primaries (IL-1β) which were then detected with anti-goat fluorescently conjugated secondary antibodies. Data collection and analysis were conducted using a FACScaliburTM flow cytometer using CellQuestTM software (Becton Dickinson). For fluorescence analysis, a bivariate dot plot of forward versus side scatter was generated, and cells were analyzed through a live cell gate.

Intracellular cytokine staining

Cells isolated as above from the spinal cords of mice were briefly restimulated in vitro for 2 hours at 37° C with 50ng/mL PMA add 1uM ionomycin and cytokines were concentrated with 1μg/ml Brefeldin A. Cells were then washed in PBS, fixed with 0.4% PFA, and permeablized with 0.1% saponin, in PBS supplemented with 0.5% BSA and 0.1% azide. Cells were incubated with antibodies in the PBS-saponin buffer, washed, resuspended in PFA, and analyzed immediately.

Bone marrow chimera

Bone marrow harvested from the femur and tibula was depleted of differentiated lymphocytes by negative selection using anti-CD4 and anti-CD8 magnetic beads (Dynal Biotech, Oslo, Norway) according to manufacture's instructions. After 45 minutes rocking at 4° C, lymphocytes bound to beads were removed and extensively washed prior to being resuspended. 2×10^6 cells were then transferred retro-orbitally to sex-matched recipients that had received a lethal irradiation dose of 1,000 rads 24 hours prior. Irradiation control mice did not received transferred cells and served as an irradiation control in each experiment. Transferred bone marrow was allowed 6 weeks for full engraftment prior to MOG immunization as described above.

Magnetic Resonance Imaging

EAE mice underwent Gd-MRI either before the onset of clinical signs, during the acute phase, or in the chronic phase. Unimmunized mice (n=2) served as controls. Mice were anesthetized using 5% isoflurane in oxygen, fitted with a custom nose cone to deliver 1% isoflurane in oxygen for maintenance, and placed in a custom holder and surface coil as described previously (25). The tail vein of the mouse was cannulated with a 28 G catheter (Strategic Applications, Inc., Libertyville, IL) prior to placement of the entire preparation in a 4.7 T MRI (Varian Inc, Palo Alto, CA). T1-weighted spin-echo images were collected before and 5 minutes after the administration of 0.02 ml bolus of 17.2 mg/ml gadolinium-diethylenetriamine pentaacetic acid (DTPA; Omniscan, GE Healthcare, Princeton, NJ) with the following parameters: TR/TE 700/14 ms, 128 × 128 matrix (zero filled to 256 × 256), 1 cm2 FOV, 1 mm slice thickness, 4 averages. Data analysis was accomplished via the creation of enhancement maps, which were created on a pixel-by-pixel basis using the equation: % Enhancement = (Spost - Spre) / Spre × 100, where Spre and Spost are the signal intensities of the pre- and post-contrast images, respectively. Regions of interest were manually traced along the ventrolateral white matter on the pre-contrast images and applied to the enhancement maps.

Statistical Analyses

A Student's t-test was used to evaluate statisitical significance in the following studies: enhancement between mice imaged during different phases of EAE and control mice, flow cytometric analyses, and Q-RT-PCR. Statistical significance was assigned to p values less than 0.05 and a single asterisk indicates p<0.05, double asterisk indicates p<0.005, and triple asterisk indicates p<0.0005.

RESULTS

Loss of CXCL12 polarity is associated with T cell entry during the induction of EAE

Initiation of inflammation within the CNS during MS and EAE is classically attributed to the entrance of myelin-specific CD4+ T cells (26, 27), which leads to demyelination. CXCL12 redistribution at the BBB occurs at the peak of EAE (7) and in active MS lesions (8). In order to define the kinetics of CXCL12 redistribution with regard to leukocyte entry, we evaluated CXCL12 expression in the spinal cords of mice during preclinical (days 11-13) and peak (days 14-16) stages of active immunization EAE (Figure 1a). Alterations in the CXCL12 expression pattern were observed in tissues collected from preclinical mice on day 11 post-immunization when there was little to no obvious inflammatory lesion formation and at the peak of EAE, as previously described (7). Loss in CXCL12 polarity at peak of disease was highly associated with the presence of classic inflammatory cuffs (Figure 1a) and analysis of 132 vessels in low-power spinal cord images from four mice revealed that 88.2% of inflamed (> 10 perivascular leukocytes) versus 0% of uninflamed (<10 perivascular leukocytes) vessels exhibited loss in CXCL12 polarity, defined as loss of basolateral expression of CXCL12 with co-localization with CD31 (Figure 1a). To examine whether CXCL12 redistribution is associated with immune cell entry into the CNS, serial spinal cord sections collected from preclinical EAE mice were examined for perivascular associated CD3+ and CD11b+ cells via immunohistochemical labeling. In vessels where loss of CXCL12 polarity was evident (Figure 1b, left panel), 100% of the serial sections analyzed had detectable T cells (Figure 1b, middle panel) whereas no macrophages were detected (Figure 1b, right panel and graph). As observed in human studies (8), arterioles retain polarized expression of CXCL12 (Figure 1b, left panel) and do not exhibit leukocytes within the perivascular space (Figure 1b, middle panel).

Figure 1. CXCL12 redistribution is associated with T cell entry and IL-1β production.

Figure 1

Spinal cords from at least four MOG-immunized mice in two separate experiments were stained for CXCL12 (red), CD31 (green), and nuclei (blue) and analyzed by confocal microscopy to evaluate CXCL12 relocation at naïve, preclinical (days 11-13), and peak (days 14-16) time-points during disease. Quantitation of CXCL12 polarity (loss of polarity=gray bars, retained polarity=black bars) in 132 vessels from 4 mice at peak of disease (a). Inflamed vessels were defined as those with >10 perivascular leukocytes. Leukocyte entry into the CNS at the preclinical time-point was determined by staining serial sections for T cells (CD3) and macrophages (CD11b). Note the entry of T cells (arrowhead and gray bar), but not CD11b cells, correlates with CXCL12 redistribution (b).

Gadolinium (Gd) enhancement does not occur during preclinical EAE

Among the possibilities for the mechanism of CXCL12 relocation would be nonspecific BBB disruption. One way to examine this in vivo, is via Gd-enhanced magnetic resonance imaging (MRI). Gd is a paramagnetic contrast agent that enters the CNS parenchyma when there is disruption of the BBB (28). Gd-MRI imaging is used in patients with neurologic diseases to detect inflammation and/or BBB compromise within the CNS (3). We performed Gd-MRI of spinal cords in unimmunzed mice and in mice at pre-clinical, peak, and chronic phases of EAE. Examination of Gd uptake revealed no enhancement in spinal cords of unimmunized mice or in mice at preclinical or chronic time-points after MOG immunization (Figure 2). These data suggest that CXCL12 relocalization detected during preclinical time-points is not due to BBB disruption but is instead likely to be occurring in response to the presence of molecules expressed by the myelin-specific T cells that initiate autoimmune inflammation.

Figure 2. Gd-MRI of mice with EAE.

Figure 2

Gd-MRI images of spinal cords of MOG-immunized mice during pre-onset (days 11-13), acute (days 14-28) and chronic (days 28-35) stages of EAE versus those obtained from an unimmunized (naïve) mouse (a). Change in signal intensity is observed in acute mouse only. Blood spinal cord barrier integrity was assessed as the percent change in signal intensity ([post – pre]/pre * 100) from spinal cord white matter at vertebral segment L3 and depicted as mean change in signal intensity ± SD for N=2-3 animals/group; *p<0.01 (b).

IL-1β induces CXCL12 relocation at the BBB

Activated T cells that traffic across the CNS endothelium have been shown to produce inflammatory cytokines such as TNF-α and IL-1β (29, 30), both of which have been observed to regulate CXCL12 expression in other contexts (9, 10, 31, 32). To determine if these cytokines play a role in the relocation of CXCL12 at the BBB, we administered IL-1β, TNF-α, and PBS intravenously to wild-type mice. Spinal cords were collected at 3, 6, and 24 hours post-treatment and analyzed by immunohistochemical staining. At 3 hours post-treatment CXCL12 remained expressed at the basolateral surface of the endothelium in all treatment groups. By 6 hours post-treatment there was a decrease in basolateral CXCL12 staining in the mice given IL-1β, without associated relocation to the lumen (data not shown). Twenty-four hours after cytokine administration, analysis revealed that IL-1β, but not TNF-α, induced CXCL12 relocation in 90.1% of vessels. The CXCL12 expression pattern induced by IL-1β was similar to that seen in vessels from MOG-immunized wild-type mice in the induction phase of EAE (Figure 3a). Administration of an equal volume of PBS did not lead to any change in the pattern of CXCL12 expression (Figure 3a). In addition, direct administration of TNF-α to the spinal cord via stereotactic injection did not induce alterations in the pattern of expression of CXCL12 at the BBB (data not shown).

Figure 3. IL-1β induces CXCL12 redistribution at the BBB.

Figure 3

CXCL12 relocation was determined by visualizing spinal cord sections double-labeled with CXCL12 (red) and CD31 (green) in mice administered cytokines (IL-1β, TNF-α, or PBS control) (a). Additionally, cytokine receptor (IL-1R, TNFR1, and wild-type control) deficient mice were immunized with MOGp35-55 and spinal cords were collected when the wild-type controls reached the peak of disease (b). Data shown is compiled from 11 to 13 images taken from 2 mice. Corresponding bar graphs depict the percentage of all vessels analyzed within a group for relocation of CXCL12, which is defined as lack of basolateral staining for CXCL12. Fluorescence intensity tracings depicting CXCL12 (red) location relative to CD31 (green) are shown below the corresponding image and the data is collected at the area of the transecting white arrow.

We confirmed that TNF-α was not required for CXCL12 relocation by examining MOG-immunized mice deficient in TNFR1, which were previously reported to be resistant to EAE (33), at the time-point where their wild-type controls reached the peak of disease. Immunostaining of spinal cord sections from TNFR1 deficient mice showed that basolateral expression of CXCL12 was lost in 92.3% of vessels analyzed while 100% of the vessels analyzed in immunized IL-1R deficient mice, which are also protected from EAE (21), had retained the basolateral expression (Figure 3b). Although TNFR1-deficient mice exhibited relocation of CXCL12 such that expression was no longer polarized, the pattern of CXCL12 staining in these mice was not identical to that observed at the peak of EAE in wild-type mice with increased co-localization with CD31 (Figure 3b). This suggests that although TNF-α is not required for CXCL12 relocation, it may be required for the establishment of the appropriate binding sites for this chemokine during the relocation process. Thus, while lack of basolateral CXCL12 expression is not sufficient to induce disease, there may be additional CXCL12 binding sites regulated by TNF-α that allow relocation of CXCL12 to sites that impact on expression of EAE.

IL-17+ γδT cells produce IL-1β during EAE

IL-1β is produced by macrophages and activated microglia (34, 35); however CXCL12 relocation occurs during the induction of EAE when initial T cell entry occurs and macrophages and activated microglia are yet not detected. To determine if T cells are the source of IL-1β, we performed extensive analyses of IL-1β production during the induction of EAE. In spinal cord sections selected from mice at the preclinical time-point, double-label immunofluorescent staining revealed that a subset of T cells was IL-1β positive (Figure 4a). Cells were isolated from the spinal cords of MOG-immunized mice at the first sign of EAE for flow cytometric analysis. Onset of disease was chosen as the earliest time-point at which adequate numbers of cells for analysis can be isolated from the spinal cord. Flow cytometric analysis of cells recovered from the spinal cord of EAE mice also showed that amongst the CD3+ T cell population, an average of 67.9% of cells were positive for IL-1β (Figure 4b). Flow cytometric analysis of CD3+ lymphocytes showed that 72.2% to 94.2% of cells in the CD8+, CD4+, and γδ+ T cell populations were IL-1β+ (Figure 4c).

Figure 4. T cells produce IL-1β in the CNS during autoimmunity.

Figure 4

Cellular sources of IL-1β in spinal cords at the induction of EAE (day 11) were analyzed by immunofluorescent confocal microscopy of serial tissue sections with anti-IL-1β and anti-CD3 or anti-CD11b. White arrowheads indicate IL-1β-positive cells (a). Flow cytometric analysis of CD3+ T cells isolated from spinal cords derived from mice exhibiting early signs of EAE shows the percentage of IL-1β positive (y-axis), CD3 positive (x-axis) cells (b) and the percentage of CD8, CD4, and γδT cells that are IL-1β positive (c). Data is compiled from 3 to 5 independent experiments and include at between 10-15 mice; *p>0.05.

MOG-immunized TNFR1 deficient mice, in which CXCL12 is relocated, have relatively robust trafficking of T cells into the perivascular space, given the absence of clinical disease, with entry of few or no CD11b+ or GR1+ cells (36). Thus, the use of these mice was ideal for determining which IL-1β-producing T cell subtype was associated with CXCL12 relocation. Cells were isolated from the spinal cords of MOG-immunized TNFR deficient mice when their wild-type controls began to show signs of disease and analyzed by flow cytometry. Almost 50% of CD3+ T cells expressed IL-1β and an analysis of the T cell subsets showed that 77% of T cells isolated were γδT cells (Figure 5a). Depletion of γδT cells has been shown to decrease the levels of IL-1β at the induction of EAE (37) and IL-17+ γδT cells have been implicated in the induction of autoimmune joint inflammation (38). We therefore further analyzed the γδT cell population isolated from wild-type mice with EAE and found a population of γδT cells that produced significantly more IL-17 than the γδ- T cells. An average of 95.1% of these γδ IL-17+ cells also expressed IL-1β (Figure 5b). In accordance with previously published data suggesting a role for IL-1β in the promotion of IL-17 during CNS autoimmunity, no IL-17 mRNA was detectable in the spinal cords recovered from MOG-immunized IL-1R deficient mice (Figure 5c).

Figure 5. IL-17-expressing γδT cells produce IL-1β.

Figure 5

Expression of IL-1β was analyzed in the CNS of MOG-immunized TNFR1 deficient mice using Q-RT-PCR (left panel) and flow cytometry (middle and right panel). Tissues were obtained at time-points when MOG-immunized wild-type controls exhibited EAE. Data are compiled from 2 experiments with 2-6 mice per group (a). Gamma-delta T cells were isolated from wild-type EAE mice and analyzed for IL-17 production by intracellular staining and flow cytometric analysis (dot plot) and the mean fluorescent intensity of IL-17 staining amongst γδT cells versus others was determined (middle bar graph). IL-1β is produced by an average of 95.1% of IL-17+ cells as measured by intercellular staining and flow cytometry. Data are compiled from 3 to 5 experiments with 10-15 mice; * indicates p>0.05 (b). Levels of IL-17 mRNA as determined by QPCR in wild-type and IL-1R deficient mice indicate that no IL-17 is present in the spinal cords of IL-1R deficient mice. Data from 2 independent experiments, with 10 mice, **p>0.005 (c).

CNS levels of IL-1R determine the clinical severity of EAE and the extent of CXCL12 relocation

While both IL-1 (22) and IL-1R (21) deficient mice are resistant to EAE induced by MOG-immunization, adoptive transfer of wild-type myelin-specific T cells into IL-1R deficient mice was not protective from EAE (21), suggesting a requirement of IL-1R within the adoptively transferred T cell population. In order to address the T cell role of IL-1R on CXCL12 relocation, CXCL12 expression patterns were evaluated in the spinal cords of mice with EAE induced by adoptive transfer of wild-type or IL-R deficient T cells derived from MOG-immunized mice into wild-type or IL-1R deficient recipients (Figure 6a). Spinal cords were harvested at the onset of disease and, of the inflamed vessels, 73% exhibited CXCL12 relocation in wild-type mice adoptively transferred with wild-type MOG-specific T cells (incidence 80%), while 51% had CXCL12 relocation in IL-1R-deficient mice adoptively transferred with wild-type MOG-specific T cells (incidence 57%) and only 20% of inflamed vessels of wild-type mice adoptively transferred with IL-1R-deficient, MOG-specific T cells (incidence 33%) exhibited relocation (Figure 6a). These data suggest that expression of IL-1R in recipient mice determines the extent of CXCL12 relocation. Because endogenous T cells also participate in the induction of adoptive transfer EAE (39), we wished to more definitively determine if the effect of IL-1R deficiency on disease severity and CXCL12 relocation was localized to the leukocyte population or the CNS. Thus, we also generated bone marrow chimeras using IL-1R deficient and wild-type mice. Chimeric mice lacking the IL-1R and reconstituted with wild-type bone marrow or vice-versa were both susceptible to EAE, although the group of IL-1R deficient mice receiving wild-type bone marrow exhibited a significant decrease in severity of disease compared to either wild-type mice receiving IL-1R deficient bone marrow or wild-type into wild-type controls (Figure 6b). Additionally, chimeras with IL-1R deficiency on irradiation-protected CNS resident cells had a reduced mean-maximal disease score compared to wild-type into wild-type controls and a delayed onset compared to both other disease groups (Table 1). As expected, IL-1R deficient into IL-1R deficient controls exhibited minimal clinical disease that was significantly decreased compared with all groups and a delay in onset compared with the wild-type into wild-type group. As in the adoptive transfer experiments, we evaluated the CXCL12 expression within spinal cord tissues of both types of chimera after MOG immunization at the first signs of clinical disease and obtained similar results (Figure 6c). Of the inflamed vessels, 42% had CXCL12 relocation in IL-1R-deficient mice reconstituted with wild-type bone marrow whereas only 17% of inflamed vessels of wild-type mice reconstituted with IL-1R-deficient bone marrow exhibited relocation. Identical staining protocols using spinal cord tissue collected from naïve IL-1R deficient mice showed that 100% of vessels analyzed displayed normal, basolateral expression of CXCL12 indicating that IL-1R deficiency does not affect the pattern of CXCL12 expression in the absence of EAE (data not shown). Although the immunohistochemical data suggest that IL-1R expression on recipient CNS cells, including the endothelial cells, determine the severity of EAE and the extent of CXCL12 relocation, the clinical disease severity data also indicate a role for IL-1R expression on infiltrating hematopoietic cells. Flow cytometric analysis of cells isolated from the spinal cords of mice with EAE indicate that the percentage of γδ T cells expressing IL-1R (83.3%) is significantly higher than the percentages of CD8 (21.7%) and CD4 (11.2%) cells expressing the IL-1R (Figure 6d).

Figure 6. IL-1R deficiency on CNS cells lessens EAE.

Figure 6

Immunohistochemical analysis of inflamed venules within spinal cord sections collected from adoptively transferred mice revealed a 2.5-fold increase in relocation of CXCL12 in wild-type mice adoptively transferred with MOG-specific, IL-1R deficient T cells (gray bar) when compared with the reverse adoptive transfer paradigm (black bar). Adoptive transfer of wild-type MOG-specific T cells into wild-type animals served as a positive control for the maximum relocation possible (white bar). Data are representative of two experiments with 3-5 mice/group (a). Bone marrow chimeric mice were immunized with MOGp35-55 to induce EAE and scored daily for clinical signs of disease. Irradiated IL-1R-deficient mice engrafted with wild-type bone marrow (squares) had less severe disease compared to wild-type mice engrafted with IL-1R deficient bone marrow (inverted triangles) and wild-type control (triangles). Irradiated IL-1R-deficient mice engrafted with IL-1R-deficient bone marrow (diamonds) exhibited the lease severe disease. Graph depicts the average clinical scores of 8-24 mice per group in up to three separate experimental groups (b). Immunohistochemical analysis of inflamed venules within spinal cord sections collected from bone marrow chimeric mice revealed a 2.5-fold increase in relocation of CXCL12 in wild-type mice engrafted with IL-1R deficient bone marrow (gray bar) compared with the reverse chimera (black bar) (c). Flow cytometric analysis of T cell subsets recovered from the spinal cords of MOG-immunized wild-type mice and stained for the IL-1R. Data is compiled from 4 to 6 experiments with between 10-15 mice; **p>0.005 and ***p>0.0005 (d).

Table I.

EAE in bone marrow chimera generated with IL-1R-deficient mice.

incidence mean max disease score standard error p-value compared to WT into IL-1R average days until onset standard error p-value compared to WT into IL-1R
WT into IL-1R 23/24 2.58 0.25 16.14 0.55
IL-1R into WT 17/21 2.86 0.36 0.53 14.47 0.47 0.033 *
WT into WT 17/17 4.06 0.13 0.000051 *** 13.24 0.44 0.00036 ***
IL-1R into IL-1R 7/8 1.00 0.12 <0.003a 15.86 0.67 0.0038b **
a

p-value compared to all other groups

b

p-value compared to WT into WT

Discussion

IL-1β has historically been associated with the induction of autoimmune neuroinflammatory diseases but few studies have uncovered down-stream effects of IL-1R signaling at the BBB. Because prior studies have demonstrated that pathologic alteration in CXCL12 expression at the BBB occurs specifically during autoimmune diseases (8) and that IL-1β can regulate CXCL12 expression in neural cells (31), we hypothesized that the inflammatory cytokine IL-1β might play a role in regulating the relocation of CXCL12 at the CNS endothelium. Using EAE, a murine model of MS, we found that CXCL12 relocation occurs at early, preclinical time-points, prior to overt BBB disruption and is associated with the perivascular infiltration of IL-1β-expressing T cells that include a population of γδ T cells that also express IL-17. We also observed that IL-1β can directly induce relocation of CXCL12 at the BBB and that decreased levels of CNS IL-1R diminish the extent of this relocation and of clinical disease severity.

In most diseases and animal models, activated microglia and macrophages are considered to be the predominant source of IL-1β in the CNS (14, 40). Because of its unique regulation and inflammatory potential, IL-1β is usually considered to be an innate cytokine as initial tissue insult leads to activation and release of this potent molecule (41). Gamma-delta T cells participate in the innate arm of the immune response via recognition of non-peptide ligands (32, 33). These cells have been detected within EAE and MS lesions (42, 43) and numbers of activated γδT cells are elevated in the peripheral blood of MS patients compared to control patients (44). Depletion of γδ T cells in mice with EAE leads to a reduction in CNS IL-1β and decreased clinical disease severity (37, 42, 45). We directly measured levels of IL-1β in T cells derived from the CNS of mice with EAE and observed that the γδ T cell fraction expressed the highest levels of this cytokine. We also observed that the majority of T cells present in the CNS of MOG-immunized TNFR1 deficient mice, which exhibit loss of basolateral expression of CXCL12, are γδT cells. However, given that TNFR1-deficient mice are resistant to EAE (33, 36), the presence of IL-1β-expressing γδT cells and CXCL12 relocation may be necessary but insufficient to induce clinical disease. Because CXCL12 expression at the BBB of MOG-immunized TNFR1-deficient mice displayed increased co-localization with CD31, it is possible that TNF-α regulates only a subset of possible CXCL12 binding sites. For example, the in vitro attachment of CXCL12 to heparan sulphate proteoglycans on cultured rheumatoid arthritis endothelial cells was recently shown to be enhanced during exposure to inflammatory cytokines (46). It is therefore possible that multiple T cell-derived cytokines act in concert to promote the perivascular egress of leukocytes during induction of autoimmunity.

Prior studies indicated that CXCL12 relocation at the BBB correlates with disease severity in MS patients and may contribute to both leukocyte entry into the CNS and egress from perivascular spaces (6, 8). The loss of CXCL12 polarity at the induction of EAE, prior to the onset of BBB disruption and clinical disease suggests this phenomenon is a precursor to the development of lesions. Indeed, use of Gd-MRI demonstrated that the time-point of CXCL12 relocation does not correlate with detectable levels of Gd enhancement. Understanding the molecular mechanism of CXCL12 relocation might therefore reveal novel therapeutic targets for the prevention of leukocyte entry early during the course of an MS exacerbation. We observed a population of γδT cells producing relatively high levels of both IL-1β and IL-17. IL-17 has been detected in both inflammatory lesions (47) and CSF (48) of MS patients and has recently been shown to be important for the induction of EAE (49). In addition, adoptive transfer of myelin-specific CD4+ Th17 cells has been shown to induce the selective upregulation of the chemokines CXCL1 and CXCL2 within the spinal cords of recipient mice (50). CXCL1 and CXCL2 are ELR+ chemokines found in EAE lesions that are potent chemoattractants for PMN (51). In EAE, the expression of these molecules was shown to be essential for the early infiltration of PMN and induction of EAE. In our study, γδ T cells were found to be a source of IL-17 and could therefore also play a role in the recruitment of PMN and eventual BBB disruption that allow the infiltration of nonspecific leukocytes during clinical onset of EAE.

Although IL-1R-deficient mice were shown to be resistant to EAE (21), these studies did not identify cellular targets of IL-1β. Induction of EAE using chimeric mice suggest that IL-1R-expressing cells within the irradiation resistant CNS determine the severity of clinical disease. This is consistent with the reduction in disease seen in mice with over-expression of IL-1β antagonists within the CNS (52). Since IL-1β, which induces CXCL12 relocation, is produced by IL-17-producing γδ T cells, it is possible that these molecules interact to promote CNS inflammation, suggesting that IL-1β and its receptor might be targets for the treatment of MS. Further studies examining the effects of IL-1β and IL-17 on the expression of CXCL12 binding receptors are currently underway. As γδ T cells represent only a portion of the T cell population identified within the CNS of mice with early EAE, they may not be required for inducing CXCL12 relocation. Studies in γδ T cell-deficient mice and/or adoptive transfer experiments utilizing IL-1β-deficent γδ T cells, will be required to directly demonstrate that these cells mediate CXCL12 relocation via their production of IL-1β.

Studies have implicated a number of different molecules in the trafficking of leukocytes at the BBB including the interaction between endothelial CD40 and CD40L expressed on T cells. Lack of CD40 activations leads to decreased parenchymal penetration of T cells within the CNS during both EAE and encephalitis due to West Nile virus with extensive perivascular localization of T cells noted in both models (53-55). CD40: CD40L interactions have also been shown to regulate cell trafficking via CXCL12 expression during joint inflammation (12). These studies suggest that CD40: CD40L interactions could potentially contribute to CXCL12 expression patterns at the BBB. Although the current study does not directly address this possibility, administration of soluble CD40 ligand did not induce CXCL12 redistribution and analysis of CXCL12 expression within the spinal cords of MOG-immunized CD40-deficient mice did not reveal alteration in CXCL12 polarity at the BBB (all our unpublished data). Thus, it is unlikely that CD40 participates in the relocation of CXCL12 during EAE.

To date, very little is known about the mechanisms involving CXCL12 regulation within the CNS or about the molecules contributing to the relocalization of CXCL12 at the BBB, which appears to be an important feature of MS. This study indicates that the early entrance of IL-1β-expressing, autoreactive T cells leads to the pathologic relocation of CXCL12 and that disruption of IL-1R signaling on CNS cells may ameliorate the extent of autoimmune disease. While further studies are needed to determine the contribution of other molecules in the relocation of CXCL12 at the BBB, the current study suggests that IL-1β and the IL-1R could potentially be targets for the prevention of new lesions during MS.

Acknowledgements

The authors would like to thank Dr. John Russell for advice in experimental design, critical feedback, and gifts of reagents. Additional thanks to Julia Sim, Dawn Koch and Mickey Croyle for technical assistance.

References

  • 1.Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16:1–13. doi: 10.1016/j.nbd.2003.12.016. [DOI] [PubMed] [Google Scholar]
  • 2.Frohman EM, Racke MK, Raine CS. Multiple sclerosis--the plaque and its pathogenesis. N Engl J Med. 2006;354:942–955. doi: 10.1056/NEJMra052130. [DOI] [PubMed] [Google Scholar]
  • 3.Filippi M, Falini A, Arnold DL, Fazekas F, Gonen O, Simon JH, Dousset V, Savoiardo M, Wolinsky JS. Magnetic resonance techniques for the in vivo assessment of multiple sclerosis pathology: consensus report of the white matter study group. J Magn Reson Imaging. 2005;21:669–675. doi: 10.1002/jmri.20336. [DOI] [PubMed] [Google Scholar]
  • 4.Goodkin DE, Rooney WD, Sloan R, Bacchetti P, Gee L, Vermathen M, Waubant E, Abundo M, Majumdar S, Nelson S, Weiner MW. A serial study of new MS lesions and the white matter from which they arise. Neurology. 1998;51:1689–1697. doi: 10.1212/wnl.51.6.1689. [DOI] [PubMed] [Google Scholar]
  • 5.Stumm RK, Rummel J, Junker V, Culmsee C, Pfeiffer M, Krieglstein J, Hollt V, Schulz S. A dual role for the SDF-1/CXCR4 chemokine receptor system in adult brain: isoform-selective regulation of SDF-1 expression modulates CXCR4-dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci. 2002;22:5865–5878. doi: 10.1523/JNEUROSCI.22-14-05865.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.McCandless EE, Zhang B, Diamond MS, Klein RS. CXCR4 antagonism increases T cell trafficking in the central nervous system and improves survival from West Nile virus encephalitis. Proc Natl Acad Sci U S A. 2008;105:11270–11275. doi: 10.1073/pnas.0800898105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.McCandless EE, Wang Q, Woerner BM, Harper JM, Klein RS. CXCL12 limits inflammation by localizing mononuclear infiltrates to the perivascular space during experimental autoimmune encephalomyelitis. J Immunol. 2006;177:8053–8064. doi: 10.4049/jimmunol.177.11.8053. [DOI] [PubMed] [Google Scholar]
  • 8.McCandless EE, Piccio L, Woerner BM, Schmidt RE, Rubin JB, Cross AH, Klein RS. Pathologic expression of CXCL12 at the blood brain barrier correlates with severity of Multiple Sclerosis. Am J Pathol. 2008;172:799–808. doi: 10.2353/ajpath.2008.070918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jung Y, Wang J, Schneider A, Sun YX, Koh-Paige AJ, Osman NI, McCauley LK, Taichman RS. Regulation of SDF-1 (CXCL12) production by osteoblasts; a possible mechanism for stem cell homing. Bone. 2006;38:497–508. doi: 10.1016/j.bone.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 10.Han Y, He T, Huang DR, Pardo CA, Ransohoff RM. TNF-alpha mediates SDF-1 alpha-induced NF-kappa B activation and cytotoxic effects in primary astrocytes. J Clin Invest. 2001;108:425–435. doi: 10.1172/JCI12629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fedyk ER, Jones D, Critchley HO, Phipps RP, Blieden TM, Springer TA. Expression of stromal-derived factor-1 is decreased by IL-1 and TNF and in dermal wound healing. J Immunol. 2001;166:5749–5754. doi: 10.4049/jimmunol.166.9.5749. [DOI] [PubMed] [Google Scholar]
  • 12.Kim KW, Cho ML, Kim HR, Ju JH, Park MK, Oh HJ, Kim JS, Park SH, Lee SH, Kim HY. Up-regulation of stromal cell-derived factor 1 (CXCL12) production in rheumatoid synovial fibroblasts through interactions with T lymphocytes: role of interleukin-17 and CD40L-CD40 interaction. Arthritis Rheum. 2007;56:1076–1086. doi: 10.1002/art.22439. [DOI] [PubMed] [Google Scholar]
  • 13.Peng H, Erdmann N, Whitney N, Dou H, Gorantla S, Gendelman HE, Ghorpade A, Zheng J. HIV-1-infected and/or immune activated macrophages regulate astrocyte SDF-1 production through IL-1beta. Glia. 2006;54:619–629. doi: 10.1002/glia.20409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gosselin D, Rivest S. Role of IL-1 and TNF in the brain: twenty years of progress on a Dr. Jekyll/Mr. Hyde duality of the innate immune system. Brain Behav Immun. 2007;21:281–289. doi: 10.1016/j.bbi.2006.12.004. [DOI] [PubMed] [Google Scholar]
  • 15.Cannella B, Raine CS. The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol. 1995;37:424–435. doi: 10.1002/ana.410370404. [DOI] [PubMed] [Google Scholar]
  • 16.de Jong BA, Huizinga TW, Bollen EL, Uitdehaag BM, Bosma GP, van Buchem MA, Remarque EJ, Burgmans AC, Kalkers NF, Polman CH, Westendorp RG. Production of IL-1beta and IL-1Ra as risk factors for susceptibility and progression of relapse-onset multiple sclerosis. J Neuroimmunol. 2002;126:172–179. doi: 10.1016/s0165-5728(02)00056-5. [DOI] [PubMed] [Google Scholar]
  • 17.Argaw AT, Zhang Y, Snyder BJ, Zhao ML, Kopp N, Lee SC, Raine CS, Brosnan CF, John GR. IL-1beta regulates blood-brain barrier permeability via reactivation of the hypoxia-angiogenesis program. J Immunol. 2006;177:5574–5584. doi: 10.4049/jimmunol.177.8.5574. [DOI] [PubMed] [Google Scholar]
  • 18.Shaftel SS, Carlson TJ, Olschowka JA, Kyrkanides S, Matousek SB, O'Banion MK. Chronic interleukin-1beta expression in mouse brain leads to leukocyte infiltration and neutrophil-independent blood brain barrier permeability without overt neurodegeneration. J Neurosci. 2007;27:9301–9309. doi: 10.1523/JNEUROSCI.1418-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Konsman JP, Vigues S, Mackerlova L, Bristow A, Blomqvist A. Rat brain vascular distribution of interleukin-1 type-1 receptor immunoreactivity: relationship to patterns of inducible cyclooxygenase expression by peripheral inflammatory stimuli. J Comp Neurol. 2004;472:113–129. doi: 10.1002/cne.20052. [DOI] [PubMed] [Google Scholar]
  • 20.Srinivasan D, Yen JH, Joseph DJ, Friedman W. Cell type-specific interleukin-1beta signaling in the CNS. J Neurosci. 2004;24:6482–6488. doi: 10.1523/JNEUROSCI.5712-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sutton C, Brereton C, Keogh B, Mills KH, Lavelle EC. A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J Exp Med. 2006;203:1685–1691. doi: 10.1084/jem.20060285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Matsuki T, Nakae S, Sudo K, Horai R, Iwakura Y. Abnormal T cell activation caused by the imbalance of the IL-1/IL-1R antagonist system is responsible for the development of experimental autoimmune encephalomyelitis. Int Immunol. 2006;18:399–407. doi: 10.1093/intimm/dxh379. [DOI] [PubMed] [Google Scholar]
  • 23.Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8:942–949. doi: 10.1038/ni1496. [DOI] [PubMed] [Google Scholar]
  • 24.Archambault AS, Sim J, McCandless EE, Klein RS, Russell JH. Region-specific regulation of inflammation and pathogenesis in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2006;181:122–132. doi: 10.1016/j.jneuroim.2006.08.012. [DOI] [PubMed] [Google Scholar]
  • 25.Budde MD, Kim JH, Liang HF, Schmidt RE, Russell JH, Cross AH, Song SK. Toward accurate diagnosis of white matter pathology using diffusion tensor imaging. Magn Reson Med. 2007;57:688–695. doi: 10.1002/mrm.21200. [DOI] [PubMed] [Google Scholar]
  • 26.Prat A, Antel J. Pathogenesis of multiple sclerosis. Curr Opin Neurol. 2005;18:225–230. doi: 10.1097/01.wco.0000169737.99040.31. [DOI] [PubMed] [Google Scholar]
  • 27.Swanborg RH. Experimental autoimmune encephalomyelitis in the rat: lessons in T-cell immunology and autoreactivity. Immunol Rev. 2001;184:129–135. doi: 10.1034/j.1600-065x.2001.1840112.x. [DOI] [PubMed] [Google Scholar]
  • 28.Brasch RC, Weinmann HJ, Wesbey GE. Contrast-enhanced NMR imaging: animal studies using gadolinium-DTPA complex. AJR Am J Roentgenol. 1984;142:625–630. doi: 10.2214/ajr.142.3.625. [DOI] [PubMed] [Google Scholar]
  • 29.Ruddle NH, Bergman CM, McGrath KM, Lingenheld EG, Grunnet ML, Padula SJ, Clark RB. An antibody to lymphotoxin and tumor necrosis factor prevents transfer of experimental allergic encephalomyelitis. J Exp Med. 1990;172:1193–1200. doi: 10.1084/jem.172.4.1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Okuda Y, Sakoda S, Yanagihara T. The pattern of cytokine gene expression in lymphoid organs and peripheral blood mononuclear cells of mice with experimental allergic encephalomyelitis. J Neuroimmunol. 1998;87:147–155. doi: 10.1016/s0165-5728(98)00070-8. [DOI] [PubMed] [Google Scholar]
  • 31.Calderon TM, Eugenin EA, Lopez L, Kumar SS, Hesselgesser J, Raine CS, Berman JW. A role for CXCL12 (SDF-1alpha) in the pathogenesis of multiple sclerosis: regulation of CXCL12 expression in astrocytes by soluble myelin basic protein. J Neuroimmunol. 2006;177:27–39. doi: 10.1016/j.jneuroim.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 32.Ueda Y, Yang K, Foster SJ, Kondo M, Kelsoe G. Inflammation controls B lymphopoiesis by regulating chemokine CXCL12 expression. J Exp Med. 2004;199:47–58. doi: 10.1084/jem.20031104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Probert L, Eugster HP, Akassoglou K, Bauer J, Frei K, Lassmann H, Fontana A. TNFR1 signalling is critical for the development of demyelination and the limitation of T-cell responses during immune-mediated CNS disease. Brain. 2000;123(Pt 10):2005–2019. doi: 10.1093/brain/123.10.2005. [DOI] [PubMed] [Google Scholar]
  • 34.Takeshita S, Takeshita F, Haddad DE, Janabi N, Klinman DM. Activation of microglia and astrocytes by CpG oligodeoxynucleotides. Neuroreport. 2001;12:3029–3032. doi: 10.1097/00001756-200110080-00010. [DOI] [PubMed] [Google Scholar]
  • 35.Beuscher HU, Gunther C, Rollinghoff M. IL-1 beta is secreted by activated murine macrophages as biologically inactive precursor. J Immunol. 1990;144:2179–2183. [PubMed] [Google Scholar]
  • 36.Gimenez MA, Sim JE, Russell JH. TNFR1-dependent VCAM-1 expression by astrocytes exposes the CNS to destructive inflammation. J Neuroimmunol. 2004;151:116–125. doi: 10.1016/j.jneuroim.2004.02.012. [DOI] [PubMed] [Google Scholar]
  • 37.Olive C. Modulation of experimental allergic encephalomyelitis in mice by immunization with a peptide specific for the gamma delta T cell receptor. Immunol Cell Biol. 1997;75:102–106. doi: 10.1038/icb.1997.14. [DOI] [PubMed] [Google Scholar]
  • 38.Roark CL, French JD, Taylor MA, Bendele AM, Born WK, O'Brien RL. Exacerbation of collagen-induced arthritis by oligoclonal, IL-17-producing gamma delta T cells. J Immunol. 2007;179:5576–5583. doi: 10.4049/jimmunol.179.8.5576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lees JR, Iwakura Y, Russell JH. Host T cells are the main producers of IL-17 within the central nervous system during initiation of experimental autoimmune encephalomyelitis induced by adoptive transfer of Th1 cell lines. J Immunol. 2008;180:8066–8072. [Google Scholar]
  • 40.McFarland HF, Martin R. Multiple sclerosis: a complicated picture of autoimmunity. Nat Immunol. 2007;8:913–919. doi: 10.1038/ni1507. [DOI] [PubMed] [Google Scholar]
  • 41.Dinarello CA. Interleukin-1. Cytokine Growth Factor Rev. 1997;8:253–265. doi: 10.1016/s1359-6101(97)00023-3. [DOI] [PubMed] [Google Scholar]
  • 42.Rajan AJ, Gao YL, Raine CS, Brosnan CF. A pathogenic role for gamma delta T cells in relapsing-remitting experimental allergic encephalomyelitis in the SJL mouse. J Immunol. 1996;157:941–949. [PubMed] [Google Scholar]
  • 43.Selmaj K, Brosnan CF, Raine CS. Colocalization of lymphocytes bearing gamma delta T-cell receptor and heat shock protein hsp65+ oligodendrocytes in multiple sclerosis. Proc Natl Acad Sci U S A. 1991;88:6452–6456. doi: 10.1073/pnas.88.15.6452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chen Z, Freedman MS. Correlation of specialized CD16(+) gammadelta T cells with disease course and severity in multiple sclerosis. J Neuroimmunol. 2007 doi: 10.1016/j.jneuroim.2007.11.010. [DOI] [PubMed] [Google Scholar]
  • 45.Rajan AJ, Klein JD, Brosnan CF. The effect of gammadelta T cell depletion on cytokine gene expression in experimental allergic encephalomyelitis. J Immunol. 1998;160:5955–5962. [PubMed] [Google Scholar]
  • 46.Santiago B, Baleux F, Palao G, Gutierrez-Canas I, Ramirez JC, Arenzana-Seisdedos F, Pablos JL. CXCL12 is displayed by rheumatoid endothelial cells through its basic amino-terminal motif on heparan sulfate proteoglycans. Arthritis Res Ther. 2006;8:R43. doi: 10.1186/ar1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M, Giuliani F, Arbour N, Becher B, Prat A. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med. 2007;13:1173–1175. doi: 10.1038/nm1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Matusevicius D, Kivisakk P, He B, Kostulas N, Ozenci V, Fredrikson S, Link H. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler. 1999;5:101–104. doi: 10.1177/135245859900500206. [DOI] [PubMed] [Google Scholar]
  • 49.Komiyama Y, Nakae S, Matsuki T, Nambu A, Ishigame H, Kakuta S, Sudo K, Iwakura Y. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol. 2006;177:566–573. doi: 10.4049/jimmunol.177.1.566. [DOI] [PubMed] [Google Scholar]
  • 50.Carlson T, Kroenke M, Rao P, Lane TE, Segal B. The Th17-ELR+ CXC chemokine pathway is essential for the development of central nervous system autoimmune disease. J Exp Med. 2008;205:811–823. doi: 10.1084/jem.20072404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Glabinski AR, Tuohy VK, Ransohoff RM. Expression of chemokines RANTES, MIP-1alpha and GRO-alpha correlates with inflammation in acute experimental autoimmune encephalomyelitis. Neuroimmunomodulation. 1998;5:166–171. doi: 10.1159/000026333. [DOI] [PubMed] [Google Scholar]
  • 52.Furlan R, Bergami A, Brambilla E, Butti E, De Simoni MG, Campagnoli M, Marconi P, Comi G, Martino G. HSV-1-mediated IL-1 receptor antagonist gene therapy ameliorates MOG(35-55)-induced experimental autoimmune encephalomyelitis in C57BL/6 mice. Gene Ther. 2007;14:93–98. doi: 10.1038/sj.gt.3302805. [DOI] [PubMed] [Google Scholar]
  • 53.Gerritse K, Laman JD, Noelle RJ, Aruffo A, Ledbetter JA, Boersma WJ, Claassen E. CD40-CD40 ligand interactions in experimental allergic encephalomyelitis and multiple sclerosis. Proc Natl Acad Sci U S A. 1996;93:2499–2504. doi: 10.1073/pnas.93.6.2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Grewal IS, Foellmer HG, Grewal KD, Xu J, Hardardottir F, Baron JL, Janeway CA, Jr., Flavell RA. Requirement for CD40 ligand in costimulation induction, T cell activation, and experimental allergic encephalomyelitis. Science. 1996;273:1864–1867. doi: 10.1126/science.273.5283.1864. [DOI] [PubMed] [Google Scholar]
  • 55.Sitati E, McCandless EE, Klein RS, Diamond MS. CD40-CD40 Ligand Interactions Promote Trafficking of CD8+ T Cells into the Brain and Protection against West Nile Virus Encephalitis. J Virol. 2007;81:9801–9811. doi: 10.1128/JVI.00941-07. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES