Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

IFN-β Inhibits T Cell Activation Capacity of Central Nervous System APCs

Ingrid Teige, Yawei Liu and Shohreh Issazadeh-Navikas
J Immunol September 15, 2006, 177 (6) 3542-3553; DOI: https://doi.org/10.4049/jimmunol.177.6.3542
Ingrid Teige
Neuroinflammation Unit, Section for Immunology, Institute for Experimental Medical Science, Lund University, Lund, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yawei Liu
Neuroinflammation Unit, Section for Immunology, Institute for Experimental Medical Science, Lund University, Lund, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shohreh Issazadeh-Navikas
Neuroinflammation Unit, Section for Immunology, Institute for Experimental Medical Science, Lund University, Lund, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

We have previously investigated the physiological effects of IFN-β on chronic CNS inflammation and shown that IFN-β−/− mice develop a more severe experimental autoimmune encephalomyelitis than their IFN-β+/− littermates. This result was shown to be associated with a higher activation state of the glial cells and a higher T cell cytokine production in the CNS. Because this state suggested a down-regulatory effect of IFN-β on CNS-specific APCs, these results were investigated further. We report that IFN-β pretreatment of astrocytes and microglia (glial cells) indeed down-modulate their capacity to activate autoreactive Th1 cells. First, we investigated the intrinsic ability of glial cells as APCs and report that glial cells prevent autoreactive Th1 cells expansion while maintaining Ag-specific T cell effector functions. However, when the glial cells are treated with IFN-β before coculture with T cells, the effector functions of T cells are impaired as IFN-γ, TNF-α, and NO productions are decreased. Induction of the T cell activation marker, CD25 is also reduced. This suppression of T cell response is cell-cell dependent, but it is not dependent on a decrease in glial expression of MHC class II or costimulatory molecules. We propose that IFN-β might exert its beneficial effects mainly by reducing the Ag-presenting capacity of CNS-specific APCs, which in turn inhibits the effector functions of encephalitogenic T cells. This affect is of importance because activation of encephalitogenic T cells within the CNS is a prerequisite for the development of a chronic progressive CNS inflammation.

The type I IFN group member IFN-β is mainly produced by fibroblasts, macrophages, and dendritic cells in response to viral infection, dsRNA, nonvertebrate unmethylated DNA containing CpG motifs, or LPS (1, 2, 3, 4, 5). IFN-β signaling through the shared IFN-α or -β receptor triggers a cascade of events affecting both cells of the immune system as well as other cell types because the receptor is widely expressed (6). IFN-β is one of the therapeutic agents commonly used for treatment of multiple sclerosis (MS),3 an inflammatory and demyelinating disease of the CNS (7, 8). There is a large amount of data available from a wide range of studies attempting to understand the basic mechanisms of action for IFN-β. Among those, high doses of IFN-β in T cell cultures have been shown to induce a Th1 to Th2 shift and to reduce T cell proliferation (9, 10, 11). In accordance, several reports indicate that a Th1-Th2 shift occur in circulating T cells of IFN-β-treated MS patients (12). It has also been reported that IFN-β treatment causes professional APCs to produce less IL-12 and more IL-10 that would favor Th2 responses over Th1 (13, 14, 15). Furthermore, it has been suggested that immune cells accessibility to the CNS could be reduced upon IFN-β treatment by several possibilities. Among them is a down-regulation of several adhesion molecules important in leukocyte migration across the blood-brain barrier (e.g., LFA-1 and VLA-4) (16, 17). Next, stabilizing functions of endothelial tight junctions on cultured cells have been demonstrated (18). In addition, the expression of matrix metalloproteinases has been shown to be inhibited by IFN-β (19). However, despite the efforts made to elucidate the subject, several reports concerning the effects of IFN-β on APCs (macrophages) and on T cells are contradictory, as both anti- and proinflammatory properties have been suggested. Regarding T cells, down-regulated T cell response and a shift in Th1-Th2 response reported by some have been challenged by others as it has been shown that IFN-β could induce a Th1 response (20, 21, 22). With reference to macrophages, IFN-β has been shown to both increase and decrease secretion of TNF-α, IL-1β, and the soluble decoy receptor IL-1Rα, depending on activation stimuli (23). Similarly, assaying PBMCs from treated MS patients show both up- and down-regulation of costimulatory molecules (24). This finding renders interpretation of data difficult and increases the demand for mimicking the in vivo situation more closely for a better understanding of the basic mechanisms by which IFN-β exerts its beneficial effects. Many of the results obtained from MS patients have been achieved through studies of cells isolated from blood samples, although IFN-β could have profound effects on a number of different cell types considering the broad expression of the IFN-α or -β receptor (6). It is presumable that IFN-β could enter the CSF and hence access the CNS tissues due to leakage across blood brain barrier caused by increased permeability during the course of disease. In addition, we have previously shown that IFN-β is indeed endogenously produced within the CNS during experimental autoimmune encephalomyelitis (EAE) inflammation and could thus affect resident cells such as potential CNS APCs (25). So far, relatively little is known regarding the effects of IFN-β on glial cells and their relative role in regulation of autoreactive T cells at the site of inflammation. Only a handful of studies addressing this issue are available that report IFN-β inhibits glial cell expression of matrix metalloproteinases 9 and 7 (26). Furthermore, IFN-β is able to reduce MHC class II expression induced by IFN-γ on microglial cells and results in lower IL-12 production by microglia induced by LPS and IFN-γ, in a similar manner to the APCs of lymphoid compartments (27).

In searching for a way to address the physiological mechanisms by which IFN-β could regulate immune response and the resulting CNS inflammation, we used IFN-β gene-deleted (IFN-β −/−) mice and EAE, the commonly used animal model for MS (25). The study revealed an augmented and chronic EAE with increased incidence in IFN-β−/− mice. This finding was associated with a significantly elevated number of activated glial cells and higher in vivo T cell cytokine production in the CNS of IFN-β−/− mice compared with their wild-type (IFN-β+/−) littermates. However, autoantigen-specific T cells isolated from peripheral lymphoid organs were similar in their production of cytokines and displayed equal encephalitogenic capacity as shown by adoptive transfer studies. These results suggested an increased activation of the encephalitogenic T cells in situ in IFN-β−/− mice compared with IFN-β+/−, resulting in an augmentation of the EAE. Hence, we have now addressed the effects of IFN-β on the Ag-presenting capacity of astroglia and microglia and report that treatment of glial cells with IFN-β alone reduces their capacity to both induce and sustain an Ag-specific T cell cytokine production. Our data indicate that one of the basic mechanisms by which IFN-β might exert its beneficial effects could be due to its ability to down-modulate the Ag-presenting capacity of CNS glial cells, resulting in a less efficient reactivation of autoreactive T cells. Secondly, IFN-β suppresses the Ag-presenting capacity of the glial cells to maintain effector functions of activated T cells.

Materials and Methods

Mice

All mice used for cell cultures were of the B10.RIII strain. For EAE experiments, IFN-β−/− mice generated as described previously (28) were backcrossed for 12 generations to the B10.RIII strain, and these mice were compared with their IFN-β+/+ littermates. All mice were bred and kept at the conventional animal facility at Lund University (Lund, Sweden). All experiments were performed in accordance with the ethical committee in Malmö-Lund, Sweden.

Glial cell cultures

Primary mixed glial cell cultures were established from the forebrain and cerebellum of 1- to 2-day-old B10.RIII mice. The tissues were carefully dissected out and freed of meninges before being placed in HBSS solution supplemented with 1 mM pyruvate and 11 mM glucose. Tissue was then chopped into smaller pieces using a razor blade and incubated with 1% trypsin (Sigma-Aldrich) for 10 min at 37°C. Thereafter, DNase and FCS were added to a final concentration of 0.12 and 0.5%, respectively, and incubated for 8 min. Finally the solution was mechanically dissociated to single cell levels using a fire-constricted Pasteur pipette. The cells were seeded at a concentration of 1.5 × 105 cells/cm2 in a 1:1 mix of DMEM and F12 medium (Invitrogen Life Technologies) supplemented with 0.16 μM/ml penicillin, 0.03 μM/ml streptomycin, and 5% FCS. Medium was changed every third to fourth day. After 7–10 days in culture, oligodendrocytes were detached from the cell monolayer by shaking on an orbital shaker. For expansion, the cells were split using a trypsin and EDTA solution (Sigma-Aldrich) upon reaching confluence. The cells were used for T cell coculture after two to three passages and consisted of a mixture of 20–30% Mac-1+ microglia and 70–80% glial fibrillary acidic protein (GFAP+) astrocytes. Pure microglial and astrocytic cultures were established as described (29). Briefly, after 8 days in culture, the mixed glial cells were vigorously shaken at 900-1000 rpm for 3 h on an orbital shaker. For microglial cultures, the floating cells were collected, washed, and reseeded in 96-well plates (Nunc) at a concentration of 1 × 104 cells/well. After adhering overnight, nonadherent or loosely attached cells were washed away. The adherent cells represented >97% pure microglial cells as determined by Mac-1+ staining, with <3% being GFAP+. For astrocyte cultures, the still adherent cells were trypsinized and reseeded in flasks and left to adhere for 30 min. Floating or loosely attached cells were recovered by mild shaking by hand and the adhesion process was repeated, this time for 1 h. Cells in the supernatant were thereafter collected, washed, and reseeded in 96-well plates (Nunc) at a concentration of 4 × 104 cells/well. These cells were >96% pure astrocytes as determined by GFAP+ staining, <4% were Mac-1+ cells. Both these cultures were used within 24–48 h after establishment for coculture with T cells.

Establishment of myelin-specific T cell lines

Two T cell lines were generated by immunizing three 8- to 12-wk-old male mice in the hind paws and tail base with a total of 200 μl of a 1:1 emulsion of 250 μg of either myelin basic protein (MBP)89–101 or myelin oligodendrocyte glycoprotein peptide 35–55 in PBS and CFA containing 100 μg of Mycobacterium tuberculosis H37Ra (Difco). Draining lymph nodes were collected day 10 postimmunization and the cells went through several rounds of Ag-specific restimulation as described (30). This generated T cell lines of the Th1 phenotype that produced large amounts of IFN-γ and IL-2 and very low amounts of IL-4 upon Ag stimulation. In the experiments in our study, both MBP89–101-specific and myelin oligodendrocyte glycoprotein peptide (35–55)-specific T cell lines have been used with very similar results. Hence, no distinction will be made between them in the following sections. In all experiments, the T cell lines had been subjected to from four to eight stimulation rounds.

Coculture of T cells and glial cells

After the last trypsinization, glial cells were seeded in 96-well plates (Nunc) at 4–5 × 104 cells/well. After 24–48 h of adherence time, the cells were either left untreated or treated for 24 h with 100 U/ml IFN-β or IFN-γ. The cultures were then washed three times and T cells were added in a 1:1 ratio. The T cell lines were established as previously discussed, and were, in a first set of experiments, in a resting state (kept in medium supplemented with IL-2 for a minimum of 2 wk) when added to the differently treated glial cultures with or without the addition of Ag. These cells were referred to as resting T cells. The concentration of the antigenic peptides was 20 μg/ml if not stated otherwise. These cocultures were pulsed with [3H]thymidine after 48 h and harvested after 24 h of further incubation on glass fiber filters. [3H]Thymidine incorporation was measured in a beta scintillation counter (Matrix 96 Direct Beta counter; Packard Instrument). Just before harvesting, supernatants were collected for cytokine analysis. In parallel, some T cells were recollected after a total of 72 h in culture, stained for different activation markers, and analyzed in a four-color BD FACSort (BD Biosciences).

In a second set of experiments, the T cells were first activated for 48 h with splenic APCs as described. They are referred to as activated T cells, which were washed thoroughly to remove Ag before being added to untreated or IFN-β- or IFN-γ-pretreated glial cells. These cultures were [3H]thymidine pulsed after 24 h in glial cocultures and harvested after 24 h of further incubation. As before, supernatants were collected and FACS staining experiments were performed in parallel.

Transwell cocultures of T cells and glial cells

Mixed glial cultures were seeded at 2.5 × 105 cells/well in 24-well plates (Nunc). After 24–48 h of adherence, 2.5 × 105 T cells activated for 48 h with splenic APCs (activated T cells) were added to the cultures in cell culture inserts (Transwells) with a pore size of 0.4 μm (BD Falcon). This method allowed soluble molecules, but not cells, to pass between the glia surface and the Transwell insert. As controls, glia and T cells plated at the same density were cultured in 24-well plates without inserts, thus allowing cell-cell contact. After 24 h in coculture, the cells were [3H]thymidine pulsed and after an additional 24 h of incubation, the cells were harvested and supernatants were collected.

Supernatant-mediated inhibition of T cell proliferation

Resting T cells were cocultured with glial cells in cell-cell contact cultures with or without Ag (as described) and supernatants were collected under sterile conditions. To investigate whether the supernatants could inhibit T cell proliferation, resting T cells were activated with Ag and splenic APCs as described. This activation was performed in new medium or medium conditioned with different ratios of the harvested supernatants. The proliferation index was calculated as proliferation in cultures with conditioned medium divided by the control cultures containing 100% fresh medium.

FACS staining and evaluation

After 24 h of treatment with IFN-γ, IFN-β, or a combination of these two cytokines (100 U/ml IFN-γ for 12 h after which 100 U/ml IFN-β was added for the remaining 12 h), glial cell cultures were washed once with PBS and detached and dissociated using a trypsin and EDTA solution (Sigma-Aldrich). Activated or resting T cells were after 48 or 72 h in coculture, respectively, collected through vigorous flushing with the pipette. After washing in FACS buffer (2% FCS in PBS, for FACS of glial cells this buffer also contained 1 mM EDTA), cells were first incubated with anti-Fc receptor Ab (24.G.2, our hybridoma collection) at 10 μg/ml. Thereafter, glial cells were incubated with biotinylated FITC-marked or PE-marked Abs against Mac-1 (M1/70), MHC class II (Y3P or 7.16.17), CD1d (1B1), B7-1 (16-10A1), B7-2 (GL1), CD40 (3/23), ICAM-1 (3E2), VCAM-1 (429), Fas ligand (Kay-10), TGF-β1 (A75-3.1), and GFAP (Zymed Laboratories). For astrocytic GFAP staining and intracellular TGF-β1 staining, cells were first fixed using 3% paraformaldehyde and thereafter permeabilized using 0.3% saponin in FACS buffer. T cells were stained with biotinylated FITC- or PE-marked Ab against CD4 (Gk1.5), TCR αβ (H57-597), CD28 (3751), CTLA-4 (UC10), CD40L (MR1), CD25 (7D4), CD69 (H1.2F3), and CD44 (Caltag Laboratories). After washing, cells were incubated with streptavidin-PE or allophycocyanin diluted 1/400 (BD Pharmingen). Annexin V and propidium iodide (BD Pharmingen) staining was used to detect apoptotic and dead cells. In both types of staining, all Abs were allowed to bind for 20 min on ice. All Abs were used at a concentration of 1–5 μg/ml and were purchased from BD Pharmingen unless stated otherwise. The cells were analyzed using a four-color FACSort (BD Biosciences). Positivity was evaluated by comparison of negative samples stained with irrelevant isotype-matched controls and streptavidin-PE or allophycocyanin. Astrocytes were gated on GFAP, microglia on Mac-1, and T cells on CD4 and TCR expression. For FACS-based proliferation studies, T cells were CFSE labeled at a concentration of 5 nM before addition to glial cells. Cells were collected after 72 h and counterstained with allophycocyanin-labeled anti-CD4 (Gk1.5). T cells labeled just before FACS analysis were used as positive controls.

ELISA

Primary Abs: anti-IFN-γ (5 μg/ml, clone R6A2; our hybridoma collection), and anti-IL-2 and anti-TNF-α (2 and 6 μg/ml, clones JES6-1A12 and G281-2626, respectively; BD Pharmingen). Secondary biotinylated Abs: anti-IFN-γ (0.6 μg/ml, clone Ani8; our hybridoma collection), and anti-IL-2 and anti-TNF-α (both used at 1 μg/ml, clones JES6-5H4 and MP6-XT3, respectively; BD Pharmingen). ELISA were performed as previously described (25)

NO assay

Nitrite was measured as the stable end product of NO and levels were quantified by a colorimetric assay based on the Griess reaction (31).

Induction and clinical evaluation of active EAE

MBP89–101 peptide (VHFFKNIVTPRTP-COOH; from Å. Engström, University of Uppsala, Uppsala, Sweden) was used to induce EAE in IFN-β−/− and IFN-β+/+ littermates. Mice were immunized s.c. at the base of the tail with 100 μl of emulsion of 250 μg of MBP89–101 peptide in PBS and CFA containing Mycobacterium tuberculosis H37Ra (Difco). In addition, each animal received 400 ng of pertussis toxin (from Bordetella pertussis; Sigma-Aldrich) dissolved in 100 μl of PBS; this mix was given i.p. at the day of immunization and 2 days later.

Mice were sacrificed at day 13 postimmunization for immunohistochemistry during the acute phase of the EAE (25).

Immunohistochemistry

Brain and spinal cord were dissected out on day 13 postimmunization and immediately embedded in OTC compound (Sakura Finetek) and snap-frozen in isopentane on dry ice. Tissues were cryosectioned in 10-μm slices and kept at −70°C until staining. For single staining, the sections were incubated with biotinylated anti-CD1d (1B1; BD Pharmingen) after which ExtrAvidin-peroxidase (Sigma-Aldrich) and diaminobenzidine (Saveen Biotech) were used for detection. Hematoxylin was used for background staining. For double staining, the tissues were again incubated with biotinylated anti-CD1d with streptavidin-Cy3 being used for detection. Tissues were simultaneously stained with FITC-labeled anti-NK1.1 (PK136; our own hybridoma collection).

Statistical analysis

When adding results from several experiments together, differences in proliferation, cytokine, and NO production were analyzed using a paired sign t test. When comparing these parameters within single experiments, the Student t test was used. Differences in FACS marker expression were analyzed with the Student paired t test. Values of p ≤ 0.05 were considered significant.

Results

The intrinsic capacity of glial cells as APCs is to prevent further expansion of T cells while maintaining T cell effector function

We were at first interested in determining whether there are differences between astrocytes and microglia in their Ag-presenting capacity, as there has been a great deal of controversy in the literature (32, 33, 34). As depicted in Fig. 1⇓, there was no major qualitative differences observed in between these two CNS-specific APCs in their Ag-presenting capacity, measured by inducing T cell proliferation (Fig. 1⇓A) and effector functions, determined by production of IFN-γ (Fig. 1⇓, B and C), TNF-α, and NO (data not shown). Furthermore, as shown in Fig. 1⇓, B and C, there were no discrepancies observed between these two APC populations with regard to effect of cytokine treatment (IFN-γ and IFN-β). Hence, we have used mixed culture of astrocytes and glial cells and referred to them as glial cells. Interestingly, our analysis of the glial cells’ Ag-presenting capacity revealed that glial cells do not allow Ag-specific T cell expansion, although they do induce Ag-specific T cells effector functions, measured by production of large amount of IL-2, IFN-γ, TNF-α, and NO (Fig. 2⇓). This result strongly indicates that the intrinsic function of CNS resident glial cells is to permit activated T cells to exert their effector functions once entering CNS, while still preventing them from further expansion in situ.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Astrocytes and microglia fail to induce T cell proliferation while sustaining T cell IFN-γ production. Glial cells cannot induce an Ag-specific T cell proliferation. A, CFSE-labeling of T cells in coculture with pure untreated astrocytes and Ag (solid black histogram) or with pure microglia and Ag (gray solid histogram) is shown. None of these cocultures show proliferating T cells. The hatched histogram shows the positive control of nondividing T cells, that is, T cells CFSE labeled just before FACS analysis. Both astrocytes (B) and microglia (C) do induce an Ag-specific IFN-γ response. Also, both cell types respond similarly to IFN-β or -γ treatment. The IFN-γ response seen in microglial cultures without exogenously added Ag could be explained by the presence of small amounts of endogenous myelin. No IFN-γ was observed in any cultures of astrocytes or microglial cells only. Data show one representative experiment each, and error bars represent SD of duplicate samples. ∗, p < 0.05; ∗∗, p < 0.01.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Glial cells inhibit T cell expansion but induce T cell effector functions. A, Mixed glial cultures Ag-specifically inhibit T cell proliferation but induce Ag-specific IL-2 (B), IFN-γ (C), TNF-α (D), and NO production (E). Data represent three to nine individual experiments taken together, and error bars represent SEM.

IFN-β negatively regulates the Ag-presenting capacity of glial cells by partly rescuing T cell expansion

It is commonly believed that development of a chronic, autoimmune CNS inflammation is the result of activation of autoreactive T cells in the peripheral lymphoid organs, which thereafter enter the bloodstream and transverse the blood brain barrier. It is also generally believed that these T cells require reactivation in situ to exert their deleterious effects (35, 36). Therefore, the function of CNS resident APC populations (glial cells) could be detrimental in the outcome of T cell reactivation in CNS. Thus, we investigated the effects of IFN-β on these cells and compared it with the known effect of a proinflammatory cytokine, IFN-γ. We chose to study both cytokine effects on resting T cells, thus determining whether glial cells could function as professional APCs, as well as on already activated T cells. These T cells were activated using conventional irradiated splenic APCs and Ag (see Materials and Methods) for 48 h and thereafter washed and cocultured with glial cells. In this way, one could mimic interaction of activated T cells entering the CNS with residual glial cells. Because there were no differences observed between specific T cell expansion using CSFE labeling compared with DNA synthesis and thymidine up-take (see Figs. 1⇑A and 2A), we use proliferation measured by thymidine up-take as read out. Surprisingly, treatment of glial cells with the proinflammatory cytokine IFN-γ leads to significant Ag-specific inhibition of T cells proliferation (Fig. 3⇓A). Similar findings were observed when activated T cells were used (Fig. 3⇓B). Importantly, none of the differently treated glial cells induced significant T cell death as measured by annexin V and propidium iodide staining (data not shown). Because these findings challenge several earlier reports regarding the APC role of glial cells in induction of T cell proliferation (32, 37, 38), we investigated what could be the reason for this discrepancy. As shown in Fig. 3⇓C, irradiation of glial cells before coculture with resting T cells results in entirely different findings, as irradiated glial cells could induce T cell proliferation. Furthermore, IFN-β treatment does not have any significant effect on T cell proliferation under this condition (Fig. 3⇓C). Very similar results were also achieved when using mitomycin-treated glial cells (data not shown). Hence, irradiation and/or mitomycin exposure completely alters the APC function of glial cells; it renders them capable of inducing Ag-specific proliferation. It might be assumed that this finding could partly be due to the prevention of T cell expansion as a result of glial cell proliferation and crowding in nonirradiated cultures. However, this was addressed by using half the number of glial and T cells for coculture and very similar results were achieved (data not shown). In addition, as shown earlier in Fig. 1⇑A, CSFE labeling of T cells before coculture with glial cells (which exclusively address T cell proliferation) verified the inability of glial cells to induce T cell proliferation.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Glia-mediated T cell proliferation arrest requires living glia. A, Addition of Ag to cocultures of nonirradiated glial cells and resting T cells gives a significantly suppressed T cell proliferation in IFN-γ-treated cultures. IFN-β treatment results in higher proliferation compared with untreated glial cells. B, The proliferation is also suppressed in cocultures of activated T cells and IFN-γ-treated glial cells (nonirradiated). C, Lethal irradiation of the glial cells just before T cell addition results in a totally different proliferation pattern. Untreated or IFN-β-treated glial cells then induce a high Ag-specific proliferation and no suppression is seen in IFN-γ-treated cultures. Data in A and B show the mean ± SEM of eight to nine experiments. Data in C show one representative experiment in which irradiated glial cells were used. Error bars represent SD of triplicate samples. ∗, p < 0.05; ∗∗, p < 0.01.

IFN-β negatively regulates the Ag-presenting capacity of glial cells leading to inhibition of T cell effector function

As stated previously, our interest was to determine the effect of IFN-β-treated glial cells on regulation and maintenance of T cell effector function. Thus, the capacity of resting and activated T cells to produce IFN-γ and TNF-α (two proinflammatory cytokines produced by these encephalitogenic Th1 T cell lines) was measured in response to coculture with IFN-β- or IFN-γ-treated glial cells. Neither pure glial cells (astrocytes and microglia) nor mixed glial cultures produced any measurable IFN-γ or TNF-α when cultured alone in the absence of T cells, regardless of exogenous cytokine stimulation (detection level 5–10 pg/ml) (data not shown). Thus, the measured IFN-γ and TNF-α in the supernatant of cocultures most likely represent the cytokine production by T cells. As depicted in Fig. 4⇓, A and C, IFN-β reduced Ag-specific IFN-γ and TNF-α production by T cells. This result was in contrast to IFN-γ, which as expected, enhanced the proinflammatory cytokine production of T cells. In cocultures using activated T cells, glial cells induced an enhanced T cell cytokine production, and this was totally prevented in IFN-β-treated cultures (Fig. 4⇓, B and D). Because it has been suggested that IFN-β can alter the phenotypic response by driving Th1 toward a Th2 phenotype, IL-4 was measured in these cultures. However, we found no increase in IL-4 production when the glial cells were pretreated with IFN-β, rather a tendency was observed toward a similar down-modulating effect resulting in lower amounts of IL-4 (data not shown). Hypothetically, elevated levels of the immunosuppressive cytokine TGF-β1 could possibly mediate the observed effects of IFN-β pretreatment. However, no TGF-β1 was detected in any supernatant, as measured by ELISA (data not shown).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

IFN-β treatment of glial cells reduces T cell production of IFN-γ and TNF-α. T cells cocultured with IFN-β-treated glial cultures induce a lower Ag-specific T cell IFN-γ (A) and TNF-α (C) response compared with untreated controls. Pretreatment with IFN-γ enhanced the cytokine response. B and D, When T cells preactivated with splenic APCs and Ag are added to glial cultures, untreated glial prolong the T cell cytokine production compared with T cells cultured alone. IFN-β pretreatment of the glial cells totally inhibits this enhancement and restores the cytokine production to values indistinguishable from T cells cultured alone. No IFN-γ or TNF-α was observed in cultures of glial cells only. Data show the mean values of eight to nine individual experiments, and error bars represent SEM of these experiments. ∗, p < 0.05; ∗∗, p < 0.01.

IFN-β treatment prevents glia-induced T cell up-regulation of CD25

Aside from cytokine production, other markers generally used to define T cell activation were also examined after coculture with IFN-β or -γ treated or untreated glial cells. CD44 expression was high in all cultures probably reflecting a memory phenotype of the encephalitogenic cell lines. CD69, in contrast, was expressed at relatively low levels in all cultures (data not shown). It is possible that investigation of the cells at a different time point would reveal distinctions that were not observed in this study as CD69 is defined as an early activation marker (39). CTLA-4 expression was very low on T cells from all cultures, whereas CD28 and CD40L were not conclusively up- or down-regulated upon coculture with differently treated glial cells (data not shown). When investigating the activation marker CD25 (the IL-2R α-chain), data well in accordance with the cytokine results were obtained. In cocultures of resting T cells and IFN-β pretreated glial cells, a down-regulation of T cell CD25 expression was observed compared with T cell cocultures with untreated glial cells. IFN-γ pretreatment increased the CD25 T cell expression. This observation was true for cultures without Ag addition as shown in Fig. 5⇓A. In the presence of Ag, a very high CD25 up-regulation was observed in all cultures and the effects of either IFN-β or IFN-γ treatment were no longer observed (data not shown). Cocultures of activated T cells with glial cells resulted in up-regulated T cell CD25 expression compared with T cells cultured alone. These data again suggest that glial cells can perpetuate the T cell effector phase and thus support the findings of cytokine production by T cells (Fig. 4⇑, B and D). In accordance, IFN-β treatment of the glial cells totally inhibited the up-regulation of CD25, while IFN-γ treatment enhanced this up-regulation even further (Fig. 5⇓B). It is important to note that no IFN-β is left in the cultures when the T cells are added, and IFN-β treatment alone does not induce an autocrine loop of IFN-β production (which was measured by ELISA, data not shown). Hence, IFN-β is not acting on the T cells, but on the glial cells in the cultures. Taken together, these data suggest that IFN-β inhibits the capacity of potential CNS APCs to function as Ag-presenting cells activating resting T cells. In addition, it prevents their capacity to sustain an ongoing T cell effector phase after Ag-specific activation.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

IFN-β treatment of glial cells inhibits the up-regulation of CD25 expression on T cells. A, IFN-β treatment of glial cells reduces the CD25 expression on subsequent cocultures with resting T cells in the absence of Ag, whereas IFN-γ treatment increases this compared with untreated glial cells. Addition of Ag induces a very strong CD25 up-regulation on the T cells with no difference between treatments (data not shown). B, Coculture of activated T cells with untreated glial cells induces up-regulation of the T cell activation marker CD25 compared with T cells culture alone. IFN-β pretreatment of the glial cells totally inhibits this up-regulation, whereas IFN-γ pretreatment increases the CD25 expression even further. Data show the mean value of three experiments. Fluorescence geometric mean values ± SEM are shown in parentheses. ∗, p < 0.05; ∗∗, p < 0.01.

The effects of glial cells on T cell responses is cell-cell contact dependent

Ag-specific activation of resting T cells is obviously dependent on MHC class II and TCR interaction, and therefore cell-cell contact. Nevertheless, we investigated whether the effects of glial cells on preactivated T cells were mediated through a membrane-bound ligand or through a soluble mediator, hence whether a Transwell system was used. As demonstrated in Fig. 6⇓, A and B, both the effects of untreated glial cells on preactivated T cell proliferation and cytokine production, as well as the relative effect of IFN-β or -γ treatment, are inhibited once the cells are physically separated by a membrane. This result strongly indicates that glial cells require direct contact with T cells to exert their function. Still, the possibility remained that this cell-cell interaction resulted in the production of a soluble mediator that could in return affect/inhibit T cell proliferation. To test this hypothesis, T cells were activated, this time with splenic APCs in the presence of different amounts of conditioned medium from glial and T cell cocultures with or without Ag. The results obtained with supernatants from Ag-containing cultures are depicted in Fig. 6⇓C and show that supernatant recovered from glia-T cell cocultures inhibit T cell proliferation in a concentration-dependent manner. However, supernatants from untreated, IFN-β- or IFN-γ-treated cultures inhibited T cell proliferation to the same extent. This outcome suggests that the differences observed between IFN-β- and IFN-γ-treated glial cells were dependent on the expression of one or more cell surface molecules yet undefined and not on soluble factors.

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Glial cell-mediated perpetuation of activated T cell cytokine responses requires cell-cell contact. A, The IFN-γ-mediated inhibition of proliferation requires cell-cell contact as demonstrated by the fact that inhibition is blocked in cultures where the T cells and glial cells are separated by a membrane. Error bars represent SD of triplicate samples. B, When activated T cells and glial cells are allowed cell-cell contact, an increase of the cytokine secretion by activated T cells is observed. This increase is totally repressed in IFN-β-treated cultures and enhanced in IFN-γ-treated cultures. When the T cells and glial cells are separated by a membrane, inhibiting direct cell-cell contact but allowing soluble molecules to pass freely, this cytokine production enhancement is blocked. Error bars represent SD of triplicate samples. C, Different concentrations of supernatants from cocultures containing glial cells, T cells, and Ag were added to T cells stimulated by splenic APCs, and proliferation was measured. Data show that there is a soluble mediator released upon cell-cell contact in glia-T cell cocultures that can inhibit proliferation to a certain extent in a concentration-dependent manner. However, the differently treated supernatants have a similar inhibitory capacity. Error bars represent SD of triplicate samples. ∗∗, p < 0.01; ∗∗∗, p < 0.001.

Glial cells inability to induce T cell proliferation is not due to high NO production or deficiency in IL-2 production

NO has previously been shown to inhibit T cell proliferation (40). Furthermore, it is well established that T cells require IL-2 for successful proliferation (41) and it has been debated whether glial cells are capable of inducing an IL-2 response (33, 42, 43, 44). As shown in Fig. 2⇑B, the glial cells induced a clear Ag-specific IL-2 response, and the T cell proliferation arrest could thus not be explained by deficiencies in IL-2 production. To investigate whether the high NO production correlated with the observed inhibition of T cell proliferation, the kinetics of these parameters were examined. Fig. 7⇓A demonstrates that proliferation increases over time and after 96 h in the IFN-γ-treated glial cell culture, the Ag-specific inhibition of proliferation is in fact overcome. Next, Fig. 7⇓B shows that the NO production is also increasing over time and reaches a peak at 96 h, correlating with the highest level of proliferation. These data suggest that the inhibition of proliferation is not due to high levels of NO production.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Proliferation arrest is not caused by high NO production. A, The kinetics of cell proliferation in glial cell (G) and T cell (T) cocultures is shown. Proliferation is expressed as a percentage of the proliferation in cultures of glial cells only. The inhibition of proliferation is Ag-dependent. The inhibition gets less efficient over time and is finally overcome at 96 h in IFN-γ-treated cultures. The kinetics of glial cell proliferation alone are not affected by the treatments (data not shown). B, All glial cultures induce an Ag-specific NO production, which increases over time. The figure shows that the highest NO levels coincide with the highest T cell proliferation. Error bars represent SD of triplicate samples in all panels.

IFN-β treatment does not decreased MHC class II or costimulatory expression on glial cells

The data presented in Fig. 6⇑C suggested that differences in expression of membrane-bound ligands could be responsible for the observed differences between T cell cocultures of untreated, IFN-β-treated, or IFN-γ-treated glia. T cells generally require signals through both MHC class II and costimulatory molecules for full activation and an effective Ag-specific response. FACS studies were performed to investigate whether IFN-β treatment caused a reduced expression of these molecules on glial cells, which could explain the achieved results. Surprisingly, no decrease was observed in any of the markers investigated in this model as result of IFN-β treatment. Microglia and astrocytes were studied separately in these experiments based on Mac-1 or GFAP expression, respectively. MHC class II was not expressed in untreated astrocytes, IFN-γ strongly up-regulated the molecule, and IFN-β alone had no effect. In accordance with previously published data (45, 46, 47), IFN-β down-regulated an IFN-γ-mediated MHC class II expression on the astrocytes (Fig. 8⇓A). The microglia population expressed low levels of MHC class II and treatment with both IFNs resulting in a MHC class II up-regulation (see Fig. 8⇓A). The costimulatory molecules B7-1, B7-2, and CD40 were also studied. Astrocytes were shown to be highly positive for B7-1, whereas negative for B7-2 and CD40 (see Fig. 8⇓, B–D). None of these molecules was affected by IFN-γ or IFN-β treatment alone, although the combination of both cytokines generated a tendency toward up-regulation of B7-2 and CD40. In the microglia population, which was weakly positive for all these molecules when untreated, both cytokines again generated higher expression of these three costimulatory molecules. An additive effect was observed with respect to B7-2 expression, in which the combination of both IFNs resulted in an even higher expression compared with any cytokine alone (see Fig. 8⇓C). ICAM-1 and VCAM-1 were also investigated. ICAM-1 was shown to be relatively highly expressed in both astrocytes and microglia, but the expression was further up-regulated by both IFN-γ and IFN-β on microglia (see Fig. 8⇓E). The pattern was similar for microglial VCAM-1 expression (see Fig. 8⇓F), whereas astrocytes expressed very high levels of VCAM that were further up-regulated by the combination treatment with both IFNs. Because glial cells have been shown to induce T cell apoptosis in a Fas ligand-dependent manner (48) and to secrete the immunosuppressive cytokine TGF-β1 (49, 50), these molecules were also studied. However, no or very low expression was observed in glial cells and there was no marked effect as a result of pretreatment with IFNs (data not shown). These results demonstrate that the suppressive effects that IFN-β treatment of glial cells exerts on both T cell activation and effector phase maintenance is not likely to be mediated through down-regulation of any of the molecules involved in classical T cell activation.

FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

IFN-β treatment of glial cells does not induce down-regulation of MHC class II or costimulatory molecules. Mixed glial cells consisted of astrocytes and microglia distinguished by expression of GFAP and Mac-1, respectively. A, Astrocytes were negative for MHC class II when untreated or IFN-β-treated, but MHC class II was strongly up-regulated upon IFN-γ stimulation. IFN-β reduced the IFN-γ-mediated up-regulation. Untreated microglia expressed low amounts of MHC class II and this effect was further enhanced when treated with either IFN-β or IFN-γ. B, Untreated astrocytes expressed high levels of B7-1, and the expression remained unchanged upon treatment. Microglia expressed medium levels of B7-1 and the expression increased upon treatment with IFN-β or IFN-γ. C, Astrocytes were negative for B7-2 in all conditions. Untreated microglia expressed medium levels of B7-2, whereas the expression was significantly up-regulated after treatment with IFN-β, IFN-γ, or both in combination. D, Astrocytes were negative for CD40 in all cultures although a nonsignificant tendency toward CD40 up-regulation was seen in cultures treated with both IFN-β and IFN-γ. Untreated microglia expressed low levels of CD40 and the expression was increased in cultures treated with IFN-β or IFN-γ. E and F, Astrocytes express very high levels of ICAM and VCAM, respectively, which is further increased by combination treatment with both IFN-γ and IFN-β. Untreated microglia expressed only low levels but up-regulated their expression upon treatment with either IFN-β or IFN-γ.

IFN-β and IFN-γ elevate CD1d expression on glial cells

We also investigated the expression of the nonclassical MHC-like molecule CD1d because we have reported that this molecule plays a crucial role in the regulation of CNS inflammation (30). Interestingly, we found that both astrocytes and microglia express relatively high levels of CD1d and this was elevated by IFN-β as well as by IFN-γ (Fig. 9⇓A). We continued to investigate how expression of CD1d is regulated on glial cells in vivo and whether lack of endogenous IFN-β influenced CD1d expression in the CNS. As shown in Fig. 9⇓B, CD1d (stained in red) is indeed up-regulated in glial cells in the inflamed CNS of EAE mice during the acute phase of the disease. The CD1+ cells are mainly astrocytes (Fig. 9⇓B), as CD1 expression colocalized with GFAP expression (data not shown). Moreover, this reaction was associated with the recruitment of NK1.1+ CD3+ cells to the CNS. Fig. 9⇓B shows NK1.1+ infiltrating cells (stained in green) in very close proximity to CD1-expressing astrocytes. Subsequent double staining with anti-CD3 confirmed the presence of NK1.1+ T cells (data not shown). However, no difference was found between IFN-β−/− mice and their IFN-β+/− littermates with respect to the regulation of CD1d expression in glial cells during the course of EAE (data not shown).

FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

Glial cells express CD1d NK1.1+ infiltrating cells in close vicinity of CD1+ glial cells in the CNS of EAE mice. A, Untreated astrocytes and microglia expressed medium levels of CD1d but the expression increased upon treatment with either IFN-β or IFN-γ. Data show the mean value ± SEM for 10 experiments. Significant difference between untreated and IFN-treated cultures is shown ∗, p < 0.05; ∗∗; p < 0.01; and ∗∗∗, p < 0.001. B, NK1.1+ infiltrating cells (green) in close interaction with CD1+ glial cells (red). The image is taken around an infiltrate from the cerebellum of an EAE-affected B10.RIII wild-type mouse on day 13 after immunization.

Discussion

It has recently been suggested that for the establishment of a chronic inflammation in the CNS mediated by encephalitogenic T cells (EAE), these T cells have to be efficiently reactivated in situ (51). This Ag-specific restimulation could be mediated by residual APCs located in the CNS. We have previously reported, using EAE model, that a deficiency in IFN-β leads to in situ activation of glial cells, the CNS-specific APCs, with a following activation of encephalitogenic Th1 cells, resulting in enhanced CNS inflammation and demyelination (25). This finding suggested that the beneficial effects of IFN-β could be a result of negative modulation of the Ag-presenting capacity of glial cells. Thus, in this study we evaluate the effect of IFN-β on the capacity of the potential CNS APCs (microglia and astrocytes) to induce and sustain activation of autoreactive Th1 cells. We also compared the effects of a proinflammatory cytokine IFN-γ.

Because the Ag-presenting capacity of glial cells has been controversial (37, 38, 52, 53) and differences have been suggested between microglia cells and astrocytes in their APC functions (32, 33, 34), we first investigated these two glial cell types separately. We found no qualitative differences in Ag-presenting capacity of these cell types, and no further differences were observed in their response to IFN-β or IFN-γ. Interestingly, we found that both astrocytes and microglia were capable of inhibiting expansion of autoreactive Th1 cells, whereas, at the same time they sustain effector functions of Th1 cells. Although this might seem contradictory at first glance, our findings are supported by previously reported in vivo data from EAE-affected animals, which suggests that autoreactive T cells infiltrating the CNS are indeed prevented from proliferating in situ (54, 55). Data presented by Ford et al. (56) are also supports our current findings. In their studies, adult microglia purified directly ex vivo induced insufficient T cell activation with inhibition of proliferation and IL-2 production despite ample production of TNF-α and IFN-γ (40, 56). Similar reports on APC capacity of astrocytes have shown that they could induce cytokine production without a corresponding proliferation (57). However, our data suggest that interaction of glial cells with Th1 cells does not render them anergic because there is no defect in IL-2 production or up-regulation of CD25 (IL-2R α-chain). In fact, interaction of T cells with untreated glial cells alone is adequate to up-regulate CD25 and IL-2 production. The observed Ag-specific inhibition of T cell proliferation in the current study is dependent on cell-cell contact, which results in release of a soluble substance that suppresses proliferation. The inhibition of T cell proliferation was only overcome in long-term (96 h) cocultures with IFN-γ-treated glial cells, which could be representative of a persistent inflammatory stimulus in vivo. In addition, when irradiated or mitomycin-treated glial cells were used, a very strong Ag-specific T cell proliferation was observed. Thus, the earlier conflicting in vitro results concerning the ability of glial cells to induce or inhibit T cell proliferation (32, 37, 57, 58) could partly be the result of the use of IFN-γ-stimulated, mitomycin-treated, or irradiated and hence damaged glial cells in the majority of previous reports (32, 37, 38, 52, 53). Prevention of proliferation reported in this study is not associated with NO production, which was suggested previously (40). Furthermore, we found no support for the involvement of TGF-β1 or Fas-Fas ligand interaction in the inhibition of T cell proliferation.

It is noteworthy to mention that only activated T cells pass the well-protected CNS (59), hence these data indicate an intrinsic ability of glial cells to permit activated T cells to exert their effector function for which they have been predestined, while at the same time inhibiting their expansion to protect the integrity of vital functions of CNS from being damaged by immune-mediated inflammation. However, our data also indicate that persisting inflammatory mediators might alter this function and bring about the glial cell capability of inducing T cell proliferation. This function could in turn lead to prolonged activation of T cells with consequent chronic neuroinflammation as seen in chronic EAE and MS.

Importantly, in this study we also show that IFN-β decreases the capacity of the glial cells to induce Ag-specific T cell effector functions, as measured by IFN-γ, NO, and TNF-α production. IFN-β thereby reduces the mediators that could lead to a chronic inflammation. This effect is also true for T cells that are already in an activated state when encountering the glial cells. Interestingly, untreated glial cells increased T cell cytokine production, as well as induced CD25 up-regulation on preactivated T cells, which speaks for an inherent capacity to sustain T cell effector functions independent of Ag specificity. This activation was entirely blocked when glial cells were pretreated with IFN-β before interaction with T cells. The fact that similar results are obtained in both of these conditions, of which the latter is devoid of Ag, strongly suggests that activation is mediated by a mechanism distinct from MHC class II and TCR interaction. In theory, there is a possibility that traces of Ag are still present in the culture medium loaded onto the splenic APCs. However, when the CD25 expression was measured on resting T cells, in which neither Ag nor APCs have been present for a minimum of 2 wk, CD25 was still up-regulated when T cells were cocultured with untreated glial cells. This up-regulation was prevented in IFN-β-treated glial cell cultures and this argues against involvement of an Ag-dependent mechanism. Taken together, our findings might shed light on the possible local effects of IFN-β in other situations than direct IFN-β administration, where there is an endogenous up-regulation of IFN-β within the CNS, such as in the event of neuroinflammation (autoimmune or viral induced) that could result in defective in situ immune regulation by glial cells with subsequent chronic neuroinflammation.

Other studies investigating how glial cells affect activated T cells have taken another approach and included both APCs from lymphoid organs and Ag in their glial-T cell cocultures (60, 61). In contrast to our results, they found that astrocytes inhibit T cell effector responses as measured by IL-2 production, CD25 up-regulation, and TCR down-regulation. Gimsa et al. (61) also suggested an astrocyte-mediated CTLA-4 up-regulation on T cells, which was not observed in the present study. The differences in experimental setups could account for this controversy. Our glial and preactivated T cell cocultures are devoid of lymphoid APCs and Ag, and this setup would mimic a different milieu where the availability of Ag and lymphoid APCs is limited.

The maintenance of T cell effector functions induced by glial cells could be mediated through costimulatory and adhesion molecules constitutively expressed on glial cells. In accordance, both the astrocytes and microglia displayed different constitutive expression patterns of B7, CD40, ICAM-1, and VCAM-1. The potential capacity of astrocytes to act as APCs has been widely debated and much of it has centered on their B7 expression (37). The B7-1 expression observed in our study is likely to be strain-specific because astrocytes from SJL mice have similarly been shown to be mainly expressing B7-1 (62, 63), whereas astrocytes from BALB/c mice are primarily expressing B7-2 (53).

On the contrary, the inhibitory effect of IFN-β is not likely to be mediated by signaling through the MHC class II-TCR pathway or costimulatory signaling. None of the molecules we investigated that are involved in these pathways were down-regulated upon IFN-β treatment in either the astrocyte or microglia population. Rather the opposite was observed, as both MHC class II and several of the costimulatory molecules were up-regulated in the microglia fraction. In agreement with our earlier reports (25, 64), no shift from Th1 to Th2 phenotype was detected as a result of IFN-β treatment of glial cells, as has been previously suggested by others (15, 65). These facts together strongly point toward a more generalized down-modulatory effect of IFN-β on the stimulatory capacity of CNS APCs. Interestingly, IFN-β was recently shown to up-regulate the inhibitory costimulatory molecule PD-L1 in dendritic cells (66) and it remains to be investigated whether IFN-β has similar affect on glial cells. In one study (27), microglia were activated with IFN-γ or LPS and the addition of IFN-β was shown to down-regulate T cell cytokine production. However, because IFN-β also reduced the IFN-γ-induced MHC class II expression, this result alone could explain the reduction in T cell activation. Our report demonstrates the sole effects of IFN-β on glial APC functions.

Because our data suggested an immunoregulatory role of glial cells on encephalitogenic T cell expansion and function, it was of particular interest to investigate the regulation of CD1d on glial cells. CD1d is known to stimulate a subset of T cells with postulated immunoregulatory functions in EAE and MS (67, 68, 69), and we have shown that mice deficient in CD1d develop augmented and a more chronic EAE (30). Interestingly, we found that both IFNs increased expression of CD1d on astrocytes and microglia. This observation is of value for future studies exploring CD1d regulation in the CNS, as this is the first report on cytokines capable of up-regulating CD1d on glial cells. We also demonstrate that CD1d is up-regulated in vivo on glial cells of EAE-affected mice. This finding is in accordance with previous reports in which it has been shown that both MS patients (70) and EAE-affected guinea pigs (71) display CD1 reactivity in the CNS. The majority of the CD1d-restricted immunoregulatory T cells express the NK cell marker NK1.1. Indeed, we detected NK1.1+ cells in close vicinity of CD1+ astrocytes, indicative of a functional CD1-dependent Ag presentation by these glial cells. Nevertheless, despite the up-regulatory effects of IFN-β on CD1d, there was no difference in CNS CD1 expression between EAE-affected IFN-β-deficient mice and their wild-type littermates. Taking the in vitro and in vivo findings together, it could be concluded that IFN-β has an effect on the induction of the CD1d molecule on glial cells, and thus could play a role in the regulation of CNS inflammation through this mechanism. However, this effect is not exclusively IFN-β-dependent and could be overcome by other factors up-regulated in an inflammatory environment, such as IFN-γ. EAE-affected IFN-β-deficient mice do have increased levels of IFN-γ in the CNS (25) and this could explain the maintained levels of CD1 expression in these mice.

In summary, we report that the intrinsic role of glial cells is to inhibit expansion of T cells while allowing their effector function. Moreover, IFN-β suppresses the capacity of these potential CNS APCs, both to induce an Ag-specific T cell cytokine response, and also to sustain an ongoing T cell effector function. The effects of IFN-β require cell-cell contact, but are not dependent on down-regulation of MHC class II or on any of the costimulatory or adhesion molecules involved in classical T cell activation. The current report might shed light on the basic mechanisms of IFN-β function during CNS inflammation and the possible partial beneficial effects of IFN-β on MS patients, as well as have implications for other neuroinflammatory and neurodegenerative disease processes where glial activation is believed to be central to the disease progression.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by grants from the Swedish Foundation for Strategic Research, Swedish Research Council-Natural Science, Swedish Research Council-Medicine, Alfred Österlund Foundation, Tore Nilson Foundation, His Majesty Gustav V Foundation, the Royal Swedish Academy of Science, Royal Physiographic Society, Lund, Sweden, the M. Bergvalls Foundation, Åke Wiberg Foundation, Börje Dahlin Foundation, Segerfalk Foundation, and the Crafoord Foundation.

  • ↵2 Address correspondence and reprint requests to Dr. Shohreh Issazadeh-Navikas, Neuroinflammation Unit, Institute for Experimental Medical Science, Biomedical Centre I13, Lund University, 221 84 Lund, Sweden. E-mail address: Shohreh.Issazadeh{at}med.lu.se

  • ↵3 Abbreviations used in this paper: MS, multiple sclerosis; EAE, experimental autoimmune encephalomyelitis; MBP, myelin basic protein; GFAP, glial fibrillary acidic protein.

  • Received July 26, 2005.
  • Accepted June 20, 2006.
  • Copyright © 2006 by The American Association of Immunologists

References

  1. ↵
    Sun, S., X. Zhang, D. F. Tough, J. Sprent. 1998. Type I interferon-mediated stimulation of T cells by CpG DNA. J. Exp. Med. 188: 2335-2342.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Pestka, S., J. A. Langer, K. C. Zoon, C. E. Samuel. 1987. Interferons and their actions. Annu. Rev. Biochem. 56: 727-777.
    OpenUrlCrossRefPubMed
  3. ↵
    Kadowaki, N., S. Antonenko, Y.-J. Liu. 2001. Distinct CpG DNA and polyinosinic-polycytidylic acid double-stranded RNA, respectively, stimulate CD11c− type 2 dendritic cell precursors and CD11c+ dendritic cells to produce type I IFN. J. Immunol. 166: 2291-2295.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Matsumoto, M., K. Funami, M. Tanabe, H. Oshiumi, M. Shingai, Y. Seto, A. Yamamoto, T. Seya. 2003. Subcellular localization of Toll-like receptor 3 in human dendritic cells. J. Immunol. 171: 3154-3162.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Hoshino, K., T. Kaisho, T. Iwabe, O. Takeuchi, S. Akira. 2002. Differential involvement of IFN-β in Toll-like receptor-stimulated dendritic cell activation. Int. Immunol. 14: 1225-1231.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Pfeffer, L. M., C. A. Dinarello, R. B. Herberman, B. R. Williams, E. C. Borden, R. Bordens, M. R. Walter, T. L. Nagabhushan, P. P. Trotta, S. Pestka. 1998. Biological properties of recombinant α-interferons: 40th anniversary of the discovery of interferons. Cancer Res. 58: 2489-2499.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Hafler, D. A., H. L. Weiner. 1989. MS: a CNS and systemic autoimmune disease. Immunol. Today 10: 104-107.
    OpenUrlCrossRefPubMed
  8. ↵
    Lassmann, H.. 1999. The pathology of multiple sclerosis and its evolution. Philos. Trans. R. Soc. Lond. B Biol. Sci. 354: 1635-1640.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Francis, G., F. Weber, P. Rieckmann. 2000. Effect of interferon β on human myelin basic protein-specific T-cell lines: comparison of IFNβ-1a and IFNβ-1b. Neurology 55: 322-323.
    OpenUrlFREE Full Text
  10. ↵
    Noronha, A., A. Toscas, M. A. Jensen. 1993. Interferon β decreases T cell activation and interferon γ production in multiple sclerosis. J. Neuroimmunol. 46: 145-153.
    OpenUrlCrossRefPubMed
  11. ↵
    Rep, M. H., R. Q. Hintzen, C. H. Polman, R. A. van Lier. 1996. Recombinant interferon-β blocks proliferation but enhances interleukin-10 secretion by activated human T-cells. J. Neuroimmunol. 67: 111-118.
    OpenUrlCrossRefPubMed
  12. ↵
    Jansson, A., J. Ernerudh, M. Kvarnstrom, C. Ekerfelt, M. Vrethem. 2003. Elispot assay detection of cytokine secretion in multiple sclerosis patients treated with interferon-β1a or glatiramer acetate compared with untreated patients. Mult. Scler. 9: 440-445.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    McRae, B. L., R. T. Semnani, M. P. Hayes, G. A. van Seventer. 1998. Type I IFNs inhibit human dendritic cell IL-12 production and Th1 cell development. J. Immunol. 160: 4298-4304.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Tuohy, V. K., M. Yu, L. Yin, P. M. Mathisen, J. M. Johnson, J. A. Kawczak. 2000. Modulation of the IL-10/IL-12 cytokine circuit by interferon-β inhibits the development of epitope spreading and disease progression in murine autoimmune encephalomyelitis. J. Neuroimmunol. 111: 55-63.
    OpenUrlCrossRefPubMed
  15. ↵
    Wang, X., M. Chen, K. P. Wandinger, G. Williams, S. Dhib-Jalbut. 2000. IFN-β-1b inhibits IL-12 production in peripheral blood mononuclear cells in an IL-10-dependent mechanism: relevance to IFN-β-1b therapeutic effects in multiple sclerosis. J. Immunol. 165: 548-557.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Muraro, P. A., L. Liberati, L. Bonanni, A. Pantalone, C. M. Caporale, C. Iarlori, G. De Luca, D. Farina, A. Lugaresi, D. Gambi. 2004. Decreased integrin gene expression in patients with MS responding to interferon-β treatment. J. Neuroimmunol. 150: 123-131.
    OpenUrlCrossRefPubMed
  17. ↵
    Calabresi, P. A., C. M. Pelfrey, L. R. Tranquill, H. Maloni, H. F. McFarland. 1997. VLA-4 expression on peripheral blood lymphocytes is downregulated after treatment of multiple sclerosis with interferon β. Neurology 49: 1111-1116.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Kraus, J., A. K. Ling, S. Hamm, K. Voigt, P. Oschmann, B. Engelhardt. 2004. Interferon-β stabilizes barrier characteristics of brain endothelial cells in vitro. Ann. Neurol. 56: 192-205.
    OpenUrlCrossRefPubMed
  19. ↵
    Galboiz, Y., S. Shapiro, N. Lahat, A. Miller. 2002. Modulation of monocytes matrix metalloproteinase-2, MT1-MMP and TIMP-2 by interferon-γ and -β: implications to multiple sclerosis. J. Neuroimmunol. 131: 191-200.
    OpenUrlCrossRefPubMed
  20. ↵
    Sinigaglia, F., D. D’Ambrosio, L. Rogge. 1999. Type I interferons and the Th1/Th2 paradigm. Dev. Comp. Immunol. 23: 657-663.
    OpenUrlCrossRefPubMed
  21. ↵
    McRae, B. L., L. J. Picker, G. A. van Seventer. 1997. Human recombinant interferon-β influences T helper subset differentiation by regulating cytokine secretion pattern and expression of homing receptors. Eur. J. Immunol. 27: 2650-2656.
    OpenUrlCrossRefPubMed
  22. ↵
    Koike, F., J. Satoh, S. Miyake, T. Yamamoto, M. Kawai, S. Kikuchi, K. Nomura, K. Yokoyama, K. Ota, T. Kanda, et al 2003. Microarray analysis identifies interferon β-regulated genes in multiple sclerosis. J. Neuroimmunol. 139: 109-118.
    OpenUrlCrossRefPubMed
  23. ↵
    Molnarfi, N., L. Gruaz, J. M. Dayer, D. Burger. 2004. Opposite effects of IFN β on cytokine homeostasis in LPS- and T cell contact-activated human monocytes. J. Neuroimmunol. 146: 76-83.
    OpenUrlCrossRefPubMed
  24. ↵
    Shapiro, S., Y. Galboiz, N. Lahat, A. Kinarty, A. Miller. 2003. The ‘immunological-synapse’ at its APC side in relapsing and secondary-progressive multiple sclerosis: modulation by interferon-β. J. Neuroimmunol. 144: 116-124.
    OpenUrlCrossRefPubMed
  25. ↵
    Teige, I., A. Treschow, A. Teige, R. Mattsson, V. Navikas, T. Leanderson, R. Holmdahl, S. Issazadeh-Navikas. 2003. IFN-β gene deletion leads to augmented and chronic demyelinating experimental autoimmune encephalomyelitis. J. Immunol. 170: 4776-4784.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Liuzzi, G. M., T. Latronico, A. Fasano, G. Carlone, P. Riccio. 2004. Interferon-β inhibits the expression of metalloproteinases in rat glial cell cultures: implications for multiple sclerosis pathogenesis and treatment. Mult. Scler. 10: 290-297.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Kawanokuchi, J., T. Mizuno, H. Kato, N. Mitsuma, A. Suzumura. 2004. Effects of interferon-β on microglial functions as inflammatory and antigen presenting cells in the central nervous system. Neuropharmacology 46: 734-742.
    OpenUrlCrossRefPubMed
  28. ↵
    Erlandsson, L., R. Blumenthal, M. L. Eloranta, H. Engel, G. Alm, S. Weiss, T. Leanderson. 1998. Interferon-β is required for interferon-α production in mouse fibroblasts. Curr. Biol. 8: 223-226.
    OpenUrlCrossRefPubMed
  29. ↵
    Aloisi, F., G. Penna, J. Cerase, B. Menendez Iglesias, L. Adorini. 1997. IL-12 production by central nervous system microglia is inhibited by astrocytes. J. Immunol. 159: 1604-1612.
    OpenUrlAbstract
  30. ↵
    Teige, A., I. Teige, S. Lavasani, R. Bockermann, E. Mondoc, R. Holmdahl, S. Issazadeh-Navikas. 2004. CD1-dependent regulation of chronic central nervous system inflammation in experimental autoimmune encephalomyelitis. J. Immunol. 172: 186-194.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Green, L. C., D. A. Wagner, J. Glogowski, P. L. Skipper, J. S. Wishnok, S. R. Tannenbaum. 1982. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal. Biochem. 126: 131-138.
    OpenUrlCrossRefPubMed
  32. ↵
    Aloisi, F., F. Ria, G. Penna, L. Adorini. 1998. Microglia are more efficient than astrocytes in antigen processing and in Th1 but not Th2 cell activation. J. Immunol. 160: 4671-4680.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Aloisi, F., F. Ria, S. Columba-Cabezas, H. Hess, G. Penna, L. Adorini. 1999. Relative efficiency of microglia, astrocytes, dendritic cells and B cells in naive CD4+ T cell priming and Th1/Th2 cell restimulation. Eur. J. Immunol. 29: 2705-2714.
    OpenUrlCrossRefPubMed
  34. ↵
    Brevig, T., T. Kristensen, J. Zimmer. 2000. Induction of human T-cell proliferation by porcine fetal brain cells: role of astrocytes and macrophages/microglia. Transplant. Proc. 32: 961-962.
    OpenUrlCrossRefPubMed
  35. ↵
    Hickey, W. F., H. Kimura. 1988. Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science 239: 290-292.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Hinrichs, D. J., K. W. Wegmann, G. N. Dietsch. 1987. Transfer of experimental allergic encephalomyelitis to bone marrow chimeras: endothelial cells are not a restricting element. J. Exp. Med. 166: 1906-1911.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Soos, J. M., T. A. Ashley, J. Morrow, J. C. Patarroyo, B. E. Szente, S. S. Zamvil. 1999. Differential expression of B7 co-stimulatory molecules by astrocytes correlates with T cell activation and cytokine production. Int. Immunol. 11: 1169-1179.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Fierz, W., B. Endler, K. Reske, H. Wekerle, A. Fontana. 1985. Astrocytes as antigen-presenting cells. I. Induction of Ia antigen expression on astrocytes by T cells via immune interferon and its effect on antigen presentation. J. Immunol. 134: 3785-3793.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Castellanos, M. del C., S. López-Giral, M. López-Cabrera, M. O. de Landázuri. 2002. Multiple cis-acting elements regulate the expression of the early T cell activation antigen CD69. Eur. J. Immunol. 32: 3108-3117.
    OpenUrlCrossRefPubMed
  40. ↵
    Juedes, A. E., N. H. Ruddle. 2001. Resident and infiltrating central nervous system APCs regulate the emergence and resolution of experimental autoimmune encephalomyelitis. J. Immunol. 166: 5168-5175.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Schwartz, R. H.. 2003. T cell anergy. Annu. Rev. Immunol. 21: 305-334.
    OpenUrlCrossRefPubMed
  42. ↵
    Xiao, B. G., A. Diab, J. Zhu, P. van der Meide, H. Link. 1998. Astrocytes induce hyporesponses of myelin basic protein-reactive T and B cell function. J. Neuroimmunol. 89: 113-121.
    OpenUrlCrossRefPubMed
  43. ↵
    Matyszak, M. K., S. Denis-Donini, S. Citterio, R. Longhi, F. Granucci, P. Ricciardi-Castagnoli. 1999. Microglia induce myelin basic protein-specific T cell anergy or T cell activation, according to their state of activation. Eur. J. Immunol. 29: 3063-3076.
    OpenUrlCrossRefPubMed
  44. ↵
    Klyushnenkova, E. N., P. Vanguri. 1997. Ia expression and antigen presentation by glia: strain and cell type-specific differences among rat astrocytes and microglia. J. Neuroimmunol. 79: 190-201.
    OpenUrlCrossRefPubMed
  45. ↵
    Devajyothi, C., I. Kalvakolanu, G. T. Babcock, H. A. Vasavada, P. H. Howe, R. M. Ransohoff. 1993. Inhibition of interferon-γ-induced major histocompatibility complex class II gene transcription by interferon-β and type β1 transforming growth factor in human astrocytoma cells: definition of cis-element. J. Biol. Chem. 268: 18794-18800.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Ransohoff, R. M., C. Devajyothi, M. L. Estes, G. Babcock, R. A. Rudick, E. M. Frohman, B. P. Barna. 1991. Interferon-β specifically inhibits interferon-γ-induced class II major histocompatibility complex gene transcription in a human astrocytoma cell line. J. Neuroimmunol. 33: 103-112.
    OpenUrlCrossRefPubMed
  47. ↵
    Satoh, J., D. W. Paty, S. U. Kim. 1995. Differential effects of β and γ interferons on expression of major histocompatibility complex antigens and intercellular adhesion molecule-1 in cultured fetal human astrocytes. Neurology 45: 367-373.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Bechmann, I., B. Steiner, U. Gimsa, G. Mor, S. Wolf, M. Beyer, R. Nitsch, F. Zipp. 2002. Astrocyte-induced T cell elimination is CD95 ligand dependent. J. Neuroimmunol. 132: 60-65.
    OpenUrlCrossRefPubMed
  49. ↵
    Dhandapani, K. M., M. Hadman, L. De Sevilla, M. F. Wade, V. B. Mahesh, D. W. Brann. 2003. Astrocyte protection of neurons: role of transforming growth factor-β signaling via a c-Jun-AP-1 protective pathway. J. Biol. Chem. 278: 43329-43339.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Flanders, K. C., R. F. Ren, C. F. Lippa. 1998. Transforming growth factor-β in neurodegenerative disease. Prog. Neurobiol. 54: 71-85.
    OpenUrlCrossRefPubMed
  51. ↵
    Kawakami, N., S. Lassmann, Z. Li, F. Odoardi, T. Ritter, T. Ziemssen, W. E. Klinkert, J. W. Ellwart, M. Bradl, K. Krivacic, et al 2004. The activation status of neuroantigen-specific T cells in the target organ determines the clinical outcome of autoimmune encephalomyelitis. J. Exp. Med. 199: 185-197.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Fontana, A., W. Fierz, H. Wekerle. 1984. Astrocytes present myelin basic protein to encephalitogenic T-cell lines. Nature 307: 273-276.
    OpenUrlCrossRefPubMed
  53. ↵
    Nikcevich, K. M., K. B. Gordon, L. Tan, S. D. Hurst, J. F. Kroepfl, M. Gardinier, T. A. Barrett, S. D. Miller. 1997. IFN-γ-activated primary murine astrocytes express B7 costimulatory molecules and prime naive antigen-specific T cells. J. Immunol. 158: 614-621.
    OpenUrlAbstract
  54. ↵
    Wekerle, H., H. Lassmann. 1994. Contra: evidence against a primary lesion in the target organ in autoimmune disease. Int. Arch. Allergy Immunol. 103: 328-331.
    OpenUrlCrossRefPubMed
  55. ↵
    Ohmori, K., Y. Hong, M. Fujiwara, Y. Matsumoto. 1992. In situ demonstration of proliferating cells in the rat central nervous system during experimental autoimmune encephalomyelitis: evidence suggesting that most infiltrating T cells do not proliferate in the target organ. Lab. Invest. 66: 54-62.
    OpenUrlPubMed
  56. ↵
    Ford, A. L., E. Foulcher, F. A. Lemckert, J. D. Sedgwick. 1996. Microglia induce CD4 T lymphocyte final effector function and death. J. Exp. Med. 184: 1737-1745.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Weber, F., E. Meinl, F. Aloisi, C. Nevinny-Stickel, E. Albert, H. Wekerle, R. Hohlfeld. 1994. Human astrocytes are only partially competent antigen presenting cells: possible implications for lesion development in multiple sclerosis. Brain 117: (Pt. 1):59-69.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Trajkovic, V., O. Vuckovic, S. Stosic-Grujicic, D. Miljkovic, D. Popadic, M. Markovic, V. Bumbasirevic, A. Backovic, I. Cvetkovic, L. Harhaji, et al 2004. Astrocyte-induced regulatory T cells mitigate CNS autoimmunity. Glia 47: 168-179.
    OpenUrlCrossRefPubMed
  59. ↵
    Wekerle, H.. 1993. T-cell autoimmunity in the central nervous system. Intervirology 35: 95-100.
    OpenUrlPubMed
  60. ↵
    Meinl, E., F. Aloisi, B. Ertl, F. Weber, R. de Waal Malefytqq, H. Wekerle, R. Hohlfeld. 1994. Multiple sclerosis: immunomodulatory effects of human astrocytes on T cells. Brain 117: 1323-1332.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Gimsa, U., A. Øren, P. Pandiyan, D. Teichmann, I. Bechmann, R. Nitsch, M. C. Brunner-Weinzierl. 2004. Astrocytes protect the CNS: antigen-specific T helper cell responses are inhibited by astrocyte-induced upregulation of CTLA-4 (CD152). J. Mol. Med. 82: 364-372.
    OpenUrlCrossRefPubMed
  62. ↵
    Girvin, A. M., K. B. Gordon, C. J. Welsh, N. A. Clipstone, S. D. Miller. 2002. Differential abilities of central nervous system resident endothelial cells and astrocytes to serve as inducible antigen-presenting cells. Blood 99: 3692-3701.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Tan, L., K. B. Gordon, J. P. Mueller, L. A. Matis, S. D. Miller. 1998. Presentation of proteolipid protein epitopes and B7-1-dependent activation of encephalitogenic T cells by IFN-γ-activated SJL/J astrocytes. J. Immunol. 160: 4271-4279.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Matheu, V., A. Treschow, I. Teige, V. Navikas, S. Issazadeh-Navikas. 2005. Local therapy with CpG motifs in a murine model of allergic airway inflammation in IFN-β knock-out mice. Respir. Res. 6: 25
    OpenUrlCrossRefPubMed
  65. ↵
    Luca, M. E., L. Visser, C. J. Lucas, L. Nagelkerken. 1999. IFN-β modulates specific T cell responses in vitro but does not affect experimental autoimmune encephalomyelitis in the SJL mouse. J. Neuroimmunol. 100: 190-196.
    OpenUrlCrossRefPubMed
  66. ↵
    Schreiner, B., M. Mitsdoerffer, B. C. Kieseier, L. Chen, H. P. Hartung, M. Weller, H. Wiendl. 2004. Interferon-β enhances monocyte and dendritic cell expression of B7–H1 (PD-L1), a strong inhibitor of autologous T-cell activation: relevance for the immune modulatory effect in multiple sclerosis. J. Neuroimmunol. 155: 172-182.
    OpenUrlCrossRefPubMed
  67. ↵
    Gausling, R., C. Trollmo, D. A. Hafler. 2001. Decreases in interleukin-4 secretion by invariant CD4−CD8− Vα24JαQ T cells in peripheral blood of patients with relapsing-remitting multiple sclerosis. Clin. Immunol. 98: 11-17.
    OpenUrlCrossRefPubMed
  68. ↵
    Miyamoto, K., S. Miyake, T. Yamamura. 2001. A synthetic glycolipid prevents autoimmune encephalomyelitis by inducing TH2 bias of natural killer T cells. Nature 413: 531-534.
    OpenUrlCrossRefPubMed
  69. ↵
    Singh, A. K., M. T. Wilson, S. Hong, D. Olivares-Villagómez, C. Du, A. K. Stanic, S. Joyce, S. Sriram, Y. Koezuka, L. Van Kaer. 2001. Natural killer T cell activation protects mice against experimental autoimmune encephalomyelitis. J. Exp. Med. 194: 1801-1811.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Battistini, L., F. R. Fischer, C. S. Raine, C. F. Brosnan. 1996. CD1b is expressed in multiple sclerosis lesions. J. Neuroimmunol. 67: 145-151.
    OpenUrlPubMed
  71. ↵
    Cipriani, B., L. Chen, K. Hiromatsu, H. Knowles, C. S. Raine, L. Battistini, S. A. Porcelli, C. F. Brosnan. 2003. Upregulation of group 1 CD1 antigen presenting molecules in guinea pigs with experimental autoimmune encephalomyelitis: an immunohistochemical study. Brain Pathol. 13: 1-9.
    OpenUrlPubMed
PreviousNext
Back to top

In this issue

The Journal of Immunology: 177 (6)
The Journal of Immunology
Vol. 177, Issue 6
15 Sep 2006
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
IFN-β Inhibits T Cell Activation Capacity of Central Nervous System APCs
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
IFN-β Inhibits T Cell Activation Capacity of Central Nervous System APCs
Ingrid Teige, Yawei Liu, Shohreh Issazadeh-Navikas
The Journal of Immunology September 15, 2006, 177 (6) 3542-3553; DOI: 10.4049/jimmunol.177.6.3542

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
IFN-β Inhibits T Cell Activation Capacity of Central Nervous System APCs
Ingrid Teige, Yawei Liu, Shohreh Issazadeh-Navikas
The Journal of Immunology September 15, 2006, 177 (6) 3542-3553; DOI: 10.4049/jimmunol.177.6.3542
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606