The contribution of local factors to the activation of immune cells infiltrating the CNS of patients with multiple sclerosis (MS) remains to be defined. The cytokine IL-15 is pivotal in the maintenance and activation of CD8 T lymphocytes, a prominent lymphocyte population found in MS lesions. We investigated whether astrocytes are a functional source of IL-15 sufficient to enhance CD8 T lymphocyte responses and whether they provide IL-15 in the inflamed CNS of patients with MS. We observed that human astrocytes in primary cultures increased surface IL-15 levels upon activation with combinations of proinflammatory cytokines. Expanded human myelin autoreactive CD8 T lymphocytes cultured with such activated astrocytes displayed elevated lytic enzyme content, NKG2D expression, and Ag-specific cytotoxicity. These functional enhancements were abrogated by anti–IL-15–blocking Abs. Immunohistochemical analysis of brain tissue sections obtained from patients with MS demonstrated colocalization for IL-15 and the astrocyte marker glial fibrillary acidic protein within white matter lesions. The majority of astrocytes (80–90%) present in demyelinating MS lesions expressed IL-15, whereas few astrocytes in normal control brain sections had detectable IL-15. IL-15 could be detected in the majority of Iba-1–expressing microglia in the control sections, albeit in lower numbers when compared with microglia/macrophages in MS lesions. Furthermore, infiltrating CD8 T lymphocytes in MS lesions were in close proximity to IL-15–expressing cells. Astrocyte production of IL-15 resulting in the activation of CD8 T lymphocytes ascribes a role for these cells as contributors to the exacerbation of tissue damage during MS pathogenesis.

Multiple sclerosis (MS) is an inflammatory disease of the CNS, pathologically characterized by activated glial cells, demyelination, and axonal loss (1). Immune cells, including macrophages and T lymphocytes, are detected within MS lesions (2). CD4 T lymphocytes are traditionally considered the main effectors in the pathogenesis of MS, but CD8 T lymphocytes have been suggested to play an important role in the tissue damage (3, 4). These cells are present in MS lesions in similar or greater amounts than their CD4 counterparts (59). CD8 T lymphocytes with polarization of their cytolytic granules are observed in close proximity to oligodendrocytes and demyelinated axons (6). These lymphocytes bear an activated effector memory phenotype, are oligoclonally expanded, and persist over time in the CNS of patients with MS (6, 912). Finally, animal models have illustrated the capacity of CD8 T lymphocytes to mediate MS-like diseases (1317).

IL-15 is essential for the development, activation, and survival of CD8 T lymphocytes and NK cells (18, 19). Monocytes/macrophages and dendritic cells have been reported as the main source of this cytokine (20, 21). Intracellular IL-15 binds to the high-affinity IL-15Rα; this complex is subsequently transported to the cell surface for an efficient cross-presentation to other cells. The surface IL-15–IL-15Rα complex stimulates cells bearing the IL-2/IL15Rβ and common γ chain (2224). Increased local expression of IL-15 has been suggested to contribute to the immunopathology of several human inflammatory diseases including rheumatoid arthritis, celiac disease, and inflammatory bowel disease (2528).

The precise contribution of IL-15 to MS immunopathogenesis has not been elucidated, although elevated IL-15 in serum and in PBMCs at both the mRNA and protein levels in patients with MS has been reported (2932). IL-15 mRNA has been detected in murine and human CNS tissue and in primary cultures of astrocytes and microglia (3336). However, IL-15 protein levels do not correlate with mRNA detection, and cell membrane rather than secreted IL-15 is crucial in mediating in vivo effects (22, 37). Thus, it is essential to document functional surface IL-15 protein expression in human CNS cells. Although the IL-15 protein has been detected on astrocytes in inflammatory mouse models (36, 38), whether these nonmyeloid cells represent an important and functional IL-15 source in human CNS diseases specifically for CD8 T cells has not been addressed.

Astrocytes are implicated as contributors to both innate and adaptive immune responses taking place in the CNS (39). Our study shows that conditions mimicking CNS inflammation as observed in MS result in a robust increase in surface-bound IL-15 on human astrocytes in vitro. Moreover, this surface expression boosted the cytotoxicity of human CD8 T lymphocytes. We also demonstrate that IL-15 expression is upregulated on astrocytes within MS lesions. Taken together, our results demonstrate that astrocytes, via the production of IL-15, participate in enhancing effector responses of CNS-infiltrating CD8 T lymphocytes and thus are potential important contributors to tissue injury in MS.

Human fetal CNS tissue was obtained from Albert Einstein College of Medicine (Bronx, NY). Their ethical review boards and McGill University’s approved the studies. Astrocytes and microglia were isolated as previously described (40) and grown in DMEM containing 10% (v/v) FBS. Adult microglia were isolated from surgical resections performed for the treatment of non–tumor-related intractable epilepsy, in accordance with guidelines of the ethical board of McGill University and as previously described (41). Cells were treated for 24 h with IL-1β (10 ng/ml; Medicorp, Montreal, Quebec, Canada), IFN-γ (200 U/ml = 3.6 ng/ml; Biosource International, Camarillo, CA), TNF (2000 U/ml = 20 ng/ml; Biosource International), combinations of those cytokines, or IFN-γ followed by LPS (100 ng/ml; Sigma-Aldrich, Oakville, Ontario, Canada).

Informed consent was obtained from healthy donors for these studies that were approved by McGill University and Centre Hospitalier de l’Université de Montréal ethical boards (Montreal, Quebec, Canada). PBMCs isolated by Ficoll density gradient were plated in RPMI 1640 containing 10% (v/v) FBS, and floating cells were removed by washing after 2 h, leaving adherent monocytes.

CD8 T cell lines specific for influenza matrix protein 1 aa 58–66 (influenza: GILGFVFTL) or myelin basic protein (MBP) aa 111–119 (MBP: SLSRFSWGA) were expanded as previously published (42). CD8 T cell lines were stimulated with autologous Ag-loaded B cells once a week, and IL-15 (10 ng/ml; PeproTech, Rocky Hill, NJ) was added both concurrently with fresh peptide loaded APCs and 3d later. CD8 T cell lines were considered specific when the amount of IFN-γ secreted after 24–36 h (ELISA from BD Biosciences, Mississauga, Ontario, Canada) in the presence of specific peptide-loaded autologous B cells was at least twice the amount secreted in the presence of irrelevant peptide-loaded B cells (Table I).

Table I.
Specificity of CD8 T cell lines as assessed by IFN-γ secretion
IFN-γ (pg/ml)
Ag–CD8 T Cell LineB Cells Plus Influenza (SI)B Cells Plus MBP (SI)
MBP line 1 32 89 (2.7) 
MBP line 2 56 146 (2.6) 
MBP line 3 54 118 (2.2) 
Influenza line 1 991 (5.9) 167 
Influenza line 2 1001 (5.0) 197 
Influenza line 3 840 (6.7) 124 
IFN-γ (pg/ml)
Ag–CD8 T Cell LineB Cells Plus Influenza (SI)B Cells Plus MBP (SI)
MBP line 1 32 89 (2.7) 
MBP line 2 56 146 (2.6) 
MBP line 3 54 118 (2.2) 
Influenza line 1 991 (5.9) 167 
Influenza line 2 1001 (5.0) 197 
Influenza line 3 840 (6.7) 124 

SI, stimulation index.

Astrocytes, microglia, and monocytes were detached and stained for surface and intracellular molecules as previously described (40) and subsequently acquired on an FACSCalibur or LSR II (BD Biosciences). Mouse mAbs directed at human protein either unconjugated or conjugated to FITC, Alexa Fluor 488, Pacific Blue, PE, PE-Cy5, or allophycocyanin were used. Surface staining targeted: IL-15, IL-15Rα (R&D Systems, Minneapolis, MN), MHC class I (MHC-I; HLA-A/B/C), CD14, and CD8 (BD Biosciences). Intracellular staining targeted: perforin, granzyme B (BD Biosciences), or glial fibrillary acidic protein (GFAP) (Molecular Probes, Invitrogen Life Technologies, Burlington, Ontario, Canada). In some experiments, dead cells were excluded using 7-aminoactinomycin D (BD Biosciences) according to the manufacturer’s instructions. Appropriate isotype controls were used in all steps. Flow cytometry analyses were performed using FlowJo (Tree Star, Ashland, OR), and the change in median fluorescence intensity (ΔMFI) was calculated by subtracting the fluorescence of the isotype from that of the stain.

Astrocytes were plated at 1 × 105 cells in 500 μl/well in a 48-well plate and allowed to grow up to ∼70% confluence prior to being activated with cytokines (IL-1β + IFN-γ) for 24 h and finally treated with monensin (5 nM) for 1 h. After two washes, blocking anti–IL-15 Ab (20 μg/ml) (clone 34559, R&D Systems) or an isotype control was added for 1 h. The blocking anti–IL-15 mAb (clone 34559) used in our experiments is a well-characterized blocking reagent, as demonstrated by several groups (20, 4345). Subsequently, 5 × 104 CD8 T lymphocytes were added per well, and the coculture was maintained for 48 h in a 500-μl volume, after which cells were harvested, then stained and analyzed by flow cytometry as described above. For some experiments, we compared two commercially available IL-15–blocking Abs (clones 34559 and 34505, R&D Systems) and obtained similar results. To test the cytotoxic activity of CD8 T lymphocytes cocultured with astrocytes, autologous B cells previously loaded with specific or control Ag were added at a 1:1 ratio concurrently with anti-CD107a PE-conjugated Ab (BD Biosciences) and monensin A (5 nM) to prevent rapid internalization of CD107a by endocytosis, as used by others (46, 47). Five hours later, cells were harvested and stained for CD8 and analyzed by flow cytometry.

Total RNA was isolated using TRIzol (Invitrogen Life Technologies) and the Qiagen RNeasy mini kit (Qiagen, Mississauga, Ontario, Canada) according to manufacturer’s instructions and as previously described (40, 41). RNA samples were treated with DNase (Qiagen) and then transcribed into cDNA using random hexaprimers (Roche, Mississauga, Ontario, Canada) with the Moloney murine leukemia virus-RT enzyme (Invitrogen Life Technologies) at 42°C. IL-15 expression level was determined by quantitative real-time PCR (qPCR) using the ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, CA). Primers and TaqMan FAM-labeled MGB probes for IL-15 were obtained from Applied Biosystems (TaqMan Gene Expression Assays). β-Actin primers and cycling parameters have been previously published (41). qPCR cycling was performed according to the ABI Prism 7000 Sequence Detection System (Applied Biosystems) default temperature settings (2 min at 50°C and 10 min at 95°C, followed by 40 cycles of 15 s at 95°C and 1 min at 60°C) in a volume of 25 μl with 1× TaqMan Universal Master Mix (Applied Biosystems). For relative mRNA expression, IL-15 amplification was normalized to endogenous control and cDNA from cells found to have high IL-15 expression.

Tissue sections of postmortem brains from donors without CNS disease and patients diagnosed clinically and confirmed by neuropathological examination as having MS were obtained from the NeuroResource Tissue Bank, University College London Institute of Neurology, London, U.K. CNS tissues were donated to the tissue bank with informed consent following ethical review by the London Research Ethics Committee, London, U.K. This study was approved by the Centre Hospitalier de l’Université de Montreal Ethical Committee. Snap-frozen coded sections (∼1-cm2 and 10-μm thick) were cut from blocks of normal control and MS brain tissues. Sections cut before and immediately after the ones used for the immunofluorescence studies were stained with Oil Red O (ORO) and hematoxylin and scored as previously described (48) (Table II). Serial sections were air-dried and fixed in cold acetone for 10 min and then blocked for nonspecific binding for 1 h in HBSS containing 2% (v/v) horse serum, 2% (v/v) FBS, 1 mM HEPES buffer, and 0.1% (w/v) sodium azide, and then blocked for endogenous biotin or biotin-binding proteins using the Blocking Kit from Vector Laboratories (Burlington, Ontario, Canada) following the manufacturer’s instructions. All Abs were diluted in HBSS containing 2% (v/v) horse serum, 2% (v/v) FBS, 1 mM HEPES buffer, and 0.1% (w/v) sodium azide, and washes were done in PBS. Staining for IL-15 was performed overnight at 4°C using monoclonal mouse anti-human IL-15 Ab (25 μg/ml; R&D Systems clone 34559). Sections were subsequently incubated for 2 h with biotinylated polyclonal goat anti-mouse Abs (DakoCytomation, Mississauga, Ontario, Canada), then incubated for 1.5 h with Cy3-conjugated streptavidin (Jackson ImmunoResearch Laboratories, West Grove, PA) and Alexa Fluor 488-conjugated anti-GFAP mouse mAb (10 μg/ml; Molecular Probes), FITC-conjugated anti-CD68 mouse mAb (2 μg/ml; Biolegend, San Diego, CA), rabbit anti–Iba-I Ab (5 μg/ml; Wako Chemicals USA, Richmond, VA), or allophycocyanin-conjugated anti-CD8 (BD Biosciences) mouse mAb for at least 2 h at room temperature. Iba-I staining was followed by Alexa Fluor 488-conjugated goat anti-rabbit Abs (Invitrogen Life Technologies), whereas CD8 staining was followed by rabbit anti-allophycocyanin Abs and FITC-conjugated donkey anti-rabbit Abs for 2 h each. Sections were incubated for 30 min with RNase (100 μg/ml; Sigma-Aldrich) and then TO-PRO-3 iodide (3.3 μM; Invitrogen Life Technologies) for 10 min. To quench tissue autofluorescence, sections were treated with 1% (w/v) Sudan black (Sigma-Aldrich) in 70% ethanol for 3 min and washed thoroughly. Control stainings were concurrently carried out on adjacent sections using appropriate primary isotype controls (for IL-15, mouse IgG1 [Sigma-Aldrich]; for GFAP, Alexa Fluor 488-conjugated mouse IgG1 [Caltag Laboratories, Burlingame, CA]; for CD68, FITC-conjugated mouse IgG2a [BD Biosciences]; for CD8, allophycocyanin-conjugated mouse IgG1 [BD Biosciences]; for Iba-1, rabbit Igs [Jackson ImmunoResearch Laboratories]) at the same concentrations. Slides were observed using a Leica confocal microscope SP5 (Leica Microsystems, Richmond Hill, Ontario, Canada). Confocal images were acquired simultaneously in different channels throughout a 4–8 μm z-stack every 0.2–0.5 μm. We validated staining specificity by lack of signal when the corresponding laser was turned off but with the others still on. Several fields (more than five) containing cells positive for the relevant specific cell marker were taken for each section and used to assess the proportion of cells expressing IL-15. Moreover, we confirmed the absence of bleed-through by re-examination of selected sections via sequential scanning.

Table II.
Description of postmortem brain sections
Block No.SexAge (y)DD (y)Cause of DeathDFT (h)Sample TypeORO ScoreaSummary Observations on ORO-Stained Sections
Female 47 – Respiratory failure 20 NC W, FV, R 0, 1 All white matter 
Female 68 – Colorectal metastatic tumor 23 NC W, OV, R 0, 0 All white matter 
Male 49 – Myocardial infarction and coronary artery thrombosis 11 NC W, PV, R 0, 0 White matter with small edge of cortical gray matter 
Female 47 20 Bronchopneumonia MS AQ, OSv, L 5, 2 Large, very active MS plaque with white matter 
Female 47 20 Bronchopneumonia MS AQ, FSv, L 4, 2 Cortical gray matter surrounding subcortical normal-appearing white matter with an active lesion in center of section 
Female 29 Bronchopneumonia 11 MS acute and SAQs, CB, L 3, 2 Abnormal-appearing white matter with eight individual areas of acute or subacute plaque 
Male 69 21 Multipleb 14 MS CQ, OV, R
WM 0, 0
0, 2 Chronic plaque along patchy white matter containing perivenular cuffing 
Female 46 22 Bronchopneumonia MS SAQ, CB, R 2, 1 Cerebellar gray and white matter with a large chronic plaque containing a small subacute area 
Female 60 34 Renal failure 24 MS AQs, TV, L 3, 4 White matter, large chronic plaque, two active plaques and one subacute plaque; large perivenular cuffs 
10 Female 49 11 Bronchopneumonia 16 MS SAQ, Pons 1, 2 Mostly chronic plaque with scattered ORO-positive macrophages; abnormal-appearing white matter 
Block No.SexAge (y)DD (y)Cause of DeathDFT (h)Sample TypeORO ScoreaSummary Observations on ORO-Stained Sections
Female 47 – Respiratory failure 20 NC W, FV, R 0, 1 All white matter 
Female 68 – Colorectal metastatic tumor 23 NC W, OV, R 0, 0 All white matter 
Male 49 – Myocardial infarction and coronary artery thrombosis 11 NC W, PV, R 0, 0 White matter with small edge of cortical gray matter 
Female 47 20 Bronchopneumonia MS AQ, OSv, L 5, 2 Large, very active MS plaque with white matter 
Female 47 20 Bronchopneumonia MS AQ, FSv, L 4, 2 Cortical gray matter surrounding subcortical normal-appearing white matter with an active lesion in center of section 
Female 29 Bronchopneumonia 11 MS acute and SAQs, CB, L 3, 2 Abnormal-appearing white matter with eight individual areas of acute or subacute plaque 
Male 69 21 Multipleb 14 MS CQ, OV, R
WM 0, 0
0, 2 Chronic plaque along patchy white matter containing perivenular cuffing 
Female 46 22 Bronchopneumonia MS SAQ, CB, R 2, 1 Cerebellar gray and white matter with a large chronic plaque containing a small subacute area 
Female 60 34 Renal failure 24 MS AQs, TV, L 3, 4 White matter, large chronic plaque, two active plaques and one subacute plaque; large perivenular cuffs 
10 Female 49 11 Bronchopneumonia 16 MS SAQ, Pons 1, 2 Mostly chronic plaque with scattered ORO-positive macrophages; abnormal-appearing white matter 
a

ORO and cuffing: scored on a scale of 0–5 for ORO and hematoxylin staining; 0 is what would be expected in normal control white matter. Data are averaged from serial, duplicate sections cut immediately before and after the section examined by immunocytochemistry.

b

1A, myocardial infarction; 1B, pneumonia; 1C, diabetes mellitus; 2, MS.

–, not applicable; AQ, active plaque; CB, cerebellum; CQ, chronic plaque; DD, disease duration; DFT, death to snap-freezing time; F, frontal; L, left; NC W, normal control white matter; O, occipital; P, parietal; R, right; SAQ, subacute plaque; Sv, subventricular; T, temporal; V, ventricular; WM, white matter.

Data handling and analysis (Student t test) were performed using Prism 3.0 (GraphPad, La Jolla, CA).

The surface-bound form of IL-15 is mainly responsible for cellular signaling to neighboring cells (22). Thus, to determine whether human CNS cells are a relevant source of this cytokine, we assessed the presence of surface IL-15 on human astrocytes. Highly enriched primary cultures of human astrocytes (>95% pure) (Fig. 1A) were either left alone or incubated for 24 h with IL-1β, IFN-γ, TNF, IFN-γ + TNF, or IFN-γ + IL-1β prior to being stained for IL-15 and IL-15Rα. A small proportion of astrocytes (1–15%) expressed surface IL-15 when left untreated or treated with individual cytokines IL-1β, IFN-γ, or TNF (Fig. 1B). However, treatment with the combinations of IFN-γ + TNF or IFN-γ + IL-1β both resulted in surface IL-15 expression in >55% of astrocytes (Fig. 1B). These observations were confirmed with astrocytes obtained from multiple donors (Fig. 1C; untreated [10.0 ± 2.9%] versus IFN-γ + TNF [55.6 ± 10.7%] or IFN-γ + IL-1β [56.4 ± 9.0%]; *p < 0.05), although the absolute percentage observed varied from donor to donor. All treatments that induced IL-15 were associated with significantly increased MHC-I expression on the astrocytes (Fig. 1D, untreated versus cytokine treatment; *p < 0.02). We also confirmed using qPCR that cytokine combinations IFN-γ + TNF and IFN-γ + IL-1β increased mRNA level coding for IL-15 in human astrocytes (Fig. 1E). A large proportion of astrocytes basally expressed IL-15Rα, and this percentage increased for some donors following cytokine treatments compared with untreated cells (Fig. 1B, 1F). We also performed ELISA to detect IL-15 in astrocyte-conditioned media, but secreted levels were below the minimum detection threshold of the assay (10 pg/ml). Moreover, when we added soluble rIL-15 to astrocytes, we could not detect surface binding of the cytokine after 5 h of incubation at 37°C. This suggests that astrocytes synthesized IL-15, which was then translocated and expressed on the cell surface bound to its IL-15Rα, similar to other cell types (22).

FIGURE 1.

Proinflammatory cytokines enhance IL-15 expression by human astrocytes. Primary cultures of human astrocytes were analyzed for IL-15 and IL-15Rα expression under basal conditions and following proinflammatory cytokine treatments. A, Typical purity of astrocytes expressing GFAP as observed by flow cytometry. B, Representative dot plots indicating the percentage of astrocytes from one donor expressing surface IL-15 and IL-15Rα either under basal conditions (untreated) or following cytokine treatment (each treatment indicated above dot plot). The combination of IFN-γ + TNF or IFN-γ + IL-1β significantly increased the percentage of astrocytes expressing surface IL-15. Quadrants are drawn according to isotype controls depicted in the leftmost panels. C, Results obtained from four astrocyte donors for the percentage of IL-15–expressing astrocytes. D, Astrocytes upregulated MHC-I expression (ΔMFI) following proinflammatory cytokine treatments. Each dot represents a distinct astrocyte donor. E, Relative mRNA expression of IL-15 as determined using qPCR for three astrocyte donors. F, Results obtained from four astrocyte donors for the percentage of IL-15Rα–expressing cells. Statistical analyses comparing untreated versus cytokine treatment. *p < 0.05.

FIGURE 1.

Proinflammatory cytokines enhance IL-15 expression by human astrocytes. Primary cultures of human astrocytes were analyzed for IL-15 and IL-15Rα expression under basal conditions and following proinflammatory cytokine treatments. A, Typical purity of astrocytes expressing GFAP as observed by flow cytometry. B, Representative dot plots indicating the percentage of astrocytes from one donor expressing surface IL-15 and IL-15Rα either under basal conditions (untreated) or following cytokine treatment (each treatment indicated above dot plot). The combination of IFN-γ + TNF or IFN-γ + IL-1β significantly increased the percentage of astrocytes expressing surface IL-15. Quadrants are drawn according to isotype controls depicted in the leftmost panels. C, Results obtained from four astrocyte donors for the percentage of IL-15–expressing astrocytes. D, Astrocytes upregulated MHC-I expression (ΔMFI) following proinflammatory cytokine treatments. Each dot represents a distinct astrocyte donor. E, Relative mRNA expression of IL-15 as determined using qPCR for three astrocyte donors. F, Results obtained from four astrocyte donors for the percentage of IL-15Rα–expressing cells. Statistical analyses comparing untreated versus cytokine treatment. *p < 0.05.

Close modal

Previous studies (3336) reported that microglia express IL-15 mRNA without evaluating whether this cytokine was present on the cell membrane. Thus, we evaluated whether human microglia also provide surface IL-15 and compared adult microglia to human peripheral monocytes, which derive from the same lineage. Adult microglia and monocytes were either untreated or stimulated for 24 h and then stained for surface IL-15, MHC-I, CD11c (for microglia), and CD14 (for monocytes) (Fig. 2A). We confirmed on numerous samples that microglia concurrently express both CD68 and CD11c (data not shown). A proportion of human adult microglia did express surface IL-15 (∼40%) (Fig. 2A) under basal culture conditions, but this expression did not increase posttreatment [IFN-γ + LPS, IFN-γ + IL-1β, IFN-γ + TNF (Fig. 2A and data not shown)] despite increased MHC-I expression, as we have previously shown (49). Surface IL-15 expression was nearly undetectable in untreated monocytes; however, following their activation with IFN-γ + LPS, a well-known combination that activates these myeloid cells, most cells expressed surface IL-15 as previously shown (20).

FIGURE 2.

Human microglia express surface IL-15. A, Monocytes (top panels) and adult microglia (bottom panels) were either untreated or activated with IFN-γ + LPS and then analyzed for CD14 or CD11c and IL-15 surface expression. Representative dot plots illustrate the percentage of IL-15–expressing CD14+ or CD11c+ cells for each cell type. Quadrants are drawn according to isotype controls (leftmost panels) for each individual cell type and treatment. B, Fetal astrocytes and microglia from the same donor were either untreated or activated with IFN-γ + TNF, IFN-γ + IL-1β, or IFN-γ + LPS (microglia) and then analyzed for IL-15 expression and CD11c (microglia) or MHC-I (astrocytes). For all panels, results shown are representative of at least two donors per cell type.

FIGURE 2.

Human microglia express surface IL-15. A, Monocytes (top panels) and adult microglia (bottom panels) were either untreated or activated with IFN-γ + LPS and then analyzed for CD14 or CD11c and IL-15 surface expression. Representative dot plots illustrate the percentage of IL-15–expressing CD14+ or CD11c+ cells for each cell type. Quadrants are drawn according to isotype controls (leftmost panels) for each individual cell type and treatment. B, Fetal astrocytes and microglia from the same donor were either untreated or activated with IFN-γ + TNF, IFN-γ + IL-1β, or IFN-γ + LPS (microglia) and then analyzed for IL-15 expression and CD11c (microglia) or MHC-I (astrocytes). For all panels, results shown are representative of at least two donors per cell type.

Close modal

We also compared IL-15 expression by fetal astrocytes and microglia from the same donor following IFN-γ + TNF or IFN-γ + IL-1β exposure (Fig. 2B). A small percentage (9.7%) of human fetal microglia isolated from the same material as human fetal astrocytes expressed IL-15 at basal levels (Fig. 2B). The proportions of fetal microglia expressing surface IL-15 increased following IFN-γ + TNF or IFN-γ + IL-1β (33.2 or 19.2%) treatment (Fig. 2B, one donor representative of three) as did astrocytes from the same donor (10.3% untreated versus 31.4 and 36.3%). Thus, both human adult and fetal microglia can provide surface IL-15.

In an effort to model the CD8 T lymphocytes found in MS lesions as closely as possible, we evaluated the effects of IL-15 on Ag-specific CD8 T lymphocytes (3). Human Ag (MBP or influenza)-specific CD8 T cell lines (see typical examples in Table I) were characterized by flow cytometry according to their effector memory profile (loss of CCR7), expression of lytic enzymes (perforin and granzyme B), and expression of NKG2D, a coactivating receptor we have previously shown to be involved in oligodendrocyte killing (40). Ag-specific CD8 T lymphocytes were cocultured with either untreated astrocytes or astrocytes pretreated with IFN-γ + IL-1β (inducing a high proportion of surface IL-15–expressing cells, as per Fig. 1) for 48 h in the presence of either an Ab against IL-15 or an isotype control. Flow cytometric data from 1 representative CD8 T cell line are illustrated in Fig. 3A–C, and data from 10 CD8 T cell lines are summarized in Fig. 3D–F. The proportion of CD8 T lymphocytes expressing granzyme B and perforin was low in untreated CD8 T lymphocytes (Fig. 3A, 6%; 3D, average of 10 CD8 T cell lines: 3.6 ± 0.5%), but increased at least 3-fold in the presence of rIL-15 (Fig. 3A, 24%, top panels; 3D, average of 10 CD8 T cell lines: 22.8 ± 1.4%). CD8 T lymphocytes cultured on untreated astrocytes showed a modest increase in the proportion of cells expressing both perforin and granzyme B (Fig. 3A, alone 6% versus untreated astrocytes: 14%; 3D, average of 10 CD8 T cell lines: 3.6 ± 0.5 versus 12.7 ± 1.0%). This effect was most likely due to low basal expression of IL-15 by untreated astrocytes because it was efficiently blocked by anti–IL-15 Ab (Fig. 3A, 7%; 3D, average of 10 CD8 T cell lines: 5.4 ± 0.6%). The ΔMFI of granzyme B was also elevated following coculture with astrocytes compared with untreated CD8 T lymphocytes (Fig. 3E, data from 10 CD8 T cell lines: alone, 135 ± 25 versus untreated astrocytes, 496 ± 60). CD8 T lymphocytes cultured on treated astrocytes in the presence of the isotype control displayed a pronounced 5-fold increase in the proportion of cells expressing perforin and granzyme B (Fig. 3A, 30%; 3D, data for 10 CD8 T cell lines: alone, 3.6 ± 0.5% versus treated astrocytes plus isotype, 28.0 ± 1.7%) and in the level of granzyme B per cell (Fig. 3E, ΔMFI of granzyme B: alone, 135 ± 25 versus treated astrocytes plus isotype, 1171 ± 208; ***p < 0.001). Culture of CD8 T lymphocytes on treated astrocytes in the presence of anti–IL-15 Ab significantly reduced the percentage of CD8 T lymphocytes expressing perforin and granzyme B (Fig. 3A, 8%; Fig. 3D, average of 10 CD8 T cell lines 8.5 ± 0.8%; ***p < 0.001 compared with isotype). Thus, surface IL-15 provided by astrocytes significantly elevated the expression of lytic enzymes by human CD8 T lymphocytes.

FIGURE 3.

IL-15 provided by human astrocytes enhances the cytotoxic profile of human CD8 T lymphocytes. MBP-specific CD8 T cells (AF) were left alone, cultured with recombinant human IL-15, or cocultured with untreated or IL-1β + IFN-γ–treated astrocytes (Treated) in the presence of an isotype control (Iso.) or an anti–IL-15 blocking Ab (α-IL15) and then stained for CD8, CCR7, NKG2D, perforin, and granzyme B. Representative dot plots gated on CD8 T cells illustrating the detection of perforin and granzyme B (A), granzyme B and CCR7 (B), or NKG2D (C) observed with one MBP-specific human CD8 T cell line. IL-15 provided as a recombinant protein or by astrocytes increased proportion of lytic enzyme-expressing cells (D) and granzyme B (E) and NKG2D (F) content on a per-cell basis (ΔMFI) by CD8 T cells. Each dot represents one distinct MBP-specific CD8 T cell line obtained from two donors. Results are representative of 18 different CD8 T cell lines generated from six donors, specific for either influenza or MBP and cultured on astrocytes from three donors. G, Ag-specific CD8 T lymphocytes were left alone, cultured with rIL-15 (+IL-15), or cocultured with IL-1β + IFN-γ–treated astrocytes in the presence of an isotype or an anti–IL-15–blocking Ab prior to the addition of anti-CD107a Ab and autologous target cells loaded with specific or control Ag. Results pooled from two different CD8 T cell lines performed in triplicate are illustrated. Statistical analyses comparing adequate groups as illustrated by lines in the figure. *p < 0.05; **p < 0.01; ***p < 0.001.

FIGURE 3.

IL-15 provided by human astrocytes enhances the cytotoxic profile of human CD8 T lymphocytes. MBP-specific CD8 T cells (AF) were left alone, cultured with recombinant human IL-15, or cocultured with untreated or IL-1β + IFN-γ–treated astrocytes (Treated) in the presence of an isotype control (Iso.) or an anti–IL-15 blocking Ab (α-IL15) and then stained for CD8, CCR7, NKG2D, perforin, and granzyme B. Representative dot plots gated on CD8 T cells illustrating the detection of perforin and granzyme B (A), granzyme B and CCR7 (B), or NKG2D (C) observed with one MBP-specific human CD8 T cell line. IL-15 provided as a recombinant protein or by astrocytes increased proportion of lytic enzyme-expressing cells (D) and granzyme B (E) and NKG2D (F) content on a per-cell basis (ΔMFI) by CD8 T cells. Each dot represents one distinct MBP-specific CD8 T cell line obtained from two donors. Results are representative of 18 different CD8 T cell lines generated from six donors, specific for either influenza or MBP and cultured on astrocytes from three donors. G, Ag-specific CD8 T lymphocytes were left alone, cultured with rIL-15 (+IL-15), or cocultured with IL-1β + IFN-γ–treated astrocytes in the presence of an isotype or an anti–IL-15–blocking Ab prior to the addition of anti-CD107a Ab and autologous target cells loaded with specific or control Ag. Results pooled from two different CD8 T cell lines performed in triplicate are illustrated. Statistical analyses comparing adequate groups as illustrated by lines in the figure. *p < 0.05; **p < 0.01; ***p < 0.001.

Close modal

CD8 T cells responding to IL-15 provided as a recombinant protein or by treated astrocytes were determined to be effector memory cells because cells that acquired granzyme B expression did not express CCR7 (Fig. 3B). We also observed that CD8 T cells cultured in the same conditions expressed elevated levels of NKG2D compared with CD8 T cells cultured alone (Fig. 3C, 3F, average 10 CD8 T cell lines ΔMFI of NKG2D: alone, 10.3 ± 1.1; treated astrocytes plus isotype control, 31.6 ± 1.9; treated astrocytes with anti–IL-15, 17.6 ± 1.0; isotype versus anti–IL-15 ***p < 0.001). These results indicate that IL-15 provided by astrocytes enhanced NKG2D levels on human CD8 T lymphocytes. Supernatants of activated astrocytes were not able to increase perforin, granzyme B, or NKG2D expression levels in CD8 T lymphocytes, suggesting that the observed effects by astrocytes were mediated by membrane-bound IL-15 (data not shown). Moreover, cocultures of astrocytes and CD8 T lymphocytes separated by a transwell did not reproduce these results, supporting the cell–cell contact dependence of our observations (data not shown). We obtained similar results with MBP- and influenza-specific CD8 T cell lines, suggesting that regardless of their Ag specificity, CD8 T lymphocytes infiltrating the CNS respond to IL-15 provided by astrocytes (Fig. 3). To confirm that astrocyte-provided IL-15 had an impact on ex vivo-purified CD8 T cells, IFN-γ + IL-1β–activated astrocytes were cocultured for 6 d with CFSE-labeled ex vivo-isolated heterologous CD8 T lymphocytes. The addition of Ab against IL-15 did significantly decrease the proportion of CD8 T cells expressing granzyme B compared with the isotype control (anti–IL-15: 12% versus isotype 21%; p < 0.05), without significantly affecting the proliferation of CD8 T cells (24 versus 21%). To rule out that only IL-15 pre-exposed T lymphocytes were sensitive to IL-15–mediated effects, cell lines cultured with IL-2 from the day of isolation were also incubated with rIL-15. The response of these cells was the same as cells cultured with IL-15 (data not shown).

To determine whether the increased content of lytic enzymes in CD8 T lymphocytes correlated with augmented cytotoxic activity, we used the well-characterized CD107a-mobilization flow cytometry-based assay (Fig. 3G). Lytic enzymes are stored in intracellular granules bearing CD107a. In the presence of appropriate target cells, these enzymes are released, resulting in the transient expression of CD107a on the surface of effector cells. The CD107a mobilization assay has been shown to correlate with degranulation and killing activity (as measured by [51Cr] release) of human CD8 T lymphocytes (46, 47, 50). To evaluate the consequences of the CD8 T cell–astrocyte interaction on the subsequent encounter of CD8 T cells with their specific target cells, we added autologous B cells loaded with either specific or control peptide in the astrocyte–CD8 T lymphocyte coculture. CD8 T lymphocytes cultured with rIL-15 showed a 3-fold greater proportion of CD107a surface-expressing cells in presence of target cells pulsed with the appropriate Ag in comparison with CD8 T lymphocytes left alone (12 versus 4%) (Fig. 3G). IL-15 provided by treated astrocytes also increased the proportion of CD8 T lymphocytes expressing CD107a reaching 10% with an isotype control; this proportion was significantly diminished when anti–IL-15–blocking Ab was included in the coculture (6%) (Fig. 3G). Importantly, degranulation of CD8 T lymphocytes was Ag-specific because the addition of control Ag-loaded target cells did not trigger any significant response even in the presence of an IL-15 source (1–4%) (Fig. 3G).

To assess whether astrocytes could provide IL-15 in vivo, especially within MS lesions, we performed immunohistochemistry on postmortem brain tissues obtained from patients with MS and normal controls (description of samples provided in Table II). Sections were stained with GFAP and IL-15–specific Abs or appropriate isotype controls. At least five distinct fields (at ×400, each field comprises 0.09 mm2) per section from three nonneurologic disease controls and six patients with MS containing GFAP-positive cells were thoroughly analyzed, and representative fields are illustrated (Fig. 4). All MS sections examined (n = 6 donors) displayed strong immunoreactivity for GFAP, especially within and at the edges of demyelinated lesions. A high proportion of astrocytes (>80–90% as assessed by quantification of 21 distinct fields from 6 MS lesions for >220 GFAP+ cells) expressed high levels of IL-15 (Fig. 4H, 4J, 4M, 4O, white arrows). IL-15 was detected in astrocytes in acute (Fig. 4, block 5) and subacute/chronic (Fig. 4, block 10) MS lesions. Colocalization of IL-15 and GFAP was confirmed by scanning the tissue at different z planes. We also detected strong IL-15 immunoreactivity in astrocytes near blood vessels with or without perivascular cuffs (data not shown). In all sections from controls, we also detected GFAP+ astrocytes. However, the majority of these cells (>80%) did not express detectable IL-15, with only a few showing weak expression (Fig. 4B, 4C, white arrow).

FIGURE 4.

Astrocytes strongly express IL-15 in MS lesions. CNS tissue from one control (block 1: AE) and lesions from two patients with MS (block 5: FJ; block 10: KO) were stained for GFAP (green: A, F, K) and IL-15 (red: B, G, L) and merge of GFAP and IL-15 (C, H, M). D, I, and N, Isotype controls. E, J, and O show a magnified view of the cell in the white box. Several IL-15–positive cells were astrocytes, as indicated by the yellow overlay in H, M, J, and O. White arrows indicate examples of astrocytes colabeled for IL-15. Orange arrowheads indicate cells in both controls and MS tissue samples that expressed IL-15 but were not astrocytes. Pictures shown are z stacks of five to eight 0.5-μm-thick layers and are representative of five fields taken from each section of three controls and seven MS donors. Original magnification ×400. Scale bar, 50 μm.

FIGURE 4.

Astrocytes strongly express IL-15 in MS lesions. CNS tissue from one control (block 1: AE) and lesions from two patients with MS (block 5: FJ; block 10: KO) were stained for GFAP (green: A, F, K) and IL-15 (red: B, G, L) and merge of GFAP and IL-15 (C, H, M). D, I, and N, Isotype controls. E, J, and O show a magnified view of the cell in the white box. Several IL-15–positive cells were astrocytes, as indicated by the yellow overlay in H, M, J, and O. White arrows indicate examples of astrocytes colabeled for IL-15. Orange arrowheads indicate cells in both controls and MS tissue samples that expressed IL-15 but were not astrocytes. Pictures shown are z stacks of five to eight 0.5-μm-thick layers and are representative of five fields taken from each section of three controls and seven MS donors. Original magnification ×400. Scale bar, 50 μm.

Close modal

We also observed IL-15 expression in GFAP-negative cells in MS sections (Fig. 4H, 4M, orange arrowheads). To identify these cells, adjacent sections were stained for IL-15 and Iba-1 or CD68, two markers characteristic of macrophages/microglia. The number of Iba-1+ cells (Fig. 5A, 5E, 5I) was proportional to the ORO scoring, a well-characterized immunohistopathological staining that detects lipids in phagocytic macrophages (Table II); in MS lesions with an ORO score >3 (blocks 4–6 and 9), >250 macrophages/microglia per mm2 were detected, whereas <100/mm2 were observed in the other sections (blocks 8 and 10). Seventy-five percent of the 450 macrophages/microglia counted from a total of 24 fields of seven MS lesions (60–90% for individual sections) expressed IL-15 (Fig. 5G, 5K), although some myeloid cells had undetectable IL-15 (Fig. 5K, orange arrow). Similar results were obtained when CD68 was used to identify macrophages/microglia (data not shown). In these sections, we also observed numerous IL-15+ cells with an astrocyte-like morphology that were not macrophages/microglia (Fig. 5G, white arrowheads), consistent with our results presented above showing colocalization of IL-15 with GFAP-expressing cells. Fewer Iba-1+ cells were detected in control sections (Fig. 5) compared with MS tissues, and a proportion of them expressed IL-15 (34 of 53 microglia counted in a total of 15 fields from the 3 control sections (64%) (Fig. 5A–C). Thus, overall, the number of Iba-1+ cells expressing detectable IL-15 levels per surface area was strongly reduced in controls compared with MS tissues.

FIGURE 5.

Microglia/macrophages express IL-15 in CNS tissue. CNS tissue from one control (block 1: AD) and two distinct patients with MS (block 9: EH; block 4: IL) containing demyelinated areas were stained for Iba-1 (green: A, E, I), IL-15 (red: B, F, J), and TO-PRO. D, H, and L, Isotype controls. Several Iba-1+ cells visible in MS lesions (E, I) expressed detectable levels of IL-15 (F, J), but non–Iba-1 cells also expressed IL-15 (G). White arrows indicate examples of microglia/macrophages colabeled for IL-15, white arrowheads point to IL-15+ cells that are not microglia/macrophages, and orange arrowheads indicate microglia/macrophages that do not express IL-15. Pictures shown are representative of five fields taken from each section of three controls and seven blocks from six MS donors. Original magnification ×400. Scale bar, 50 μm.

FIGURE 5.

Microglia/macrophages express IL-15 in CNS tissue. CNS tissue from one control (block 1: AD) and two distinct patients with MS (block 9: EH; block 4: IL) containing demyelinated areas were stained for Iba-1 (green: A, E, I), IL-15 (red: B, F, J), and TO-PRO. D, H, and L, Isotype controls. Several Iba-1+ cells visible in MS lesions (E, I) expressed detectable levels of IL-15 (F, J), but non–Iba-1 cells also expressed IL-15 (G). White arrows indicate examples of microglia/macrophages colabeled for IL-15, white arrowheads point to IL-15+ cells that are not microglia/macrophages, and orange arrowheads indicate microglia/macrophages that do not express IL-15. Pictures shown are representative of five fields taken from each section of three controls and seven blocks from six MS donors. Original magnification ×400. Scale bar, 50 μm.

Close modal

We investigated the presence of CD8 T lymphocytes in the vicinity of IL-15–expressing cells (Fig. 6) and noted whether these T cells were located in the CNS parenchyma or within perivascular cuffs. Although the number of CD8 T lymphocytes per surface area varied between donors, CD8 T cells were present in the CNS parenchyma of all the MS lesions examined (Fig. 6D) and in perivascular cuffs (Fig. 6G). Moreover, CD8 T cells were observed outside of blood vessels, only where cells displayed evident IL-15 expression (Fig. 6F, 6I). In control sections, no CD8 T lymphocytes could be observed in the CNS parenchyma, and only rare CD8 T lymphocytes were associated with blood vessels (Fig. 6A).

FIGURE 6.

CD8 T lymphocytes are present in close vicinity to abundant IL-15 in MS lesions. CNS tissue from one control (block 2: AC) and two patients with MS (block 6: DF; block 9: GI) were stained for CD8 (green: A, D, G), IL-15 (red: B, E, H), and TO-PRO (labeling the nuclei in blue). C, F, and I, Merge of CD8, IL-15, and TO-PRO. Few CD8 T lymphocytes were detected in controls, and they were not located in the parenchyma. Numerous CD8 T lymphocytes were found in MS lesions close to IL-15–positive cells, especially in perivascular cuffs (GI) and in the parenchyma (DF). Pictures shown are representative of five fields taken from each section of the three blocks from as many controls and seven blocks from six MS donors. Original magnification ×200. Scale bar, 75 μm.

FIGURE 6.

CD8 T lymphocytes are present in close vicinity to abundant IL-15 in MS lesions. CNS tissue from one control (block 2: AC) and two patients with MS (block 6: DF; block 9: GI) were stained for CD8 (green: A, D, G), IL-15 (red: B, E, H), and TO-PRO (labeling the nuclei in blue). C, F, and I, Merge of CD8, IL-15, and TO-PRO. Few CD8 T lymphocytes were detected in controls, and they were not located in the parenchyma. Numerous CD8 T lymphocytes were found in MS lesions close to IL-15–positive cells, especially in perivascular cuffs (GI) and in the parenchyma (DF). Pictures shown are representative of five fields taken from each section of the three blocks from as many controls and seven blocks from six MS donors. Original magnification ×200. Scale bar, 75 μm.

Close modal

Our study demonstrates that human astrocytes in a proinflammatory milieu express sufficient surface IL-15 so as to enhance effector functions of Ag-specific CD8 T lymphocytes. Although IL-15 mRNA and protein expression in murine and human CNS cells has been reported (3336), our results are the first, to our knowledge, to demonstrate that surface IL-15 expressed by human glial cells is functional and acts on human CD8 T lymphocytes. Our in situ data specifically document the prominent expression of IL-15 in MS lesions. In addition, CD8 T lymphocytes are localized to areas with abundant IL-15–expressing cells in MS lesions.

In our in vitro studies, treatment with single cytokines (IFN-γ, IL-1β, or TNF) did not alter IL-15 levels on astrocytes, although these treatments did increase MHC-I expression (Fig. 1D). We conclude that such treatments did not trigger either the appropriate signaling cascade or amount of intracellular signaling to induce surface IL-15 expression (51). However, IFN-γ + TNF and IFN-γ + IL-1β combinations significantly increased the proportion of astrocytes expressing surface IL-15, and these cytokines have been shown to induce other immune mediators in human primary astrocytes (52, 53). Activated T lymphocytes and macrophages/microglia, abundantly present in MS lesions (1), are classic in vivo sources of these cytokines (1, 5457).

Our in vitro results demonstrate that activated human astrocytes augment the expression of multiple effector molecules (granzyme B, perforin, NKG2D) and MHC-I–restricted Ag-specific cytotoxicity by CD8 T lymphocytes in a contact-dependent manner (Fig. 3). Thus, local IL-15 may enhance the propensity of parenchymal CD8 T cells to lyse MHC-I–expressing target cells such as oligodendrocytes, the cellular target of MS demyelination. Our previous work demonstrates that oligodendrocytes in MS lesions express NKG2D ligands (40). Moreover, we have shown that the same cytokine combinations (e.g., IFN-γ + TNF) that increase surface IL-15 on human astrocytes, leading to elevated NKG2D and lytic enzyme expression by CD8 T lymphocytes, augment NKG2D ligands on human oligodendrocytes (40). Thus, the concomitant presence within MS lesions of IL-15–expressing astrocytes and NKG2D ligand-expressing oligodendrocytes may enhance CD8 T lymphocyte effector functions and consequently exacerbate the CD8 T lymphocyte-mediated killing of oligodendrocytes. Supernatants from activated astrocytes, or insertion of a transwell separating both cell types, did not reproduce the coculture results. Cells from both immune (monocytes and dendritic cells) (21, 5860) and nonimmune origin (synovial fibroblasts and endothelial cells) (26, 61, 62) have previously been shown to activate T lymphocytes via contact-dependent IL-15–mediated mechanisms. Our functional studies were conducted using fetal astrocytes, as we are unable to isolate adult astrocytes in sufficient number or purity. However, our immunohistochemistry data demonstrate that human adult astrocytes express significant amounts of IL-15, especially in MS lesions (Fig. 4).

Our immunohistochemistry studies show that IL-15 expression is significantly increased in MS lesions compared with controls and that astrocytes are major sources of these augmented cytokine levels (Fig. 4). Moreover, enhanced IL-15 levels were observed in both acute (blocks 4–6 and 9) and subacute/chronic (blocks 7, 8, and 10) MS lesions, suggesting that this cytokine is upregulated for extended periods of time within the inflamed CNS during MS. Astrocytes are the most abundant glial cell type within the CNS. Moreover, these cells are strategically localized at the blood–brain barrier and thus can interact with incoming immune cells, including CD8 T lymphocytes as soon as they enter the CNS. Several functions have been attributed to astrocytes (53). Our data also indicate that the numerous macrophages and microglia in MS lesions provide IL-15 to infiltrating immune cells, such as CD8 T lymphocytes, in the CNS parenchyma. Moreover, several studies have documented the capacity of human monocytes/macrophages for providing functional IL-15 to T cells (20, 23), supporting the notion that CNS macrophages and most likely microglia do the same. In line with our findings, astrocytes have been shown to be the main source of IL-15 in LPS-injected mice, with reactive microglia being an additional source (38). Dendritic cells, found mainly in the perivascular spaces in MS lesions, could serve as an additional source of IL-15 (21). Finally, CD8 T cells detected in the parenchyma and in perivascular cuffs of MS lesions were in close proximity to IL-15–expressing cells (Fig. 6).

Blockade of systemic IL-15 (or IL-15 signaling receptor) in animal models featuring an inflammatory autoimmune response (e.g., rheumatoid arthritis, diabetes, psoriasis) has been shown to decrease disease development and severity (6366). Recently, Gomez-Nicola and colleagues (67) observed aggravated experimental autoimmune encephalomyelitis in IL-15 knockout mice compared with wild-type littermates, although the maximum clinical score (1.6) they obtained for wild-type animals was low compared with other reports. However, because IL-15 null mice displayed marked reductions in the numbers of NK and memory CD8 T cells (68), indicating an abnormal immune system, interpretations of the role IL-15 using these mice should be made with extreme caution. Other groups have demonstrated that nonmicroglial CNS cells, especially astrocytes via NF-κB signaling, respond to CNS inflammation and play a deleterious role in autoimmune CNS inflammation during experimental autoimmune encephaylomyelitis (69, 70). We have previously shown that human astrocytes upon TLR3 ligation produce chemokines and cytokines (i.e., IL-6) (41) that can activate T cells. Our data assign a role to these cells as stimulators of infiltrating CD8 T lymphocytes in the inflamed CNS during MS. Moreover, we are currently investigating whether other factors derived from astrocytes contribute in maintaining or dampening T cell activation. Although not addressed in this study, IL-15 could participate in other aspects of neuroinflammation given its chemoattractive and prosurvival properties (71, 72). For example, IL-15 has been previously shown to be a potent T cell chemoattractant during rheumatoid arthritis, resulting in enhanced T cell migration to inflamed joints (73, 74). Its prosurvival properties could contribute to aspects of neuroinflammation.

Studies in humans support a role for IL-15 in the target organs of rheumatoid arthritis (26, 62, 74) and celiac disease (7578). In the latter, prolonged exposure of CD8 T lymphocytes to IL-15 in the inflamed gut epithelium greatly enhances their cytotoxicity toward target cells. This was suggested to be a mechanism at the basis of the tissue destruction observed in patients with celiac disease. Our data support the proposal that a similar mechanism could potentially be at work in the inflamed CNS of patients with MS. In conclusion, we demonstrate that in the target organ of MS pathogenesis, astrocytes via the production of IL-15 contribute to perpetuating the activation of damaging CD8 T lymphocytes.

We thank Ellie McCrea, Diane Beauseigle, Manon Blain, Caroline Lambert, and Janet Laganière for technical assistance. We also thank Drs. André Oliver and Jeffrey Hall for providing adult human CNS tissues for the in vitro studies, Dr. Alexandre Prat for access to the Leica confocal microscope and the LSRII flow cytometer, and Drs. Christine Vande Velde and Réjean Lapointe for scientific discussion.

Disclosures The authors have no financial conflicts of interest.

This work was supported by grants from the Multiple Sclerosis Society of Canada (MSSC) (to N.A. and J.P.A.). N.A. holds a Donald Paty Career Development Award from the MSSC and a Chercheur-Boursier salary award from the Fonds de la Recherche en Santé du Québec. P.S. was supported by a Canadian Graduate Scholarships doctoral research award from the Canadian Institutes of Health Research. C.L.P. was supported by the Canadian Institutes of Health Research Neuroinflammation Training Program and holds a studentship from the MSSC.

Abbreviations used in this paper:

not applicable

AQ

active plaque

CB

cerebellum

CQ

chronic plaque

DD

disease duration

DFT

death to snap-freezing time

F

frontal

GFAP

glial fibrillary acidic protein

L

left

MBP

myelin basic protein

ΔMFI

change in median fluorescence intensity

MHC-I

MHC class I

MS

multiple sclerosis

NC W

normal control white matter

O

occipital

ORO

Oil Red O

P

parietal

qPCR

quantitative real-time PCR

R

right

SAQ

subacute plaque

SI

stimulation index

Sv

subventricular

T

temporal

V

ventricular

WM

white matter.

1
Sospedra
M.
,
Martin
R.
.
2005
.
Immunology of multiple sclerosis.
Annu. Rev. Immunol.
23
:
683
747
.
2
Noseworthy
J. H.
,
Lucchinetti
C.
,
Rodriguez
M.
,
Weinshenker
B. G.
.
2000
.
Multiple sclerosis.
N. Engl. J. Med.
343
:
938
952
.
3
Friese
M. A.
,
Fugger
L.
.
2005
.
Autoreactive CD8+ T cells in multiple sclerosis: a new target for therapy?
Brain
128
:
1747
1763
.
4
Zozulya
A. L.
,
Wiendl
H.
.
2008
.
The role of CD8 suppressors versus destructors in autoimmune central nervous system inflammation.
Hum. Immunol.
69
:
797
804
.
5
Hauser
S. L.
,
Bhan
A. K.
,
Gilles
F.
,
Kemp
M.
,
Kerr
C.
,
Weiner
H. L.
.
1986
.
Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions.
Ann. Neurol.
19
:
578
587
.
6
Neumann
H.
,
Medana
I. M.
,
Bauer
J.
,
Lassmann
H.
.
2002
.
Cytotoxic T lymphocytes in autoimmune and degenerative CNS diseases.
Trends Neurosci.
25
:
313
319
.
7
Lassmann
H.
2004
.
Recent neuropathological findings in MS—implications for diagnosis and therapy.
J. Neurol.
251
(
Suppl 4
):
IV2
IV5
.
8
Gay
F. W.
,
Drye
T. J.
,
Dick
G. W.
,
Esiri
M. M.
.
1997
.
The application of multifactorial cluster analysis in the staging of plaques in early multiple sclerosis. Identification and characterization of the primary demyelinating lesion.
Brain
120
:
1461
1483
.
9
Babbe
H.
,
Roers
A.
,
Waisman
A.
,
Lassmann
H.
,
Goebels
N.
,
Hohlfeld
R.
,
Friese
M.
,
Schröder
R.
,
Deckert
M.
,
Schmidt
S.
, et al
.
2000
.
Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction.
J. Exp. Med.
192
:
393
404
.
10
Jacobsen
M.
,
Cepok
S.
,
Quak
E.
,
Happel
M.
,
Gaber
R.
,
Ziegler
A.
,
Schock
S.
,
Oertel
W. H.
,
Sommer
N.
,
Hemmer
B.
.
2002
.
Oligoclonal expansion of memory CD8+ T cells in cerebrospinal fluid from multiple sclerosis patients.
Brain
125
:
538
550
.
11
Skulina
C.
,
Schmidt
S.
,
Dornmair
K.
,
Babbe
H.
,
Roers
A.
,
Rajewsky
K.
,
Wekerle
H.
,
Hohlfeld
R.
,
Goebels
N.
.
2004
.
Multiple sclerosis: brain-infiltrating CD8+ T cells persist as clonal expansions in the cerebrospinal fluid and blood.
Proc. Natl. Acad. Sci. USA
101
:
2428
2433
.
12
Rus
H.
,
Pardo
C. A.
,
Hu
L.
,
Darrah
E.
,
Cudrici
C.
,
Niculescu
T.
,
Niculescu
F.
,
Mullen
K. M.
,
Allie
R.
,
Guo
L.
, et al
.
2005
.
The voltage-gated potassium channel Kv1.3 is highly expressed on inflammatory infiltrates in multiple sclerosis brain.
Proc. Natl. Acad. Sci. USA
102
:
11094
11099
.
13
Huseby
E. S.
,
Liggitt
D.
,
Brabb
T.
,
Schnabel
B.
,
Ohlén
C.
,
Goverman
J.
.
2001
.
A pathogenic role for myelin-specific CD8(+) T cells in a model for multiple sclerosis.
J. Exp. Med.
194
:
669
676
.
14
Sun
D.
,
Whitaker
J. N.
,
Huang
Z.
,
Liu
D.
,
Coleclough
C.
,
Wekerle
H.
,
Raine
C. S.
.
2001
.
Myelin antigen-specific CD8+ T cells are encephalitogenic and produce severe disease in C57BL/6 mice.
J. Immunol.
166
:
7579
7587
.
15
Saxena
A.
,
Bauer
J.
,
Scheikl
T.
,
Zappulla
J.
,
Audebert
M.
,
Desbois
S.
,
Waisman
A.
,
Lassmann
H.
,
Liblau
R. S.
,
Mars
L. T.
.
2008
.
Cutting edge: Multiple sclerosis-like lesions induced by effector CD8 T cells recognizing a sequestered antigen on oligodendrocytes.
J. Immunol.
181
:
1617
1621
.
16
Zehntner
S. P.
,
Brisebois
M.
,
Tran
E.
,
Owens
T.
,
Fournier
S.
.
2003
.
Constitutive expression of a costimulatory ligand on antigen-presenting cells in the nervous system drives demyelinating disease.
FASEB J.
17
:
1910
1912
.
17
Na
S. Y.
,
Cao
Y.
,
Toben
C.
,
Nitschke
L.
,
Stadelmann
C.
,
Gold
R.
,
Schimpl
A.
,
Hünig
T.
.
2008
.
Naive CD8 T-cells initiate spontaneous autoimmunity to a sequestered model antigen of the central nervous system.
Brain
131
:
2353
2365
.
18
Ma
A.
,
Koka
R.
,
Burkett
P.
.
2006
.
Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis.
Annu. Rev. Immunol.
24
:
657
679
.
19
Waldmann
T. A.
2006
.
The biology of interleukin-2 and interleukin-15: implications for cancer therapy and vaccine design.
Nat. Rev. Immunol.
6
:
595
601
.
20
Neely
G. G.
,
Robbins
S. M.
,
Amankwah
E. K.
,
Epelman
S.
,
Wong
H.
,
Spurrell
J. C.
,
Jandu
K. K.
,
Zhu
W.
,
Fogg
D. K.
,
Brown
C. B.
,
Mody
C. H.
.
2001
.
Lipopolysaccharide-stimulated or granulocyte-macrophage colony-stimulating factor-stimulated monocytes rapidly express biologically active IL-15 on their cell surface independent of new protein synthesis.
J. Immunol.
167
:
5011
5017
.
21
Ferlazzo
G.
,
Pack
M.
,
Thomas
D.
,
Paludan
C.
,
Schmid
D.
,
Strowig
T.
,
Bougras
G.
,
Muller
W. A.
,
Moretta
L.
,
Münz
C.
.
2004
.
Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs.
Proc. Natl. Acad. Sci. USA
101
:
16606
16611
.
22
Budagian
V.
,
Bulanova
E.
,
Paus
R.
,
Bulfone-Paus
S.
.
2006
.
IL-15/IL-15 receptor biology: a guided tour through an expanding universe.
Cytokine Growth Factor Rev.
17
:
259
280
.
23
Dubois
S.
,
Mariner
J.
,
Waldmann
T. A.
,
Tagaya
Y.
.
2002
.
IL-15Ralpha recycles and presents IL-15 In trans to neighboring cells.
Immunity
17
:
537
547
.
24
Stonier
S. W.
,
Schluns
K. S.
.
2010
.
Trans-presentation: a novel mechanism regulating IL-15 delivery and responses.
Immunol. Lett.
127
:
85
92
.
25
Fehniger
T. A.
,
Caligiuri
M. A.
.
2001
.
Interleukin 15: biology and relevance to human disease.
Blood
97
:
14
32
.
26
McInnes
I. B.
,
Leung
B. P.
,
Sturrock
R. D.
,
Field
M.
,
Liew
F. Y.
.
1997
.
Interleukin-15 mediates T cell-dependent regulation of tumor necrosis factor-alpha production in rheumatoid arthritis.
Nat. Med.
3
:
189
195
.
27
Mention
J. J.
,
Ben Ahmed
M.
,
Bègue
B.
,
Barbe
U.
,
Verkarre
V.
,
Asnafi
V.
,
Colombel
J. F.
,
Cugnenc
P. H.
,
Ruemmele
F. M.
,
McIntyre
E.
, et al
.
2003
.
Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease.
Gastroenterology
125
:
730
745
.
28
Sakai
T.
,
Kusugami
K.
,
Nishimura
H.
,
Ando
T.
,
Yamaguchi
T.
,
Ohsuga
M.
,
Ina
K.
,
Enomoto
A.
,
Kimura
Y.
,
Yoshikai
Y.
.
1998
.
Interleukin 15 activity in the rectal mucosa of inflammatory bowel disease.
Gastroenterology
114
:
1237
1243
.
29
Pashenkov
M.
,
Mustafa
M.
,
Kivisäkk
P.
,
Link
H.
.
1999
.
Levels of interleukin-15-expressing blood mononuclear cells are elevated in multiple sclerosis.
Scand. J. Immunol.
50
:
302
308
.
30
Blanco-Jerez
C.
,
Plaza
J. F.
,
Masjuan
J.
,
Orensanz
L. M.
,
Alvarez-Cermeño
J. C.
.
2002
.
Increased levels of IL-15 mRNA in relapsing—remitting multiple sclerosis attacks.
J. Neuroimmunol.
128
:
90
94
.
31
Kivisäkk
P.
,
Matusevicius
D.
,
He
B.
,
Söderström
M.
,
Fredrikson
S.
,
Link
H.
.
1998
.
IL-15 mRNA expression is up-regulated in blood and cerebrospinal fluid mononuclear cells in multiple sclerosis (MS).
Clin. Exp. Immunol.
111
:
193
197
.
32
Vaknin-Dembinsky
A.
,
Brass
S. D.
,
Brass
S.
,
Gandhi
R.
,
Weiner
H. L.
.
2008
.
Membrane bound IL-15 is increased on CD14 monocytes in early stages of MS.
[Published erratum appears in 2009 J. Neuroimmunol. 207: 122.]
J. Neuroimmunol.
195
:
135
139
.
33
Hanisch
U. K.
,
Lyons
S. A.
,
Prinz
M.
,
Nolte
C.
,
Weber
J. R.
,
Kettenmann
H.
,
Kirchhoff
F.
.
1997
.
Mouse brain microglia express interleukin-15 and its multimeric receptor complex functionally coupled to Janus kinase activity.
J. Biol. Chem.
272
:
28853
28860
.
34
Kurowska
M.
,
Rudnicka
W.
,
Maślińska
D.
,
Maśliński
W.
.
2002
.
Expression of IL-15 and IL-15 receptor isoforms in select structures of human fetal brain.
Ann. N. Y. Acad. Sci.
966
:
441
445
.
35
Lee
Y. B.
,
Satoh
J.
,
Walker
D. G.
,
Kim
S. U.
.
1996
.
Interleukin-15 gene expression in human astrocytes and microglia in culture.
Neuroreport
7
:
1062
1066
.
36
Gómez-Nicola
D.
,
Valle-Argos
B.
,
Suardíaz
M.
,
Taylor
J. S.
,
Nieto-Sampedro
M.
.
2008
.
Role of IL-15 in spinal cord and sciatic nerve after chronic constriction injury: regulation of macrophage and T-cell infiltration.
J. Neurochem.
107
:
1741
1752
.
37
McInnes
I. B.
,
Gracie
J. A.
.
2004
.
Interleukin-15: a new cytokine target for the treatment of inflammatory diseases.
Curr. Opin. Pharmacol.
4
:
392
397
.
38
Gómez-Nicola
D.
,
Valle-Argos
B.
,
Pita-Thomas
D. W.
,
Nieto-Sampedro
M.
.
2008
.
Interleukin 15 expression in the CNS: blockade of its activity prevents glial activation after an inflammatory injury.
Glia
56
:
494
505
.
39
Nair
A.
,
Frederick
T. J.
,
Miller
S. D.
.
2008
.
Astrocytes in multiple sclerosis: a product of their environment.
Cell. Mol. Life Sci.
65
:
2702
2720
.
40
Saikali
P.
,
Antel
J. P.
,
Newcombe
J.
,
Chen
Z.
,
Freedman
M.
,
Blain
M.
,
Cayrol
R.
,
Prat
A.
,
Hall
J. A.
,
Arbour
N.
.
2007
.
NKG2D-mediated cytotoxicity toward oligodendrocytes suggests a mechanism for tissue injury in multiple sclerosis.
J. Neurosci.
27
:
1220
1228
.
41
Jack
C. S.
,
Arbour
N.
,
Manusow
J.
,
Montgrain
V.
,
Blain
M.
,
McCrea
E.
,
Shapiro
A.
,
Antel
J. P.
.
2005
.
TLR signaling tailors innate immune responses in human microglia and astrocytes.
J. Immunol.
175
:
4320
4330
.
42
Arbour
N.
,
Lapointe
R.
,
Saikali
P.
,
McCrea
E.
,
Regen
T.
,
Antel
J. P.
.
2006
.
A new clinically relevant approach to expand myelin specific T cells.
J. Immunol. Methods
310
:
53
61
.
43
Krutzik
S. R.
,
Hewison
M.
,
Liu
P. T.
,
Robles
J. A.
,
Stenger
S.
,
Adams
J. S.
,
Modlin
R. L.
.
2008
.
IL-15 links TLR2/1-induced macrophage differentiation to the vitamin D-dependent antimicrobial pathway.
J. Immunol.
181
:
7115
7120
.
44
Schlaepfer
E.
,
Speck
R. F.
.
2008
.
Anti-HIV activity mediated by natural killer and CD8+ cells after toll-like receptor 7/8 triggering.
PLoS One
3
:
e1999
.
45
Correia
M. P.
,
Cardoso
E. M.
,
Pereira
C. F.
,
Neves
R.
,
Uhrberg
M.
,
Arosa
F. A.
.
2009
.
Hepatocytes and IL-15: a favorable microenvironment for T cell survival and CD8+ T cell differentiation.
J. Immunol.
182
:
6149
6159
.
46
Rubio
V.
,
Stuge
T. B.
,
Singh
N.
,
Betts
M. R.
,
Weber
J. S.
,
Roederer
M.
,
Lee
P. P.
.
2003
.
Ex vivo identification, isolation and analysis of tumor-cytolytic T cells.
Nat. Med.
9
:
1377
1382
.
47
Suni
M. A.
,
Maino
V. C.
,
Maecker
H. T.
.
2005
.
Ex vivo analysis of T-cell function.
Curr. Opin. Immunol.
17
:
434
440
.
48
Li
H.
,
Cuzner
M. L.
,
Newcombe
J.
.
1996
.
Microglia-derived macrophages in early multiple sclerosis plaques.
Neuropathol. Appl. Neurobiol.
22
:
207
215
.
49
Jack
C. S.
,
Arbour
N.
,
Blain
M.
,
Meier
U. C.
,
Prat
A.
,
Antel
J. P.
.
2007
.
Th1 polarization of CD4+ T cells by Toll-like receptor 3-activated human microglia.
J. Neuropathol. Exp. Neurol.
66
:
848
859
.
50
Kohrt
H. E.
,
Shu
C. T.
,
Stuge
T. B.
,
Holmes
S. P.
,
Weber
J.
,
Lee
P. P.
.
2005
.
Rapid assessment of recognition efficiency and functional capacity of antigen-specific T-cell responses.
J. Immunother.
28
:
297
305
.
51
Waldmann
T. A.
,
Tagaya
Y.
.
1999
.
The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK cell differentiation and host response to intracellular pathogens.
Annu. Rev. Immunol.
17
:
19
49
.
52
Jana
M.
,
Anderson
J. A.
,
Saha
R. N.
,
Liu
X.
,
Pahan
K.
.
2005
.
Regulation of inducible nitric oxide synthase in proinflammatory cytokine-stimulated human primary astrocytes.
Free Radic. Biol. Med.
38
:
655
664
.
53
Williams
A.
,
Piaton
G.
,
Lubetzki
C.
.
2007
.
Astrocytes—friends or foes in multiple sclerosis?
Glia
55
:
1300
1312
.
54
Basu
A.
,
Krady
J. K.
,
Levison
S. W.
.
2004
.
Interleukin-1: a master regulator of neuroinflammation.
J. Neurosci. Res.
78
:
151
156
.
55
Probert
L.
,
Selmaj
K.
.
1997
.
TNF and related molecules: trends in neuroscience and clinical applications.
J. Neuroimmunol.
72
:
113
117
.
56
Owens
T.
2002
.
Identification of new therapeutic targets for prevention of CNS inflammation.
Expert Opin. Ther. Targets
6
:
203
215
.
57
Benveniste
E. N.
1998
.
Cytokine actions in the central nervous system.
Cytokine Growth Factor Rev.
9
:
259
275
.
58
Brilot
F.
,
Strowig
T.
,
Roberts
S. M.
,
Arrey
F.
,
Münz
C.
.
2007
.
NK cell survival mediated through the regulatory synapse with human DCs requires IL-15Ralpha.
J. Clin. Invest.
117
:
3316
3329
.
59
Carson
W. E.
,
Ross
M. E.
,
Baiocchi
R. A.
,
Marien
M. J.
,
Boiani
N.
,
Grabstein
K.
,
Caligiuri
M. A.
.
1995
.
Endogenous production of interleukin 15 by activated human monocytes is critical for optimal production of interferon-gamma by natural killer cells in vitro.
J. Clin. Invest.
96
:
2578
2582
.
60
Dubsky
P.
,
Saito
H.
,
Leogier
M.
,
Dantin
C.
,
Connolly
J. E.
,
Banchereau
J.
,
Palucka
A. K.
.
2007
.
IL-15-induced human DC efficiently prime melanoma-specific naive CD8+ T cells to differentiate into CTL.
Eur. J. Immunol.
37
:
1678
1690
.
61
Miranda-Carús
M. E.
,
Balsa
A.
,
Benito-Miguel
M.
,
Pérez de Ayala
C.
,
Martín-Mola
E.
.
2004
.
IL-15 and the initiation of cell contact-dependent synovial fibroblast-T lymphocyte cross-talk in rheumatoid arthritis: effect of methotrexate.
J. Immunol.
173
:
1463
1476
.
62
Oppenheimer-Marks
N.
,
Brezinschek
R. I.
,
Mohamadzadeh
M.
,
Vita
R.
,
Lipsky
P. E.
.
1998
.
Interleukin 15 is produced by endothelial cells and increases the transendothelial migration of T cells In vitro and in the SCID mouse-human rheumatoid arthritis model In vivo.
J. Clin. Invest.
101
:
1261
1272
.
63
Ferrari-Lacraz
S.
,
Zanelli
E.
,
Neuberg
M.
,
Donskoy
E.
,
Kim
Y. S.
,
Zheng
X. X.
,
Hancock
W. W.
,
Maslinski
W.
,
Li
X. C.
,
Strom
T. B.
,
Moll
T.
.
2004
.
Targeting IL-15 receptor-bearing cells with an antagonist mutant IL-15/Fc protein prevents disease development and progression in murine collagen-induced arthritis.
J. Immunol.
173
:
5818
5826
.
64
Villadsen
L. S.
,
Schuurman
J.
,
Beurskens
F.
,
Dam
T. N.
,
Dagnaes-Hansen
F.
,
Skov
L.
,
Rygaard
J.
,
Voorhorst-Ogink
M. M.
,
Gerritsen
A. F.
,
van Dijk
M. A.
, et al
.
2003
.
Resolution of psoriasis upon blockade of IL-15 biological activity in a xenograft mouse model.
J. Clin. Invest.
112
:
1571
1580
.
65
Chong
M. M.
,
Chen
Y.
,
Darwiche
R.
,
Dudek
N. L.
,
Irawaty
W.
,
Santamaria
P.
,
Allison
J.
,
Kay
T. W.
,
Thomas
H. E.
.
2004
.
Suppressor of cytokine signaling-1 overexpression protects pancreatic beta cells from CD8+ T cell-mediated autoimmune destruction.
J. Immunol.
172
:
5714
5721
.
66
Demirci
G.
,
Strom
T. B.
,
Li
X. C.
.
2003
.
Islet allograft rejection in nonobese diabetic mice involves the common gamma-chain and CD28/CD154-dependent and -independent mechanisms.
J. Immunol.
171
:
3878
3885
.
67
Gomez-Nicola
D.
,
Spagnolo
A.
,
Guaza
C.
,
Nieto-Sampedro
M.
.
2010
.
Aggravated experimental autoimmune encephalomyelitis in IL-15 knockout mice.
Exp. Neurol.
222
:
235
242
.
68
Kennedy
M. K.
,
Glaccum
M.
,
Brown
S. N.
,
Butz
E. A.
,
Viney
J. L.
,
Embers
M.
,
Matsuki
N.
,
Charrier
K.
,
Sedger
L.
,
Willis
C. R.
, et al
.
2000
.
Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice.
J. Exp. Med.
191
:
771
780
.
69
van Loo
G.
,
De Lorenzi
R.
,
Schmidt
H.
,
Huth
M.
,
Mildner
A.
,
Schmidt-Supprian
M.
,
Lassmann
H.
,
Prinz
M. R.
,
Pasparakis
M.
.
2006
.
Inhibition of transcription factor NF-kappaB in the central nervous system ameliorates autoimmune encephalomyelitis in mice.
Nat. Immunol.
7
:
954
961
.
70
Brambilla
R.
,
Persaud
T.
,
Hu
X.
,
Karmally
S.
,
Shestopalov
V. I.
,
Dvoriantchikova
G.
,
Ivanov
D.
,
Nathanson
L.
,
Barnum
S. R.
,
Bethea
J. R.
.
2009
.
Transgenic inhibition of astroglial NF-kappa B improves functional outcome in experimental autoimmune encephalomyelitis by suppressing chronic central nervous system inflammation.
J. Immunol.
182
:
2628
2640
.
71
Petrovas
C.
,
Mueller
Y. M.
,
Dimitriou
I. D.
,
Bojczuk
P. M.
,
Mounzer
K. C.
,
Witek
J.
,
Altman
J. D.
,
Katsikis
P. D.
.
2004
.
HIV-specific CD8+ T cells exhibit markedly reduced levels of Bcl-2 and Bcl-xL.
J. Immunol.
172
:
4444
4453
.
72
Huntington
N. D.
,
Puthalakath
H.
,
Gunn
P.
,
Naik
E.
,
Michalak
E. M.
,
Smyth
M. J.
,
Tabarias
H.
,
Degli-Esposti
M. A.
,
Dewson
G.
,
Willis
S. N.
, et al
.
2007
.
Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1.
Nat. Immunol.
8
:
856
863
.
73
Perera
L. P.
,
Goldman
C. K.
,
Waldmann
T. A.
.
1999
.
IL-15 induces the expression of chemokines and their receptors in T lymphocytes.
J. Immunol.
162
:
2606
2612
.
74
McInnes
I. B.
,
al-Mughales
J.
,
Field
M.
,
Leung
B. P.
,
Huang
F. P.
,
Dixon
R.
,
Sturrock
R. D.
,
Wilkinson
P. C.
,
Liew
F. Y.
.
1996
.
The role of interleukin-15 in T-cell migration and activation in rheumatoid arthritis.
Nat. Med.
2
:
175
182
.
75
Hüe
S.
,
Mention
J. J.
,
Monteiro
R. C.
,
Zhang
S.
,
Cellier
C.
,
Schmitz
J.
,
Verkarre
V.
,
Fodil
N.
,
Bahram
S.
,
Cerf-Bensussan
N.
,
Caillat-Zucman
S.
.
2004
.
A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease.
Immunity
21
:
367
377
.
76
Jabri
B.
,
Sollid
L. M.
.
2006
.
Mechanisms of disease: immunopathogenesis of celiac disease.
Nat. Clin. Pract. Gastroenterol. Hepatol.
3
:
516
525
.
77
Meresse
B.
,
Chen
Z.
,
Ciszewski
C.
,
Tretiakova
M.
,
Bhagat
G.
,
Krausz
T. N.
,
Raulet
D. H.
,
Lanier
L. L.
,
Groh
V.
,
Spies
T.
, et al
.
2004
.
Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease.
Immunity
21
:
357
366
.
78
Meresse
B.
,
Curran
S. A.
,
Ciszewski
C.
,
Orbelyan
G.
,
Setty
M.
,
Bhagat
G.
,
Lee
L.
,
Tretiakova
M.
,
Semrad
C.
,
Kistner
E.
, et al
.
2006
.
Reprogramming of CTLs into natural killer-like cells in celiac disease.
J. Exp. Med.
203
:
1343
1355
.