Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2007 Nov;122(3):409-17.
doi: 10.1111/j.1365-2567.2007.02655.x. Epub 2007 Jul 6.

Innate immunity modulates autoimmunity: type 1 interferon-beta treatment in multiple sclerosis promotes growth and function of regulatory invariant natural killer T cells through dendritic cell maturation

Affiliations

Innate immunity modulates autoimmunity: type 1 interferon-beta treatment in multiple sclerosis promotes growth and function of regulatory invariant natural killer T cells through dendritic cell maturation

Gianluigi Gigli et al. Immunology. 2007 Nov.

Abstract

Type 1 interferon-beta (T1IFN-beta) is an innate cytokine and the first-choice therapy for multiple sclerosis (MS). It is still unclear how T1IFN-beta, whose main function is to promote innate immunity during infections, plays a beneficial role in autoimmune disease. Here we show that T1IFN-beta promoted the expansion and function of invariant natural killer (iNKT) cells, an innate T-cell subset with strong immune regulatory properties that is able to prevent autoimmune disease in pre-clinical models of MS and type 1 diabetes. Specifically, we observed that T1IFN-beta treatment significantly increased the percentages of Valpha24(+) NKT cells in peripheral blood mononuclear cells of MS patients. Furthermore, iNKT cells of T1IFN-beta-treated individuals showed a dramatically improved secretion of cytokines (interleukins 4 and 5 and interferon-gamma) in response to antigenic stimulation compared to iNKT cells isolated from the same patients before T1IFN-beta treatment. The effect of T1IFN-beta on iNKT cells was mediated through the modulation of myeloid dendritic cells (DCs). In fact, DCs modulated in vivo or in vitro by T1IFN-beta were more efficient antigen-presenting cells for iNKT cells. Such a modulatory effect of T1IFN-beta was associated with up-regulation on DCs of key costimulatory molecules for iNKT (i.e. CD80, CD40 and CD1d). Our data identified the iNKT cell/DC pathway as a new target for the immune regulatory effect of T1IFNs in autoimmune diseases and provide a possible mechanism to explain the clinical efficacy of T1IFN-beta in MS.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Percentage of Vα24+ CD3+ NKT cells increased in PBMC of MS patients after treatment with T1IFN-β. (a) Cumulative data of percentages of iNKT in healthy individuals and MS patients before and after treatment with T1IFN-β are presented. Total peripheral blood mononuclear cells (PBMC) were stained with PE-conjugated anti-CD3 and FITC-conjugated anti-Vα24 monoclonal antibodies and analysed by flow cytometry. Percentages of CD3+ Vα24+ iNKT cells significantly increased in T1IFN-β-treated patients (n = 8) compared to PBMC samples from untreated individuals (untreated MS n = 8 and healthy controls, n = 8) (non-parametric Mann–Whitney test, P = 0·04). (b) Percentages of Vα24+ CD3+ iNKT cells increased progressively in the PBMC of MS patients after treatment with T1IFN-β. PBMC samples of each patient (open squares) and controls (closed squares) collected at different time-points were stained and analysed in a single experiment as in (b) (c) The majority of invariant Vα24+ T cells were αGalCer/CD1d+. Total PBMC of MS patients (n = 3) and controls (n = 3) were double-stained with PE-conjugated αGalCer-loaded CD1d tetramers and FITC-conjugated anti-Vα24 mAb. The small percentage (5–15%) of Vα24+ T cells that were αGalCer/CD1d could represent CD1d-restricted iNKT cells that did not recognize αGalCer as their glycolipid antigen.
Figure 2
Figure 2
T1IFN-β treatment in MS patients promoted iNKT cell function. The iNKT cells purified from T1IFN-β-treated MS patients (T1IFN-β MS; n = 8) secreted significantly larger amounts of IL-4 (P ≤ 0·01), IL-5 (P ≤ 0·01) and IFN-γ (P ≤ 0·05) compared to their counterparts from untreated healthy controls (n = 8) or the same MS patients before they started treatment (untreated MS; n = 8). IL-10 secretion increased slightly, but not significantly, in T1IFN-β-treated MS patients compared to untreated controls or MS patients (P ≤ 0·09). Vα24+ iNKT cells were expanded by culturing PBMC with αGalCer (100 ng/ml) and cytokines (recombinant human IL-7 and IL-15) and purified by magnetic bead separation with anti-Vα24 monoclonal antibody. iNKT cells (2 × 104 per well) were plated on a 96-well plate and stimulated with 2 × 105αGalCer-pulsed DCs. Supernatants were collected after 7 days and cytokines were simultaneously measured in the supernatants of iNKT cell cultures by a multiplexed flow cytometric assay (Cytometric Bead Array).
Figure 3
Figure 3
T1IFN-β did not directly enhance NKT cell proliferation and cytokine secretion. The addition of T1IFN-β to iNKT cell cultures inhibited both antigen-specific proliferation and cytokine secretion. Purified Vα24+ iNKT cells (2 × 104; previously expanded in vitro from PBMC of untreated individuals) were stimulated with unpulsed (– αGalCer) or antigen-pulsed (+ αGalCer), irradiated DCs (2 × 105) in 96-well plates with/without rhT1IFN-β (1000 U/ml). (a) Proliferation was measured by 3[H]thymidine incorporation in the last 16 hr of a 3-day culture. (b) Supernatants were collected after 7 days and cytokine release (IL-5) was measured by CBA assay.
Figure 4
Figure 4
T1IFN-β acted on DC by increasing their stimulatory capacity upon iNKT cells. (a) Pre-conditioning of monocytes with T1IFN-β in treated MS patients favoured differentiation of DCs with an enhanced antigen-presenting capacity upon iNKT cells compared to DCs from untreated MS patients or healthy controls (P ≤ 0·05). PBMC were collected from MS patients before (MS) and 3 months after treatment with T1IFN-β (T1IFN-β/MS) and kept for 2 hr at 37°, in 5% CO2 in RPMI-1640 with 10% fetal calf serum. Adherent cells were cultured for 5 days with rhGM-CSF (400 U/ml) and rhIL-4 (200 U/ml) to differentiate DCs. DCs were either unpulsed or were pulsed with the iNKT cell antigen (± αGalCer, 100 ng/ml), irradiated and cultured at 2 × 105 cells/well in a 96-well plate together with 2 × 104 purified Vα24+ iNKT cells previously expanded from autologous PBMC. Supernatants were collected after 7 days and iNKT cell stimulation was evaluated as the mean of cytokine release (IL-5 and IFN-γ) measured by CBA assay. Data are presented as mean ± SEM of duplicate determinations (SEM <5% are not shown). One representative experiment out of two is shown. (b) DCs differentiated in vitro in the presence of T1IFN-β were more effective antigen-presenting cells for iNKT cells compared to non-T1IFN-β-modulated DCs (P ≤ 0·01). DCs were differentiated from PBMC of untreated individuals (healthy donors) with rhGM-CSF and rhIL-4 with/without rhT1IFN-β, pulsed with αGalCer (100 ng/ml), irradiated and added to autologous purified Vα24+ iNKT cells. Unpulsed DCs were used as negative control (– αGalCer). Supernatants were collected after 7 days and iNKT cell stimulation was evaluated as mean of cytokine release (IL-5 and IFN-γ). Data are presented as mean ± SEM of duplicate determinations. One representative experiment out of three is shown.
Figure 5
Figure 5
T1IFN-β induced semi-maturation of resting DCs. (a) T1IFN-β up-regulated the expression of CD1d and maturation markers (CD80, CD40) on resting immature DCs. Myeloid DCs were derived from PBMC of healthy individuals with rhGM-CSF (400 U/ml) and rhIL-4 (200 U/ml) in the presence (open histogram) or absence (shaded histogram) of rhT1IFN-β (1000 U/ml). Expression of CD11c, maturation markers CD80 and CD40 and CD1d was determined by flow cytometric analysis. CD80, CD40 and CD1d histograms refer to gated CD11c+ cells. The graph at the right side of each histogram represents cumulative data of three separate determinations ±SEM. (b) T1IFN-β-conditioned DCs were not fully mature and did not secrete cytokines (IL-12 and IL-10). Cytokine release by immature DCs derived from PBMC without (iDC) or with rhT1IFN-β (T1IFN-β iDC) was assessed by CBA on supernatants of 5-day DC cultures. As positive controls, we used fully mature DCs (mDC) obtained by adding LPS (1 μg/ml) to the last 24 hr of culture.

Similar articles

Cited by

References

    1. The IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing–remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology. 1993;43:655–61. - PubMed
    1. Paty D, Li DK UBC/MRI Study Group and the IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing–remitting multiple sclerosis. II. MRI analysis of a multicenter, randomized, double-blind, placebo controlled trial. Neurology. 1993;43:662–7. - PubMed
    1. Hafler DA, Slavik JM, Anderson DE, O'Connor KC, De Jager P, Baecher-Allan C. Multiple sclerosis. Immunol Rev. 2005;204:208–31. - PubMed
    1. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type 1 interferons (α/β) in immunity and autoimmunity. Annu Rev Immunol. 2005;23:307–36. - PubMed
    1. Biron CA. Interferons α and β as immune regulators – a new look. Immunity. 2001;14:661–4. - PubMed

Publication types

-