Three donors that were used in experiments are shown; (C) Treg were depleted within PBMC of HD (black) or MS (reddish) and replaced with the same amount of Treg from an independent 3rd HD. Treg, but was abolished by anti-IL-6 receptor antibodies. However, magnitude and lethality of GvHD induced by MS T cells was significantly decreased after interferon-beta therapy and the reaction was prevented by Treg activation [17,18,19]. Until today such analysis were restricted to studies or were resolved in mouse models. Several potential drugs were recognized in the experimental autoimmune encephalomyelitis (EAE), the mouse model for MS, but translational studies exhibited only minor success rates [20,21]. To overcome limitations of such experimental mouse models, humanized mice have been developed. The transfer of human cells into immunodeficient mice allows the modulation of human immune responses [22,23]. In these mice, Zayoud detected antigen-specific responses of human T cells after the transfer of human PBMC from healthy donors (HD) and subsequent O-Phospho-L-serine subcutaneous immunization with myelin oligodendrocyte glycoprotein [24]. Here, we analyzed the influence of IFN- therapy on T cell immune regulation in regard to Treg control and observed that MS-related impaired suppression of T cells through Treg was significantly restored following IFN- treatment both and is limited. Especially, modern therapeutics for the targeted modulation are often epitope and species-specific. Therefore, we focused on the evaluation of a humanized mouse model allowing analysis and modulation of autoaggressive T cells from MS patients suppressor assays, we observed that CD4+ and CD8+ T cells from therapy-naive MS patients were resistant to Treg-mediated suppression of impartial third healthy individuals when compared to T cells from HD (Physique 1). Interestingly, this feature was independent of the diseases course, because no differences were observed between MS patients in relapse in comparison to those with a stable disease course (Supplemental Table S1). O-Phospho-L-serine Open in a separate window Physique 1 Treg resistant T cells of MS patients escaped control of functional active Treg. Treg-depleted PBMC from therapy-naive MS patients (MS, reddish) or healthy donors (HD, black) were cocultured with or without Treg and stimulated with anti-CD3 mAb. Proliferation was determined by 3H-Tdr incorporation on day three and displayed as mean SEM of triplicate measurements. (Left) bars symbolize imply SEM of triplicates of one representative experiment (of = 28); (Right) curves show proliferation in presence of different Treg numbers of five different donors, 0.05, ** 0.01 are shown. 2.2. O-Phospho-L-serine Transfer of PBMC from Therapy-Naive MS Patients into Immunodeficient Mice Resulted in an Accelerated Systemic Inflammation that Is Not Controlled by Activated Treg Thus, hyperactivated T cells in peripheral blood of therapy-naive MS patients are inefficiently controlled by functional active Treg. To further analyze this phenomenon resulting in disease prevention. Indeed, systemic inflammation after HD PBMC transfer into immunodeficient NOD/mice was prevented by activated third donor Treg, whereas mice engrafted with PBMC of therapy-naive MS patients developed an accelerated course of systemic inflammation. Mice showed early lethality that could not be ameliorated by activated Treg demonstrating that Treg resistance of MS T effector cells also occured (Physique 2C). Despite the injection of functional active Treg, all mice died within 18 days, demonstrating an increased aggressive activity of MS T effector cells and their resistance to Treg-mediated control. Open in a separate window Physique 2 Transferring CXCL12 human PBMC of MS patients into newborn immunodeficient mice resulted in a severe systemic inflammation without protection by activated Treg. (A) Plan: Treg were depleted within PBMC and replaced by the same amount of Treg from an independent HD. Subsequently 5 106 CD25-depleted PBMC were transferred intraperitoneally into newborn immunodeficient mice; (B) To control Treg function we cocultured them with allogeneic PBMC (ratio 1:1) and stimulated with anti-CD3 mAb. T cell proliferation was determined by 3H-Tdr incorporation on day three and displayed as mean SEM of triplicate measurements. Three donors that were used in experiments are shown; (C) Treg were depleted within PBMC of HD (black) or MS (reddish) and replaced with the same amount of Treg from an independent 3rd HD. Subsequently, 5 106 cells were injected with or without gp120 (5 g/mouse) intraperitoneally into.