scholarly journals Innate immunomodulation with MIS416: mechanism of action in experimental autoimmune encephalomyelitis

2021 ◽  
Author(s):  
◽  
Madeleine P. J. White

<p>Multiple sclerosis (MS) is an immune-mediated disease in which self-reacting T lymphocytes enter the central nervous system (CNS) and direct the damage of the myelin sheaths, which protect nerve axons. While there is no cure for MS currently, specific disease-modifying therapies are available that target the relapsing-remitting form of MS. However, these therapies are not effective in progressive forms of MS, which affect ~50% of MS patients in New Zealand, and thus there is an urgent need for novel treatments to be developed to treat these patients. MIS416 is a microparticle, which targets phagocytic cells by activating cytosolic receptors NOD2 and Toll-like receptor 9, and has recently completed a phase 2a trial in the treatment of progressive MS with promising results. The aim of this thesis was to elucidate the mechanism(s) by which MIS416 modified autoimmune disease using an animal model of MS, experimental autoimmune encephalomyelitis (EAE).  We determined that weekly intravenous doses of MIS416 were effective at reducing disease in a chronic model of MS, EAE, and we used this dosing regimen to further understand mechanisms involved in MIS416-induced EAE protection. It was found that MIS416 treatment induced a number of peripheral immune changes which had the potential to alter a T cell-mediated autoimmune disease, including increased levels of serum NO and IFN-γ, increased numbers of Tregs and macrophages as well as increased expression of PD-L1, an anti-proliferative ligand. Additionally, we also determined that MIS416 treatment significantly reduced T cell proliferation in vitro and in vivo.  To investigate which of these peripheral changes were involved in MIS416-mediated protection from EAE, we assessed each change individually in our disease model. We found that the central immune-modulating factor responsible for the protective effects of MIS416 treatment was IFN-γ (White, Webster, O'Sullivan, Stone, & La Flamme, 2014) as IFN-γ-/- mice treated with MIS416 were not protected from EAE. This protective role of MIS416-induced IFN-γ was likely to have been mediated through downstream effects given that in the absence of IFN-γ many of these changes were not seen. In particular, the macrophage population did not expand in MIS416-treated IFN-γ-/- mice, and the expression of the immunoregulatory ligand, PD-L1, was not enhanced suggesting that macrophage expressed PD-L1 may be one of the ways in which MIS416-induced IFN-γ altered EAE. While NO was found to contribute to the MIS416-mediated suppression of T cell proliferation ex vivo and IFN-γ-/- mice did not have elevated levels of NO, the in vivo inhibition of iNOS by aminoguanidine did not ablate MIS416 protection suggesting that these pathways were not central to disease protection. Additionally, MIS416 treatment was shown to increase the number and function of Tregs, and in the absence of IFN-γ this expansion was reduced. Altogether, this research indicated that IFN-γ was key to MIS416-mediated protection possibly through an expansion of immunoregulatory macrophage populations, enhanced PD-L1 expression, and enhanced Treg numbers and function.  In addition to these peripheral immune effects, we found that MIS416 treatment also altered cellular trafficking to the CNS. MIS416-treated EAE mice had reduced CNS infiltration as measured by both flow cytometry and histology compared to untreated EAE mice, and MIS416 treatment also reduced the EAE-induced permeability of the blood brain barrier. Furthermore, our study determined that MIS416-induced trafficking of immune cells to the CNS in the absence of EAE, and this trafficking also occurred in an IFN-γ-dependent manner. Given the suppressive nature of cells found within the periphery of MIS416-treated mice, the cells which trafficked to the CNS in response to MIS416 treatment may have had beneficial roles in EAE by suppressing T cell responses from within the CNS.  In summary, the work outlined in this thesis revealed that MIS416 treatment induced an immunoregulatory state that was capable of suppressing T cell proliferation and reducing EAE disease. Moreover, this research highlighted that in the right context, a pro-inflammatory cytokine such as IFN-γ may be protective in autoimmune disease. Finally, when these findings are applied more widely, they indicate that the immune modulations induced by MIS416 may have potential in other diseases mediated by immune dysregulation in addition to MS.</p>

2021 ◽  
Author(s):  
◽  
Madeleine P. J. White

<p>Multiple sclerosis (MS) is an immune-mediated disease in which self-reacting T lymphocytes enter the central nervous system (CNS) and direct the damage of the myelin sheaths, which protect nerve axons. While there is no cure for MS currently, specific disease-modifying therapies are available that target the relapsing-remitting form of MS. However, these therapies are not effective in progressive forms of MS, which affect ~50% of MS patients in New Zealand, and thus there is an urgent need for novel treatments to be developed to treat these patients. MIS416 is a microparticle, which targets phagocytic cells by activating cytosolic receptors NOD2 and Toll-like receptor 9, and has recently completed a phase 2a trial in the treatment of progressive MS with promising results. The aim of this thesis was to elucidate the mechanism(s) by which MIS416 modified autoimmune disease using an animal model of MS, experimental autoimmune encephalomyelitis (EAE).  We determined that weekly intravenous doses of MIS416 were effective at reducing disease in a chronic model of MS, EAE, and we used this dosing regimen to further understand mechanisms involved in MIS416-induced EAE protection. It was found that MIS416 treatment induced a number of peripheral immune changes which had the potential to alter a T cell-mediated autoimmune disease, including increased levels of serum NO and IFN-γ, increased numbers of Tregs and macrophages as well as increased expression of PD-L1, an anti-proliferative ligand. Additionally, we also determined that MIS416 treatment significantly reduced T cell proliferation in vitro and in vivo.  To investigate which of these peripheral changes were involved in MIS416-mediated protection from EAE, we assessed each change individually in our disease model. We found that the central immune-modulating factor responsible for the protective effects of MIS416 treatment was IFN-γ (White, Webster, O'Sullivan, Stone, & La Flamme, 2014) as IFN-γ-/- mice treated with MIS416 were not protected from EAE. This protective role of MIS416-induced IFN-γ was likely to have been mediated through downstream effects given that in the absence of IFN-γ many of these changes were not seen. In particular, the macrophage population did not expand in MIS416-treated IFN-γ-/- mice, and the expression of the immunoregulatory ligand, PD-L1, was not enhanced suggesting that macrophage expressed PD-L1 may be one of the ways in which MIS416-induced IFN-γ altered EAE. While NO was found to contribute to the MIS416-mediated suppression of T cell proliferation ex vivo and IFN-γ-/- mice did not have elevated levels of NO, the in vivo inhibition of iNOS by aminoguanidine did not ablate MIS416 protection suggesting that these pathways were not central to disease protection. Additionally, MIS416 treatment was shown to increase the number and function of Tregs, and in the absence of IFN-γ this expansion was reduced. Altogether, this research indicated that IFN-γ was key to MIS416-mediated protection possibly through an expansion of immunoregulatory macrophage populations, enhanced PD-L1 expression, and enhanced Treg numbers and function.  In addition to these peripheral immune effects, we found that MIS416 treatment also altered cellular trafficking to the CNS. MIS416-treated EAE mice had reduced CNS infiltration as measured by both flow cytometry and histology compared to untreated EAE mice, and MIS416 treatment also reduced the EAE-induced permeability of the blood brain barrier. Furthermore, our study determined that MIS416-induced trafficking of immune cells to the CNS in the absence of EAE, and this trafficking also occurred in an IFN-γ-dependent manner. Given the suppressive nature of cells found within the periphery of MIS416-treated mice, the cells which trafficked to the CNS in response to MIS416 treatment may have had beneficial roles in EAE by suppressing T cell responses from within the CNS.  In summary, the work outlined in this thesis revealed that MIS416 treatment induced an immunoregulatory state that was capable of suppressing T cell proliferation and reducing EAE disease. Moreover, this research highlighted that in the right context, a pro-inflammatory cytokine such as IFN-γ may be protective in autoimmune disease. Finally, when these findings are applied more widely, they indicate that the immune modulations induced by MIS416 may have potential in other diseases mediated by immune dysregulation in addition to MS.</p>


2001 ◽  
Vol 6 (12) ◽  
pp. 1055-1066 ◽  
Author(s):  
Keigyou Yoh ◽  
Takehiko Sugawara ◽  
Hozumi Motohashi ◽  
Yousuke Takahama ◽  
Akio Koyama ◽  
...  

Blood ◽  
1997 ◽  
Vol 90 (11) ◽  
pp. 4513-4521 ◽  
Author(s):  
Dieter Körholz ◽  
Ursula Banning ◽  
Halvard Bönig ◽  
Markus Grewe ◽  
Marion Schneider ◽  
...  

Abstract Interleukin-15 (IL-15) is a potent T-cell stimulating factor, which has recently been used for pre-clinical in vivo immunotherapy. Here, the IL-15 effect on CD3-stimulated peripheral human T cells was investigated. IL-15 induced a significant T-cell proliferation and upregulated CD25 expression. IL-15 significantly enhanced T-cell production of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-10. Between 10- and 100-fold greater concentrations of IL-15 were necessary to reach a biological effect equivalent to that of IL-2. Blockade of IL-2 binding to the high-affinity IL-2 receptor did not affect the IL-15 effects, suggesting that IL-15 did not act by inducing endogenous IL-2. Exogenously administered IL-10 significantly reduced the IL-15 and IL-2–mediated IFN-γ and TNF-α production, whereas T-cell proliferation and CD25 expression were not affected. The inhibitory effects of exogenously administered IL-10 on T-cell cytokine production appeared indirect, and are likely secondary to decreased IL-12 production by accessory cells. Inhibition of endogenous IL-10 binding to the IL-10 receptor significantly increased IFN-γ and TNF-α release from T cells. These data suggest that endogenous IL-10 can regulate activated T-cell production of IFN-γ and TNF-α via a paracrine negative feedback loop. The observations of this study could be of relevance for the therapeutic use of IL-15 in vivo.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4698-4698
Author(s):  
Myoung Woo Lee ◽  
Dae Seong Kim ◽  
Hye Jin Kim ◽  
Meong Hi Son ◽  
Soo Hyun Lee ◽  
...  

Abstract Abstract 4698 Background: It is important to overcome the limitations such as graft rejection and graft versus host disease (GvHD) in allogeneic hematopoietic stem cell transplantation. Mesenchymal stem cells (MSCs), which evoke only minimal immune reactivity, may have anti-inflammatory and immunomodulatory effects. Purpose: In this study, we aimed to identify the immunomodulatory properties of human MSCs and to elucidate the possible mechanism of their properties for clinical treatment of allogeneic conflicts using MSCs. Materials & Methods: We conducted a comparative analysis about the immunomodulatory properties of MSCs derived from adult human tissues, including bone marrow (BM), adipose tissues (AT), umbilical cord blood (CB), and cord Wharton's jelly (WJ), in vitro and in vivo models. Results: AT-MSCs, CB-MSCs, and WJ-MSCs effectively suppressed phytohemagglutinin (PHA)-induced T-cell proliferation as effectively as did BM-MSCs. Levels of interferon (IFN)-g secreted from activated T-cells increased over time, but these levels were significantly reduced when cocultured with each type of MSCs. In addition, expression of indoleamine 2,3-dioxygenase (IDO) increased in MSCs treated with IFN-γ via JAK/STAT1 signaling pathways. Treatment with anti-IFN-g antibodies, JAK1/2 inhibitor or STAT1 siRNA restored PHA-induced T-cell proliferation. Use of an antagonist, 1-methyl-L-tryptophan, also restored PHA-induced T-cell proliferation, suggesting that IDO contributes to IFN-g-induced immunosuppression in MSCs. Moreover, infusion of IFN-g-treated MSCs decreased symptoms for human peripheral blood-derived mononuclear cells-induced GvHD in NOD/SCID mice, which resulted in an increase of survival rate of in vivo GvHD model. Conclusion: These data indicate that IFN-γ produced by activated T-cells is correlated with induction of IDO expression in MSCs by IFN-γ receptor/JAK/STAT1 pathway, which resulted in suppression of T-cell proliferation. Our findings suggest that MSCs derived from BM, AT, CB, or WJ could be used for clinical treatment of allogeneic conflicts. Disclosures: No relevant conflicts of interest to declare.


2022 ◽  
Vol 12 ◽  
Author(s):  
Lili Tang ◽  
Ge Li ◽  
Yang Zheng ◽  
Chunmei Hou ◽  
Yang Gao ◽  
...  

Tim-3, an immune checkpoint inhibitor, is widely expressed on the immune cells and contributes to immune tolerance. However, the mechanisms by which Tim-3 induces immune tolerance remain to be determined. Major histocompatibility complex II (MHC-II) plays a key role in antigen presentation and CD4+T cell activation. Dysregulated expressions of Tim-3 and MHC-II are associated with the pathogenesis of many autoimmune diseases including multiple sclerosis. Here we demonstrated that, by suppressing MHC-II expression in macrophages via the STAT1/CIITA pathway, Tim-3 inhibits MHC-II-mediated autoantigen presentation and CD4+T cell activation. As a result, overexpression or blockade of Tim-3 signaling in mice with experimental autoimmune encephalomyelitis (EAE) inhibited or increased MHC-II expression respectively and finally altered clinical outcomes. We thus identified a new mechanism by which Tim-3 induces immune tolerance in vivo and regulating the Tim-3-MHC-II signaling pathway is expected to provide a new solution for multiple sclerosis treatment.


Sign in / Sign up

Export Citation Format

Share Document