In vivo demonstration of T cell migration and elimination in CNS autoimmune disease. Both transgenetically labelled MBP-specific and OVA-specific T cells undergo apoptosis during experimental autoimmune encephalomyelitis

1998 ◽  
Vol 90 (1) ◽  
pp. 80
Author(s):  
J. Bauer ◽  
M. Bradl ◽  
W.F. Hickey ◽  
C. Linington ◽  
H. Wekerle ◽  
...  
2016 ◽  
Vol 214 (1) ◽  
pp. 209-226 ◽  
Author(s):  
Xiaoyan Xu ◽  
Lei Han ◽  
Guixian Zhao ◽  
Shengjie Xue ◽  
Yunzhen Gao ◽  
...  

Directional autoreactive CD4+ T cell migration into the central nervous system plays a critical role in multiple sclerosis. Recently, DOCK8 was identified as a guanine-nucleotide exchange factor (GEF) for Cdc42 activation and has been associated with human mental retardation. Little is known about whether DOCK8 is related to multiple sclerosis (MS) and how to restrict its GEF activity. Using two screening systems, we found that LRCH1 competes with Cdc42 for interaction with DOCK8 and restrains T cell migration. In response to chemokine stimulation, PKCα phosphorylates DOCK8 at its three serine sites, promoting DOCK8 separation from LRCH1 and translocation to the leading edge to guide T cell migration. Point mutations at the DOCK8 serine sites block chemokine- and PKCα-induced T cell migration. Importantly, Dock8 mutant mice or Lrch1 transgenic mice were protected from MOG (35–55) peptide–induced experimental autoimmune encephalomyelitis (EAE), whereas Lrch1-deficient mice displayed a more severe phenotype. Notably, DOCK8 expression was markedly increased in PBMCs from the acute phase of MS patients. Together, our study demonstrates LRCH1 as a novel effector to restrain PKCα–DOCK8–Cdc42 module–induced T cell migration and ameliorate EAE.


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>


1995 ◽  
Vol 182 (2) ◽  
pp. 357-366 ◽  
Author(s):  
S J Khoury ◽  
L Gallon ◽  
W Chen ◽  
K Betres ◽  
M E Russell ◽  
...  

Experimental autoimmune encephalomyelitis (EAE), an experimental model for the study of multiple sclerosis, is an autoimmune disease of the central nervous system that can be induced in a number of species by immunization with myelin basic protein (MBP). MBP-reactive CD4+ T cells, predominantly expressing the V beta 8.2 T cell receptor (TCR), migrate from the peripheral lymphoid organs and initiate the inflammatory response in the brain. We have previously shown that a single intrathymic injection of MBP or its major encephalitogenic peptide (p71-90), but not a nonencephalitogenic peptide (p21-40), induces antigen-specific systemic tolerance and inhibits the induction of EAE in Lewis rats. In this study, we investigated the mechanisms of induction and maintenance of acquired thymic tolerance in this model. First, we investigated which thymic cell is responsible for "induction" of systemic tolerance. Thymic dendritic-enriched cells, isolated by plastic adherence, when incubated in vitro with p71-90 and injected intravenously into Lewis rats, were capable of preventing the development of EAE, but his protection was lost in thymectomized recipients. In addition, intravenous injection of thymic dendritic cells isolated from animals that had been previously injected intrathymically with p71-90 but not p21-40 also prevented the development of EAE. Second, to determine the "effector" mechanisms involved in acquired thymic tolerance, we compared TCR expression in the brains of animals with actively induced EAE with TCR expression in animals that received intrathymic injection of p71-90 or p21-40. Using a semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) technique, we found increased expression of CD4 and V beta 8.2 message in brains of immunized animals compared with those of naive animals. In animals intrathymically injected with p71-90 but not p21-40, CD4 and V beta 8.2 transcript levels were significantly reduced compared with immunized controls. Immunohistologic studies of brain tissue and spleens with specific V beta 8.2 and control V beta 10 monoclonal antibodies confirmed these observations in vivo. These findings, taken together with recent data demonstrating that activated T cells circulate through the thymus, suggest that interaction of thymic dendritic cells with specific TCR of activated peripheral T cells can lead to inactivation of these antigen-specific cells and confirm the role of V beta 8.2-expressing T cells in EAE.


2011 ◽  
Vol 2011 ◽  
pp. 1-6 ◽  
Author(s):  
Ani Grigorian ◽  
Michael Demetriou

Multiple sclerosis (MS) is an inflammatory demyelinating and neurodegenerative disease initiated by autoreactive T cells. Mgat5, a gene in the Asn (N-) linked protein glycosylation pathway, associates with MS severity and negatively regulates experimental autoimmune encephalomyelitis (EAE) and spontaneous inflammatory demyelination in mice. N-glycan branching by Mgat5 regulates interaction of surface glycoproteins with galectins, forming a molecular lattice that differentially controls the concentration of surface glycoproteins. T-cell receptor signaling, T-cell proliferation, TH1 differentiation, and CTLA-4 endocytosis are inhibited by Mgat5 branching. Non-T cells also contribute to MS pathogenesis and express abundant Mgat5 branched N-glycans. Here we explore whether Mgat5 deficiency in myelin-reactive T cells is sufficient to promote demyelinating disease. Adoptive transfer of myelin-reactive Mgat5−/− T cells into Mgat5+/+ versus Mgat5−/− recipients revealed more severe EAE in the latter, suggesting that Mgat5 branching deficiency in recipient naive T cells and/or non-T cells contribute to disease pathogenesis.


Sign in / Sign up

Export Citation Format

Share Document