Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.853
Filter
1.
J Neuroinflammation ; 21(1): 146, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824594

ABSTRACT

T cells play an important role in the acquired immune response, with regulatory T cells (Tregs) serving as key players in immune tolerance. Tregs are found in nonlymphoid and damaged tissues and are referred to as "tissue Tregs". They have tissue-specific characteristics and contribute to immunomodulation, homeostasis, and tissue repair through interactions with tissue cells. However, important determinants of Treg tissue specificity, such as antigen specificity, tissue environment, and pathology, remain unclear. In this study, we analyzed Tregs in the central nervous system of mice with ischemic stroke and experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. The gene expression pattern of brain Tregs in the EAE model was more similar to that of ischemic stroke Tregs in the brain than to that of spinal cord Tregs. In addition, most T-cell receptors (TCRs) with high clonality were present in both the brain and spinal cord. Furthermore, Gata3+ and Rorc+ Tregs expressed TCRs recognizing MOG in the spinal cord, suggesting a tissue environment conducive to Rorc expression. Tissue-specific chemokine/chemokine receptor interactions in the spinal cord and brain influenced Treg localization. Finally, spinal cord- or brain-derived Tregs had greater anti-inflammatory capacities in EAE mice, respectively. Taken together, these findings suggest that the tissue environment, rather than pathogenesis or antigen specificity, is the primary determinant of the tissue-specific properties of Tregs. These findings may contribute to the development of novel therapies to suppress inflammation through tissue-specific Treg regulation.


Subject(s)
Brain , Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Spinal Cord , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , Mice , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Female , Disease Models, Animal
2.
J Extracell Vesicles ; 13(6): e12446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38844736

ABSTRACT

Dendritic cells (DCs) are essential orchestrators of immune responses and represent potential targets for immunomodulation in autoimmune diseases. Human amniotic fluid secretome is abundant in immunoregulatory factors, with extracellular vesicles (EVs) being a significant component. However, the impact of these EVs on dendritic cells subsets remain unexplored. In this study, we investigated the interaction between highly purified dendritic cell subsets and EVs derived from amniotic fluid stem cell lines (HAFSC-EVs). Our results suggest that HAFSC-EVs are preferentially taken up by conventional dendritic cell type 2 (cDC2) through CD29 receptor-mediated internalization, resulting in a tolerogenic DC phenotype characterized by reduced expression and production of pro-inflammatory mediators. Furthermore, treatment of cDC2 cells with HAFSC-EVs in coculture systems resulted in a higher proportion of T cells expressing the regulatory T cell marker Foxp3 compared to vehicle-treated control cells. Moreover, transfer of HAFSC-EV-treated cDC2s into an EAE mouse model resulted in the suppression of autoimmune responses and clinical improvement. These results suggest that HAFSC-EVs may serve as a promising tool for reprogramming inflammatory cDC2s towards a tolerogenic phenotype and for controlling autoimmune responses in the central nervous system, representing a potential platform for the study of the effects of EVs in DC subsets.


Subject(s)
Amniotic Fluid , Dendritic Cells , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Extracellular Vesicles , Multiple Sclerosis , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Amniotic Fluid/cytology , Amniotic Fluid/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Multiple Sclerosis/therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Female , Stem Cells/metabolism , Stem Cells/cytology , Mice, Inbred C57BL
3.
Sci Rep ; 14(1): 13146, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849434

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating disease affecting the central nervous system (CNS). T helper (Th) 17 cells are involved in the pathogenesis of MS and its animal model of experimental autoimmune encephalomyelitis (EAE) by infiltrating the CNS and producing effector molecules that engage resident glial cells. Among these glial cells, astrocytes have a central role in coordinating inflammatory processes by responding to cytokines and chemokines released by Th17 cells. In this study, we examined the impact of pathogenic Th17 cells on astrocytes in vitro and in vivo. We identified that Th17 cells reprogram astrocytes by driving transcriptomic changes partly through a Janus Kinase (JAK)1-dependent mechanism, which included increased chemokines, interferon-inducible genes, and cytokine receptors. In vivo, we observed a region-specific heterogeneity in the expression of cell surface cytokine receptors on astrocytes, including those for IFN-γ, IL-1, TNF-α, IL-17, TGFß, and IL-10. Additionally, these receptors were dynamically regulated during EAE induced by adoptive transfer of myelin-reactive Th17 cells. This study overall provides evidence of Th17 cell reprogramming of astrocytes, which may drive changes in the astrocytic responsiveness to cytokines during autoimmune neuroinflammation.


Subject(s)
Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Janus Kinase 1 , Myelin-Oligodendrocyte Glycoprotein , Receptors, Cytokine , Th17 Cells , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Animals , Astrocytes/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Mice , Receptors, Cytokine/metabolism , Receptors, Cytokine/genetics , Janus Kinase 1/metabolism , Mice, Inbred C57BL , Cytokines/metabolism , Cellular Reprogramming , Female , Cells, Cultured
4.
Int Immunopharmacol ; 134: 112246, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759372

ABSTRACT

BACKGROUND: A wide array of histone deacetylase (HDAC) inhibitors and aryl hydrocarbon receptor (AHR) agonists commonly arrest experimental autoimmune encephalomyelitis (EAE). However, it is not known whether HDAC inhibition is linked to the AHR signaling pathway in EAE. METHODS: We investigated how the pan-HDAC inhibitor SB939 (pracinostat) exerted immunoregulatory action in the myelin oligodendrocyte glycoprotein 35-55 (MOG35-55)-induced EAE mouse model by evaluating changes in of signal transducer and activator of transcription 3 (STAT3) acetylation and the expression of indoleamine 2,3-dioxygenase 1 (IDO1) and AHR in inflamed spinal cords during EAE evolution. We proved the involvement of IDO1 and the AHR in SB939-mediated immunosuppression using Ido1-/- and Ahr-/- mice. RESULTS: Administration with SB939 halted EAE progression, which depended upon IDO1 expression in neurons of the central nervous system (CNS). Our in vitro and in vivo studies demonstrated that SB939 sustained the interleukin-6-induced acetylation of STAT3, resulting in the stable transcriptional activation of Ido1. The therapeutic effect of SB939 also required the AHR, which is expressed mainly in CD4+ T cells and macrophages in CNS disease lesions. Finally, SB939 was shown to markedly reduce the proliferation of CD4+ T cells in inflamed neuronal tissues but not in the spleen or draining lymph nodes. CONCLUSIONS: Overall, our results suggest that IDO1 tryptophan metabolites produced by neuronal cells may act on AHR in pathogenic CD4+ T cells in a paracrine fashion in the CNS and that the specific induction of IDO1 expression in neurons at disease-afflicted sites can be considered a therapeutic approach to block the progression of multiple sclerosis without affecting systemic immunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Histone Deacetylase Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Mice, Inbred C57BL , Mice, Knockout , Neurons , STAT3 Transcription Factor , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Mice , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Female , Spinal Cord/pathology , Spinal Cord/metabolism , Spinal Cord/immunology , Spinal Cord/drug effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Central Nervous System/immunology , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Disease Progression , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Peptide Fragments/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Interleukin-6/metabolism , Interleukin-6/genetics
5.
Int Immunopharmacol ; 135: 112326, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38796967

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disorder of the central nervous system. Recent research has revealed that mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), containing specific miRNAs, possess immunomodulatory properties and have demonstrated therapeutic potential in the treatment of MS. This study aimed to investigate the role MSC-EVs, containing microRNA-181a-5p (miR-181a-5p) in both experimental autoimmune encephalomyelitis (EAE), an established animal model of MS, and lipopolysaccharide-stimulated BV2 microglia. We evaluated clinical symptoms and inflammatory responses in EAE mice following intrathecal injections of MSC-EVs. MSC-EVs containing miR-181a-5p were co-cultured with microglia to explore their impact on inflammation and cell pyroptosis. We validated the interaction between miR-181a-5p and its downstream regulators and conducted in vivo verification by injecting manipulated EVs containing miR-181a-5p into EAE mice. Our results demonstrated that MSC-EVs, containing miR-181a-5p reduced the clinical symptoms of EAE mice. Furthermore, we observed downregulation of miR-181a-5p in EAE model mice, and its expression was restored after treatment with MSC-EVs, which corresponded to suppressed microglial inflammation and pyroptosis. Additionally, EVs containing miR-181a-5p mitigated spinal cord injury and demyelination in EAE mice. Mechanistically, ubiquitin-specific protease 15 (USP15) exhibited high expression in EAE mice, and miR-181a-5p was specifically targeted and bound to USP15, thereby regulating the RelA/NEK7 axis. In conclusion, MSC-EVs containing miR-181a-5p inhibit microglial inflammation and pyroptosis through the USP15-mediated RelA/NEK7 axis, thus alleviating the clinical symptoms of EAE. These findings present a potential therapeutic approach for the treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Extracellular Vesicles , Mice, Inbred C57BL , MicroRNAs , Microglia , Animals , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Mice , Microglia/metabolism , Female , Mesenchymal Stem Cells/metabolism , Pyroptosis , Cell Line , Multiple Sclerosis/therapy , Humans , Disease Models, Animal , Lipopolysaccharides , Demyelinating Diseases/therapy
6.
J Neuroinflammation ; 21(1): 144, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822334

ABSTRACT

Cumulative evidence has established that Interferon (IFN)-γ has both pathogenic and protective roles in Multiple Sclerosis and the animal model, Experimental Autoimmune Encephalomyelitis (EAE). However, the underlying mechanisms to the beneficial effects of IFN-γ are not well understood. In this study, we found that IFN-γ exerts therapeutic effects on chronic, relapsing-remitting, and chronic progressive EAE models. The frequency of regulatory T (Treg) cells in spinal cords from chronic EAE mice treated with IFN-γ was significantly increased with no effect on Th1 and Th17 cells. Consistently, depletion of FOXP3-expressing cells blocked the protective effects of IFN-γ, indicating that the therapeutic effect of IFN-γ depends on the presence of Treg cells. However, IFN-γ did not trigger direct in vitro differentiation of Treg cells. In vivo administration of blocking antibodies against either interleukin (IL)-10, transforming growth factor (TGF)-ß or program death (PD)-1, revealed that the protective effects of IFN-γ in EAE were also dependent on TGF-ß and PD-1, but not on IL-10, suggesting that IFN-γ might have an indirect role on Treg cells acting through antigen-presenting cells. Indeed, IFN-γ treatment increased the frequency of a subset of splenic CD11b+ myeloid cells expressing TGF-ß-Latency Associated Peptide (LAP) and program death ligand 1 (PD-L1) in a signal transducer and activator of transcription (STAT)-1-dependent manner. Furthermore, splenic CD11b+ cells from EAE mice preconditioned in vitro with IFN-γ and myelin oligodendrocyte glycoprotein (MOG) peptide exhibited a tolerogenic phenotype with the capability to induce conversion of naïve CD4+ T cells mediated by secretion of TGF-ß. Remarkably, adoptive transfer of splenic CD11b+ cells from IFN-γ-treated EAE mice into untreated recipient mice ameliorated clinical symptoms of EAE and limited central nervous system infiltration of mononuclear cells and effector helper T cells. These results reveal a novel cellular and molecular mechanism whereby IFN-γ promotes beneficial effects in EAE by endowing splenic CD11b+ myeloid cells with tolerogenic and therapeutic activities.


Subject(s)
CD11b Antigen , Encephalomyelitis, Autoimmune, Experimental , Interferon-gamma , Mice, Inbred C57BL , Myeloid Cells , Spleen , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Interferon-gamma/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Spleen/immunology , CD11b Antigen/metabolism , Female , Myelin-Oligodendrocyte Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Transforming Growth Factor beta/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Forkhead Transcription Factors/metabolism , Disease Models, Animal
7.
Sci Immunol ; 9(95): eabq1558, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701190

ABSTRACT

Steroid resistance poses a major challenge for the management of autoimmune neuroinflammation. T helper 17 (TH17) cells are widely implicated in the pathology of steroid resistance; however, the underlying mechanisms are unknown. In this study, we identified that interleukin-1 receptor (IL-1R) blockade rendered experimental autoimmune encephalomyelitis (EAE) mice sensitive to dexamethasone (Dex) treatment. Interleukin-1ß (IL-1ß) induced a signal transducer and activator of transcription 5 (STAT5)-mediated steroid-resistant transcriptional program in TH17 cells, which promoted inflammatory cytokine production and suppressed Dex-induced anti-inflammatory genes. TH17-specific deletion of STAT5 ablated the IL-1ß-induced steroid-resistant transcriptional program and rendered EAE mice sensitive to Dex treatment. IL-1ß synergized with Dex to promote the STAT5-dependent expression of CD69 and the development of central nervous system (CNS)-resident CD69+ TH17 cells. Combined IL-1R blockade and Dex treatment ablated CNS-resident TH17 cells, reduced EAE severity, and prevented relapse. CD69+ tissue-resident TH17 cells were also detected in brain lesions of patients with multiple sclerosis. These findings (i) demonstrate that IL-1ß-STAT5 signaling in TH17 cells mediates steroid resistance and (ii) identify a therapeutic strategy for reversing steroid resistance in TH17-mediated CNS autoimmunity.


Subject(s)
Dexamethasone , Encephalomyelitis, Autoimmune, Experimental , Interleukin-1beta , STAT5 Transcription Factor , Th17 Cells , Animals , Th17 Cells/immunology , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/immunology , Mice , Interleukin-1beta/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Mice, Inbred C57BL , Drug Resistance , Signal Transduction/immunology , Mice, Knockout , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/drug therapy , Female , Humans
8.
Yakugaku Zasshi ; 144(5): 489-496, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692922

ABSTRACT

The tumor necrosis factor receptor (TNFR)-associated factor (TRAF) family of molecules are intracellular adaptors that regulate cellular signaling through members of the TNFR and Toll-like receptor superfamily. Mammals have seven TRAF molecules numbered sequentially from TRAF1 to TRAF7. Although TRAF5 was identified as a potential regulator of TNFR superfamily members, the in vivo function of TRAF5 has not yet been fully elucidated. We identified an unconventional role of TRAF5 in interleukin-6 (IL-6) receptor signaling involving CD4+ T cells. Moreover, TRAF5 binds to the signal-transducing glycoprotein 130 (gp130) receptor for IL-6 and inhibits the activity of the janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway. In addition, Traf5-deficient CD4+ T cells exhibit significantly enhanced IL-6-driven differentiation of T helper 17 (Th17) cells, which exacerbates neuroinflammation in experimental autoimmune encephalomyelitis. Furthermore, TRAF5 demonstrates a similar activity to gp130 for IL-27, another cytokine of the IL-6 family. Additionally, Traf5-deficient CD4+ T cells display significantly increased IL-27-mediated differentiation of Th1 cells, which increases footpad swelling in delayed-type hypersensitivity response. Thus, TRAF5 functions as a negative regulator of gp130 in CD4+ T cells. This review aimed to explain how TRAF5 controls the differentiation of CD4+ T cells and discuss how the expression of TRAF5 in T cells and other cell types can influence the development and progression of autoimmune and inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes , Encephalomyelitis, Autoimmune, Experimental , Signal Transduction , TNF Receptor-Associated Factor 5 , Humans , Animals , TNF Receptor-Associated Factor 5/genetics , TNF Receptor-Associated Factor 5/metabolism , TNF Receptor-Associated Factor 5/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Cytokine Receptor gp130/physiology , Cytokine Receptor gp130/metabolism , Th17 Cells/immunology , Interleukin-6/metabolism , Interleukin-6/physiology , Cell Differentiation , Receptors, Interleukin-6/physiology , Receptors, Interleukin-6/metabolism , Janus Kinases/metabolism , Janus Kinases/physiology , STAT Transcription Factors/physiology , STAT Transcription Factors/metabolism , Mice
9.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692923

ABSTRACT

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Subject(s)
Adaptor Proteins, Signal Transducing , Lymphocyte Activation , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes , ZAP-70 Protein-Tyrosine Kinase , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , CD3 Complex , Cell Adhesion , Cell Movement , Chemokine CXCL12/physiology , Chemokine CXCL12/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Inflammation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Propionibacterium acnes/physiology , Propionibacterium acnes/immunology , Receptors, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/physiology
10.
CNS Neurosci Ther ; 30(5): e14736, 2024 05.
Article in English | MEDLINE | ID: mdl-38739106

ABSTRACT

AIMS: Multiple sclerosis (MS) is a neuroinflammatory demyelinating disease. Microglia are reportedly involved in the pathogenesis of MS. However, the key molecules that control the inflammatory activity of microglia in MS have not been identified. METHODS: Experimental autoimmune encephalomyelitis (EAE) mice were randomized into CD22 blockade and control groups. The expression levels of microglial CD22 were measured by flow cytometry, qRT-PCR, and immunofluorescence. The effects of CD22 blockade were examined via in vitro and in vivo studies. RESULTS: We detected increased expression of microglial CD22 in EAE mice. In addition, an in vitro study revealed that lipopolysaccharide upregulated the expression of CD22 in microglia and that CD22 blockade modulated microglial polarization. Moreover, an in vivo study demonstrated that CD22 blockade aggravated EAE in mice and promoted microglial M1 polarization. CONCLUSION: Collectively, our study indicates that CD22 may be protective against EAE and may play a critical role in the maintenance of immune homeostasis in EAE mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Microglia , Sialic Acid Binding Ig-like Lectin 2 , Animals , Female , Mice , Cell Polarity/drug effects , Cell Polarity/physiology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein/immunology
11.
Int J Mol Sci ; 25(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38732169

ABSTRACT

Infections may affect the course of autoimmune inflammatory diseases of the central nervous system (CNS), such as multiple sclerosis (MS). Infections with lactate dehydrogenase-elevating virus (LDV) protected mice from developing experimental autoimmune encephalomyelitis (EAE), a mouse counterpart of MS. Uninfected C57BL/6 mice immunized with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) experienced paralysis and lost weight at a greater rate than mice who had previously been infected with LDV. LDV infection decreased the presentation of the MOG peptide by CD11b+CD11c+ dendritic cells (DC) to pathogenic T lymphocytes. When comparing non-infected mice to infected mice, the histopathological examination of the CNS showed more areas of demyelination and CD45+ and CD3+, but not Iba1+ cell infiltration. These results suggest that the protective effect of LDV infection against EAE development is mediated by a suppression of myelin antigen presentation by a specific DC subset to autoreactive T lymphocytes. Such a mechanism might contribute to the general suppressive effect of infections on autoimmune diseases known as the hygiene hypothesis.


Subject(s)
Dendritic Cells , Encephalomyelitis, Autoimmune, Experimental , Lactate dehydrogenase-elevating virus , Multiple Sclerosis , Myelin-Oligodendrocyte Glycoprotein , Animals , Female , Mice , Antigen Presentation/immunology , Cardiovirus Infections/immunology , CD11b Antigen/metabolism , CD11b Antigen/immunology , CD11c Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/virology , Lactate dehydrogenase-elevating virus/immunology , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Front Immunol ; 15: 1391949, 2024.
Article in English | MEDLINE | ID: mdl-38765015

ABSTRACT

Dimethyl fumarate (DMF, Tecfidera) is an oral drug utilized to treat relapsing-remitting multiple sclerosis (MS). DMF treatment reduces disease activity in MS. Gastrointestinal discomfort is a common adverse effect of the treatment with DMF. This study aimed to investigate the effect of DMF administration in the gut draining lymph nodes cells of C57BL6/J female mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. We have demonstrated that the treatment with DMF (7.5 mg/kg) significantly reduces the severity of EAE. This reduction of the severity is accompanied by the increase of both proinflammatory and anti-inflammatory mechanisms at the beginning of the treatment. As the treatment progressed, we observed an increasing number of regulatory Foxp3 negative CD4 T cells (Tr1), and anti-inflammatory cytokines such as IL-27, as well as the reduction of PGE2 level in the mesenteric lymph nodes of mice with EAE. We provide evidence that DMF induces a gradual anti-inflammatory response in the gut draining lymph nodes, which might contribute to the reduction of both intestinal discomfort and the inflammatory response of EAE. These findings indicate that the gut is the first microenvironment of action of DMF, which may contribute to its effects of reducing disease severity in MS patients.


Subject(s)
Dimethyl Fumarate , Encephalomyelitis, Autoimmune, Experimental , Lymph Nodes , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Animals , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymph Nodes/immunology , Lymph Nodes/drug effects , Mice , Female , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Mesentery , Cytokines/metabolism , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Disease Models, Animal
13.
Sci Immunol ; 9(95): eadk0865, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701189

ABSTRACT

Dysregulated B cell cytokine production contributes to pathogenesis of immune-mediated diseases including multiple sclerosis (MS); however, the underlying mechanisms are poorly understood. In this study we investigated how cytokine secretion by pro-inflammatory (GM-CSF-expressing) and anti-inflammatory (IL-10-expressing) B cells is regulated. Pro-inflammatory human B cells required increased oxidative phosphorylation (OXPHOS) compared with anti-inflammatory B cells. OXPHOS reciprocally modulated pro- and anti-inflammatory B cell cytokines through regulation of adenosine triphosphate (ATP) signaling. Partial inhibition of OXPHOS or ATP-signaling including with BTK inhibition resulted in an anti-inflammatory B cell cytokine shift, reversed the B cell cytokine imbalance in patients with MS, and ameliorated neuroinflammation in a myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalitis mouse model. Our study identifies how pro- and anti-inflammatory cytokines are metabolically regulated in B cells and identifies ATP and its metabolites as a "fourth signal" that shapes B cell responses and is a potential target for restoring the B cell cytokine balance in autoimmune diseases.


Subject(s)
B-Lymphocytes , Cytokines , Encephalomyelitis, Autoimmune, Experimental , Inflammation , Multiple Sclerosis , Oxidative Phosphorylation , Animals , Multiple Sclerosis/immunology , Humans , Cytokines/immunology , Cytokines/metabolism , Mice , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Female , Male , Mice, Inbred C57BL , Adult , Adenosine Triphosphate/metabolism , Middle Aged
14.
Biol Sex Differ ; 15(1): 41, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750588

ABSTRACT

BACKGROUND: Differences in immune responses between women and men are leading to a strong sex bias in the incidence of autoimmune diseases that predominantly affect women, such as multiple sclerosis (MS). MS manifests in more than twice as many women, making sex one of the most important risk factor. However, it is incompletely understood which genes contribute to sex differences in autoimmune incidence. To address that, we conducted a gene expression analysis in female and male human spleen and identified the transmembrane protein CD99 as one of the most significantly differentially expressed genes with marked increase in men. CD99 has been reported to participate in immune cell transmigration and T cell regulation, but sex-specific implications have not been comprehensively investigated. METHODS: In this study, we conducted a gene expression analysis in female and male human spleen using the Genotype-Tissue Expression (GTEx) project dataset to identify differentially expressed genes between women and men. After successful validation on protein level of human immune cell subsets, we assessed hormonal regulation of CD99 as well as its implication on T cell regulation in primary human T cells and Jurkat T cells. In addition, we performed in vivo assays in wildtype mice and in Cd99-deficient mice to further analyze functional consequences of differential CD99 expression. RESULTS: Here, we found higher CD99 gene expression in male human spleens compared to females and confirmed this expression difference on protein level on the surface of T cells and pDCs. Androgens are likely dispensable as the cause shown by in vitro assays and ex vivo analysis of trans men samples. In cerebrospinal fluid, CD99 was higher on T cells compared to blood. Of note, male MS patients had lower CD99 levels on CD4+ T cells in the CSF, unlike controls. By contrast, both sexes had similar CD99 expression in mice and Cd99-deficient mice showed equal susceptibility to experimental autoimmune encephalomyelitis compared to wildtypes. Functionally, CD99 increased upon human T cell activation and inhibited T cell proliferation after blockade. Accordingly, CD99-deficient Jurkat T cells showed decreased cell proliferation and cluster formation, rescued by CD99 reintroduction. CONCLUSIONS: Our results demonstrate that CD99 is sex-specifically regulated in healthy individuals and MS patients and that it is involved in T cell costimulation in humans but not in mice. CD99 could potentially contribute to MS incidence and susceptibility in a sex-specific manner.


The immune system protects us from bacterial and viral infections and impacts the outcome of many diseases. Thus, understanding immunological processes is crucial to unravel pathogenic mechanisms and to develop new therapeutic treatment options. Sex is a biological variable affecting immunity and it is known that females and males differ in their immunological responses. Women mount stronger immune responses leading to more rapid control of infections and greater vaccine efficacy compared to men. However, this enhanced immune responsiveness is accompanied by female preponderance and susceptibility to autoimmune diseases like systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis (MS). MS sex ratio varies around 2:1 to 3:1 with a steadily increasing incidence in female MS patients making sex one of the top risk factors for developing MS. However, the underlying biological mechanisms including sex hormones as well as genetic and epigenetic factors and their complex interplay remain largely unknown. Here, we discovered the gene and its encoded protein CD99 to be differentially expressed between women and men with men showing increased expression on many immune cell subsets including T cells. Since T cells are key contributors to MS pathogenesis, we examined the role of CD99 on T cells of healthy individuals and MS patients. We were able to identify CD99-mediated T cell regulation, which might contribute to sex differences in MS susceptibility and incidence indicating the importance to include sex as a biological variable. Of note, these differences were not reproduced in mice showing the necessity of functional research in humans.


Subject(s)
12E7 Antigen , Multiple Sclerosis , Sex Characteristics , T-Lymphocytes , Animals , Female , Male , Humans , 12E7 Antigen/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Mice, Inbred C57BL , Jurkat Cells , Spleen/metabolism , Spleen/immunology , Species Specificity , Mice , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice, Knockout , Adult
15.
Eur J Immunol ; 54(6): e2350548, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38634287

ABSTRACT

Transforming growth factor beta (TGF-ß) signaling is essential for a balanced immune response by mediating the development and function of regulatory T cells (Tregs) and suppressing autoreactive T cells. Disruption of this balance can result in autoimmune diseases, including multiple sclerosis (MS). MicroRNAs (miRNAs) targeting TGF-ß signaling have been shown to be upregulated in naïve CD4 T cells in MS patients, resulting in a limited in vitro generation of human Tregs. Utilizing the murine model experimental autoimmune encephalomyelitis, we show that perinatal administration of miRNAs, which target the TGF-ß signaling pathway, enhanced susceptibility to central nervous system (CNS) autoimmunity. Neonatal mice administered with these miRNAs further exhibited reduced Treg frequencies with a loss in T cell receptor repertoire diversity following the induction of experimental autoimmune encephalomyelitis in adulthood. Exacerbated CNS autoimmunity as a result of miRNA overexpression in CD4 T cells was accompanied by enhanced Th1 and Th17 cell frequencies. These findings demonstrate that increased levels of TGF-ß-associated miRNAs impede the development of a diverse Treg population, leading to enhanced effector cell activity, and contributing to an increased susceptibility to CNS autoimmunity. Thus, TGF-ß-targeting miRNAs could be a risk factor for MS, and recovering optimal TGF-ß signaling may restore immune homeostasis in MS patients.


Subject(s)
Autoimmunity , Central Nervous System , Encephalomyelitis, Autoimmune, Experimental , MicroRNAs , Multiple Sclerosis , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Transforming Growth Factor beta , MicroRNAs/genetics , MicroRNAs/immunology , Animals , T-Lymphocytes, Regulatory/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Transforming Growth Factor beta/metabolism , Mice , Signal Transduction/immunology , Autoimmunity/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , Humans , Central Nervous System/immunology , Th17 Cells/immunology , Mice, Inbred C57BL , Th1 Cells/immunology , Cell Differentiation/immunology , Female
16.
Sci Adv ; 10(17): eadk1045, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38657065

ABSTRACT

T helper 17 (TH17) cells are implicated in autoimmune diseases, and several metabolic processes are shown to be important for their development and function. In this study, we report an essential role for sphingolipids synthesized through the de novo pathway in TH17 cell development. Deficiency of SPTLC1, a major subunit of serine palmitoyl transferase enzyme complex that catalyzes the first and rate-limiting step of de novo sphingolipid synthesis, impaired glycolysis in differentiating TH17 cells by increasing intracellular reactive oxygen species (ROS) through enhancement of nicotinamide adenine dinucleotide phosphate oxidase 2 activity. Increased ROS leads to impaired activation of mammalian target of rapamycin C1 and reduced expression of hypoxia-inducible factor 1-alpha and c-Myc-induced glycolytic genes. SPTLCI deficiency protected mice from developing experimental autoimmune encephalomyelitis and experimental T cell transfer colitis. Our results thus show a critical role for de novo sphingolipid biosynthetic pathway in shaping adaptive immune responses with implications in autoimmune diseases.


Subject(s)
Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental , Serine C-Palmitoyltransferase , Sphingolipids , Th17 Cells , Animals , Sphingolipids/metabolism , Sphingolipids/biosynthesis , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/cytology , Mice , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Reactive Oxygen Species/metabolism , Glycolysis , Mice, Knockout , Colitis/metabolism , Colitis/pathology , Mice, Inbred C57BL
17.
J Immunol ; 212(11): 1647-1657, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38578274

ABSTRACT

Long-term therapeutic outcomes of multiple sclerosis (MS) remain hindered by the chronic nature of immune cell stimulation toward self-antigens. Development of novel methods to target and deplete autoreactive T lymphocytes remains an attractive target for therapeutics for MS. We developed a programmed cell death 1 (PD-1)-targeted radiolabeled mAb and assessed its ability to deplete activated PD-1+ T lymphocytes in vitro and its ability to reduce disease burden of the myelin oligodendrocyte glycoprotein 35-55 experimental autoimmune encephalomyelitis (EAE) model in C57BL/6 mice. We also investigated the upregulation of PD-1 on infiltrating lymphocytes in an animal model of MS. Finally, we demonstrate the (to our knowledge) first reported positron-emission tomography/computed tomography imaging of activated PD-1+ cells in the EAE animal model of MS. We found that the 177Lu radioisotope-labeled anti-PD-1 mAb demonstrated significant in vitro cytotoxicity toward activated CD4+PD-1+ T lymphocytes and led to significant reduction in overall disease progression in the EAE animal model. Our results show high expression of PD-1 on infiltrating lymphocytes in the spinal cords of EAE diseased animals. Positron-emission tomography/computed tomography imaging of the anti-PD-1 mAb demonstrated significant uptake in the cervical draining lymph nodes highlighting accumulation of activated lymphocytes. Targeted depletion of T lymphocytes using T cell activation markers such as PD-1 may present a novel method to reduce autoimmune attack and inflammation in autoimmune diseases such as MS. Development of multimodal nuclear theranostic agents may present the opportunity to monitor T cell activation via imaging radioisotopes and simultaneously treat MS using therapeutic radioisotopes.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Lymphocyte Activation , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , Mice , Lymphocyte Activation/immunology , Antibodies, Monoclonal , T-Lymphocytes/immunology , Female , Disease Models, Animal , Multiple Sclerosis/immunology , Multiple Sclerosis/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Humans
18.
Eur J Immunol ; 54(6): e2350761, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38566526

ABSTRACT

In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Endothelial Cells , Macrophages , Mice, Inbred C57BL , Mice, Knockout , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Macrophages/immunology , Macrophages/metabolism , Endothelial Cells/metabolism , Endothelial Cells/immunology , Spinal Cord/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , CD4-Positive T-Lymphocytes/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal
19.
Nat Biomed Eng ; 8(5): 611-627, 2024 May.
Article in English | MEDLINE | ID: mdl-38561491

ABSTRACT

Butyrate-a metabolite produced by commensal bacteria-has been extensively studied for its immunomodulatory effects on immune cells, including regulatory T cells, macrophages and dendritic cells. However, the development of butyrate as a drug has been hindered by butyrate's poor oral bioavailability, owing to its rapid metabolism in the gut, its low potency (hence, necessitating high dosing), and its foul smell and taste. Here we report that the oral bioavailability of butyrate can be increased by esterifying it to serine, an amino acid transporter that aids the escape of the resulting odourless and tasteless prodrug (O-butyryl-L-serine, which we named SerBut) from the gut, enhancing its systemic uptake. In mice with collagen-antibody-induced arthritis (a model of rheumatoid arthritis) and with experimental autoimmune encephalomyelitis (a model of multiple sclerosis), we show that SerBut substantially ameliorated disease severity, modulated key immune cell populations systemically and in disease-associated tissues, and reduced inflammatory responses without compromising the global immune response to vaccination. SerBut may become a promising therapeutic for autoimmune and inflammatory diseases.


Subject(s)
Arthritis, Experimental , Biological Availability , Butyrates , Prodrugs , Serine , Animals , Prodrugs/pharmacology , Prodrugs/therapeutic use , Prodrugs/pharmacokinetics , Prodrugs/chemistry , Mice , Serine/metabolism , Butyrates/pharmacology , Butyrates/therapeutic use , Butyrates/chemistry , Butyrates/administration & dosage , Administration, Oral , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Female
20.
Sci Immunol ; 9(94): eadh0085, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669317

ABSTRACT

Thymic negative selection of the T cell receptor (TCR) repertoire is essential for establishing self-tolerance and acquired allograft tolerance following organ transplantation. However, it is unclear whether and how peripheral clonal deletion of alloreactive T cells induces transplantation tolerance. Here, we establish that programmed cell death protein 1 (PD-1) is a hallmark of alloreactive T cells and is associated with clonal expansion after alloantigen encounter. Moreover, we found that diphtheria toxin receptor (DTR)-mediated ablation of PD-1+ cells reshaped the TCR repertoire through peripheral clonal deletion of alloreactive T cells and promoted tolerance in mouse transplantation models. In addition, by using PD-1-specific depleting antibodies, we found that antibody-mediated depletion of PD-1+ cells prevented heart transplant rejection and the development of experimental autoimmune encephalomyelitis (EAE) in humanized PD-1 mice. Thus, these data suggest that PD-1 is an attractive target for peripheral clonal deletion and induction of immune tolerance.


Subject(s)
Clonal Deletion , Immune Tolerance , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Animals , Programmed Cell Death 1 Receptor/immunology , Mice , Clonal Deletion/immunology , Immune Tolerance/immunology , Humans , Encephalomyelitis, Autoimmune, Experimental/immunology , Heart Transplantation , T-Lymphocytes/immunology , Mice, Knockout , Mice, Inbred BALB C , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...