Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(93): eadd4818, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38427718

ABSTRACT

T follicular helper (TFH) cells are essential for effective antibody responses, but deciphering the intrinsic wiring of mouse TFH cells has long been hampered by the lack of a reliable protocol for their generation in vitro. We report that transforming growth factor-ß (TGF-ß) induces robust expression of TFH hallmark molecules CXCR5 and Bcl6 in activated mouse CD4+ T cells in vitro. TGF-ß-induced mouse CXCR5+ TFH cells are phenotypically, transcriptionally, and functionally similar to in vivo-generated TFH cells and provide critical help to B cells. The study further reveals that TGF-ß-induced CXCR5 expression is independent of Bcl6 but requires the transcription factor c-Maf. Classical TGF-ß-containing T helper 17 (TH17)-inducing conditions also yield separate CXCR5+ and IL-17A-producing cells, highlighting shared and distinct cell fate trajectories of TFH and TH17 cells. We demonstrate that excess IL-2 in high-density T cell cultures interferes with the TGF-ß-induced TFH cell program, that TFH and TH17 cells share a common developmental stage, and that c-Maf acts as a switch factor for TFH versus TH17 cell fates in TGF-ß-rich environments in vitro and in vivo.


Subject(s)
T-Lymphocytes, Helper-Inducer , Transforming Growth Factor beta , Animals , Mice , Transforming Growth Factor beta/metabolism , B-Lymphocytes , CD4-Positive T-Lymphocytes , Cell Differentiation , Proto-Oncogene Proteins c-maf/metabolism
2.
Nature ; 627(8003): 407-415, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383779

ABSTRACT

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Subject(s)
Aquaporin 4 , Autoantibodies , Autoantigens , B-Lymphocytes , Immune Tolerance , Neuromyelitis Optica , Animals , Humans , Mice , AIRE Protein , Aquaporin 4/deficiency , Aquaporin 4/genetics , Aquaporin 4/immunology , Aquaporin 4/metabolism , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/immunology , Germinal Center/cytology , Germinal Center/immunology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thyroid Epithelial Cells/immunology , Thyroid Epithelial Cells/metabolism , Transcriptome
3.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33879600

ABSTRACT

BACKGROUND: High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective. METHODS: IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1. CONTROL: Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice. RESULTS: Our therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages. CONCLUSION: IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.


Subject(s)
Cancer Vaccines/administration & dosage , Interleukin-6/metabolism , Neoplasms/drug therapy , Oligodeoxyribonucleotides/administration & dosage , Papillomavirus E7 Proteins/administration & dosage , Tumor-Associated Macrophages/drug effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Female , Injections, Subcutaneous , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Oligodeoxyribonucleotides/immunology , Papillomavirus E7 Proteins/immunology , Phenotype , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Signal Transduction , Tumor Burden/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
4.
J Clin Invest ; 130(9): 4587-4600, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32484796

ABSTRACT

Th cells integrate signals from their microenvironment to acquire distinct specialization programs for efficient clearance of diverse pathogens or for immunotolerance. Ionic signals have recently been demonstrated to affect T cell polarization and function. Sodium chloride (NaCl) was proposed to accumulate in peripheral tissues upon dietary intake and to promote autoimmunity via the Th17 cell axis. Here, we demonstrate that high-NaCl conditions induced a stable, pathogen-specific, antiinflammatory Th17 cell fate in human T cells in vitro. The p38/MAPK pathway, involving NFAT5 and SGK1, regulated FoxP3 and IL-17A expression in high-NaCl conditions. The NaCl-induced acquisition of an antiinflammatory Th17 cell fate was confirmed in vivo in an experimental autoimmune encephalomyelitis (EAE) mouse model, which demonstrated strongly reduced disease symptoms upon transfer of T cells polarized in high-NaCl conditions. However, NaCl was coopted to promote murine and human Th17 cell pathogenicity, if T cell stimulation occurred in a proinflammatory and TGF-ß-low cytokine microenvironment. Taken together, our findings reveal a context-dependent, dichotomous role for NaCl in shaping Th17 cell pathogenicity. NaCl might therefore prove beneficial for the treatment of chronic inflammatory diseases in combination with cytokine-blocking drugs.


Subject(s)
Cellular Microenvironment/drug effects , Cytokines/immunology , MAP Kinase Signaling System/drug effects , Sodium Chloride, Dietary/pharmacology , Th17 Cells/immunology , Animals , Cellular Microenvironment/immunology , Cytokines/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Transgenic , Th17 Cells/pathology
5.
J Immunol ; 204(4): 747-751, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924653

ABSTRACT

IL-6 binds to the IL-6R α-chain (IL-6Rα) and signals via the signal transducer gp130. Recently, IL-6 was found to also bind to the cell surface glycoprotein CD5, which would then engage gp130 in the absence of IL-6Rα. However, the biological relevance of this alternative pathway is under debate. In this study, we developed a mouse model, in which murine IL-6 is overexpressed in a CD11c-Cre-dependent manner. Transgenic mice developed a lethal immune dysregulation syndrome with increased numbers of Ly-6G+ neutrophils and Ly-6Chi monocytes/macrophages. IL-6 overexpression promoted activation of CD4+ T cells while suppressing CD5+ B-1a cell development. However, additional ablation of IL-6Rα protected IL-6-overexpressing mice from IL-6-triggered inflammation and fully phenocopied IL-6Rα-deficient mice without IL-6 overexpression. Mechanistically, IL-6Rα deficiency completely prevented downstream activation of STAT3 in response to IL-6. Altogether, our data clarify that IL-6Rα is the only biologically relevant receptor for IL-6 in mice.


Subject(s)
Interleukin-6/immunology , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Animals , Mice , Mice, Transgenic
6.
Nat Neurosci ; 22(10): 1731-1742, 2019 10.
Article in English | MEDLINE | ID: mdl-31501572

ABSTRACT

Mitochondria vary in morphology and function in different tissues; however, little is known about their molecular diversity among cell types. Here we engineered MitoTag mice, which express a Cre recombinase-dependent green fluorescent protein targeted to the outer mitochondrial membrane, and developed an isolation approach to profile tagged mitochondria from defined cell types. We determined the mitochondrial proteome of the three major cerebellar cell types (Purkinje cells, granule cells and astrocytes) and identified hundreds of mitochondrial proteins that are differentially regulated. Thus, we provide markers of cell-type-specific mitochondria for the healthy and diseased mouse and human central nervous systems, including in amyotrophic lateral sclerosis and Alzheimer's disease. Based on proteomic predictions, we demonstrate that astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neuronal mitochondria. We also characterize cell-type differences in mitochondrial calcium buffering via the mitochondrial calcium uniporter (Mcu) and identify regulator of microtubule dynamics protein 3 (Rmdn3) as a determinant of endoplasmic reticulum-mitochondria proximity in Purkinje cells. Our approach enables exploring mitochondrial diversity in many in vivo contexts.


Subject(s)
Brain/cytology , Mitochondria/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Calcium Signaling/genetics , Calcium Signaling/physiology , Cells, Cultured , Cerebellum/cytology , Fatty Acids/metabolism , Humans , Mice , Mice, Transgenic , Mitochondrial Membranes/metabolism , Proteomics , Purkinje Cells/metabolism
7.
Nat Rev Drug Discov ; 17(6): 395-412, 2018 06.
Article in English | MEDLINE | ID: mdl-29725131

ABSTRACT

Interleukin-6 (IL-6) is a pivotal cytokine with a diverse repertoire of physiological functions that include regulation of immune cell proliferation and differentiation. Dysregulation of IL-6 signalling is associated with inflammatory and lymphoproliferative disorders such as rheumatoid arthritis and Castleman disease, and several classes of therapeutics have been developed that target components of the IL-6 signalling pathway. So far, monoclonal antibodies against IL-6 or IL-6 receptor (IL-6R) and Janus kinases (JAK) inhibitors have been successfully developed for the treatment of autoimmune diseases such as rheumatoid arthritis. However, clinical trials of agents targeting IL-6 signalling have also raised questions about the diseases and patient populations for which such agents have an appropriate benefit-risk profile. Knowledge from clinical trials and advances in our understanding of the complexities of IL-6 signalling, including the potential to target an IL-6 trans-signalling pathway, are now indicating novel opportunities for therapeutic intervention. In this Review, we overview the roles of IL-6 in health and disease and analyse progress with several approaches of inhibiting IL-6-signalling, with the aim of illuminating when and how to apply IL-6 blockade.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Interleukin-6/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Clinical Trials as Topic , Humans , Receptors, Interleukin-6/metabolism
9.
Nat Immunol ; 18(1): 74-85, 2017 01.
Article in English | MEDLINE | ID: mdl-27893700

ABSTRACT

The cellular sources of interleukin 6 (IL-6) that are relevant for differentiation of the TH17 subset of helper T cells remain unclear. Here we used a novel strategy for the conditional deletion of distinct IL-6-producing cell types to show that dendritic cells (DCs) positive for the signaling regulator Sirpα were essential for the generation of pathogenic TH17 cells. Using their IL-6 receptor α-chain (IL-6Rα), Sirpα+ DCs trans-presented IL-6 to T cells during the process of cognate interaction. While ambient IL-6 was sufficient to suppress the induction of expression of the transcription factor Foxp3 in T cells, trans-presentation of IL-6 by DC-bound IL-6Rα (called 'IL-6 cluster signaling' here) was needed to prevent premature induction of interferon-γ (IFN-γ) expression in T cells and to generate pathogenic TH17 cells in vivo. Our findings should guide therapeutic approaches for the treatment of TH17-cell-mediated autoimmune diseases.


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-6 Receptor alpha Subunit/genetics , Interleukin-6/metabolism , Th17 Cells/immunology , Animals , Autoimmunity , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
10.
J Autoimmun ; 56: 34-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25282335

ABSTRACT

Optic neuritis is a common inflammatory manifestation of multiple sclerosis (MS). In experimental autoimmune encephalomyelitis (EAE), the optic nerve is affected as well. Here, we investigated whether autoimmune inflammation in the optic nerve is distinct from inflammation in other parts of the central nervous system (CNS). In our study, inflammatory infiltrates in the optic nerve and the brain were characterized by a high fraction of Ly6G(+) granulocytes whereas in the spinal cord, macrophage infiltrates were predominant. At the peak of disease, IL-17 mRNA abundance was highest in the optic nerve as compared with other parts of the CNS. The ratio of IL-17 vs IFN-γ producing CD4(+) T cells was higher in the optic nerve and brain than in the spinal cord and more effector CD4(+) T cells were committed to the Th17 transcriptional program in the optic nerve than in the spinal cord. IL-17 producing γδ T cells but rather not Ly6G(+) granulocytes themselves contributed to IL-17 production. Optical coherence tomography (OCT) studies on murine eyes revealed a decline in thickness of the retinal nerve fiber layer (RNFL) and the common layer of ganglion cells and inner plexiform layer (GCL+) after the recovery from motor symptoms indicating that autoimmune inflammation induced a significant atrophy of optic nerve fibers during EAE. Neutralization of IL-17 by treatment with anti-IL-17 antibodies reduced but did not abrogate motor symptoms of EAE. However, RNFL and GCL+ atrophy were completely prevented by blocking IL-17. Thus, the optic nerve compartment is particularly prone to support IL-17 mediated inflammatory responses during CNS autoimmunity and structural integrity of the retina can be preserved by neutralizing IL-17.


Subject(s)
Antibodies, Neutralizing/pharmacology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-17/antagonists & inhibitors , Optic Nerve/immunology , Optic Nerve/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/administration & dosage , Atrophy , Autoimmunity , Brain/immunology , Brain/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Leukocyte Common Antigens/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mice , Optic Neuritis/diagnosis , Optic Neuritis/drug therapy , Optic Neuritis/immunology , Optic Neuritis/pathology , Rats , Spinal Cord/immunology , Spinal Cord/pathology , Tomography, Optical Coherence
11.
Immunity ; 41(5): 722-36, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25464853

ABSTRACT

Regulatory T (Treg) cells maintain immune homeostasis and prevent inflammatory and autoimmune responses. During development, thymocytes bearing a moderately self-reactive T cell receptor (TCR) can be selected to become Treg cells. Several observations suggest that also in the periphery mature Treg cells continuously receive self-reactive TCR signals. However, the importance of this inherent autoreactivity for Treg cell biology remains poorly defined. To address this open question, we genetically ablated the TCR of mature Treg cells in vivo. These experiments revealed that TCR-induced Treg lineage-defining Foxp3 expression and gene hypomethylation were uncoupled from TCR input in mature Treg cells. However, Treg cell homeostasis, cell-type-specific gene expression and suppressive function critically depend on continuous triggering of their TCR.


Subject(s)
Autoimmunity/immunology , Forkhead Transcription Factors/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , DNA Methylation/immunology , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Inflammation/immunology , Interferon Regulatory Factors/biosynthesis , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism , Thymocytes/cytology
12.
PLoS One ; 9(10): e109199, 2014.
Article in English | MEDLINE | ID: mdl-25296340

ABSTRACT

Transgenic expression of B- and T-cell receptors (BCRs and TCRs, respectively) has been a standard tool to study lymphocyte development and function in vivo. The generation of transgenic mice is time-consuming and, therefore, a faster method to study the biology of defined lymphocyte receptors in vivo would be highly welcome. Using 2A peptide-linked multicistronic retroviral vectors to transduce stem cells, TCRs can be expressed rapidly in mice of any background. We aimed at adopting this retrogenic technology to the in vivo expression of BCRs. Using a well characterised BCR specific for hen egg lysozyme (HEL), we achieved surface expression of the retrogenically encoded BCR in a Rag-deficient pro B-cell line in vitro. In vivo, retrogenic BCRs were detectable only intracellularly but not on the surface of B cells from wild type or Rag2-deficient mice. This data, together with the fact that no BCR retrogenic mouse model has been published in the 7 years since the method was originally published for TCRs, strongly suggests that achieving BCR-expression in vivo with retrogenic technology is highly challenging if not impossible.


Subject(s)
Receptors, Antigen, B-Cell/metabolism , Animals , Cell Line , Cells, Cultured , Mice , Mice, Transgenic , Muramidase/genetics , Muramidase/metabolism , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Transfection
13.
Nat Commun ; 5: 3770, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24796719

ABSTRACT

Central nervous system (CNS) autoimmunity is regulated by the balance of pro-inflammatory cytokines and IL-10. Here we identify the transcriptional regulator Blimp1 as crucial to induce IL-10 in inflammatory T helper cells. Pre-committed Th17 cells respond to IL-27 and IL-12 by upregulating Blimp1 and adopt a Tr-1-like phenotype characterized by IL-10 and IFN-γ production. Accordingly, Blimp1-deficient effector T cells fail to produce IL-10, and deficiency in Tr-1 cell function leads to uncontrolled Th17 cell-driven CNS pathology without the need to stabilize the Th17 phenotype with IL-23. IL-23 counteracts IL-27 and IL-12-mediated effects on Tr-1-development reinforcing the pro-inflammatory phenotype of Th17 cells. Thus, the balance of IL-23 vs IL-12/IL-27 signals into CD4(+) effector T cells determines whether tissue inflammation is perpetuated or resolves.


Subject(s)
Inflammation/prevention & control , Interleukin-12/metabolism , Interleukin-23/metabolism , Interleukins/metabolism , Repressor Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , Humans , Interferon-gamma/metabolism , Positive Regulatory Domain I-Binding Factor 1
14.
Acta Neuropathol Commun ; 2: 27, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24606807

ABSTRACT

INTRODUCTION: Natalizumab blocks α4-integrins and is a prototypic agent for a series of anti-inflammatory drugs that impair trafficking of immune cells into the CNS. However, modulation of the access of immune cells to the CNS is associated with impaired immune surveillance and detrimental viral infections of the CNS. Here, we explored the potency of cellular immune responses within the CNS to protect against viral encephalitis in mice with T cell conditional disruption of VLA-4 integrin (α4ß1) expression. RESULTS: While VLA-4 expression in virus specific Th1 cells is non-redundant for their ability to access the CNS, α4-integrin deficient Th17 cells enter the CNS compartment and generate an inflammatory milieu upon intrathecal vaccinia virus (VV) infection. However, in contrast to Th1 cells that can adopt direct cytotoxic properties, Th17 cells fail to clear the virus due to insufficient Eomes induced perforin-1 expression. CONCLUSION: The quality of the intrathecal cellular antiviral response under conditions of impaired VLA-4 function jeopardizes host protection. Our functional in vivo data extend our mechanistic understanding of anti-viral immunity in the CNS and help to estimate the risk potential of upcoming therapeutic agents that target the trafficking of immune cells into distinct anatomical compartments.


Subject(s)
Adoptive Transfer/methods , Integrin alpha4beta1/immunology , Meningoencephalitis/immunology , Meningoencephalitis/prevention & control , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies/therapeutic use , CD4 Antigens/genetics , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Freund's Adjuvant/toxicity , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/immunology , Homeodomain Proteins/genetics , Integrin alpha4beta1/genetics , Meningoencephalitis/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Perforin/genetics , Perforin/metabolism , Pore Forming Cytotoxic Proteins/genetics , Retroviridae Infections/complications
15.
J Clin Invest ; 123(1): 247-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23221338

ABSTRACT

IL-17-producing CD8+ T (Tc17) cells are detectible in multiple sclerosis (MS) lesions; however, their contribution to the disease is unknown. To identify functions of Tc17 cells, we induced EAE, a murine model of MS, in mice lacking IFN regulatory factor 4 (IRF4). IRF4-deficient mice failed to generate Tc17 and Th17 cells and were resistant to EAE. After adoptive transfer of WT CD8+ T cells and subsequent immunization for EAE induction in these mice, the CD8+ T cells developed a Tc17 phenotype in the periphery but could not infiltrate the CNS. Similarly, transfer of small numbers of WT CD4+ T cells alone did not evoke EAE, but when transferred together with CD8+ T cells, IL-17-producing CD4+ (Th17) T cells accumulated in the CNS and mice developed severe disease. Th17 accumulation and development of EAE required IL-17A production by CD8+ T cells, suggesting that Tc17 cells are required to promote CD4+ T cell-mediated induction of EAE. Accordingly, patients with early-stage MS harbored a greater number of Tc17 cells in the cerebrospinal fluid than in peripheral blood. Our results reveal that Tc17 cells contribute to the initiation of CNS autoimmunity in mice and humans by supporting Th17 cell pathogenicity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Knockout , Th17 Cells/metabolism , Th17 Cells/pathology , Th17 Cells/transplantation
16.
J Immunol ; 187(12): 6346-56, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079988

ABSTRACT

Plasmacytoid dendritic cells (PDCs) have been shown to present Ags and to contribute to peripheral immune tolerance and to Ag-specific adaptive immunity. However, modulation of adaptive immune responses by selective Ag targeting to PDCs with the aim of preventing autoimmunity has not been investigated. In the current study, we demonstrate that in vivo Ag delivery to murine PDCs via the specifically expressed surface molecule sialic acid binding Ig-like lectin H (Siglec-H) inhibits Th cell and Ab responses in the presence of strong immune stimulation in an Ag-specific manner. Correlating with sustained low-level MHC class II-restricted Ag presentation on PDCs, Siglec-H-mediated Ag delivery induced a hyporesponsive state in CD4(+) T cells leading to reduced expansion and Th1/Th17 cell polarization without conversion to Foxp3(+) regulatory T cells or deviation to Th2 or Tr1 cells. Siglec-H-mediated delivery of a T cell epitope derived from the autoantigen myelin oligodendrocyte glycoprotein to PDCs effectively delayed onset and reduced disease severity in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis by interfering with the priming phase without promoting the generation or expansion of myelin oligodendrocyte glycoprotein-specific Foxp3(+) regulatory T cells. We conclude that Ag delivery to PDCs can be harnessed to inhibit Ag-specific immune responses and prevent Th cell-dependent autoimmunity.


Subject(s)
Antigen Presentation/immunology , Autoantigens/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins/physiology , Receptors, Cell Surface/physiology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Myelin Proteins/immunology , Myelin Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein , T-Lymphocytes, Helper-Inducer/metabolism
17.
J Exp Med ; 208(12): 2465-76, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-22025301

ABSTRACT

The integrin α4ß1 (VLA-4) is used by encephalitogenic T cells to enter the central nervous system (CNS). However, both Th1 and Th17 cells are capable of inducing experimental autoimmune encephalomyelitis (EAE), and the molecular cues mediating the infiltration of Th1 versus Th17 cells into the CNS have not yet been defined. We investigated how blocking of α4 integrins affected trafficking of Th1 and Th17 cells into the CNS during EAE. Although antibody-mediated inhibition of α4 integrins prevented EAE when MOG(35-55)-specific Th1 cells were adoptively transferred, Th17 cells entered the brain, but not the spinal cord parenchyma, irrespective of α4 blockade. Accordingly, T cell-conditional α4-deficient mice were not resistant to actively induced EAE but showed an ataxic syndrome with predominantly supraspinal infiltrates of IL-23R(+)CCR6(+)CD4(+) T cells. The entry of α4-deficient Th17 cells into the CNS was abolished by blockade of LFA-1 (αLß2 integrin). Thus, Th1 cells preferentially infiltrate the spinal cord via an α4 integrin-mediated mechanism, whereas the entry of Th17 cells into the brain parenchyma occurs in the absence of α4 integrins but is dependent on the expression of αLß2. These observations have implications for the understanding of lesion localization, immunosurveillance, and drug design in multiple sclerosis.


Subject(s)
Cell Movement/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Adoptive Transfer , Analysis of Variance , Animals , Cell Differentiation/immunology , Flow Cytometry , Histological Techniques , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Th1 Cells/immunology
18.
Eur J Immunol ; 41(3): 833-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21287545

ABSTRACT

PI3Ks control signal transduction triggered by growth factors and G-protein-coupled receptors and regulate an array of biological processes, including cellular proliferation, differentiation, survival and migration. Herein, we investigated the role of PI3Kγ in the pathogenesis of EAE. We show that, in the absence of PI3Kγ expression, clinical signs of EAE were delayed and mitigated. PI3Kγ-deficient myelin oligodendrocyte glycoprotein (MOG)(35-55) -specific CD4(+) T cells appeared later in the secondary lymphoid organs and in the CNS than their WT counterparts. Transfer of WT CD4(+) cells into PI3Kγ(-/-) mice prior to MOG(35-55) immunisation restored EAE severity to WT levels, supporting the relevance of PI3Kγ expression in Th cells for the pathogenesis of EAE; however, PI3Kγ was dispensable for Th1 and Th17 differentiation, thus excluding an altered expression of these pathogenetically relevant cytokines as the cause for ameliorated EAE in PI3Kγ(-/-) mice. These findings demonstrate that PI3Kγ contributes to the development of autoimmune CNS inflammation.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/deficiency , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Adoptive Transfer , Animals , Cell Differentiation , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Glycoproteins/immunology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Phosphoinositide-3 Kinase Inhibitors , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Time Factors
19.
Immunity ; 33(3): 351-63, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20832339

ABSTRACT

Mice that lack interleukin-23 (IL-23) are resistant to T cell-mediated autoimmunity. Although IL-23 is a maturation factor for T helper 17 (Th17) cells, a subset of γδ T cells expresses the IL-23 receptor (IL-23R) constitutively. Using IL-23R reporter mice, we showed that γδ T cells were the first cells to respond to IL-23 during experimental autoimmune encephalomyelitis (EAE). Although γδ T cells produced Th17 cell-associated cytokines in response to IL-23, their major function was to prevent the development of regulatory T (Treg) cell responses. IL-23-activated γδ T cells rendered αß effector T cells refractory to the suppressive activity of Treg cells and also prevented the conversion of conventional T cells into Foxp3(+) Treg cells in vivo. Thus, IL-23, which by itself has no direct effect on Treg cells, is able to disarm Treg cell responses and promote antigen-specific effector T cell responses via activating γδ T cells.


Subject(s)
Autoimmunity , Interleukin-23/physiology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Encephalomyelitis, Autoimmune, Experimental/etiology , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Receptors, Antigen, T-Cell/physiology , Receptors, Interleukin/physiology , Interleukin-22
20.
Blood ; 115(19): 3899-906, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20200353

ABSTRACT

Members of the Toll/interleukin-1 receptor (TIR) family are of importance for host defense and inflammation. Here we report that the TIR-family member interleukin-33R (IL-33R) cross-activates the receptor tyrosine kinase c-Kit in human and murine mast cells. The IL-33R-induced activation of signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase 1/2 (Erk1/2), protein kinase B (PKB), and Jun NH(2)-terminal kinase 1 (JNK1) depends on c-Kit and is required to elicit optimal effector functions. Costimulation with the c-Kit ligand stem cell factor (SCF) is necessary for IL-33-induced cytokine production in primary mast cells. The structural basis for this cross-activation is the complex formation between c-Kit, IL-33R, and IL-1R accessory protein (IL-1RAcP). We found that c-Kit and IL-1RAcP interact constitutively and that IL-33R joins this complex upon ligand binding. Our findings support a model in which signals from seemingly disparate receptors are integrated for full cellular responses.


Subject(s)
Interleukin-1 Receptor Accessory Protein/metabolism , Interleukins/metabolism , Mast Cells/metabolism , Proto-Oncogene Proteins c-kit/physiology , Receptors, Interleukin/metabolism , Signal Transduction , Animals , Blotting, Western , Bone Marrow/metabolism , Cells, Cultured , Cytokines/metabolism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Integrases/metabolism , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Stem Cell Factor/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...