Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Cell ; 163(1): 160-73, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406376

ABSTRACT

Focal adhesion kinase (FAK) promotes anti-tumor immune evasion. Specifically, the kinase activity of nuclear-targeted FAK in squamous cell carcinoma (SCC) cells drives exhaustion of CD8(+) T cells and recruitment of regulatory T cells (Tregs) in the tumor microenvironment by regulating chemokine/cytokine and ligand-receptor networks, including via transcription of Ccl5, which is crucial. These changes inhibit antigen-primed cytotoxic CD8(+) T cell activity, permitting growth of FAK-expressing tumors. Mechanistically, nuclear FAK is associated with chromatin and exists in complex with transcription factors and their upstream regulators that control Ccl5 expression. Furthermore, FAK's immuno-modulatory nuclear activities may be specific to cancerous squamous epithelial cells, as normal keratinocytes do not have nuclear FAK. Finally, we show that a small-molecule FAK kinase inhibitor, VS-4718, which is currently in clinical development, also drives depletion of Tregs and promotes a CD8(+) T cell-mediated anti-tumor response. Therefore, FAK inhibitors may trigger immune-mediated tumor regression, providing previously unrecognized therapeutic opportunities.


Subject(s)
Carcinoma, Squamous Cell/immunology , Chemokine CCL5/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape , Aminopyridines/administration & dosage , Animals , Carcinoma, Squamous Cell/metabolism , Chemokine CCL5/immunology , Disease Models, Animal , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Keratinocytes/metabolism , Mice , Mice, Nude , Skin Neoplasms/metabolism , Transcription, Genetic
2.
Immunity ; 44(5): 1114-26, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27192577

ABSTRACT

Regulatory T (Treg) cells expressing Foxp3 transcripton factor are essential for immune homeostasis. They arise in the thymus as a separate lineage from conventional CD4(+)Foxp3(-) T (Tconv) cells. Here, we show that the thymic development of Treg cells depends on the expression of their endogenous cognate self-antigen. The formation of these cells was impaired in mice lacking this self-antigen, while Tconv cell development was not negatively affected. Thymus-derived Treg cells were selected by self-antigens in a specific manner, while autoreactive Tconv cells were produced through degenerate recognition of distinct antigens. These distinct modes of development were associated with the expression of T cell receptor of higher functional avidity for self-antigen by Treg cells than Tconv cells, a difference subsequently essential for the control of autoimmunity. Our study documents how self-antigens define the repertoire of thymus-derived Treg cells to subsequently endow this cell type with the capacity to undermine autoimmune attack.


Subject(s)
CTLA-4 Antigen/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/metabolism , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/immunology , Animals , Autoantigens/immunology , CTLA-4 Antigen/genetics , Cells, Cultured , Clonal Selection, Antigen-Mediated , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/genetics
3.
Eur J Immunol ; 52(8): 1335-1349, 2022 08.
Article in English | MEDLINE | ID: mdl-35579560

ABSTRACT

CD4+ FOXP3+ Tregs are currently explored to develop cell therapies against immune-mediated disorders, with an increasing focus on antigen receptor-engineered Tregs. Deciphering their mode of action is necessary to identify the strengths and limits of this approach. Here, we addressed this issue in an autoimmune disease of the CNS, EAE. Following disease induction, autoreactive Tregs upregulated LAG-3 and CTLA-4 in LNs, while IL-10 and amphiregulin (AREG) were increased in CNS Tregs. Using genetic approaches, we demonstrated that IL-10, CTLA-4, and LAG-3 were nonredundantly required for the protective function of antigen receptor-engineered Tregs against EAE in cell therapy whereas AREG was dispensable. Treg-derived IL-10 and CTLA-4 were both required to suppress acute autoreactive CD4+ T-cell activation, which correlated with disease control. These molecules also affected the accumulation in the recipients of engineered Tregs themselves, underlying complex roles for these molecules. Noteworthy, despite the persistence of the transferred Tregs and their protective effect, autoreactive T cells eventually accumulated in the spleen of treated mice. In conclusion, this study highlights the remarkable power of antigen receptor-engineered Tregs to appropriately provide multiple suppressive factors nonredundantly necessary to prevent autoimmune attacks.


Subject(s)
Autoimmunity , Immune System Diseases , Animals , CTLA-4 Antigen , Cell- and Tissue-Based Therapy , Forkhead Transcription Factors/genetics , Interleukin-10 , Mice , Receptors, Antigen , T-Lymphocytes, Regulatory
4.
Neuropathol Appl Neurobiol ; 49(1): e12851, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36181265

ABSTRACT

AIMS: Axonal injury in multiple sclerosis (MS) and experimental models is most frequently detected in acutely demyelinating lesions. We recently reported a compensatory neuronal response, where mitochondria move to the acutely demyelinated axon and increase the mitochondrial content following lysolecithin-induced demyelination. We termed this homeostatic phenomenon, which is also evident in MS, the axonal response of mitochondria to demyelination (ARMD). The aim of this study is to determine whether ARMD is consistently evident in experimental demyelination and how its perturbation relates to axonal injury. METHODS: In the present study, we assessed axonal mitochondrial content as well as axonal mitochondrial respiratory chain complex IV activity (cytochrome c oxidase or COX) of axons and related these to axonal injury in nine different experimental disease models. We used immunofluorescent histochemistry as well as sequential COX histochemistry followed by immunofluorescent labelling of mitochondria and axons. RESULTS: We found ARMD a consistent and robust phenomenon in all experimental disease models. The increase in mitochondrial content within demyelinated axons, however, was not always accompanied by a proportionate increase in complex IV activity, particularly in highly inflammatory models such as experimental autoimmune encephalomyelitis (EAE). Axonal complex IV activity inversely correlated with the extent of axonal injury in experimental disease models. CONCLUSIONS: Our findings indicate that ARMD is a consistent and prominent feature and emphasise the importance of complex IV activity in the context of ARMD, especially in autoimmune inflammatory demyelination, paving the way for the development of novel neuroprotective therapies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Multiple Sclerosis/pathology , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Neurons/pathology , Mitochondria/pathology
5.
Eur J Immunol ; 49(1): 112-120, 2019 01.
Article in English | MEDLINE | ID: mdl-30485411

ABSTRACT

T cell adaptation is an important peripheral tolerogenic process which ensures that the T cell population can respond effectively to pathogens but remains tolerant to self-antigens. We probed the mechanisms of T cell adaptation using an experimental autoimmune encephalomyelitis (EAE) model in which the fate of autopathogenic T cells could be followed. We demonstrated that immunisation with a high dose of myelin basic protein (MBP) peptide and complete Freund's adjuvant failed to effectively initiate EAE, in contrast to low dose MBP peptide immunisation which readily induced disease. The proportion of autopathogenic CD4+ T cells in the central nervous system (CNS) of mice immunised with a high dose of MBP peptide was not significantly different to mice immunised with a low dose. However, autopathogenic T cells in mice immunised with high dose MBP peptide had an unresponsive phenotype in ex vivo recall assays. Importantly, whilst expression of PD-1 was increased on adapted CD4+ T cells within the CNS, loss of PD-1 function did not prevent the development of the unresponsive state. The lack of a role for PD-1 in the acquisition of the adapted state stands in striking contrast to the reported functional importance of PD-1 in T cell unresponsiveness in other disease models.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Autoantigens/immunology , Cells, Cultured , Clonal Anergy , Disease Models, Animal , Humans , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/immunology , Peptide Fragments/immunology , Up-Regulation
6.
Acta Neuropathol ; 140(2): 143-167, 2020 08.
Article in English | MEDLINE | ID: mdl-32572598

ABSTRACT

Axonal loss is the key pathological substrate of neurological disability in demyelinating disorders, including multiple sclerosis (MS). However, the consequences of demyelination on neuronal and axonal biology are poorly understood. The abundance of mitochondria in demyelinated axons in MS raises the possibility that increased mitochondrial content serves as a compensatory response to demyelination. Here, we show that upon demyelination mitochondria move from the neuronal cell body to the demyelinated axon, increasing axonal mitochondrial content, which we term the axonal response of mitochondria to demyelination (ARMD). However, following demyelination axons degenerate before the homeostatic ARMD reaches its peak. Enhancement of ARMD, by targeting mitochondrial biogenesis and mitochondrial transport from the cell body to axon, protects acutely demyelinated axons from degeneration. To determine the relevance of ARMD to disease state, we examined MS autopsy tissue and found a positive correlation between mitochondrial content in demyelinated dorsal column axons and cytochrome c oxidase (complex IV) deficiency in dorsal root ganglia (DRG) neuronal cell bodies. We experimentally demyelinated DRG neuron-specific complex IV deficient mice, as established disease models do not recapitulate complex IV deficiency in neurons, and found that these mice are able to demonstrate ARMD, despite the mitochondrial perturbation. Enhancement of mitochondrial dynamics in complex IV deficient neurons protects the axon upon demyelination. Consequently, increased mobilisation of mitochondria from the neuronal cell body to the axon is a novel neuroprotective strategy for the vulnerable, acutely demyelinated axon. We propose that promoting ARMD is likely to be a crucial preceding step for implementing potential regenerative strategies for demyelinating disorders.


Subject(s)
Demyelinating Diseases/pathology , Mitochondria/pathology , Multiple Sclerosis/pathology , Nerve Degeneration/pathology , Neuroprotection/physiology , Animals , Axons/pathology , Humans , Mice , Organelle Biogenesis
7.
J Immunol ; 200(3): 1169-1187, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29263216

ABSTRACT

The disposal of apoptotic bodies by professional phagocytes is crucial to effective inflammation resolution. Our ability to improve the disposal of apoptotic bodies by professional phagocytes is impaired by a limited understanding of the molecular mechanisms that regulate the engulfment and digestion of the efferocytic cargo. Macrophages are professional phagocytes necessary for liver inflammation, fibrosis, and resolution, switching their phenotype from proinflammatory to restorative. Using sterile liver injury models, we show that the STAT3-IL-10-IL-6 axis is a positive regulator of macrophage efferocytosis, survival, and phenotypic conversion, directly linking debris engulfment to tissue repair.


Subject(s)
Interleukin-10/metabolism , Interleukin-6/metabolism , Liver Cirrhosis/pathology , Liver/injuries , Macrophages/immunology , Phagocytosis/immunology , STAT3 Transcription Factor/metabolism , Adoptive Transfer , Animals , Apoptosis/immunology , Humans , Liver/pathology , Macrophages/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/immunology , Regeneration/physiology , Zebrafish/embryology
8.
Nature ; 507(7492): 366-370, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24572363

ABSTRACT

B lymphocytes have critical roles as positive and negative regulators of immunity. Their inhibitory function has been associated primarily with interleukin 10 (IL-10) because B-cell-derived IL-10 can protect against autoimmune disease and increase susceptibility to pathogens. Here we identify IL-35-producing B cells as key players in the negative regulation of immunity. Mice in which only B cells did not express IL-35 lost their ability to recover from the T-cell-mediated demyelinating autoimmune disease experimental autoimmune encephalomyelitis (EAE). In contrast, these mice displayed a markedly improved resistance to infection with the intracellular bacterial pathogen Salmonella enterica serovar Typhimurium as shown by their superior containment of the bacterial growth and their prolonged survival after primary infection, and upon secondary challenge, compared to control mice. The increased immunity found in mice lacking IL-35 production by B cells was associated with a higher activation of macrophages and inflammatory T cells, as well as an increased function of B cells as antigen-presenting cells (APCs). During Salmonella infection, IL-35- and IL-10-producing B cells corresponded to two largely distinct sets of surface-IgM(+)CD138(hi)TACI(+)CXCR4(+)CD1d(int)Tim1(int) plasma cells expressing the transcription factor Blimp1 (also known as Prdm1). During EAE, CD138(+) plasma cells were also the main source of B-cell-derived IL-35 and IL-10. Collectively, our data show the importance of IL-35-producing B cells in regulation of immunity and highlight IL-35 production by B cells as a potential therapeutic target for autoimmune and infectious diseases. This study reveals the central role of activated B cells, particularly plasma cells, and their production of cytokines in the regulation of immune responses in health and disease.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity/immunology , Interleukins/metabolism , Salmonella Infections/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD40 Antigens/immunology , Female , Humans , Interleukin-10/metabolism , Interleukins/immunology , Lymphocyte Activation , Macrophages/cytology , Macrophages/immunology , Male , Mice , Plasma Cells/immunology , Plasma Cells/metabolism , Salmonella Infections/microbiology , T-Lymphocytes/immunology , Toll-Like Receptor 4/immunology
9.
J Allergy Clin Immunol ; 141(1): 152-162, 2018 01.
Article in English | MEDLINE | ID: mdl-28583370

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are both forms of eczema and are common inflammatory skin diseases with a central role of T cell-derived IL-22 in their pathogenesis. Although prostaglandin (PG) E2 is known to promote inflammation, little is known about its role in processes related to AD and ACD development, including IL-22 upregulation. OBJECTIVES: We sought to investigate whether PGE2 has a role in IL-22 induction and development of ACD, which has increased prevalence in patients with AD. METHODS: T-cell cultures and in vivo sensitization of mice with haptens were used to assess the role of PGE2 in IL-22 production. The involvement of PGE2 receptors and their downstream signals was also examined. The effects of PGE2 were evaluated by using the oxazolone-induced ACD mouse model. The relationship of PGE2 and IL-22 signaling pathways in skin inflammation were also investigated by using genomic profiling in human lesional AD skin. RESULTS: PGE2 induces IL-22 from T cells through its receptors, E prostanoid receptor (EP) 2 and EP4, and involves cyclic AMP signaling. Selective deletion of EP4 in T cells prevents hapten-induced IL-22 production in vivo, and limits atopic-like skin inflammation in the oxazolone-induced ACD model. Moreover, both PGE2 and IL-22 pathway genes were coordinately upregulated in human AD lesional skin but were at less than significant detection levels after corticosteroid or UVB treatments. CONCLUSIONS: Our results define a crucial role for PGE2 in promoting ACD by facilitating IL-22 production from T cells.


Subject(s)
Dermatitis, Allergic Contact/immunology , Dinoprostone/immunology , Interleukins/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/pathology , Dinoprostone/genetics , Humans , Interleukins/genetics , Mice , Mice, Knockout , Skin/pathology , T-Lymphocytes/pathology , Interleukin-22
10.
Thorax ; 73(11): 1081-1084, 2018 11.
Article in English | MEDLINE | ID: mdl-29574419

ABSTRACT

Acute lung injury is a neutrophil-dominant, life-threatening disease without effective therapies and better understanding of the pathophysiological mechanisms involved is an urgent need. Here we show that interleukin (IL)-22 is produced from innate lymphoid cells (ILC) and is responsible for suppression of experimental lung neutrophilic inflammation. Blocking prostaglandin E2 (PGE2) synthesis reduces lung ILCs and IL-22 production, resulting in exacerbation of lung neutrophilic inflammation. In contrast, activation of the PGE2 receptor EP4 prevents acute lung inflammation. We thus demonstrate a mechanism for production of innate IL-22 in the lung during acute injury, highlighting potential therapeutic strategies for control of lung neutrophilic inflammation by targeting the PGE2/ILC/IL-22 axis.


Subject(s)
Dinoprostone/pharmacology , Immunity, Innate/drug effects , Interleukins/biosynthesis , Lymphocytes/metabolism , Pneumonia/prevention & control , Animals , Disease Models, Animal , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Interleukin-22
11.
Immunol Cell Biol ; 96(10): 1049-1059, 2018 11.
Article in English | MEDLINE | ID: mdl-29758102

ABSTRACT

Inflammatory bowel disease (IBD) is a condition of chronic inflammatory intestinal disorder with increasing prevalence but limited effective therapies. The purine metabolic pathway is involved in various inflammatory processes including IBD. However, the mechanisms through which purine metabolism modulates IBD remain to be established. Here, we found that mucosal expression of genes involved in the purine metabolic pathway is altered in patients with active ulcerative colitis (UC), which is associated with elevated gene expression signatures of the group 3 innate lymphoid cell (ILC3)-interleukin (IL)-22 pathway. In mice, blockade of ectonucleotidases (NTPDases), critical enzymes for purine metabolism by hydrolysis of extracellular adenosine 5'-triphosphate (eATP) into adenosine, exacerbates dextran-sulfate sodium-induced intestinal injury. This exacerbation of colitis is associated with reduction of colonic IL-22-producing ILC3s, which afford essential protection against intestinal inflammation, and is rescued by exogenous IL-22. Mechanistically, activation of ILC3s for IL-22 production is reciprocally mediated by eATP and adenosine. These findings reveal that the NTPDase-mediated balance between eATP and adenosine regulates ILC3 cell function to provide protection against intestinal injury and suggest potential therapeutic strategies for treating IBD by targeting the purine-ILC3 axis.


Subject(s)
Colitis/etiology , Colitis/metabolism , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Purines/metabolism , Animals , Biomarkers , Colitis/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Transcriptome
12.
Proc Natl Acad Sci U S A ; 111(8): E784-93, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516158

ABSTRACT

Peptide immunotherapy (PIT) offers realistic prospects for the treatment of allergic diseases, including allergic asthma. Much is understood of the behavior of naive T cells in response to PIT. However, treatment of patients with ongoing allergic disease requires detailed understanding of the responses of allergen-experienced T cells. CD62L expression by allergen-experienced T cells corresponds to effector/effector memory (CD62L(lo)) and central memory (CD62L(hi)) subsets, which vary with allergen exposure (e.g., during, or out with, pollen season). The efficacy of PIT on different T helper 2 (Th2) cell memory populations is unknown. We developed a murine model of PIT in allergic airway inflammation (AAI) driven by adoptively transferred, traceable ovalbumin-experienced Th2 cells. PIT effectively suppressed AAI driven by unfractionated Th2 cells. Selective transfer of CD62L(hi) and CD62L(lo) Th2 cells revealed that these two populations behaved differently from one another and from previously characterized (early deletional) responses of naive CD4(+) T cells to PIT. Most notably, allergen-reactive CD62L(lo) Th2 cells were long-lived within the lung after PIT, before allergen challenge, in contrast to CD62L(hi) Th2 cells. Despite this, PIT was most potent against CD62L(lo) Th2 cells in protecting from AAI, impairing their ability to produce Th2 cytokines, whereas this capacity was heightened in PIT-treated CD62L(hi) Th2 cells. We conclude that Th2 cells do not undergo an early deletional form of tolerance after PIT. Moreover, memory Th2 subsets respond differently to PIT. These findings have implications for the clinical translation of PIT in different allergic scenarios.


Subject(s)
Hypersensitivity/drug therapy , Hypersensitivity/immunology , Immunologic Memory/immunology , Immunotherapy/methods , Ovalbumin/immunology , Peptide Fragments/immunology , Th2 Cells/immunology , Animals , Bronchoalveolar Lavage , Flow Cytometry , Hypersensitivity/pathology , L-Selectin/immunology , Lung/pathology , Mice , Mice, Transgenic , Ovalbumin/therapeutic use , Peptide Fragments/therapeutic use , Th2 Cells/cytology
13.
Immunology ; 146(2): 194-205, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26190495

ABSTRACT

Foxp3(+) regulatory T (Treg) cells prevent the development of autoimmunity and immunopathology, as well as maintaining homeostasis and tolerance to commensal microorganisms. The suppressive activity of Treg cells is their defining characteristic, generating great interest in their therapeutic potential. However, suppressive and effector functions are not entirely exclusive. Considerable evidence points to the ability of supposedly anti-inflammatory Foxp3-expressing Treg cells to also express transcription factors that have been characterized as cardinal drivers of T effector cell function. We will consider the mounting evidence that Treg cells can function in non-suppressive capacities and review the impetus for this functional change, its relevance to developing immune and autoimmune responses and its significance to the development of Treg-based therapies.


Subject(s)
Cytokines/immunology , Forkhead Transcription Factors/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Receptors, Cytokine/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/immunology , Animals , Cytokines/genetics , Cytokines/metabolism , Forkhead Transcription Factors/metabolism , Genetic Markers , Genetic Predisposition to Disease , Humans , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Inflammation/therapy , Inflammation Mediators/metabolism , Phenotype , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Eur J Immunol ; 44(11): 3342-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25168419

ABSTRACT

Interest in manipulating the immunosuppressive powers of Foxp3-expressing T regulatory cells as an immunotherapy has been tempered by their reported ability to produce proinflammatory cytokines when manipulated in vitro, or in vivo. Understanding processes that can limit this potentially deleterious effect of Treg cells in a therapeutic setting is therefore important. Here, we have studied this using induced (i) Treg cells in which de novo Foxp3 expression is driven by TCR-stimulation in vitro in the presence of TGF-ß. We show that iTreg cells can produce significant amounts of three proinflammatory cytokines (IFN-γ, GM-CSF and TNF-α) upon secondary TCR stimulation. GM-CSF is a critical T-cell derived cytokine for the induction of EAE in mice. Despite their apparent capacity to produce GM-CSF, myelin autoantigen-responsive iTreg cells were unable to provoke EAE. Instead, they maintained strong suppressive function in vivo, preventing EAE induction by their CD4+ Foxp3- counterparts. We identified that although iTreg cells maintained the ability to produce IFN-γ and TNF-α in vivo, their ability to produce GM-CSF was selectively degraded upon antigen stimulation under inflammatory conditions. Furthermore, we show that IL-6 and IL-27 individually, or IL-2 and TGF-ß in combination, can mediate the selective loss of GM-CSF production by iTreg cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Inflammation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/biosynthesis , Immunotherapy , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-12/pharmacology , Interleukin-2/pharmacology , Interleukin-6/pharmacology , Interleukins/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Th1 Cells , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
15.
J Immunol ; 190(3): 881-5, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267024

ABSTRACT

Mice lacking IL-6 are resistant to autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE), which is driven by CNS-reactive CD4(+) T cells. There are multiple cellular sources of IL-6, but the critical source in EAE has been uncertain. Using cell-specific IL-6 deficiency in models of EAE induced by active immunization, passive transfer, T cell transfer, and dendritic cell transfer, we show that neither the pathogenic T cells nor CNS-resident cells are required to produce IL-6. Instead, the requirement for IL-6 was restricted to the early stages of T cell activation and was entirely controlled by dendritic cell-derived IL-6. This reflected the loss of IL-6R expression by T cells over time. These data explain why blockade of IL-6R only achieves protection against EAE if used at the time of T cell priming. The implications for therapeutic manipulation of IL-6 signaling in human T cell-driven autoimmune conditions are considered.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-6/immunology , Adoptive Transfer , Animals , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Crosses, Genetic , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Histocompatibility Antigens Class II/immunology , Immunization, Passive , Interleukin-6/deficiency , Interleukin-6/metabolism , Lymphocyte Activation , Lymphokines/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/immunology , Specific Pathogen-Free Organisms , T-Cell Antigen Receptor Specificity
16.
Proc Natl Acad Sci U S A ; 109(35): 14134-9, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22891339

ABSTRACT

Reestablishing self-tolerance in autoimmunity is thought to depend on self-reactive regulatory T cells (Tregs). Exploiting these antigen-specific regulators is hampered by the obscure nature of disease-relevant autoantigens. We have uncovered potent disease-suppressive Tregs recognizing Heat Shock Protein (Hsp) 70 self-antigens, enabling selective activity in inflamed tissues. Hsp70 is a major contributor to the MHC class II ligandome. Here we show that a conserved Hsp70 epitope (B29) is present in murine MHC class II and that upon transfer, B29-induced CD4(+)CD25(+)Foxp3(+) T cells suppress established proteoglycan-induced arthritis in mice. These self-antigen-specific Tregs were activated in vivo, and when using Lymphocyte Activation Gene-3 as a selection marker, as few as 4,000 cells sufficed. Furthermore, depletion of transferred Tregs abrogated disease suppression. Transferred cells exhibited a stable phenotype and were found in joints and draining lymph nodes up to 2 mo after transfer. Given that (i) B29 administration by itself suppressed disease, (ii) our findings were made with wild-type (T-cell receptor nontransgenic) Tregs, and (iii) the B29 human homolog is presented by HLA class II, we are nearing translation of antigen-specific Treg activation as a promising intervention for chronic inflammatory diseases.


Subject(s)
Arthritis/immunology , Arthritis/therapy , Epitopes, T-Lymphocyte/immunology , HSP70 Heat-Shock Proteins/pharmacology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Adoptive Transfer/methods , Animals , Arthritis/metabolism , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/therapy , Autoimmunity/immunology , Epitopes, T-Lymphocyte/metabolism , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Immunization/methods , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Stress, Physiological/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Nat Rev Immunol ; 2(7): 487-98, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12094223

ABSTRACT

The immune system must avoid aggressive T-cell responses against self-antigens. But, paradoxically, exposure to self-peptides seems to have an important role in positive selection in the thymus and the maintenance of a broad T-cell repertoire in the periphery. Recent experiments have highlighted situations that allow high-avidity self-reactive T cells to avoid negative selection in the thymus. Accumulating evidence indicates that other, non-deleting mechanisms control the avidity with which T cells recognize self-antigens--a phenomenon that is known as 'tuning'. This might maximize the peripheral T-cell repertoire by allowing the survival of T cells that can respond to self, but only at concentrations that are not normally reached in vivo.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes/immunology , Animals , Humans
18.
Eur J Immunol ; 43(11): 2818-23, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23878019

ABSTRACT

T cells that produce both IL-17 and IFN-γ, and co-express ROR-γt and T-bet, are often found at sites of autoimmune inflammation. However, it is unknown whether this co-expression of T-bet with ROR-γt is a prerequisite for immunopathology. We show here that T-bet is not required for the development of Th17-driven experimental autoimmune encephalomyelitis (EAE). The disease was not impaired in T-bet(-/-) mice and was associated with low IFN-γ production and elevated IL-17 production among central nervous system (CNS) infiltrating CD4(+) T cells. T-bet(-/-) Th17 cells generated in the presence of IL-6/TGF-ß/IL-1 and IL-23 produced GM-CSF and high levels of IL-17 and induced disease upon transfer to naïve mice. Unlike their WT counterparts, these T-bet(-/-) Th17 cells did not exhibit an IL-17→IFN-γ switch upon reencounter with antigen in the CNS, indicating that this functional change is not critical to disease development. In contrast, T-bet was absolutely required for the pathogenicity of myelin-responsive Th1 cells. T-bet-deficient Th1 cells failed to accumulate in the CNS upon transfer, despite being able to produce GM-CSF. Therefore, T-bet is essential for establishing Th1-mediated inflammation but is not required to drive IL-23-induced GM-CSF production, or Th17-mediated autoimmune inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Cells, Cultured , Central Nervous System/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-23/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics
19.
J Immunol ; 188(5): 2093-101, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22287719

ABSTRACT

CFA is a strong adjuvant capable of stimulating cellular immune responses. Paradoxically, adjuvant immunotherapy by prior exposure to CFA or live mycobacteria suppresses the severity of experimental autoimmune encephalomyelitis (EAE) and spontaneous diabetes in rodents. In this study, we investigated immune responses during adjuvant immunotherapy of EAE. Induction of EAE in CFA-pretreated mice resulted in a rapid influx into the draining lymph nodes (dLNs) of large numbers of CD11b(+)Gr-1(+) myeloid cells, consisting of immature cells with ring-shaped nuclei, macrophages, and neutrophils. Concurrently, a population of mycobacteria-specific IFN-γ-producing T cells appeared in the dLNs. Immature myeloid cells in dLNs expressed the chemokines CXCL10 and CXCL16 in an IFN-γ-dependent manner. Subsequently, CD4(+) T cells coexpressing the cognate chemokine receptors CXCR3 and CXCR6 and myelin oligodendrocyte glycoprotein (MOG)-specific CD4(+) T cells accumulated within the chemokine-expressing dLNs, rather than within the CNS. Migration of CD4(+) T cells toward dLN cells was abolished by depleting the CD11b(+) cells and was also mediated by the CD11b(+) cells alone. In addition to altering the distribution of MOG-specific T cells, adjuvant treatment suppressed development of MOG-specific IL-17. Thus, adjuvant immunotherapy of EAE requires IFN-γ, which suppresses development of the Th17 response, and diverts autoreactive T cells away from the CNS toward immature myeloid cells expressing CXCL10 and CXCL16 in the lymph nodes.


Subject(s)
Cell Differentiation/immunology , Chemokine CXCL10/biosynthesis , Chemokine CXCL6/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Myelin Proteins/immunology , Myeloid Cells/immunology , Receptors, CXCR3/biosynthesis , Receptors, CXCR/biosynthesis , T-Lymphocyte Subsets/immunology , Animals , Cell Migration Inhibition/immunology , Chemokine CXCL16 , Encephalomyelitis, Autoimmune, Experimental/microbiology , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/therapeutic use , Interferon-gamma/physiology , Interferon-gamma/therapeutic use , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , Myelin-Oligodendrocyte Glycoprotein , Myeloid Cells/microbiology , Myeloid Cells/pathology , Receptors, CXCR6 , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
20.
Immunol Rev ; 233(1): 146-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20192998

ABSTRACT

B lymphocytes contribute to immunity through production of antibodies, antigen presentation to T cells, and secretion of cytokines. B cells are generally considered in autoimmune diseases as drivers of pathogenesis. This view is certainly justified, given the successful utilization of the B cell-depleting reagent rituximab in patients with rheumatoid arthritis or other autoimmune pathologies. In a number of cases, however, the depletion of B cells led to an exacerbation of symptoms in patients with autoimmune disorders. In a similar manner, mice lacking B cells can develop an aggravated course of disease in several autoimmune models. These paradoxical observations are now explained by the concept that activated B cells can suppress immune responses through the production of cytokines, especially interleukin-10. Here, we review the stimulatory signals that induce interleukin-10 secretion and suppressive functions in B cells and the phenotype of the B cells with such characteristics. Finally, we formulate a model explaining how this process of immune regulation by activated B cells can confer advantageous properties to the immune system in its combat with pathogens. Altogether, this review proposes that B-cell-mediated regulation is a fundamental property of the immune system, with features of great interest for the development of new cell-based therapies for autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity , B-Lymphocytes/immunology , Immune Tolerance , Lymphocyte Activation , Toll-Like Receptors/immunology , Animals , Autoimmune Diseases/therapy , Disease Models, Animal , Humans , Interleukin-10/immunology , Mice , Phenotype , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL