Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
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
2.
Immunity ; 46(6): 943-956, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636961

ABSTRACT

The central nervous system (CNS) and its meningeal coverings accommodate a diverse myeloid compartment that includes parenchymal microglia and perivascular macrophages, as well as choroid plexus and meningeal macrophages, dendritic cells, and granulocytes. These myeloid populations enjoy an intimate relationship with the CNS, where they play an essential role in both health and disease. Although the importance of these cells is clearly recognized, their exact function in the CNS continues to be explored. Here, we review the subsets of myeloid cells that inhabit the parenchyma, meninges, and choroid plexus and discuss their roles in CNS homeostasis. We also discuss the role of these cells in various neurological pathologies, such as autoimmunity, mechanical injury, neurodegeneration, and infection. We highlight the neuroprotective nature of certain myeloid cells by emphasizing their therapeutic potential for the treatment of neurological conditions.


Subject(s)
Autoimmune Diseases/immunology , Choroid Plexus/immunology , Infections/immunology , Myeloid Cells/physiology , Neurodegenerative Diseases/immunology , Neuroimmunomodulation , Wounds and Injuries/immunology , Animals , Central Nervous System , Humans , Meninges/immunology , Neuroprotection
4.
Gut ; 73(1): 92-104, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37595983

ABSTRACT

OBJECTIVE: Wheat has become a main staple globally. We studied the effect of defined pro-inflammatory dietary proteins, wheat amylase trypsin inhibitors (ATI), activating intestinal myeloid cells via toll-like receptor 4, in experimental autoimmune encephalitis (EAE), a model of multiple sclerosis (MS). DESIGN: EAE was induced in C57BL/6J mice on standardised dietary regimes with defined content of gluten/ATI. Mice received a gluten and ATI-free diet with defined carbohydrate and protein (casein/zein) content, supplemented with: (a) 25% of gluten and 0.75% ATI; (b) 25% gluten and 0.19% ATI or (c) 1.5% purified ATI. The effect of dietary ATI on clinical EAE severity, on intestinal, mesenteric lymph node, splenic and central nervous system (CNS) subsets of myeloid cells and lymphocytes was analysed. Activation of peripheral blood mononuclear cells from patients with MS and healthy controls was compared. RESULTS: Dietary ATI dose-dependently caused significantly higher EAE clinical scores compared with mice on other dietary regimes, including on gluten alone. This was mediated by increased numbers and activation of pro-inflammatory intestinal, lymph node, splenic and CNS myeloid cells and of CNS-infiltrating encephalitogenic T-lymphocytes. Expectedly, ATI activated peripheral blood monocytes from both patients with MS and healthy controls. CONCLUSIONS: Dietary wheat ATI activate murine and human myeloid cells. The amount of ATI present in an average human wheat-based diet caused mild intestinal inflammation, which was propagated to extraintestinal sites, leading to exacerbation of CNS inflammation and worsening of clinical symptoms in EAE. These results support the importance of the gut-brain axis in inflammatory CNS disease.


Subject(s)
Multiple Sclerosis , Humans , Animals , Mice , Trypsin Inhibitors/pharmacology , Trypsin Inhibitors/chemistry , Triticum/chemistry , Amylases , Leukocytes, Mononuclear , Mice, Inbred C57BL , Inflammation , Central Nervous System , Glutens , Diet
5.
EMBO J ; 36(22): 3292-3308, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28963396

ABSTRACT

Microglia are resident macrophages of the central nervous system that contribute to homeostasis and neuroinflammation. Although known to play an important role in brain development, their exact function has not been fully described. Here, we show that in contrast to healthy adult and inflammation-activated cells, neonatal microglia show a unique myelinogenic and neurogenic phenotype. A CD11c+ microglial subset that predominates in primary myelinating areas of the developing brain expresses genes for neuronal and glial survival, migration, and differentiation. These cells are the major source of insulin-like growth factor 1, and its selective depletion from CD11c+ microglia leads to impairment of primary myelination. CD11c-targeted toxin regimens induced a selective transcriptional response in neonates, distinct from adult microglia. CD11c+ microglia are also found in clusters of repopulating microglia after experimental ablation and in neuroinflammation in adult mice, but despite some similarities, they do not recapitulate neonatal microglial characteristics. We therefore identify a unique phenotype of neonatal microglia that deliver signals necessary for myelination and neurogenesis.


Subject(s)
Brain/cytology , Brain/embryology , Microglia/metabolism , Myelin Sheath/metabolism , Neurogenesis , Aging/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/ultrastructure , CD11c Antigen/metabolism , Cell Aggregation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Neural Plate/metabolism , Up-Regulation/genetics
6.
Immunity ; 37(2): 264-75, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22902234

ABSTRACT

Mature dendritic cells (DCs) are established as unrivaled antigen-presenting cells (APCs) in the initiation of immune responses, whereas steady-state DCs induce peripheral T cell tolerance. Using various genetic approaches, we depleted CD11c(+) DCs in mice and induced autoimmune CNS inflammation. Unexpectedly, mice lacking DCs developed aggravated disease compared to control mice. Furthermore, when we engineered DCs to present a CNS-associated autoantigen in an induced manner, we found robust tolerance that prevented disease, which coincided with an upregulation of the PD-1 receptor on antigen-specific T cells. Additionally, we showed that PD-1 was necessary for DC-mediated induction of regulatory T cells. Our results show that a reduction of DCs interferes with tolerance, resulting in a stronger inflammatory response, and that other APC populations could compensate for the loss of immunogenic APC function in DC-depleted mice.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/immunology , Autoantigens/immunology , Autoimmunity/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD11c Antigen , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
7.
J Allergy Clin Immunol ; 146(3): 652-666.e11, 2020 09.
Article in English | MEDLINE | ID: mdl-32445838

ABSTRACT

BACKGROUND: Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE: We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS: Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS: Different adjuvants induce distinct IgG+ GC B-cell responses with specific transcriptomes and expression levels of the α2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3- follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-γ+ TFH1 cells, IL-6/IL-23-dependent IL-17A+ TFH17 cells, and high ratios of TFH cells to Foxp3+ follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION: This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Germinal Center/immunology , Immunoglobulin G/immunology , Alum Compounds/administration & dosage , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cytokines/immunology , Female , Freund's Adjuvant/administration & dosage , Glycosylation , Lipopolysaccharides/administration & dosage , Mice, Inbred C57BL , Mice, Knockout , Mineral Oil/administration & dosage , Mycobacterium tuberculosis/immunology , Ovalbumin/administration & dosage , Polysorbates/administration & dosage , Squalene/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vaccination
8.
EMBO J ; 35(1): 89-101, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26612827

ABSTRACT

Multiple sclerosis is the most frequent chronic inflammatory disease of the CNS. The entry and survival of pathogenic T cells in the CNS are crucial for the initiation and persistence of autoimmune neuroinflammation. In this respect, contradictory evidence exists on the role of the most potent type of antigen-presenting cells, dendritic cells. Applying intravital two-photon microscopy, we demonstrate the gatekeeper function of CNS professional antigen-presenting CD11c(+) cells, which preferentially interact with Th17 cells. IL-17 expression correlates with expression of GM-CSF by T cells and with accumulation of CNS CD11c(+) cells. These CD11c(+) cells are organized in perivascular clusters, targeted by T cells, and strongly express the inflammatory chemokines Ccl5, Cxcl9, and Cxcl10. Our findings demonstrate a fundamental role of CNS CD11c(+) cells in the attraction of pathogenic T cells into and their survival within the CNS. Depletion of CD11c(+) cells markedly reduced disease severity due to impaired enrichment of pathogenic T cells within the CNS.


Subject(s)
Antigen-Presenting Cells/physiology , Brain/pathology , CD11c Antigen/analysis , Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/chemistry , Brain/immunology , Cell Movement , Dendritic Cells/chemistry , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , Mice, Inbred C57BL , T-Lymphocytes/physiology , Th17 Cells/physiology
9.
J Immunol ; 201(11): 3218-3228, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30355786

ABSTRACT

17ß-Estradiol (E2) suppresses the development of experimental autoimmune encephalomyelitis (EAE) through estrogen receptor (ER) α, yet the cellular targets remain elusive. We have used an adoptive transfer model of myelin oligodendrocyte glycoprotein-specific CD4+ T cells from 2D2 TCR transgenic mice. We show that in the recipient mice, ERα expression in bystander CD4+ T cells, rather than in cognate 2D2 T cells, is required for the inhibition of Th17 cell differentiation by E2. Coadministration of estrogen-primed WT, but not ERα-deficient CD4+ T cells, with naive 2D2 T cells lacking ERα inhibited the development of Th17 cell-mediated EAE. Suppression of Th17 cells and protection from EAE were maintained when ERα was deleted in Foxp3+ regulatory T cells. We showed that in vivo PD-L1 blockade alleviated the anti-inflammatory action of E2 and that PD-1 expression on cognate but not bystander T cells was required for the E2-dependent inhibition of Th17 differentiation. In cotransfer experiments, we found that only WT but not PD-1KO 2D2 T cells were amenable to E2-dependent inhibition of Th17 differentiation. These results support the conclusion that the restriction of Th17 cell development by E2-primed bystander CD4+ T cells requires cell-intrinsic PD-1 signaling within cognate T cells rather than induction of regulatory 2D2 T cells through PD-1 engagement. Altogether, our results indicate that pregnancy-level concentrations of estrogen signal in conventional Foxp3neg CD4+ T cells to limit the differentiation of cognate Th17 cells through a trans-acting mechanism of suppression that requires a functional PD-1/PD-L1 regulatory axis.


Subject(s)
Bystander Effect , Central Nervous System/immunology , Estrogens/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , Autoimmunity , B7-H1 Antigen/immunology , CD4 Antigens/metabolism , Cell Differentiation , Disease Models, Animal , Estrogen Receptor alpha/genetics , Female , Forkhead Transcription Factors/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 114(8): E1480-E1489, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167776

ABSTRACT

TGF-ß is an anti-inflammatory cytokine whose signaling is negatively controlled by Smad7. Previously, we established a role for Smad7 in the generation of autoreactive T cells; however, the function of Smad7 in dendritic cells (DCs) remains elusive. Here, we demonstrate that DC-specific Smad7 deficiency resulted in elevated expression of the transcription factors Batf3 and IRF8, leading to increased frequencies of CD8+CD103+ DCs in the spleen. Furthermore, Smad7-deficient DCs expressed higher levels of indoleamine 2,3-dioxygenase (IDO), an enzyme associated with tolerance induction. Mice devoid of Smad7 specifically in DCs are resistant to the development of experimental autoimmune encephalomyelitis (EAE) as a result of an increase of protective regulatory T cells (Tregs) and reduction of encephalitogenic effector T cells in the central nervous system. In agreement, inhibition of IDO activity or depletion of Tregs restored disease susceptibility. Intriguingly, when Smad7-deficient DCs also lacked the IFN-γ receptor, the mice regained susceptibility to EAE, demonstrating that IFN-γ signaling in DCs mediates their tolerogenic function. Our data indicate that Smad7 expression governs splenic DC subset differentiation and is critical for the promotion of their efficient function in immunity.


Subject(s)
Autoimmunity/physiology , Dendritic Cells/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism
11.
Proc Natl Acad Sci U S A ; 111(32): 11810-5, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25074917

ABSTRACT

The prevalence of allergic asthma and other atopic diseases has reached epidemic proportions in large parts of the developed world. The gradual loss of the human indigenous microbiota has been held responsible for this trend. The bacterial pathogen Helicobacter pylori is a constituent of the normal gastric microbiota whose presence has been inversely linked to allergy and asthma in humans and experimental models. Here we show that oral or i.p. tolerization with H. pylori extract prevents the airway hyperresponsiveness, bronchoalveolar eosinophilia, pulmonary inflammation, and Th2 cytokine production that are hallmarks of allergen-induced asthma in mice. Asthma protection is not conferred by extracts from other enteropathogens and requires a heat-sensitive H. pylori component and the DC-intrinsic production of IL-10. The basic leucine zipper ATF-like 3 (BATF3)-dependent CD103(+)CD11b(-) dendritic cell lineage is enriched in the lungs of protected mice and strictly required for protection. Two H. pylori persistence determinants, the γ-glutamyl-transpeptidase GGT and the vacuolating cytotoxin VacA, are required and sufficient for asthma protection and can be administered in purified form to prevent asthma. In conclusion, we provide preclinical evidence for the concept that the immunomodulatory properties of H. pylori can be exploited for tolerization strategies aiming to prevent allergen-induced asthma.


Subject(s)
Asthma/microbiology , Asthma/therapy , Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Immunologic Factors/therapeutic use , Interleukin-10/immunology , Repressor Proteins/immunology , Allergens/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Asthma/immunology , Bacterial Proteins/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Disease Models, Animal , Humans , Immune Tolerance , Interleukin-18/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/deficiency , Repressor Proteins/genetics , T-Lymphocytes, Regulatory/immunology , gamma-Glutamyltransferase/immunology
12.
Arterioscler Thromb Vasc Biol ; 34(12): 2658-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25341795

ABSTRACT

OBJECTIVE: Interleukin (IL)-17A is regarded as an important cytokine to drive psoriasis, an inflammatory skin disease marked by increased cardiovascular mortality. We aimed to test the hypothesis that overproduction of IL-17A in the skin leading to dermal inflammation may systemically cause vascular dysfunction in psoriasis-like skin disease. APPROACH AND RESULTS: Conditional overexpression of IL-17A in keratinocytes caused severe psoriasis-like skin inflammation in mice (K14-IL-17A(ind/+) mice), associated with increased reactive oxygen species formation and circulating CD11b(+) inflammatory leukocytes in blood, with endothelial dysfunction, increased systolic blood pressure, left ventricular hypertrophy, and reduced survival compared with controls. In K14-IL-17A(ind/+) mice, immunohistochemistry and flow cytometry revealed increased vascular production of the nitric oxide/superoxide reaction product peroxynitrite and infiltration of the vasculature with myeloperoxidase(+)CD11b(+)GR1(+)F4/80(-) cells accompanied by increased expression of the inducible nitric oxide synthase and the nicotinamide dinucleotide phosphate (NADPH) oxidase, nox2. Neutrophil depletion by anti-GR-1 antibody injections reduced oxidative stress in blood and vessels. Neutralization of tumor necrosis factor-α and IL-6 (both downstream of IL-17A) reduced skin lesions, attenuated oxidative stress in heart and blood, and partially improved endothelial dysfunction in K14-IL-17A(ind/+) mice. CONCLUSIONS: Dermal overexpression of IL-17A induces systemic endothelial dysfunction, vascular oxidative stress, arterial hypertension, and increases mortality mainly driven by myeloperoxidase(+)CD11b(+)GR1(+)F4/80(-) inflammatory cells. Depletion of the GR-1(+) immune cells or neutralization of IL-17A downstream cytokines by biologicals attenuates the vascular phenotype in K14-IL-17A(ind/+) mice.


Subject(s)
Interleukin-17/physiology , Psoriasis/etiology , Psoriasis/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Aorta/pathology , Cardiovascular Diseases/etiology , Case-Control Studies , Disease Models, Animal , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Female , Humans , Hypertension/etiology , Hypertension/immunology , Hypertension/physiopathology , Interleukin-17/genetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/physiology , Keratinocytes/immunology , Keratinocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myocytes, Cardiac/pathology , Neutrophils/pathology , Neutrophils/physiology , Nitric Oxide/metabolism , Psoriasis/complications , Reactive Oxygen Species/metabolism , Risk Factors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Up-Regulation , Vasculitis/etiology , Vasculitis/immunology , Vasculitis/physiopathology
13.
Biol Blood Marrow Transplant ; 20(11): 1696-704, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25132527

ABSTRACT

Graft-versus-host disease (GVHD) is a frequent life-threatening complication after allogeneic hematopoietic stem cell transplantation (HSCT) and induced by donor-derived T cells that become activated by host antigen-presenting cells. To address the relevance of host dendritic cell (DC) populations in this disease, we used mouse strains deficient in CD11c(+) or CD8α(+) DC populations in a model of acute GVHD where bone marrow and T cells from BALB/c donors were transplanted into C57BL/6 hosts. Surprisingly, a strong increase in GVHD-related mortality was observed in the absence of CD11c(+) cells. Likewise, Batf3-deficient (Batf3(-/-)) mice that lack CD8α(+) DCs also displayed a strongly increased GVHD-related mortality. In the absence of CD8α(+) DCs, we detected an increased activation of the remaining DC populations after HSCT, leading to an enhanced priming of allogeneic T cells. Importantly, this was associated with reduced numbers of regulatory T cells and transforming growth factor-ß levels, indicating an aggravated failure of peripheral tolerance mechanisms after HSCT in the absence of CD8α(+) DCs. In summary, our results indicate a critical role of CD8α(+) DCs as important inducers of regulatory T cell-mediated tolerance to control DC activation and T cell priming in the initiation phase of GVHD.


Subject(s)
Bone Marrow Transplantation/methods , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft vs Host Disease/prevention & control , Transplantation, Homologous/methods , Animals , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
14.
Brain ; 136(Pt 4): 1048-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23518712

ABSTRACT

Laquinimod is an orally administered compound that is under investigation in relapsing-remitting multiple sclerosis. To understand the mechanism by which laquinimod exerts its clinical effects, we have performed human and murine studies assessing its immunomodulatory properties. In experimental autoimmune encephalomyelitis, the therapeutic administration of laquinimod beginning during the recovery of SJL mice, prevented further relapses as expected and strongly reduced infiltration of CD4+ and CD8+ T cells in the central nervous system. We hypothesized that this beneficial effect was mediated by dendritic cells, since we and others found a modulation of different dendritic cell subsets under treatment. According to the findings on antigen-presenting cells in the murine system, we found a reduced capacity of human monocyte-derived dendritic cells treated with therapeutic concentrations of laquinimod, upon maturation with lipopolysaccharide, to induce CD4+ T cell proliferation and secretion of pro-inflammatory cytokines. Furthermore, laquinimod treatment of mature dendritic cells resulted in a decreased chemokine production by both murine and human dendritic cells, associated with a decreased monocyte chemo-attraction. In laquinimod-treated patients with multiple sclerosis we consistently found reduced chemokine and cytokine secretion by conventional CD1c+ dendritic cells upon lipopolysaccharide stimulation. Similarly to the animal model of relapsing-remitting multiple sclerosis, dendritic cell subsets were altered in patients upon laquinimod treatment, as the number of conventional CD1c+ and plasmacytoid CD303+ dendritic cells were decreased within peripheral blood mononuclear cells. Moreover, laquinimod treatment in patients with multiple sclerosis and mice modified the maturation of dendritic cells demonstrated by an upregulation of CD86 expression in vivo. Our data suggest that inhibition of the NF-κB pathway is responsible for the changes observed in dendritic cell maturation and functions. These findings indicate that laquinimod exhibits its disease-modulating activity in multiple sclerosis by downregulating immunogenicity of dendritic cell responses. We suggest that monitoring dendritic cell properties in multiple sclerosis should be implemented in future therapeutic trials.


Subject(s)
Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/pathology , Quinolones/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis, Relapsing-Remitting/immunology , NF-kappa B/drug effects , NF-kappa B/immunology , Quinolones/administration & dosage
15.
J Allergy Clin Immunol ; 130(3): 781-797.e11, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22935591

ABSTRACT

BACKGROUND: Allergic contact dermatitis is one of the most common occupational diseases. A main protective mechanism in those who do not develop allergic contact dermatitis is tolerance induction by repeated exposure to low doses of contact allergen, which is termed low zone tolerance (LZT). The mechanisms that determine the tolerance induction in subjects with LZT are still elusive. OBJECTIVE: We performed analysis of the role of CD4(+)CD25(+) forkhead box protein 3 (FOXP3)-positive regulatory T (Treg) cells and dendritic cells (DCs) in mice with LZT. METHODS: Mechanisms of tolerance induction were analyzed in a murine model of LZT by using FOXP3 and IL-10 reporter mice, as well as mice that allow the selective depletion of Treg cells or DCs. RESULTS: Depletion of CD4(+)CD25(+)FOXP3(+) Treg cells during tolerance induction completely abolishes the development of LZT, resulting in a pronounced contact hypersensitivity response. Adoptive transfer experiments, depletion studies, and use of cell type-specific deficient mice revealed that IL-10 production is critical for the suppressor function of Treg cells in mice with LZT and that tolerogenic CD8(+)CD11c(+) DCs located in the skin-draining lymph nodes are essential for LZT. In the absence of Treg cells, DCs did not develop tolerogenic functions, indicating that activated IL-10(+) Treg cells might imprint the tolerogenic DC phenotype. Cell communication analysis revealed that the education of tolerogenic DCs might involve a direct interaction with Treg cells mediated by gap junctions. Subsequently, induction of tolerogenic CD11c(+) DCs leads to the generation of hapten-specific CD8(+) Treg cells, which protect against contact hypersensitivity. CONCLUSIONS: Our data demonstrate critical interactions between CD4(+)CD25(+)FOXP3(+) Treg cells and tolerogenic CD8(+)CD11c(+) DCs during the induction of LZT.


Subject(s)
Cell Communication , Dendritic Cells/physiology , Dermatitis, Allergic Contact/prevention & control , Immune Tolerance , T-Lymphocytes, Regulatory/physiology , Animals , CD11c Antigen/analysis , Dermatitis, Allergic Contact/immunology , Forkhead Transcription Factors/analysis , Interleukin-10/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, CCR7/analysis
16.
Cell Rep ; 42(4): 112378, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37060566

ABSTRACT

The signals controlling marginal zone (MZ) and follicular (FO) B cell development remain incompletely understood. Here, we show that AKT orchestrates MZ B cell formation in mice and humans. Genetic models that increase AKT signaling in B cells or abolish its impact on FoxO transcription factors highlight the AKT-FoxO axis as an on-off switch for MZ B cell formation in mice. In humans, splenic immunoglobulin (Ig) D+CD27+ B cells, proposed as an MZ B cell equivalent, display higher AKT signaling than naive IgD+CD27- and memory IgD-CD27+ B cells and develop in an AKT-dependent manner from their precursors in vitro, underlining the conservation of this developmental pathway. Consistently, CD148 is identified as a receptor indicative of the level of AKT signaling in B cells, expressed at a higher level in MZ B cells than FO B cells in mice as well as humans.


Subject(s)
B-Lymphocytes , Proto-Oncogene Proteins c-akt , Humans , Mice , Animals , Lymphoid Tissue , Signal Transduction , Spleen
17.
Circulation ; 124(12): 1370-81, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21875910

ABSTRACT

BACKGROUND: Angiotensin II (ATII), a potent vasoconstrictor, causes hypertension, promotes infiltration of myelomonocytic cells into the vessel wall, and stimulates both vascular and inflammatory cell NADPH oxidases. The predominant source of reactive oxygen species, eg, vascular (endothelial, smooth muscle, adventitial) versus phagocytic NADPH oxidase, and the role of myelomonocytic cells in mediating arterial hypertension have not been defined yet. METHODS AND RESULTS: Angiotensin II (1 mg · kg(-1) · d(-1) for 7 days) increased the number of both CD11b(+)Gr-1(low)F4/80(+) macrophages and CD11b(+)Gr-1(high)F4/80(-) neutrophils in mouse aorta (verified by flow cytometry). Selective ablation of lysozyme M-positive (LysM(+)) myelomonocytic cells by low-dose diphtheria toxin in mice with inducible expression of the diphtheria toxin receptor (LysM(iDTR) mice) reduced the number of monocytes in the circulation and limited ATII-induced infiltration of these cells into the vascular wall, whereas the number of neutrophils was not reduced. Depletion of LysM(+) cells attenuated ATII-induced blood pressure increase (measured by radiotelemetry) and vascular endothelial and smooth muscle dysfunction (assessed by aortic ring relaxation studies) and reduced vascular superoxide formation (measured by chemiluminescence, cytochrome c assay, and oxidative fluorescence microtopography) and the expression of NADPH oxidase subunits gp91(phox) and p67(phox) (assessed by Western blot and mRNA reverse-transcription polymerase chain reaction). Adoptive transfer of wild-type CD11b(+)Gr-1(+) monocytes into depleted LysM(iDTR) mice reestablished ATII-induced vascular dysfunction, oxidative stress, and arterial hypertension, whereas transfer of CD11b(+)Gr-1(+) neutrophils or monocytes from gp91(phox) or ATII receptor type 1 knockout mice did not. CONCLUSIONS- Infiltrating monocytes with a proinflammatory phenotype and macrophages rather than neutrophils appear to be essential for ATII-induced vascular dysfunction and arterial hypertension.


Subject(s)
Hypertension/immunology , Monocytes/metabolism , Muramidase/immunology , Muramidase/metabolism , Vasculitis/immunology , Angiotensin II/pharmacology , Animals , CD11b Antigen/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Gene Expression/immunology , Hypertension/chemically induced , Hypertension/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Muramidase/genetics , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Receptors, Chemokine/metabolism , Respiratory Burst/physiology , Vasculitis/chemically induced , Vasculitis/metabolism , Vasoconstrictor Agents/pharmacology
19.
J Invest Dermatol ; 142(3 Pt A): 559-570.e6, 2022 03.
Article in English | MEDLINE | ID: mdl-34536483

ABSTRACT

Jak inhibitors are increasingly used in dermatology. Despite broad inhibitory effects on cytokine signaling cascades, they only modestly increase the risk for infectious diseases. To address the molecular mechanisms underlying this unexpected clinical observation, we investigated how tofacintib (tofa), a first-in-class Jak inhibitor, regulates host defense responses in toll-like receptor 4-activated human macrophages. Specifically, we asked whether tofa inhibits anti-inflammatory IL-10 signaling, thereby counteracting the downregulation of inflammatory, host-protective pathways. We found that tofa blocked macrophage responses to IL-10 at the level of signal transducer and activator of transcription 3 phosphorylation. Furthermore, toll-like receptor 4-induced, autocrine/paracrine IL-10/IL-10R activation promoted the expression of hepcidin, the master regulator of iron metabolism, resulting in intracellular iron sequestration. In contrast, autocrine/paracrine IL-10/IL-10R activation repressed the expression of cathelicidin antimicrobial peptide as well as antigen-presenting molecules, thus together, inducing a pathogen-favoring environment. Although tofa further repressed cathelicidin, it prevented the induction of intracellular HAMP and restored the expression of antigen-presentation molecules in toll-like receptor 4-activated macrophages. Our study supports the concept that induction of IL-10/IL-10R signaling drives a complex immune evasion strategy of intracellular microbes. Moreover, we conclude that tofa has diverging effects on macrophage host response pathways, and we identify the toll-like receptor 4-IL-10-signal transducer and activator of transcription 3-HAMP axis as a potential therapeutic target to counteract immune evasion.


Subject(s)
Janus Kinase Inhibitors , Toll-Like Receptor 4 , Humans , Interleukin-10/metabolism , Iron/metabolism , Janus Kinase Inhibitors/pharmacology , Macrophages/metabolism , Piperidines , Pyrimidines , STAT3 Transcription Factor/metabolism , Toll-Like Receptor 4/metabolism
20.
Cell Rep ; 38(13): 110565, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35354043

ABSTRACT

Interleukin (IL)-10 is considered a prototypical anti-inflammatory cytokine, significantly contributing to the maintenance and reestablishment of immune homeostasis. Accordingly, it has been shown in the intestine that IL-10 produced by Tregs can act on effector T cells, thereby limiting inflammation. Herein, we investigate whether this role also applies to IL-10 produced by T cells during central nervous system (CNS) inflammation. During neuroinflammation, both CNS-resident and -infiltrating cells produce IL-10; yet, as IL-10 has a pleotropic function, the exact contribution of the different cellular sources is not fully understood. We find that T-cell-derived IL-10, but not other relevant IL-10 sources, can promote inflammation in experimental autoimmune encephalomyelitis. Furthermore, in the CNS, T-cell-derived IL-10 acts on effector T cells, promoting their survival and thereby enhancing inflammation and CNS autoimmunity. Our data indicate a pro-inflammatory role of T-cell-derived IL-10 in the CNS.


Subject(s)
Interleukin-10 , T-Lymphocytes , Animals , CD4-Positive T-Lymphocytes , Cell Survival , Central Nervous System , Inflammation , Interleukin-10/physiology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL