Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Immunity ; 56(10): 2358-2372.e5, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37699392

ABSTRACT

Lung-resident memory B cells (lung-BRMs) differentiate into plasma cells after reinfection, providing enhanced pulmonary protection. Here, we investigated the determinants of lung-BRM differentiation upon influenza infection. Kinetic analyses revealed that influenza nucleoprotein (NP)-specific BRMs preferentially differentiated early after infection and required T follicular helper (Tfh) cell help. BRM differentiation temporally coincided with transient interferon (IFN)-γ production by Tfh cells. Depletion of IFN-γ in Tfh cells prevented lung-BRM differentiation and impaired protection against heterosubtypic infection. IFN-γ was required for expression of the transcription factor T-bet by germinal center (GC) B cells, which promoted differentiation of a CXCR3+ GC B cell subset that were precursors of lung-BRMs and CXCR3+ memory B cells in the mediastinal lymph node. Absence of IFN-γ signaling or T-bet in GC B cells prevented CXCR3+ pre-memory precursor development and hampered CXCR3+ memory B cell differentiation and subsequent lung-BRM responses. Thus, Tfh-cell-derived IFN-γ is critical for lung-BRM development and pulmonary immunity, with implications for vaccination strategies targeting BRMs.


Subject(s)
Influenza, Human , T-Lymphocytes, Helper-Inducer , Humans , Interferon-gamma/metabolism , Memory B Cells , T Follicular Helper Cells/metabolism , Germinal Center , Cell Differentiation , Receptors, CXCR3/metabolism
2.
Cell Mol Immunol ; 20(6): 651-665, 2023 06.
Article in English | MEDLINE | ID: mdl-37046042

ABSTRACT

Defective interleukin-6 (IL-6) signaling has been associated with Th2 bias and elevated IgE levels. However, the underlying mechanism by which IL-6 prevents the development of Th2-driven diseases remains unknown. Using a model of house dust mite (HDM)-induced Th2 cell differentiation and allergic airway inflammation, we showed that IL-6 signaling in allergen-specific T cells was required to prevent Th2 cell differentiation and the subsequent IgE response and allergic inflammation. Th2 cell lineage commitment required strong sustained IL-2 signaling. We found that IL-6 turned off IL-2 signaling during early T-cell activation and thus inhibited Th2 priming. Mechanistically, IL-6-driven inhibition of IL-2 signaling in responding T cells was mediated by upregulation of Suppressor Of Cytokine Signaling 3 (SOCS3). This mechanism could be mimicked by pharmacological Janus Kinase-1 (JAK1) inhibition. Collectively, our results identify an unrecognized mechanism that prevents the development of unwanted Th2 cell responses and associated diseases and outline potential preventive interventions.


Subject(s)
Interleukin-6 , Th2 Cells , Humans , Th2 Cells/metabolism , Interleukin-2 , Inflammation , Immunoglobulin E , Cytokines/metabolism , Suppressor of Cytokine Signaling 3 Protein
3.
Cell Rep ; 37(13): 110178, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965421

ABSTRACT

Lipopolysaccharide (LPS) can either promote or prevent T helper 2 (Th2) cell allergic responses. However, the underlying mechanism remains unknown. We show here that LPS activity switches from pro-pathogenic to protective depending on the production of granulocyte-macrophage colony-stimulating factor (GM-CSF) by non-classical monocytes. In the absence of GM-CSF, LPS can favor pathogenic Th2 cell responses by supporting the trafficking of lung-migratory dendritic cells (mDC2s) into the lung-draining lymph node. However, when non-classical monocytes produce GM-CSF, LPS and GM-CSF synergize to differentiate monocyte-derived DCs from classical Ly6Chi monocytes that instruct mDC2s for Th2 cell suppression. Importantly, only allergens with cysteine protease activity trigger GM-CSF production by non-classical monocytes. Hence, the therapeutic effect of LPS is restricted to allergens with this enzymatic activity. Treatment with GM-CSF, however, restores the protective effects of LPS. Thus, GM-CSF produced by non-classical monocytes acts as a rheostat that fine-tunes the pathogenic and therapeutic functions of LPS.


Subject(s)
Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hypersensitivity/immunology , Inflammation/immunology , Lipopolysaccharides/pharmacology , Monocytes/immunology , Th2 Cells/immunology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hypersensitivity/etiology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology
4.
Sci Immunol ; 6(63): eabg6895, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34516781

ABSTRACT

CD8+ T cell responses to pulmonary challenges are primed by lung migratory dendritic cells (mDCs), which capture antigens in the lungs and migrate to the lung-draining mediastinal lymph node (med-LN) to activate T cells. The lungs and the spleen are not connected by the lymphatic vasculature. Thus, the current paradigm suggests that, in response to respiratory virus infections that are restricted to the respiratory tract, priming of T cell responses by lung mDCs takes place entirely in the med-LN. Our results challenge this "LN-centric" paradigm by demonstrating that, during influenza virus infection, lung mDCs egress the med-LN and traffic to the spleen, where they prime influenza-specific CD8+ T cells. CD8+ T cells primed in the spleen are transcriptionally distinct and have enhanced ability to differentiate into long-lived memory cells compared with med-LN­primed counterparts. Thus, our data identify a lung mDC trafficking pathway that connects the lungs with the spleen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Spleen/immunology , Animals , Cell Movement/immunology , Dendritic Cells/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, T-Lymphoid/immunology
5.
Sci Immunol ; 4(39)2019 09 13.
Article in English | MEDLINE | ID: mdl-31519812

ABSTRACT

Sustained T cell receptor (TCR) stimulation is required for maintaining germinal center T follicular helper (GC-TFH) cells. Paradoxically, TCR activation induces interleukin-2 receptor (IL-2R) expression and IL-2 production, thereby initiating a feedback loop of IL-2 signaling that normally inhibits TFH cells. It is unclear how GC-TFH cells can receive prolonged TCR signaling without succumbing to the detrimental effects of IL-2. Using an influenza infection model, we show here that GC-TFH cells secreted large amounts of IL-2 but responded poorly to it. To maintain their IL-2 hyporesponsiveness, GC-TFH cells required intrinsic IL-6 signaling. Mechanistically, we found that IL-6 inhibited up-regulation of IL-2Rß (CD122) by preventing association of STAT5 with the Il2rb locus, thus allowing GC-TFH cells to receive sustained TCR signaling and produce IL-2 without initiating a TCR/IL-2 inhibitory feedback loop. Collectively, our results identify a regulatory mechanism that controls the generation of GC-TFH cells.


Subject(s)
Germinal Center/immunology , Interleukin-2/immunology , Interleukin-6/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Germinal Center/cytology , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Interleukin-6/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/cytology
6.
Immunity ; 50(1): 225-240.e4, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30635238

ABSTRACT

Infants have a higher risk of developing allergic asthma than adults. However, the underlying mechanism remains unknown. We show here that sensitization of mice with house-dust mites (HDMs) in the presence of low-dose lipopolysaccharide (LPS) prevented T helper 2 (Th2) cell allergic responses in adult, but not infant, mice. Mechanistically, adult CD11b+ migratory dendritic cells (mDCs) upregulated the transcription factor T-bet in response to tumor necrosis factor-α (TNF-α), which was rapidly induced after HDM + LPS sensitization. Consequently, adult CD11b+ mDCs produced interleukin-12 (IL-12), which prevented Th2 cell development by promoting T-bet upregulation in responding T cells. Conversely, infants failed to induce TNF-α after HDM + LPS sensitization. Therefore, CD11b+ mDCs failed to upregulate T-bet and did not secrete IL-12 and Th2 cell responses normally developed in infant mice. Thus, the availability of TNF-α dictates the ability of CD11b+ mDCs to suppress allergic Th2-cell responses upon dose-dependent endotoxin sensitization and is a key mediator governing susceptibility to allergic airway inflammation in infant mice.


Subject(s)
Dendritic Cells/physiology , Hypersensitivity/immunology , Inflammation/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Adult , Animals , Animals, Newborn , Antigens, Dermatophagoides , Cell Differentiation , Humans , Immunization , Infant , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae/immunology , T-Box Domain Proteins/metabolism
7.
Nat Immunol ; 18(11): 1249-1260, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28892471

ABSTRACT

Interleukin 2 (IL-2) promotes Foxp3+ regulatory T (Treg) cell responses, but inhibits T follicular helper (TFH) cell development. However, it is not clear how IL-2 affects T follicular regulatory (TFR) cells, a cell type with properties of both Treg and TFH cells. Using an influenza infection model, we found that high IL-2 concentrations at the peak of the infection prevented TFR cell development by a Blimp-1-dependent mechanism. However, once the immune response resolved, some Treg cells downregulated CD25, upregulated Bcl-6 and differentiated into TFR cells, which then migrated into the B cell follicles to prevent the expansion of self-reactive B cell clones. Thus, unlike its effects on conventional Treg cells, IL-2 inhibits TFR cell responses.


Subject(s)
Interleukin-2/pharmacology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Cell Movement/genetics , Cell Movement/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling/methods , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Interleukin-2/administration & dosage , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
8.
J Immunol ; 199(7): 2377-2387, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28848064

ABSTRACT

B cell lymphoma-6 (Bcl-6) is a transcriptional repressor that is required for the differentiation of T follicular helper (TFH) cell populations. Currently, the molecular mechanisms underlying the transcriptional regulation of Bcl-6 expression are unclear. In this study, we have identified the Ikaros zinc finger transcription factors Aiolos and Ikaros as novel regulators of Bcl-6. We found that increased expression of Bcl-6 in CD4+ Th cell populations correlated with enhanced enrichment of Aiolos and Ikaros at the Bcl6 promoter. Furthermore, overexpression of Aiolos or Ikaros, but not the related family member Eos, was sufficient to induce Bcl6 promoter activity. Intriguingly, STAT3, a known Bcl-6 transcriptional regulator, physically interacted with Aiolos to form a transcription factor complex capable of inducing the expression of Bcl6 and the TFH-associated cytokine receptor Il6ra Importantly, in vivo studies revealed that the expression of Aiolos was elevated in Ag-specific TFH cells compared with that observed in non-TFH effector Th cells generated in response to influenza infection. Collectively, these data describe a novel regulatory mechanism through which STAT3 and the Ikaros zinc finger transcription factors Aiolos and Ikaros cooperate to regulate Bcl-6 expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Ikaros Transcription Factor/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Ikaros Transcription Factor/genetics , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-6/metabolism , STAT3 Transcription Factor/genetics , T-Lymphocytes, Helper-Inducer/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...