Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 654
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 18(5): 563-572, 2017 05.
Article in English | MEDLINE | ID: mdl-28346410

ABSTRACT

Variable strengths of signaling via the T cell antigen receptor (TCR) can produce divergent outcomes, but the mechanism of this remains obscure. The abundance of the transcription factor IRF4 increases with TCR signal strength, but how this would induce distinct types of responses is unclear. We compared the expression of genes in the TH2 subset of helper T cells to enhancer occupancy by the BATF-IRF4 transcription factor complex at varying strengths of TCR stimulation. Genes dependent on BATF-IRF4 clustered into groups with distinct TCR sensitivities. Enhancers exhibited a spectrum of occupancy by the BATF-IRF4 ternary complex that correlated with the sensitivity of gene expression to TCR signal strength. DNA sequences immediately flanking the previously defined AICE motif controlled the affinity of BATF-IRF4 for direct binding to DNA. Analysis by the chromatin immunoprecipitation-exonuclease (ChIP-exo) method allowed the identification of a previously unknown high-affinity AICE2 motif at a human single-nucleotide polymorphism (SNP) of the gene encoding the immunomodulatory receptor CTLA-4 that was associated with resistance to autoimmunity. Thus, the affinity of different enhancers for the BATF-IRF4 complex might underlie divergent signaling outcomes in response to various strengths of TCR signaling.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CTLA-4 Antigen/genetics , Enhancer Elements, Genetic/genetics , Interferon Regulatory Factors/metabolism , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell/metabolism , Th2 Cells/physiology , Animals , Autoimmunity/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Genetic Predisposition to Disease , Humans , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Polymorphism, Single Nucleotide , Protein Binding/genetics , Signal Transduction/genetics
2.
Nat Immunol ; 18(3): 344-353, 2017 03.
Article in English | MEDLINE | ID: mdl-28114290

ABSTRACT

Although master transcription factors (TFs) are key to the development of specific T cell subsets, whether additional transcriptional regulators are induced by the same stimuli that dominantly repress the development of other, non-specific T cell lineages has not been fully elucidated. Through the use of regulatory T cells (Treg cells) induced by transforming growth factor-ß (TGF-ß), we identified the TF musculin (MSC) as being critical for the development of induced Treg cells (iTreg cells) by repression of the T helper type 2 (TH2) transcriptional program. Loss of MSC reduced expression of the Treg cell master TF Foxp3 and induced TH2 differentiation even under iTreg-cell-differentiation conditions. MSC interrupted binding of the TF GATA-3 to the locus encoding TH2-cell-related cytokines and diminished intrachromosomal interactions within that locus. MSC-deficient (Msc-/-) iTreg cells were unable to suppress TH2 responses, and Msc-/- mice spontaneously developed gut and lung inflammation with age. MSC therefore enforced Foxp3 expression and promoted the unidirectional induction of iTreg cells by repressing the TH2 developmental program.


Subject(s)
Cell Differentiation , Inflammation , Intestinal Mucosa/immunology , Pneumonia/immunology , T-Lymphocytes, Regulatory/physiology , Th2 Cells/physiology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Cells, Cultured , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics , Transcription, Genetic , Transforming Growth Factor beta/metabolism
3.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34353913

ABSTRACT

The type 2 helper effector program is driven by the master transcription factor GATA3 and can be expressed by subsets of both innate lymphoid cells (ILCs) and adaptive CD4+ T helper (Th) cells. While ILC2s and Th2 cells acquire their type 2 differentiation program under very different contexts, the distinct regulatory mechanisms governing this common program are only partially understood. Here we show that the differentiation of ILC2s, and their concomitant high level of GATA3 expression, are controlled by a Gata3 enhancer, Gata3 +674/762, that plays only a minimal role in Th2 cell differentiation. Mice lacking this enhancer exhibited defects in several but not all type 2 inflammatory responses, depending on the respective degree of ILC2 and Th2 cell involvement. Our study provides molecular insights into the different gene regulatory pathways leading to the acquisition of the GATA3-driven type 2 helper effector program in innate and adaptive lymphocytes.


Subject(s)
Enhancer Elements, Genetic , GATA3 Transcription Factor/genetics , Lymphocytes/physiology , Animals , Cell Differentiation/genetics , Female , GATA3 Transcription Factor/metabolism , Homeostasis/genetics , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/physiopathology , Lymphocytes/cytology , Male , Mice, Inbred C57BL , Mice, Transgenic , Strongyloidiasis/parasitology , Strongyloidiasis/physiopathology , Th2 Cells/pathology , Th2 Cells/physiology
4.
Proc Natl Acad Sci U S A ; 117(40): 24620-24626, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32883878

ABSTRACT

The reduced development of COVID-19 for children compared to adults provides some tantalizing clues on the pathogenesis and transmissibility of this pandemic virus. First, ACE2, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor, is reduced in the respiratory tract in children. Second, coronavirus associated with common colds in children may offer some protection, due to cross-reactive humoral immunity and T cell immunity between common coronaviruses and SARS-CoV-2. Third, T helper 2 immune responses are protective in children. Fourth, surprisingly, eosinophilia, associated with T helper 2, may be protective. Fifth, children generally produce lower levels of inflammatory cytokines. Finally, the influence of the downturn in the global economy, the impact of living in quarters among families who are the most at risk, and factors including the openings of some schools, are considered. Those most disadvantaged socioeconomically may suffer disproportionately with COVID-19.


Subject(s)
Betacoronavirus/metabolism , Coronavirus Infections/immunology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Respiratory Mucosa/metabolism , Respiratory Tract Infections/immunology , Angiotensin-Converting Enzyme 2 , COVID-19 , Child , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Host-Pathogen Interactions/immunology , Humans , Pandemics , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Respiratory Tract Infections/virology , SARS-CoV-2 , Th2 Cells/physiology
5.
Lancet ; 395(10221): 371-383, 2020 02 01.
Article in English | MEDLINE | ID: mdl-32007172

ABSTRACT

Asthma is a disease of reversible airflow obstruction characterised clinically by wheezing, shortness of breath, and coughing. Increases in airway type 2 cytokine activity, including interleukin-4 (IL-4), IL-5, and IL-13, are now established biological mechanisms in asthma. Inhaled corticosteroids have been the foundation for asthma treatment, in a large part because they decrease airway type 2 inflammation. However, inhaled or systemic corticosteroids are ineffective treatments in many patients with asthma and few treatment options exist for patients with steroid resistant asthma. Although mechanisms for corticosteroid refractory asthma are likely to be numerous, the development of a new class of biologic agents that target airway type 2 inflammation has provided a new model for treating some patients with corticosteroid refractory asthma. The objective of this Therapeutic paper is to summarise the new type 2 therapeutics, with an emphasis on the biological rationale and clinical efficacy of this new class of asthma therapeutics.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Biological Products/therapeutic use , Adult , Biomarkers/metabolism , Clinical Trials, Phase III as Topic , Cytokines/antagonists & inhibitors , Cytokines/physiology , Eosinophils/physiology , Forecasting , Humans , Indoleacetic Acids/therapeutic use , Interleukin-4/antagonists & inhibitors , Interleukin-5/antagonists & inhibitors , Omalizumab/therapeutic use , Pyridines/therapeutic use , Th2 Cells/physiology , Treatment Outcome
6.
Nat Immunol ; 10(9): 929-32, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19692989

ABSTRACT

The authors recount their discovery of how pathogen-induced interleukin 12 production leads to T(H)1 T cell polarization. Simultaneously they discovered the suppressive cytokine interleukin 10 inhibits antigen-presenting cells, thus regulating development of T(H)1 cells.


Subject(s)
Antigen-Presenting Cells/physiology , Interleukin-10/physiology , Interleukin-12/physiology , Th1 Cells/physiology , Animals , Cell Differentiation , Cell Polarity , Humans , Interferon-gamma/biosynthesis , Macrophages/physiology , Th2 Cells/physiology
7.
Nat Immunol ; 10(9): 992-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19648923

ABSTRACT

The differentiation of activated CD4(+) T cells into the T helper type 1 (T(H)1) or T(H)2 fate is regulated by cytokines and the transcription factors T-bet and GATA-3. Whereas interleukin 12 (IL-12) produced by antigen-presenting cells initiates the T(H)1 fate, signals that initiate the T(H)2 fate are not completely characterized. Here we show that early GATA-3 expression, required for T(H)2 differentiation, was induced by T cell factor 1 (TCF-1) and its cofactor beta-catenin, mainly from the proximal Gata3 promoter upstream of exon 1b. This activity was induced after T cell antigen receptor (TCR) stimulation and was independent of IL-4 receptor signaling through the transcription factor STAT6. Furthermore, TCF-1 blocked T(H)1 fate by negatively regulating interferon-gamma (IFN-gamma) expression independently of beta-catenin. Thus, TCF-1 initiates T(H)2 differentiation of activated CD4(+) T cells by promoting GATA-3 expression and suppressing IFN-gamma expression.


Subject(s)
GATA3 Transcription Factor/genetics , Interferon-gamma/biosynthesis , T Cell Transcription Factor 1/physiology , Th2 Cells/physiology , Animals , Cell Differentiation , Interleukin-12/biosynthesis , Interleukin-4/physiology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/physiology , Receptors, Notch/physiology , beta Catenin/physiology
8.
Inflamm Res ; 70(5): 539-541, 2021 May.
Article in English | MEDLINE | ID: mdl-33811487

ABSTRACT

OBJECTIVE: Histamine derived from mast cells and basophils plays important roles in inducing allergic symptoms. Although T cells also produce histamine, the involvement of the histamine produced from T cells has remained enigmatic. We sought to reveal the roles of T helper 2 (Th2) cell-derived histamine in nasal allergic disorders. METHODS: The histamine production from Th2 cells was measured by EIA. The mRNA expression of histidine decarboxylase (HDC) was measured by real-time PCR. To investigate the roles of Th2 cell-derived histamine in vivo, we analyzed an antigen-specific Th2 cell transfer mouse model. RESULTS: Th2 cells produced histamine by T cell receptor stimulation, and these properties were specific for Th2 cells, but not Th1 cells and naïve CD4 T cells. The histamine produced from Th2 cells was involved in the infiltrations of Th2 cells in response to antigen exposure. CONCLUSION: These results suggest that Th2 cell-derived histamine play important roles in nasal allergic disorders.


Subject(s)
Histamine/immunology , Nasal Mucosa/immunology , Rhinitis, Allergic/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Cell Movement , Histidine Decarboxylase/genetics , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Th2 Cells/physiology , Th2 Cells/transplantation
9.
J Immunol ; 200(5): 1865-1875, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29374078

ABSTRACT

IL-10 is a pleiotropic cytokine with multifaceted functions in establishing immune homeostasis. Although expressed by Th1 and Th2 cells, conventional Th1 cells produce marginal levels of IL-10 compared with their Th2 counterparts. In this study, we investigated the epigenetic mechanisms of Il-10 gene expression in Th1 cells. Bioinformatics EMBOSS CpG plot analysis and bisulfite pyrosequencing revealed three CpG DNA methylation sites in the Il-10 gene locus. Progressive DNA methylation at all of the CpG regions of interest (ROIs) established a repressive program of Il-10 gene expression in Th1 cells. Interestingly, Th1 cells treated with IL-12 and IL-27 cytokines, thereby mimicking a chronic inflammatory condition in vivo, displayed a significant increase in IL-10 production that was accompanied by selective DNA demethylation at ROI 3 located in intron 3. IL-10-producing T cells isolated from lymphocytic choriomeningitis virus-infected mice also showed enhanced DNA demethylation at ROI 3. Binding of STAT1 and STAT3 to demethylated ROI 3 enhanced IL-10 expression in an IL-12/IL-27-dependent manner. Accordingly, CD4+ T cells isolated from STAT1- or STAT3-knockout mice were significantly defective in IL-10 production. Our data suggest that, although stably maintained DNA methylation at the promoter may repress IL-10 expression in Th1 cells, locus-specific reversible DNA demethylation may serve as a threshold platform to control transient Il-10 gene expression.


Subject(s)
DNA Methylation/genetics , Interleukin-10/genetics , Th1 Cells/physiology , Animals , CD4-Positive T-Lymphocytes/physiology , Cell Line , CpG Islands/genetics , Epigenesis, Genetic/genetics , HEK293 Cells , Humans , Interleukin-27/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/genetics , Th2 Cells/physiology
10.
Int J Neurosci ; 130(8): 788-803, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31906749

ABSTRACT

Purpose of the study: Multiple sclerosis is a CD4+ T cell mediated autoimmune disease characterized by inflammatory demyelination in the central nervous system. Acetylcholine (ACh) has been reported to be released by T lymphocytes and plays as an inflammation and immune regulator through the participation of T cells. However, both attenuated and aggravated effects of ACh in inflammation were found. The aim of this study is to further investigate the role of ACh in experimental autoimmune encephalomyelitis (EAE).Materials and methods: The left cervical vagotomy was performed to inhibit ACh release with the sham-operation as control. ACh in cerebral cortex and splenocytes culture supernatants of EAE mice were determined. Interleukin-6, interferon-γ, interleukin-4 and interleukin-17A in brain and splenocytes culture supernatants were evaluated by enzyme-linked immunosorbent assay. The proportion of CD4+ T cells and subsets were assessed by flow cytometry.Results: Compared with the sham-operation group, improved clinical and pathological parameters as well as decreased interleukin-6, interferon-γ, interleukin-4 and interleukin-17A were found in EAE mice with vagotomy suppressing the ACh. Marked reductions of CD4+ and CD4+ChAT+ cells, as well as significant decrease in Th1 with a bias to Th2 in Th1/Th2 balance and increased ChAT+Th2 proportion in the spleen were also observed in vagotomized mice.Conclusions: These findings emphasize that inhibiting ACh release by vagotomy can ameliorate the exacerbation of EAE through suppressing CD4+ T cells proliferation and regulating the differentiation of Th1, Th2 and Th17.


Subject(s)
Acetylcholine/physiology , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , T Follicular Helper Cells/physiology , Acetylcholine/metabolism , Animals , Cell Culture Techniques , Cerebral Cortex/metabolism , Mice , Spleen/metabolism , Th1 Cells/physiology , Th17 Cells/physiology , Th2 Cells/physiology , Vagotomy
11.
J Allergy Clin Immunol ; 144(5): 1143-1148, 2019 11.
Article in English | MEDLINE | ID: mdl-31703761

ABSTRACT

Technical advances in single-cell RNA sequencing (scRNA-seq) render it possible to examine the transcriptomes of single cells in patients with allergic inflammation with high resolution in the context of their specific microenvironment, treatment, and disease status. Using a recently published scRNA-seq study of tissue T cells as an example, we introduce the major pipeline steps, illustrate the options of scRNA-seq platforms, summarize new knowledge gained from this study, and provide directions for future research. The presented scRNA-seq study elucidated the T-cell heterogeneity present in an allergic inflammatory tissue focused on eosinophilic esophagitis, a prototypic, chronic, allergic disease, which provided a unique opportunity to probe the pathogenesis of allergic inflammation at the tissue level through readily available endoscopically procured biopsy specimens. scRNA-seq analysis identified 8 populations of CD3+ T cells and defined 2 disease-specific populations of CD3+CD4+ T cells, including a markedly activated type 2 cytokine-producing pathogenic cell population distinguished by expression of the short-chain fatty acid receptor free fatty acid receptor 3 and a population of regulatory T cells. In addition to presenting and interpreting new findings within the prior literature, we postulate about future single-cell next-generation sequencing platforms in this burgeoning field.


Subject(s)
Eosinophilic Esophagitis/immunology , Hypersensitivity/immunology , Inflammation/immunology , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory/physiology , Th2 Cells/physiology , Animals , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, RNA
12.
Int Arch Allergy Immunol ; 178(3): 281-290, 2019.
Article in English | MEDLINE | ID: mdl-30763933

ABSTRACT

BACKGROUND: TNF-TNFR2 signaling has been indicated to be involved in CD4+ T lymphocyte differentiation. However, its role in allergic airway inflammation is not well understood. OBJECTIVES: The aim of this study was to investigate the role of TNF-TNFR2 signaling in allergic airway inflammation. METHODS AND RESULTS: In this study, we used an allergen-induced asthma model to show that TNF-TNFR2 signaling alleviated allergic airway inflammation by reducing the airway infiltration of eosinophils and neutrophils. Activated TNF-TNFR2 signaling decreased the expression of Th2 and Th17 cytokines in serum and bronchoalveolar lavage fluid. Furthermore, TNF-TNFR2 signaling inhibited Th2 and Th17 polarization but promoted Th1 and CD4+CD25+ T cell differentiation in vivo. CONCLUSIONS: Our study indicates that TNF-TNFR2 signaling alleviates allergic airway inflammation through inhibition of Th2 and Th17 cell differentiation.


Subject(s)
Asthma/etiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Signal Transduction/physiology , Th17 Cells/physiology , Th2 Cells/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Polarity , Female , Mice , Mice, Inbred BALB C , Th17 Cells/immunology , Th2 Cells/immunology
13.
J Immunol ; 198(5): 2165-2171, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28100679

ABSTRACT

IL-10 is an immunoregulatory cytokine that has broad effects across the immune system. In Th cell subsets, Th2 cells produce considerable amounts of IL-10. The transcription factors that regulate IL-10 production in Th2 cells are still incompletely described. In this study, we demonstrate that the ETS family transcription factor ETS variant (Etv)5 regulates IL-10 production in Th2 cells. T cell-specific Etv5-deficient and littermate control mice demonstrated that IL-10 production and gene expression were significantly decreased in the absence of Etv5. In an Aspergillus fumigatus extract-induced inflammation model, IL-10-producing CD4+ T cells in bronchoalveolar lavage and lung were significantly decreased in mice that lacked Etv5 in T cells, compared with control mice. We showed that Etv5 directly binds to the Il10 locus conserved noncoding sequence 3 site and that it activates gene expression in a luciferase reporter assay and following retroviral transduction. Etv5 deficiency did not affect the expression of other transcription factors known to be important for expression of IL-10, including Jun family members, GATA3, E4BP4, and IFN regulatory factor 4. However, in the absence of Etv5, binding of these transcription factors to the Il10 locus was dramatically reduced. Ectopic Etv5 expression in Th2 cells that lack Etv5 restored IL-10 production and the binding of IL-10-inducing transcription factors including E4BP4, IFN regulatory factor 4, and GATA3. Taken together, we conclude that Etv5 plays a crucial role in regulating IL-10 production in Th2 cells by facilitating the binding of IL-10-inducing transcription factors at the Il10 locus.


Subject(s)
DNA-Binding Proteins/metabolism , Hypersensitivity/immunology , Interleukin-10/metabolism , Th2 Cells/immunology , Th2 Cells/physiology , Transcription Factors/metabolism , Allergens/immunology , Animals , Antigens, Fungal/immunology , Aspergillus fumigatus/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Interleukin-10/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Protein Binding , Transcription Factors/genetics
14.
J Immunol ; 198(10): 3927-3938, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28424240

ABSTRACT

Histone acetyltransferases (HATs) regulate inducible transcription in multiple cellular processes and during inflammatory and immune response. However, the functions of general control nonrepressed-protein 5 (Gcn5), an evolutionarily conserved HAT from yeast to human, in immune regulation remain unappreciated. In this study, we conditionally deleted Gcn5 (encoded by the Kat2a gene) specifically in T lymphocytes by crossing floxed Gcn5 and Lck-Cre mice, and demonstrated that Gcn5 plays important roles in multiple stages of T cell functions including development, clonal expansion, and differentiation. Loss of Gcn5 functions impaired T cell proliferation, IL-2 production, and Th1/Th17, but not Th2 and regulatory T cell differentiation. Gcn5 is recruited onto the il-2 promoter by interacting with the NFAT in T cells upon TCR stimulation. Interestingly, instead of directly acetylating NFAT, Gcn5 catalyzes histone H3 lysine H9 acetylation to promote IL-2 production. T cell-specific suppression of Gcn5 partially protected mice from myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an experimental model for human multiple sclerosis. Our study reveals previously unknown physiological functions for Gcn5 and a molecular mechanism underlying these functions in regulating T cell immunity. Hence Gcn5 may be an important new target for autoimmune disease therapy.


Subject(s)
Histone Acetyltransferases/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Gene Expression Regulation , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Interleukin-2/deficiency , Interleukin-2/genetics , Interleukin-2/immunology , Mice , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th1 Cells/physiology , Th2 Cells/immunology , Th2 Cells/physiology
15.
J Allergy Clin Immunol ; 142(2): 415-423, 2018 08.
Article in English | MEDLINE | ID: mdl-29103994

ABSTRACT

BACKGROUND: Various clinical, biologic, or physiologic markers of asthma have been used to identify patient clusters and potential targets for therapy. However, these identifiers frequently overlap among the different asthma groups. For instance, both eosinophil and neutrophil counts are often increased in the airways of asthmatic patients despite their typical association with type 2 and type 17 immune response, respectively. OBJECTIVES: We sought to determine whether inflammatory gene expression is related to patterns of airway inflammation and lung function and identify molecular markers for neutrophilic asthma. METHODS: Expression levels of 17 genes characterizing type 1, type 2, and type 17 lymphocytes were measured in sputum samples from 48 participants with asthma. The relationships between gene expression levels and sputum cell differentials or measures of pulmonary function were examined by using partial least squares regression. RESULTS: Gene expression levels were strongly associated with cell differentials, explaining 71% of variation in eosinophil counts and 64% of variation in neutrophil counts. The 3 genes with the strongest relationships to sputum neutrophil counts were IL1R1 (standardized regression coefficient [ß] = +0.27, P = .005), IL1RAP (ß = +0.32, P = .0004), and IL4R (ß = +0.29, P = .002). Higher expression levels of IL1R1, IL1RAP, and IL4R were associated with reduced FEV1/forced vital capacity ratio (ß = -0.11, -0.08, and -0.10; P = .005, .07, and .05). CONCLUSION: IL-1 receptor appears to be a marker of neutrophilic inflammation and airflow obstruction in patients with asthma, who have a wide range of disease severity. The IL-1 pathway might contribute to airway neutrophilia and is a potential therapeutic target in patients with neutrophilic asthma.


Subject(s)
Asthma/genetics , Neutrophils/physiology , Receptors, Interleukin-1/metabolism , Sputum/metabolism , Th1 Cells/physiology , Th17 Cells/physiology , Th2 Cells/physiology , Adult , Asthma/immunology , Biomarkers/metabolism , Eosinophils , Female , Gene Expression Regulation , Humans , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-4 Receptor alpha Subunit/genetics , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive , Receptors, Interleukin-1/genetics , Spirometry , Young Adult
16.
Int J Mol Sci ; 20(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627390

ABSTRACT

Mast cells (MC) are resident tissue cells found primarily at the interphase between tissues and the environment. These evolutionary old cells store large amounts of proteases within cytoplasmic granules, and one of the most abundant of these proteases is tryptase. To look deeper into the question of their in vivo targets, we have analyzed the activity of the human MC tryptase on 69 different human cytokines and chemokines, and the activity of the mouse tryptase (mMCP-6) on 56 mouse cytokines and chemokines. These enzymes were found to be remarkably restrictive in their cleavage of these potential targets. Only five were efficiently cleaved by the human tryptase: TSLP, IL-21, MCP3, MIP-3b, and eotaxin. This strict specificity indicates a regulatory function of these proteases and not primarily as unspecific degrading enzymes. We recently showed that the human MC chymase also had a relatively strict specificity, indicating that both of these proteases have regulatory functions. One of the most interesting regulatory functions may involve controlling excessive TH2-mediated inflammation by cleaving several of the most important TH2-promoting inflammatory cytokines, including IL-18, IL-33, TSLP, IL-15, and IL-21, indicating a potent negative feedback loop on TH2 immunity.


Subject(s)
Mast Cells/physiology , Th2 Cells/immunology , Tryptases/physiology , Animals , Catalytic Domain , Chemokines/metabolism , Cytokines/metabolism , Feedback, Physiological , Humans , Mice , Th2 Cells/physiology , Tryptases/genetics , Tryptases/metabolism
17.
Eur J Immunol ; 47(5): 841-847, 2017 05.
Article in English | MEDLINE | ID: mdl-28271497

ABSTRACT

Schistosomiasis is a nontransplacental helminth infection. Chronic infection during pregnancy suppresses allergic airway responses in offspring. We addressed the question whether in utero exposure to chronic schistosome infection (Reg phase) in mice affects B-cell and T-cell development. Therefore, we focused our analyses on T-cell differentiation capacity induced by epigenetic changes in promoter regions of signature cytokines in offspring. Here, we show that naïve T cells from offspring of schistosome infected female mice had a strong capacity to differentiate into TH 1 cells, whereas TH 2 differentiation was impaired. In accordance, reduced levels of histone acetylation of the IL-4 promoter regions were observed in naïve T cells. To conclude, our mouse model revealed distinct epigenetic changes within the naïve T-cell compartment affecting TH 2 and TH 1 cell differentiation in offspring of mothers with chronic helminth infection. These findings could eventually help understand how helminths alter T-cell driven immune responses induced by allergens, bacterial or viral infections, as well as vaccines.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Lymphocyte Activation , Pregnancy Complications, Parasitic/immunology , Schistosomiasis/immunology , T-Lymphocytes/physiology , Acetylation , Animals , Chronic Disease , Cytokines/genetics , Cytokines/immunology , Female , Histones/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mothers , Pregnancy , Promoter Regions, Genetic , Schistosomiasis/parasitology , T-Lymphocytes/immunology , Th1 Cells/immunology , Th1 Cells/physiology , Th2 Cells/immunology , Th2 Cells/physiology
18.
Allergy ; 73(12): 2342-2351, 2018 12.
Article in English | MEDLINE | ID: mdl-29777594

ABSTRACT

BACKGROUND: Basophils are commonly associated with allergic responses because of their ability to produce large amounts of pro-Th2 cytokines and histamine. However, the mechanisms through which bone marrow-resident basophils (BMRB) become fully competent cytokine and histamine producers in response to IgE crosslinking are poorly understood. Here, we sought to determine the role of IL-3 in promoting pro-Th2 basophils. METHODS: BMRB and basophils exposed to IL-3 in vitro and in vivo were evaluated for their production of Th2 cytokines and histamine in response to FcεRI crosslinking on both protein and gene expression levels. In vivo relevance of our findings was assessed in a model of ovalbumin-induced allergic asthma using IL-3-deficient and wild-type mice in a protocol of adoptive basophil transfer. RESULTS: We show that BMRB and basophils previously exposed to IL-3 differ in their ability to generate cytokines (IL-4, IL-6, IL-13, and GM-CSF) and histamine in response to FcεRI crosslinking, reflecting two stages of maturation. Exposure to IL-3 initiated an autocrine loop of endogenous IL-3 production that enhanced histamine and cytokine production upon FcεRI crosslinking. This increased responsiveness required calcium flux and was dependent on calcineurin and store-operated calcium channels. Our findings are of pathophysiological relevance, as assessed by the failure of IL-3-deficient mice to develop airway hyperreactivity, which could be restored by adoptive transfer of IL-3-derived basophils recovered from wild-type mice. CONCLUSION: IL-3-dependent basophils promote Th2 allergic AHR, which designates the IL-3/basophil axis as a promising therapeutic target for the treatment of basophil-dependent asthma.


Subject(s)
Interleukin-3/immunology , Respiratory Hypersensitivity/etiology , Animals , Basophils , Bone Marrow Cells , Cytokines/metabolism , Histamine/metabolism , Inflammation/pathology , Mice , Respiratory Hypersensitivity/pathology , Th2 Cells/immunology , Th2 Cells/physiology
19.
FASEB J ; 31(2): 519-525, 2017 02.
Article in English | MEDLINE | ID: mdl-27756768

ABSTRACT

In this study, we strove to substantiate the ability of linc-MAF-4 to act as a regulator of pathogenesis during multiple sclerosis (MS). We recruited 34 patients who were diagnosed with MS according to the revised McDonald criteria. Six patients with MS and 5 healthy volunteers contributed peripheral blood mononuclear cells for microarray analysis. Subsequent knockdown and overexpression of linc-MAF-4 in naive CD4+ T cells from the additional 28 patients with MS was performed to track changes in CD4+ T-cell subsets and their function, as well as to confirm results from the prior microarray analysis. Expression of linc-MAF-4 increased significantly in peripheral blood mononuclear cells of patients with MS compared with those of control participants. In addition, linc-MAF-4 regulated encephalitogenic T helper (Th)1-cell differentiation in patients with MS. Transfection of synthetic linc-MAF-4 into naive CD4+ T cells facilitated Th1-cell differentiation and inhibited Th2-cell differentiation by directly inhibiting MAF, which is a Th2-cell transcription factor. Linc-MAF-4 also promoted activation of CD4+ T cells from patients with MS. Expression level of linc-MAF-4 correlated with the annual relapse rate in patients with MS. Our results suggest that linc-MAF-4 is involved in the pathogenesis of MS, specifically via regulation of encephalitogenic T cells.-Zhang, F., Liu, G., Wei, C., Gao, C., Hao, J. Linc-MAF-4 regulates Th1/Th2 differentiation and is associated with the pathogenesis of multiple sclerosis by targeting MAF.


Subject(s)
Gene Expression Regulation/physiology , Multiple Sclerosis/metabolism , RNA, Long Noncoding/metabolism , Th1 Cells/physiology , Th2 Cells/physiology , Adolescent , Adult , Down-Regulation/physiology , Female , Humans , Long Interspersed Nucleotide Elements , Male , Middle Aged , Multiple Sclerosis/genetics , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/genetics , Up-Regulation , Young Adult
20.
Curr Opin Pulm Med ; 24(1): 50-55, 2018 01.
Article in English | MEDLINE | ID: mdl-29036019

ABSTRACT

PURPOSE OF REVIEW: Severe asthma is a heterogeneous disease that can be classified into phenotypes and endotypes based upon clinical or biological characteristics. Interleukin (IL)-4 and IL-13 play a key role in type 2 (T2) asthma. This article reviews the signaling pathway of IL-4 and IL-13 and highlights its targeted therapy in severe asthma. RECENT FINDINGS: Several clinical trials of biologics targeting the IL-4/IL-13 pathway have recently been completed. In patients with severe, uncontrolled asthma, targeting IL-13 alone with biologics including lebrikizumab and tralokinumab has not shown consistent reduction in asthma exacerbations. Simultaneous targeting of both IL-4 and IL-13 by blocking IL-4 receptor α using dupilumab has yielded more consistent results in reducing asthma exacerbations and improving lung function, especially in patients with increased blood eosinophils. Other biomarkers of T2 inflammation such as exhaled nitric oxide and serum periostin may also predict response to biologics targeting the IL-4/IL-13 pathway. SUMMARY: No biologic targeting the IL-4/IL-13 pathway is currently available for treatment of asthma, but emerging data suggest that biologics targeting IL-4 and IL-13 together may benefit patients with T2 high asthma. Additional data are needed about long-term efficacy and safety prior to incorporating these drugs into routine clinical practice.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Molecular Targeted Therapy , Precision Medicine , Receptors, Interleukin-13/antagonists & inhibitors , Receptors, Interleukin-4/antagonists & inhibitors , Signal Transduction/drug effects , Asthma/immunology , Asthma/physiopathology , Biomarkers/analysis , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Phenotype , Severity of Illness Index , Th2 Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL