Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.043
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 705-725, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340571

ABSTRACT

The discovery of CD4+ T cell subset-defining master transcription factors and framing of the Th1/Th2 paradigm ignited the CD4+ T cell field. Advances in in vivo experimental systems, however, have revealed that more complex lineage-defining transcriptional networks direct CD4+ T cell differentiation in the lymphoid organs and tissues. This review focuses on the layers of fate decisions that inform CD4+ T cell differentiation in vivo. Cytokine production by antigen-presenting cells and other innate cells influences the CD4+ T cell effector program [e.g., T helper type 1 (Th1), Th2, Th17]. Signals downstream of the T cell receptor influence whether individual clones bearing hallmarks of this effector program become T follicular helper cells, supporting development of B cells expressing specific antibody isotypes, or T effector cells, which activate microbicidal innate cells in tissues. These bifurcated, parallel axes allow CD4+ T cells to augment their particular effector program and prevent disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cytokines/metabolism , Humans , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
2.
Cell ; 176(4): 882-896.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30639098

ABSTRACT

T helper type 2 (Th2) cells are important regulators of mammalian adaptive immunity and have relevance for infection, autoimmunity, and tumor immunology. Using a newly developed, genome-wide retroviral CRISPR knockout (KO) library, combined with RNA-seq, ATAC-seq, and ChIP-seq, we have dissected the regulatory circuitry governing activation and differentiation of these cells. Our experiments distinguish cell activation versus differentiation in a quantitative framework. We demonstrate that these two processes are tightly coupled and are jointly controlled by many transcription factors, metabolic genes, and cytokine/receptor pairs. There are only a small number of genes regulating differentiation without any role in activation. By combining biochemical and genetic data, we provide an atlas for Th2 differentiation, validating known regulators and identifying factors, such as Pparg and Bhlhe40, as part of the core regulatory network governing Th2 helper cell fates.


Subject(s)
Receptor Cross-Talk/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genome , High-Throughput Nucleotide Sequencing , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/metabolism
3.
Nat Immunol ; 22(11): 1367-1374, 2021 11.
Article in English | MEDLINE | ID: mdl-34686862

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) represent innate homologs of type 2 helper T cells (TH2) that participate in immune defense and tissue homeostasis through production of type 2 cytokines. While T lymphocytes metabolically adapt to microenvironmental changes, knowledge of human ILC2 metabolism is limited, and its key regulators are unknown. Here, we show that circulating 'naive' ILC2s have an unexpected metabolic profile with a higher level of oxidative phosphorylation (OXPHOS) than natural killer (NK) cells. Accordingly, ILC2s are severely reduced in individuals with mitochondrial disease (MD) and impaired OXPHOS. Metabolomic and nutrient receptor analysis revealed ILC2 uptake of amino acids to sustain OXPHOS at steady state. Following activation with interleukin-33 (IL-33), ILC2s became highly proliferative, relying on glycolysis and mammalian target of rapamycin (mTOR) to produce IL-13 while continuing to fuel OXPHOS with amino acids to maintain cellular fitness and proliferation. Our results suggest that proliferation and function are metabolically uncoupled in human ILC2s, offering new strategies to target ILC2s in disease settings.


Subject(s)
Cell Proliferation , Cytokines/metabolism , Energy Metabolism , Immunity, Innate , Lymphocyte Activation , Mitochondrial Diseases/metabolism , Th2 Cells/metabolism , Amino Acids, Branched-Chain/metabolism , Arginine/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Energy Metabolism/drug effects , Humans , Immunity, Innate/drug effects , Interleukin-33/pharmacology , Lymphocyte Activation/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/immunology , Phenotype , Th2 Cells/drug effects , Th2 Cells/immunology
4.
Nat Immunol ; 22(12): 1538-1550, 2021 12.
Article in English | MEDLINE | ID: mdl-34795444

ABSTRACT

The signals driving the adaptation of type 2 dendritic cells (DC2s) to diverse peripheral environments remain mostly undefined. We show that differentiation of CD11blo migratory DC2s-a DC2 population unique to the dermis-required IL-13 signaling dependent on the transcription factors STAT6 and KLF4, whereas DC2s in lung and small intestine were STAT6-independent. Similarly, human DC2s in skin expressed an IL-4 and IL-13 gene signature that was not found in blood, spleen and lung DCs. In mice, IL-13 was secreted homeostatically by dermal innate lymphoid cells and was independent of microbiota, TSLP or IL-33. In the absence of IL-13 signaling, dermal DC2s were stable in number but remained CD11bhi and showed defective activation in response to allergens, with diminished ability to support the development of IL-4+GATA3+ helper T cells (TH), whereas antifungal IL-17+RORγt+ TH cells were increased. Therefore, homeostatic IL-13 fosters a noninflammatory skin environment that supports allergic sensitization.


Subject(s)
Cell Communication , Cell Differentiation , Interleukin-13/metabolism , Langerhans Cells/metabolism , Skin/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Allergens/pharmacology , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cells, Cultured , Databases, Genetic , Humans , Interleukin-13/genetics , Langerhans Cells/drug effects , Langerhans Cells/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction , Skin/cytology , Skin/drug effects , Skin/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Transcriptome
5.
Nat Immunol ; 21(12): 1528-1539, 2020 12.
Article in English | MEDLINE | ID: mdl-33020661

ABSTRACT

Mutations that impact immune cell migration and result in immune deficiency illustrate the importance of cell movement in host defense. In humans, loss-of-function mutations in DOCK8, a guanine exchange factor involved in hematopoietic cell migration, lead to immunodeficiency and, paradoxically, allergic disease. Here, we demonstrate that, like humans, Dock8-/- mice have a profound type 2 CD4+ helper T (TH2) cell bias upon pulmonary infection with Cryptococcus neoformans and other non-TH2 stimuli. We found that recruited Dock8-/-CX3CR1+ mononuclear phagocytes are exquisitely sensitive to migration-induced cell shattering, releasing interleukin (IL)-1ß that drives granulocyte-macrophage colony-stimulating factor (GM-CSF) production by CD4+ T cells. Blocking IL-1ß, GM-CSF or caspase activation eliminated the type-2 skew in mice lacking Dock8. Notably, treatment of infected wild-type mice with apoptotic cells significantly increased GM-CSF production and TH2 cell differentiation. This reveals an important role for cell death in driving type 2 signals during infection, which may have implications for understanding the etiology of type 2 CD4+ T cell responses in allergic disease.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Biomarkers , Caspases/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Susceptibility , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Signal Transduction
6.
Nat Immunol ; 21(6): 671-683, 2020 06.
Article in English | MEDLINE | ID: mdl-32424366

ABSTRACT

Urinary tract infections (UTIs) typically evoke prompt and vigorous innate bladder immune responses, including extensive exfoliation of the epithelium. To explain the basis for the extraordinarily high recurrence rates of UTIs, we examined adaptive immune responses in mouse bladders. We found that, following each bladder infection, a highly T helper type 2 (TH2)-skewed immune response directed at bladder re-epithelialization is observed, with limited capacity to clear infection. This response is initiated by a distinct subset of CD301b+OX40L+ dendritic cells, which migrate into the bladder epithelium after infection before trafficking to lymph nodes to preferentially activate TH2 cells. The bladder epithelial repair response is cumulative and aberrant as, after multiple infections, the epithelium was markedly thickened and bladder capacity was reduced relative to controls. Thus, recurrence of UTIs and associated bladder dysfunction are the outcome of the preferential focus of the adaptive immune response on epithelial repair at the expense of bacterial clearance.


Subject(s)
Cystitis/etiology , Cystitis/metabolism , Lymphocyte Activation/immunology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Bacterial Load , Biomarkers , Cell Line , Cystitis/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Mice , Mice, Knockout , Mucous Membrane/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Urinary Tract Infections/etiology , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Wound Healing/genetics , Wound Healing/immunology
7.
Immunity ; 55(2): 254-271.e7, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139352

ABSTRACT

Allergic immunity is orchestrated by group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells prominently arrayed at epithelial- and microbial-rich barriers. However, ILC2s and Th2 cells are also present in fibroblast-rich niches within the adventitial layer of larger vessels and similar boundary structures in sterile deep tissues, and it remains unclear whether they undergo dynamic repositioning during immune perturbations. Here, we used thick-section quantitative imaging to show that allergic inflammation drives invasion of lung and liver non-adventitial parenchyma by ILC2s and Th2 cells. However, during concurrent type 1 and type 2 mixed inflammation, IFNγ from broadly distributed type 1 lymphocytes directly blocked both ILC2 parenchymal trafficking and subsequent cell survival. ILC2 and Th2 cell confinement to adventitia limited mortality by the type 1 pathogen Listeria monocytogenes. Our results suggest that the topography of tissue lymphocyte subsets is tightly regulated to promote appropriately timed and balanced immunity.


Subject(s)
Inflammation/immunology , Interferon-gamma/immunology , Lymphocyte Subsets/immunology , Th2 Cells/immunology , Animals , Cell Death/immunology , Cell Movement/immunology , Hypersensitivity/immunology , Immunity, Innate , Interleukin-33/immunology , Interleukin-5/metabolism , Listeria monocytogenes , Listeriosis/immunology , Listeriosis/mortality , Liver/immunology , Lung/immunology , Lymphocyte Subsets/metabolism , Lysophospholipids/immunology , Mice , Parenchymal Tissue/immunology , Sphingosine/analogs & derivatives , Sphingosine/immunology , Th1 Cells/immunology , Th2 Cells/metabolism
8.
Cell ; 164(1-2): 141-155, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26774822

ABSTRACT

The DENN domain is an evolutionary conserved protein module found in all eukaryotes and serves as an exchange factor for Rab-GTPases to regulate diverse cellular functions. Variants in DENND1B are associated with development of childhood asthma and other immune disorders. To understand how DENND1B may contribute to human disease, Dennd1b(-/-) mice were generated and exhibit hyper-allergic responses following antigen challenge. Dennd1b(-/-) TH2, but not other TH cells, exhibit delayed receptor-induced T cell receptor (TCR) downmodulation, enhanced TCR signaling, and increased production of effector cytokines. As DENND1B interacts with AP-2 and Rab35, TH2 cells deficient in AP-2 or Rab35 also exhibit enhanced TCR-mediated effector functions. Moreover, human TH2 cells carrying asthma-associated DENND1B variants express less DENND1B and phenocopy Dennd1b(-/-) TH2 cells. These results provide a molecular basis for how DENND1B, a previously unrecognized regulator of TCR downmodulation in TH2 cells, contributes to asthma pathogenesis and how DENN-domain-containing proteins may contribute to other human disorders.


Subject(s)
Asthma/immunology , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Th2 Cells/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Female , Guanine Nucleotide Exchange Factors/genetics , Humans , Hypersensitivity/immunology , Lymphocyte Activation , Mice , Polymorphism, Single Nucleotide , Th2 Cells/metabolism , rab GTP-Binding Proteins/genetics
9.
Immunity ; 54(11): 2595-2610.e7, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34506733

ABSTRACT

Fungal airway infection (airway mycosis) is an important cause of allergic airway diseases such as asthma, but the mechanisms by which fungi trigger asthmatic reactions are poorly understood. Here, we leverage wild-type and mutant Candida albicans to determine how this common fungus elicits characteristic Th2 and Th17 cell-dependent allergic airway disease in mice. We demonstrate that rather than proteinases that are essential virulence factors for molds, C. albicans instead promoted allergic airway disease through the peptide toxin candidalysin. Candidalysin activated platelets through the Von Willebrand factor (VWF) receptor GP1bα to release the Wnt antagonist Dickkopf-1 (Dkk-1) to drive Th2 and Th17 cell responses that correlated with reduced lung fungal burdens. Platelets simultaneously precluded lethal pulmonary hemorrhage resulting from fungal lung invasion. Thus, in addition to hemostasis, platelets promoted protection against C. albicans airway mycosis through an antifungal pathway involving candidalysin, GP1bα, and Dkk-1 that promotes Th2 and Th17 responses.


Subject(s)
Blood Platelets/immunology , Candida albicans/physiology , Candidiasis/complications , Candidiasis/immunology , Disease Susceptibility , Host-Pathogen Interactions/immunology , Hypersensitivity/complications , Hypersensitivity/immunology , T-Lymphocyte Subsets/immunology , Blood Platelets/metabolism , Hypersensitivity/metabolism , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
10.
Nat Immunol ; 18(10): 1160-1172, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28783152

ABSTRACT

Regulatory T cells (Treg cells) perform two distinct functions: they maintain self-tolerance, and they support organ homeostasis by differentiating into specialized tissue Treg cells. We found that epigenetic modifications defined the molecular characteristics of tissue Treg cells. Tagmentation-based whole-genome bisulfite sequencing revealed more than 11,000 regions that were methylated differentially in pairwise comparisons of tissue Treg cell populations and lymphoid T cells. Similarities in the epigenetic landscape led to the identification of a common tissue Treg cell population that was present in many organs and was characterized by gain and loss of DNA methylation that included many gene sites associated with the TH2 subset of helper T cells, such as the gene encoding cytokine IL-33 receptor ST2, as well as the production of tissue-regenerative factors. Furthermore, the ST2-expressing population was dependent on the transcriptional regulator BATF and could be expanded by IL-33. Thus, tissue Treg cells integrate multiple waves of epigenetic reprogramming that define their tissue-restricted specialization.


Subject(s)
DNA Methylation , Genome-Wide Association Study , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cluster Analysis , Computational Biology/methods , CpG Islands , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Transgenic , Molecular Sequence Annotation , Organ Specificity/genetics , Organ Specificity/immunology , Promoter Regions, Genetic , Th2 Cells/metabolism , Transcription Initiation Site , Transcriptome
11.
Immunity ; 50(2): 362-377.e6, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30709738

ABSTRACT

Regulatory T (Treg) cells maintain immune tolerance through the master transcription factor forkhead box P3 (FOXP3), which is crucial for Treg cell function and homeostasis. We identified an IPEX (immune dysregulation polyendocrinopathy enteropathy X-linked) syndrome patient with a FOXP3 mutation in the domain swap interface of the protein. Recapitulation of this Foxp3 variant in mice led to the development of an autoimmune syndrome consistent with an unrestrained T helper type 2 (Th2) immune response. Genomic analysis of Treg cells by RNA-sequencing, Foxp3 chromatin immunoprecipitation followed by high-throughput DNA sequencing (ChIP-sequencing), and H3K27ac-HiChIP revealed a specific de-repression of the Th2 transcriptional program leading to the generation of Th2-like Treg cells that were unable to suppress extrinsic Th2 cells. Th2-like Treg cells showed increased intra-chromosomal interactions in the Th2 locus, leading to type 2 cytokine production. These findings identify a direct role for Foxp3 in suppressing Th2-like Treg cells and implicate additional pathways that could be targeted to restrain Th2 trans-differentiated Treg cells.


Subject(s)
Forkhead Transcription Factors/immunology , Mutation , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Child , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/metabolism , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism
12.
Nature ; 604(7905): 337-342, 2022 04.
Article in English | MEDLINE | ID: mdl-35355021

ABSTRACT

Decades of work have elucidated cytokine signalling and transcriptional pathways that control T cell differentiation and have led the way to targeted biologic therapies that are effective in a range of autoimmune, allergic and inflammatory diseases. Recent evidence indicates that obesity and metabolic disease can also influence the immune system1-7, although the mechanisms and effects on immunotherapy outcomes remain largely unknown. Here, using two models of atopic dermatitis, we show that lean and obese mice mount markedly different immune responses. Obesity converted the classical type 2 T helper (TH2)-predominant disease associated with atopic dermatitis to a more severe disease with prominent TH17 inflammation. We also observed divergent responses to biologic therapies targeting TH2 cytokines, which robustly protected lean mice but exacerbated disease in obese mice. Single-cell RNA sequencing coupled with genome-wide binding analyses revealed decreased activity of nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) in TH2 cells from obese mice relative to lean mice. Conditional ablation of PPARγ in T cells revealed that PPARγ is required to focus the in vivo TH response towards a TH2-predominant state and prevent aberrant non-TH2 inflammation. Treatment of obese mice with a small-molecule PPARγ agonist limited development of TH17 pathology and unlocked therapeutic responsiveness to targeted anti-TH2 biologic therapies. These studies reveal the effects of obesity on immunological disease and suggest a precision medicine approach to target the immune dysregulation caused by obesity.


Subject(s)
Dermatitis, Atopic , PPAR gamma , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Mice , Obesity/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Precision Medicine , Sequence Analysis, RNA , Th2 Cells/metabolism
13.
Nat Immunol ; 16(8): 859-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26098997

ABSTRACT

The receptor NLRP3 is involved in the formation of the NLRP3 inflammasome that activates caspase-1 and mediates the release of interleukin 1ß (IL-1ß) and IL-18. Whether NLRP3 can shape immunological function independently of inflammasomes is unclear. We found that NLRP3 expression in CD4(+) T cells specifically supported a T helper type 2 (TH2) transcriptional program in a cell-intrinsic manner. NLRP3, but not the inflammasome adaptor ASC or caspase-1, positively regulated a TH2 program. In TH2 cells, NLRP3 bound the Il4 promoter and transactivated it in conjunction with the transcription factor IRF4. Nlrp3-deficient TH2 cells supported melanoma tumor growth in an IL-4-dependent manner and also promoted asthma-like symptoms. Our results demonstrate the ability of NLRP3 to act as a key transcription factor in TH2 differentiation.


Subject(s)
Carrier Proteins/immunology , Cell Differentiation/immunology , Th2 Cells/immunology , Trans-Activators/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Binding/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
14.
Immunity ; 49(4): 709-724.e8, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291028

ABSTRACT

B cells thwart antigenic aggressions by releasing immunoglobulin M (IgM), IgG, IgA, and IgE, which deploy well-understood effector functions. In contrast, the role of secreted IgD remains mysterious. We found that some B cells generated IgD-secreting plasma cells following early exposure to external soluble antigens such as food proteins. Secreted IgD targeted basophils by interacting with the CD44-binding protein galectin-9. When engaged by antigen, basophil-bound IgD increased basophil secretion of interleukin-4 (IL-4), IL-5, and IL-13, which facilitated the generation of T follicular helper type 2 cells expressing IL-4. These germinal center T cells enhanced IgG1 and IgE but not IgG2a and IgG2b responses to the antigen initially recognized by basophil-bound IgD. In addition, IgD ligation by antigen attenuated allergic basophil degranulation induced by IgE co-ligation. Thus, IgD may link B cells with basophils to optimize humoral T helper type 2-mediated immunity against common environmental soluble antigens.


Subject(s)
Basophils/immunology , Galectins/immunology , Hyaluronan Receptors/immunology , Immunoglobulin D/immunology , Th2 Cells/immunology , Animals , Basophils/metabolism , Cell Line, Tumor , Cells, Cultured , Galectins/genetics , Galectins/metabolism , Gene Expression Profiling/methods , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunoglobulin D/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred BALB C , Protein Binding , Th2 Cells/metabolism
15.
Immunity ; 49(6): 1034-1048.e8, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566881

ABSTRACT

Single-nucleotide polymorphisms in ETS1 are associated with systemic lupus erythematosus (SLE). Ets1-/- mice develop SLE-like symptoms, suggesting that dysregulation of this transcription factor is important to the onset or progression of SLE. We used conditional deletion approaches to examine the impact of Ets1 expression in different immune cell types. Ets1 deletion on CD4+ T cells, but not B cells or dendritic cells, resulted in the SLE autoimmunity, and this was associated with the spontaneous expansion of T follicular helper type 2 (Tfh2) cells. Ets1-/- Tfh2 cells exhibited increased expression of GATA-3 and interleukin-4 (IL-4), which induced IgE isotype switching in B cells. Neutralization of IL-4 reduced Tfh2 cell frequencies and ameliorated disease parameters. Mechanistically, Ets1 suppressed signature Tfh and Th2 cell genes, including Cxcr5, Bcl6, and Il4ra, thus curbing the terminal Tfh2 cell differentiation process. Tfh2 cell frequencies in SLE patients correlated with disease parameters, providing evidence for the relevance of these findings to human disease.


Subject(s)
Cell Differentiation/immunology , Lupus Erythematosus, Systemic/immunology , Proto-Oncogene Protein c-ets-1/immunology , Th2 Cells/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression/immunology , Gene Expression Profiling , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Th2 Cells/metabolism
16.
Immunity ; 49(5): 886-898.e5, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30446383

ABSTRACT

Pathogenic Th17 (pTh17) cells drive inflammation and immune-pathology, but whether pTh17 cells are a Th17 cell subset whose generation is under specific molecular control remains unaddressed. We found that Ras p21 protein activator 3 (RASA3) was highly expressed by pTh17 cells relative to non-pTh17 cells and was required specifically for pTh17 generation in vitro and in vivo. Mice conditionally deficient for Rasa3 in T cells showed less pathology during experimental autoimmune encephalomyelitis. Rasa3-deficient T cells acquired a Th2 cell-biased program that dominantly trans-suppressed pTh17 cell generation via interleukin 4 production. The Th2 cell bias of Rasa3-deficient T cells was due to aberrantly elevated transcription factor IRF4 expression. RASA3 promoted proteasome-mediated IRF4 protein degradation by facilitating interaction of IRF4 with E3-ubiquitin ligase Cbl-b. Therefore, a RASA3-IRF4-Cbl-b pathway specifically directs pTh17 cell generation by balancing reciprocal Th17-Th2 cell programs. These findings indicate that a distinct molecular program directs pTh17 cell generation and reveals targets for treating pTh17 cell-related pathology and diseases.


Subject(s)
Cell Differentiation/genetics , GTPase-Activating Proteins/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Animals , Autoimmunity , Biomarkers , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , GTPase-Activating Proteins/metabolism , Gene Expression , Immunophenotyping , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice , Proteolysis , RNA, Messenger , Th17 Cells/immunology , Th2 Cells/immunology
17.
Proc Natl Acad Sci U S A ; 121(28): e2319994121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38959032

ABSTRACT

Upon encountering allergens, CD4+ T cells differentiate into IL-4-producing Th2 cells in lymph nodes, which later transform into polyfunctional Th2 cells producing IL-5 and IL-13 in inflamed tissues. However, the precise mechanism underlying their polyfunctionality remains elusive. In this study, we elucidate the pivotal role of NRF2 in polyfunctional Th2 cells in murine models of allergic asthma and in human Th2 cells. We found that an increase in reactive oxygen species (ROS) in immune cells infiltrating the lungs is necessary for the development of eosinophilic asthma and polyfunctional Th2 cells in vivo. Deletion of the ROS sensor NRF2 specifically in T cells, but not in dendritic cells, significantly abolished eosinophilia and polyfunctional Th2 cells in the airway. Mechanistically, NRF2 intrinsic to T cells is essential for inducing optimal oxidative phosphorylation and glycolysis capacity, thereby driving Th2 cell polyfunctionality independently of IL-33, partially by inducing PPARγ. Treatment with an NRF2 inhibitor leads to a substantial decrease in polyfunctional Th2 cells and subsequent eosinophilia in mice and a reduction in the production of Th2 cytokines from peripheral blood mononuclear cells in asthmatic patients. These findings highlight the critical role of Nrf2 as a spatial and temporal metabolic hub that is essential for polyfunctional Th2 cells, suggesting potential therapeutic implications for allergic diseases.


Subject(s)
Asthma , NF-E2-Related Factor 2 , Reactive Oxygen Species , Th2 Cells , NF-E2-Related Factor 2/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Mice , Asthma/immunology , Asthma/metabolism , Humans , Reactive Oxygen Species/metabolism , PPAR gamma/metabolism , Oxidative Phosphorylation , Glycolysis , Lung/immunology , Lung/metabolism , Mice, Knockout , Disease Models, Animal , Female , Cytokines/metabolism , Mice, Inbred C57BL , Interleukin-33/metabolism , Eosinophilia/immunology , Eosinophilia/metabolism
18.
Nat Immunol ; 15(12): 1162-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25362490

ABSTRACT

MicroRNAs (miRNAs) exert powerful effects on immunological function by tuning networks of target genes that orchestrate cell activity. We sought to identify miRNAs and miRNA-regulated pathways that control the type 2 helper T cell (TH2 cell) responses that drive pathogenic inflammation in asthma. Profiling miRNA expression in human airway-infiltrating T cells revealed elevated expression of the miRNA miR-19a in asthma. Modulating miR-19 activity altered TH2 cytokine production in both human and mouse T cells, and TH2 cell responses were markedly impaired in cells lacking the entire miR-17∼92 cluster. miR-19 promoted TH2 cytokine production and amplified inflammatory signaling by direct targeting of the inositol phosphatase PTEN, the signaling inhibitor SOCS1 and the deubiquitinase A20. Thus, upregulation of miR-19a in asthma may be an indicator and a cause of increased TH2 cytokine production in the airways.


Subject(s)
Asthma/immunology , Cytokines/biosynthesis , MicroRNAs/immunology , Th2 Cells/immunology , Animals , Asthma/genetics , Asthma/metabolism , Bronchoalveolar Lavage Fluid/cytology , Clinical Trials as Topic , Flow Cytometry , High-Throughput Screening Assays , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Transgenic , Multiplex Polymerase Chain Reaction , Th2 Cells/metabolism , Up-Regulation
19.
Nat Immunol ; 15(10): 947-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25129370

ABSTRACT

The transcription factor ThPOK promotes CD4(+) T cell differentiation in the thymus. Here, using a mouse strain that allows post-thymic gene deletion, we show that ThPOK maintains CD4(+) T lineage integrity and couples effector differentiation to environmental cues after antigenic stimulation. ThPOK preserved the integrity and amplitude of effector responses and was required for proper differentiation of types 1 and 2 helper T cells in vivo by restraining the expression and function of Runx3, a nuclear factor crucial for cytotoxic T cell differentiation. The transcription factor LRF acts redundantly with ThPOK to prevent the transdifferentiation of mature CD4(+) T cells into CD8(+) T cells. As such, the ThPOK-LRF transcriptional module was essential for CD4(+) T cell integrity and responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/immunology
20.
Immunity ; 47(4): 710-722.e6, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045902

ABSTRACT

Gastro-intestinal helminth infections trigger the release of interleukin-33 (IL-33), which induces type-2 helper T cells (Th2 cells) at the site of infection to produce IL-13, thereby contributing to host resistance in a T cell receptor (TCR)-independent manner. Here, we show that, as a prerequisite for IL-33-induced IL-13 secretion, Th2 cells required the expression of the epidermal growth factor receptor (EGFR) and of its ligand, amphiregulin, for the formation of a signaling complex between T1/ST2 (the IL-33R) and EGFR. This shared signaling complex allowed IL-33 to induce the EGFR-mediated activation of the MAP-kinase signaling pathway and consequently the expression of IL-13. Lack of EGFR expression on T cells abrogated IL-13 expression in infected tissues and impaired host resistance. EGFR expression on Th2 cells was TCR-signaling dependent, and therefore, our data reveal a mechanism by which antigen presentation controls the innate effector function of Th2 cells at the site of inflammation.


Subject(s)
ErbB Receptors/immunology , Interleukin-13/immunology , Interleukin-33/immunology , Receptors, Antigen, T-Cell/immunology , Th2 Cells/immunology , Amphiregulin/immunology , Amphiregulin/metabolism , Animals , Cell Line , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , HEK293 Cells , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , MAP Kinase Signaling System/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/physiology , Nocardia/immunology , Nocardia/physiology , Nocardia Infections/immunology , Nocardia Infections/metabolism , Nocardia Infections/microbiology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL