Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Publication year range
1.
Immunity ; 55(4): 623-638.e5, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385697

ABSTRACT

The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.


Subject(s)
Alarmins , Intestinal Mucosa , Acylation , Alarmins/immunology , Anthelmintics/immunology , Biomarkers, Tumor , Cytokines , DNA-Binding Proteins , Helminthiasis/immunology , Humans , Hyperplasia , Inflammation , Interleukin-33 , Intestinal Mucosa/immunology , Mebendazole , N-Acetylglucosaminyltransferases/immunology , Pore Forming Cytotoxic Proteins , STAT6 Transcription Factor/immunology
2.
Nat Immunol ; 14(11): 1190-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056746

ABSTRACT

Although intergenic long noncoding RNAs (lincRNAs) have been linked to gene regulation in various tissues, little is known about lincRNA transcriptomes in the T cell lineages. Here we identified 1,524 lincRNA clusters in 42 T cell samples, from early T cell progenitors to terminally differentiated helper T cell subsets. Our analysis revealed highly dynamic and cell-specific expression patterns for lincRNAs during T cell differentiation. These lincRNAs were located in genomic regions enriched for genes that encode proteins with immunoregulatory functions. Many were bound and regulated by the key transcription factors T-bet, GATA-3, STAT4 and STAT6. We found that the lincRNA LincR-Ccr2-5'AS, together with GATA-3, was an essential component of a regulatory circuit in gene expression specific to the TH2 subset of helper T cells and was important for the migration of TH2 cells.


Subject(s)
Gene Expression Regulation/immunology , Precursor Cells, T-Lymphoid/metabolism , RNA, Long Noncoding/genetics , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Animals , Cell Differentiation , Cell Movement , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Genetic Loci , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Protein Binding , RNA, Long Noncoding/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Transcriptome/immunology
3.
Immunity ; 42(3): 512-23, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25769611

ABSTRACT

Oral immunotherapy has had limited success in establishing tolerance in food allergy, reflecting failure to elicit an effective regulatory T (Treg) cell response. We show that disease-susceptible (Il4ra(F709)) mice with enhanced interleukin-4 receptor (IL-4R) signaling exhibited STAT6-dependent impaired generation and function of mucosal allergen-specific Treg cells. This failure was associated with the acquisition by Treg cells of a T helper 2 (Th2)-cell-like phenotype, also found in peripheral-blood allergen-specific Treg cells of food-allergic children. Selective augmentation of IL-4R signaling in Treg cells induced their reprogramming into Th2-like cells and disease susceptibility, whereas Treg-cell-lineage-specific deletion of Il4 and Il13 was protective. IL-4R signaling impaired the capacity of Treg cells to suppress mast cell activation and expansion, which in turn drove Th2 cell reprogramming of Treg cells. Interruption of Th2 cell reprogramming of Treg cells might thus provide candidate therapeutic strategies in food allergy.


Subject(s)
Food Hypersensitivity/immunology , Genetic Predisposition to Disease , Immunity, Mucosal , Receptors, Cell Surface/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Adolescent , Allergens/immunology , Animals , Cellular Reprogramming/immunology , Child , Child, Preschool , Female , Food Hypersensitivity/genetics , Food Hypersensitivity/pathology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gene Expression Regulation , Humans , Immune Tolerance , Infant , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-4/immunology , Male , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Transgenic , Receptors, Cell Surface/genetics , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/pathology , Th2 Cells/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
4.
Blood ; 137(9): 1166-1180, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33150420

ABSTRACT

The terminal differentiation of B cells into antibody-secreting cells (ASCs) is a critical component of adaptive immune responses. However, it is a very sensitive process, and dysfunctions lead to a variety of lymphoproliferative neoplasias including germinal center-derived lymphomas. To better characterize the late genomic events that drive the ASC differentiation of human primary naive B cells, we used our in vitro differentiation system and a combination of RNA sequencing and Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC sequencing). We discovered 2 mechanisms that drive human terminal B-cell differentiation. First, after an initial response to interleukin-4 (IL-4), cells that were committed to an ASC fate downregulated the CD23 marker and IL-4 signaling, whereas cells that maintained IL-4 signaling did not differentiate. Second, human CD23- cells also increased IRF4 protein to levels required for ASC differentiation, but they did that independently of the ubiquitin-mediated degradation process previously described in mice. Finally, we showed that CD23- cells carried the imprint of their previous activated B-cell status, were precursors of plasmablasts, and had a phenotype similar to that of in vivo preplasmablasts. Altogether, our results provide an unprecedented genomic characterization of the fate decision between activated B cells and plasmablasts, which provides new insights into the pathological mechanisms that drive lymphoma biology.


Subject(s)
B-Lymphocytes/immunology , Interferon Regulatory Factors/immunology , Interleukin-4/immunology , Plasma Cells/immunology , Receptors, IgE/immunology , STAT6 Transcription Factor/immunology , Cells, Cultured , Humans , Lymphocyte Activation , Lymphoma/immunology , Signal Transduction
5.
Eur J Immunol ; 51(2): 433-444, 2021 02.
Article in English | MEDLINE | ID: mdl-33067820

ABSTRACT

Infection with parasitic worms (helminths) alters host immune responses and can inhibit pathogenic inflammation. Helminth infection promotes a strong Th2 and T regulatory response while suppressing Th1 and Th17 function. Th2 responses are largely dependent on transcriptional programs directed by Stat6-signaling. We examined the importance of intact T cell Stat6 signaling on helminth-induced suppression of murine colitis that results from T cell transfer into immune-deficient mice. Colonization with the intestinal nematode Heligmosomoides polygyrus bakeri resolves WT T cell transfer colitis. However, if the transferred T cells lack intact Stat6 then helminth exposure failed to attenuate colitis or suppress MLN T cell IFN-γ or IL17 production. Loss of Stat6 signaling resulted in decreased IL10 and increased IFN-γ co-expression by IL-17+ T cells. We also transferred T cells from mice with constitutive T cell expression of activated Stat6 (Stat6VT). These mice developed a severe eosinophilic colitis that also was not attenuated by helminth infection. These results show that T cell expression of intact but regulated Stat6 signaling is required for helminth infection-associated regulation of pathogenic intestinal inflammation.


Subject(s)
Colitis/immunology , Nematospiroides dubius/immunology , STAT6 Transcription Factor/immunology , Signal Transduction/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Colitis/parasitology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Th1 Cells/parasitology , Th17 Cells/immunology , Th17 Cells/parasitology , Th2 Cells/immunology , Th2 Cells/parasitology
6.
Clin Immunol ; 229: 108784, 2021 08.
Article in English | MEDLINE | ID: mdl-34126239

ABSTRACT

OBJECTIVE: Osteoarthritis (OA), the leading cause of joint failure, is characterized by breakdown of articular cartilage and remodeling of subchondral bone in synovial joints. Despite the high prevalence and debilitating effects of OA, no disease-modifying drugs exist. Increasing evidence, including genetic variants of the interleukin 4 (IL-4) and IL-4 receptor genes, implicates a role for IL-4 in OA, however, the mechanism underlying IL-4 function in OA remains unknown. Here, we investigated the role of IL-4 in OA pathogenesis. METHODS: Il4-, myeloid-specific-Il4ra-, and Stat6-deficient and control mice were subjected to destabilization of the medial meniscus to induce OA. Macrophages, osteoclasts, and synovial explants were stimulated with IL-4 in vitro, and their function and expression profiles characterized. RESULTS: Mice lacking IL-4, IL-4Ra in myeloid cells, or STAT6 developed exacerbated cartilage damage and osteophyte formation relative to WT controls. In vitro analyses revealed that IL-4 downregulates osteoarthritis-associated genes, enhances macrophage phagocytosis of cartilage debris, and inhibits osteoclast differentiation and activation via the type I receptor. CONCLUSION: Our findings demonstrate that IL-4 protects against osteoarthritis in a myeloid and STAT6-dependent manner. Further, IL-4 can promote an immunomodulatory microenvironment in which joint-resident macrophages polarize towards an M2 phenotype and efficiently clear pro-inflammatory debris, and osteoclasts maintain a homeostatic level of activity in subchondral bone. These findings support a role for IL-4 modulation of myeloid cell types in maintenance of joint health and identify a pathway that could provide therapeutic benefit for osteoarthritis.


Subject(s)
Interleukin-4/immunology , Macrophages/immunology , Osteoarthritis/prevention & control , Osteoclasts/immunology , Animals , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interleukin-4/deficiency , Interleukin-4/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/immunology , Osteoarthritis/pathology , Osteoclasts/pathology , Phagocytosis , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Signal Transduction/immunology
7.
Eur J Immunol ; 50(7): 1044-1056, 2020 07.
Article in English | MEDLINE | ID: mdl-32108934

ABSTRACT

Repeated inhalation of airborne conidia derived from the fungus Aspergillus fumigatus (Af) can lead to a severe eosinophil-dominated inflammatory condition of the lung termed allergic bronchopulmonary aspergillosis (ABPA). ABPA affects about 5 million individuals worldwide and the mechanisms regulating lung pathology in ABPA are poorly understood. Here, we used a mouse model of ABPA to investigate the role of eosinophils and T cell-derived IL-4/IL-13 for induction of allergic lung inflammation. Selective deletion of IL-4/IL-13 in T cells blunted the Af-induced lung eosinophilia and further resulted in lower expression of STAT6-regulated chemokines and effector proteins such as Arginase 1, Relm-α, Relm-ß, and Muc5a/c. Eosinophil-deficient ΔdblGata mice showed lower IL-4 expression in the lung and the number of Th2 cells in the lung parenchyma was reduced. However, expression of the goblet cell markers Clca1 and Muc5a/c, abundance of mucin-positive cells, as well as weight gain of lungs were comparable between Af-challenged ΔdblGata and WT mice. Based on these results, we conclude that T cell-derived IL-4/IL-13 is essential for Af-induced lung eosinophilia and inflammation while eosinophils may play a more subtle immunomodulatory role and should not simply be regarded as pro-inflammatory effector cells in ABPA.


Subject(s)
Aspergillosis, Allergic Bronchopulmonary/immunology , Aspergillus fumigatus/immunology , Eosinophils/immunology , Lung/immunology , Th2 Cells/immunology , Animals , Aspergillosis, Allergic Bronchopulmonary/genetics , Aspergillosis, Allergic Bronchopulmonary/pathology , Disease Models, Animal , Eosinophils/pathology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/immunology , Lung/pathology , Mice , Mice, Knockout , Mucin 5AC/genetics , Mucin 5AC/immunology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Th2 Cells/pathology
8.
Blood ; 133(13): 1489-1494, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30696620

ABSTRACT

Persistent NF-κB activation is a hallmark of the malignant Hodgkin/Reed-Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL). Genomic lesions, Epstein-Barr virus infection, soluble factors, and tumor-microenvironment interactions contribute to this activation. Here, in an unbiased approach to identify the cHL cell-secreted key factors for NF-κB activation, we have dissected the secretome of cultured cHL cells by chromatography and subsequent mass spectrometry. We identified lymphotoxin-α (LTA) as the causative factor for autocrine and paracrine activation of canonical and noncanonical NF-κB in cHL cell lines. In addition to inducing NF-κB, LTA promotes JAK2/STAT6 signaling. LTA and its receptor TNFRSF14 are transcriptionally activated by noncanonical NF-κB, creating a continuous feedback loop. Furthermore, LTA shapes the expression of cytokines, receptors, immune checkpoint ligands and adhesion molecules, including CSF2, CD40, PD-L1/PD-L2, and VCAM1. Comparison with single-cell gene-activity profiles of human hematopoietic cells showed that LTA induces genes restricted to the lymphoid lineage, as well as those largely restricted to the myeloid lineage. Thus, LTA sustains autocrine NF-κB activation, impacts activation of several signaling pathways, and drives expression of genes essential for microenvironmental interactions and lineage ambiguity. These data provide a robust rationale for targeting LTA as a treatment strategy for cHL patients.


Subject(s)
Hodgkin Disease/immunology , Janus Kinase 2/immunology , Lymphotoxin-alpha/immunology , NF-kappa B/immunology , STAT6 Transcription Factor/immunology , Cell Line , Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Humans , Lymphotoxin-alpha/genetics , Reed-Sternberg Cells/immunology , Reed-Sternberg Cells/metabolism , Signal Transduction , Transcriptional Activation
9.
Inflamm Res ; 70(2): 183-192, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33386422

ABSTRACT

OBJECTIVE: Microglia/macrophage activation is previously reported to be involved in various ocular diseases. However, the separate role of M1/M2 phenotype microglia/macrophage in the pathological process of oxygen-induced retinopathy (OIR) remains unknown. In this research, we explored the role and regulatory mechanism of M1/M2 microglia/macrophage in OIR in C57BL/6J mice. Furthermore, we demonstrated the time phase of M1/M2 shifting of microglia/macrophage during the natural process of OIR, which is very essential for further investigations. MATERIALS AND METHODS: C57BL/6j pups were exposed to hyperoxia environment from postnatal 7(P7) to P12 then returned to normoxia. The mice were then euthanized, and the eyes were harvested at a series of time points for further investigation. The M1/M2 phenotype microglia/macrophage activity was presented by immunofluorescent staining and real-time quantitative polymerase chain reaction (qPCR). The NF-κb-STAT3 signaling and IL-4-STAT6-PPAR-γ signaling pathway activity was examined by western blot analysis. RESULTS: The microglia/macrophage were activated when the OIR model was set up after P12. The M1 microglia/macrophage activation was found in neovascularization (NV) tufts in both central and peripheral retina, which started from P12 when the mice were returned to normoxia environment and peaked at P17. During this period of time, the NF-κb-STAT3 signaling pathway was activated, resulting in the upregulated M1 phenotype microglia/macrophage polarization, along with the enhanced inflammatory cytokine expression including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1ß. Consequently, the NV tufts were observed from P12 and the volume continued to increase until P17. However, the M2 phenotype microglia/macrophage activity took over during the late phase of OIR started from P17. The IL-4-STAT6-PPAR-γ signaling activity was upregulated from P17 and peaked at P20, inducing M2 phenotype microglia polarization, which consequently led to the inhibition of inflammatory cytokines and spontaneous regression of NV tufts. CONCLUSIONS: Microglia/macrophage participate actively in the natural process of OIR in mice, and two phenotypes exert different functions. Treatment modulating microglia/macrophage polarize toward M2 phenotype might be a novel and promising method for ocular neovascular diseases such as retinopathy of prematurity (ROP), wet age-related macular degeneration (wAMD), and diabetic retinopathy (DR).


Subject(s)
Macrophages/immunology , Microglia/immunology , Retinal Diseases/immunology , Animals , Cytokines/immunology , Mice, Inbred C57BL , NF-kappa B/immunology , Oxygen , PPAR gamma/immunology , Phenotype , Retina/immunology , STAT3 Transcription Factor/immunology , STAT6 Transcription Factor/immunology , Signal Transduction
10.
Immunology ; 159(4): 441-449, 2020 04.
Article in English | MEDLINE | ID: mdl-31957000

ABSTRACT

Signaling by Kit has been extensively studied in hematopoietic cells and is essential for the survival, proliferation and maintenance of hematopoietic stem and progenitor cells. In addition to the activation of intrinsic signaling pathways, Kit has been shown to interact with lineage-restricted type I cytokine receptors and produce cross signals, e.g. erythropoietin receptor, interleukin-7 receptor (IL-7R), IL-3R. Based on the earlier studies, we hypothesize that Kit activate other type I cytokine receptors in a cell-specific manner and execute cell-specific function. To investigate other Kit-activated receptors, we tested Kit and IL-4R cross-receptor activation in murine bone-marrow-derived mast cells, which express both Kit and IL-4R at the surface level. Kit upon activation by Kit ligand (KL), activated IL-4Rα, γC , and signal transducer and activator of transcription 6 independent of its cognate ligand IL-4. Though KL and IL-4 are individually mitogenic, combinations of KL and IL-4 synergistically promoted mast cell proliferation. Furthermore, inhibition of lipid raft formation by methyl-ß-cyclodextrin resulted in loss of synergistic proliferation. Together the data suggest IL-4R as a novel Kit-activated receptor. Such cross-receptor activations are likely to be a universal mechanism of Kit signaling in hematopoiesis.


Subject(s)
Interleukin-4/pharmacology , Mast Cells/drug effects , Proto-Oncogene Proteins c-kit/genetics , Receptors, Interleukin-4/genetics , STAT6 Transcription Factor/genetics , Stem Cell Factor/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Proliferation/drug effects , Gene Expression Regulation , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mast Cells/cytology , Mast Cells/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proto-Oncogene Proteins c-kit/immunology , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-4/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , STAT6 Transcription Factor/immunology , Signal Transduction , Stem Cell Factor/genetics , Stem Cell Factor/immunology , beta-Cyclodextrins/pharmacology
11.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L888-L899, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32130032

ABSTRACT

We have previously demonstrated that upregulation of Sonic hedgehog (SHH) expression in allergic airway epithelia essentially contributes to the goblet cell metaplasia and mucous hypersecretion. However, the mechanism underlying the upregulation of SHH expression remains completely unknown. In cultured human airway epithelial cells, IL-4/IL-13 but not IL-5 robustly induces the mRNA and protein expression of SHH and in turn activates SHH signaling by promoting the JAK/STAT6-controlling transcription of SHH gene. Moreover, intratracheal instillation of IL-4 and/or IL-13 robustly activates STAT6 and concomitantly upregulates SHH expression in mouse airway epithelia, whereas, in Club cell 10-kDa protein (CC10)-positive airway epithelial cells of children with asthma, activated STAT6 closely correlates with the increased expression of SHH and high activity of SHH signaling. Finally, intratracheal inhibition of STAT6 by AS-1517499 significantly diminished the allergen-induced upregulation of SHH expression, goblet cell phenotypes, and airway hyperresponsiveness, in an ovalbumin- or house dust mite-induced mouse model with allergic airway inflammation,. Together, upregulation of SHH expression by IL-4/IL-13-induced JAK/STAT6 signaling contributes to allergic airway epithelial remodeling, and this study thus provides insight into how morphogen signaling is coordinated with Th2 cytokine pathways to regulate tissue remodeling in chronic airway diseases.


Subject(s)
Asthma/genetics , Hedgehog Proteins/genetics , Interleukin-13/genetics , Interleukin-4/genetics , Respiratory Mucosa/immunology , Animals , Anti-Asthmatic Agents/pharmacology , Asthma/chemically induced , Asthma/drug therapy , Asthma/pathology , Cell Line , Child , Female , Gene Expression Regulation , Goblet Cells/drug effects , Goblet Cells/immunology , Goblet Cells/pathology , Hedgehog Proteins/immunology , Humans , Interleukin-13/immunology , Interleukin-13/pharmacology , Interleukin-4/immunology , Interleukin-4/pharmacology , Interleukin-5/genetics , Interleukin-5/immunology , Janus Kinases/genetics , Janus Kinases/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Primary Cell Culture , Pyrimidines/pharmacology , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Signal Transduction , Transcription, Genetic , Uteroglobin/genetics , Uteroglobin/immunology
12.
Eur J Immunol ; 49(9): 1421-1432, 2019 09.
Article in English | MEDLINE | ID: mdl-31144294

ABSTRACT

Infection of mice with the gastrointestinal helminth Nippostrongylus brasiliensis elicits profound local proliferation and accumulation of type 2 innate lymphoid cells (ILC2s) in the lung. The regulation of ILC2 proliferation and accumulation in the lung is poorly understood. Using T cell-specific IL-4/IL-13-deficient mice, we demonstrate that IL-4/IL-13 secretion from Th2 cells promotes proliferation and expansion of the ILC2 population in the lung of N. brasiliensis-infected mice. Competitive mixed BM chimeras containing normal and STAT6-deficient ILC2s further indicated that ILC2s have to respond directly to IL-4/IL-13 for this effect while STAT6 is not required for IL-13 production in ILC2s. In addition, expression of a constitutively active form of STAT6 in ILC2s was sufficient to promote their proliferation in uninfected mice. The expression of MHC class II in ILC2s appeared to be enhanced by STAT6 signaling supporting the concept that Th2 cells and ILC2s can communicate in an antigen-dependent manner resulting in a Th2-regulated accumulation of ILC2s in the lung during an acute type 2 immune response. Based on our observations, targeting the STAT6 pathway in ILC2s could help to develop new treatments to dampen ILC2 proliferation in the lung and thereby ameliorate ILC2-mediated allergic inflammation.


Subject(s)
Interleukin-13/immunology , Interleukin-4/immunology , Lung/immunology , STAT6 Transcription Factor/immunology , Signal Transduction/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Asthma/immunology , Cell Proliferation/physiology , Cytokines/immunology , Immunity, Innate/immunology , Inflammation/immunology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL
13.
Clin Exp Allergy ; 50(1): 5-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31505066

ABSTRACT

The Th2 cytokines interleukin 4 (IL-4) and IL-13 and the heterodimeric IL-4 receptor (IL-4R) complexes that they interact with play a key role in the pathogenesis of allergic disorders. Dupilumab is a humanized IgG4 monoclonal antibody that targets the IL-4 receptor alpha chain (IL-4Rα), common to both IL-4R complexes: type 1 (IL-4Rα/γc; IL-4 specific) and type 2 (IL-4Rα/IL-13Rα1; IL-4 and IL-13 specific). In this review, we detail the current state of knowledge of the different signalling pathways coupled to the IL-4R complexes and examine the possible mechanisms of Dupilumab action and survey its clinical efficacy in different allergic disorders. The development of Dupilumab and the widening spectrum of its clinical applications is relevant to the current emphasis on precision medicine approaches to the blockade of pathways involved in allergic diseases.


Subject(s)
Anti-Allergic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Dermatitis, Atopic/drug therapy , Eosinophilic Esophagitis/drug therapy , Food Hypersensitivity/drug therapy , Interleukin-13/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4/immunology , Anaphylaxis/immunology , Asthma/immunology , Chronic Disease , Dermatitis, Atopic/immunology , Eosinophilic Esophagitis/immunology , Food Hypersensitivity/immunology , Humans , Inflammation/immunology , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Nasal Polyps/drug therapy , Nasal Polyps/immunology , Rhinitis, Allergic/drug therapy , Rhinitis, Allergic/immunology , STAT6 Transcription Factor/immunology , Sinusitis/drug therapy , Sinusitis/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
14.
Nat Immunol ; 9(12): 1399-406, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18978793

ABSTRACT

Toll-like receptor (TLR) signaling in macrophages is required for antipathogen responses, including the biosynthesis of nitric oxide from arginine, and is essential for immunity to Mycobacterium tuberculosis, Toxoplasma gondii and other intracellular pathogens. Here we report a 'loophole' in the TLR pathway that is advantageous to these pathogens. Intracellular pathogens induced expression of the arginine hydrolytic enzyme arginase 1 (Arg1) in mouse macrophages through the TLR pathway. In contrast to diseases dominated by T helper type 2 responses in which Arg1 expression is greatly increased by interleukin 4 and 13 signaling through the transcription factor STAT6, TLR-mediated Arg1 induction was independent of the STAT6 pathway. Specific elimination of Arg1 in macrophages favored host survival during T. gondii infection and decreased lung bacterial load during tuberculosis infection.


Subject(s)
Arginase/immunology , Bacterial Infections/immunology , Macrophages/immunology , Macrophages/microbiology , Toll-Like Receptors/immunology , Animals , Arginase/metabolism , CCAAT-Enhancer-Binding Protein-beta/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Immunoblotting , Immunohistochemistry , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Toll-Like Receptors/metabolism
15.
Nat Immunol ; 9(12): 1347-55, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18997793

ABSTRACT

Transcription factor Foxp3 is critical for generating regulatory T cells (T(reg) cells). Transforming growth factor-beta (TGF-beta) induces Foxp3 and suppressive T(reg) cells from naive T cells, whereas interleukin 6 (IL-6) inhibits the generation of inducible T(reg) cells. Here we show that IL-4 blocked the generation of TGF-beta-induced Foxp3(+) T(reg) cells and instead induced a population of T helper cells that produced IL-9 and IL-10. The IL-9(+)IL-10(+) T cells demonstrated no regulatory properties despite producing abundant IL-10. Adoptive transfer of IL-9(+)IL-10(+) T cells into recombination-activating gene 1-deficient mice induced colitis and peripheral neuritis, the severity of which was aggravated if the IL-9(+)IL-10(+) T cells were transferred with CD45RB(hi) CD4(+) effector T cells. Thus IL-9(+)IL-10(+) T cells lack suppressive function and constitute a distinct population of helper-effector T cells that promote tissue inflammation.


Subject(s)
Forkhead Transcription Factors/immunology , Interleukin-4/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta/metabolism
16.
Inflamm Res ; 69(10): 1027-1037, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32671438

ABSTRACT

OBJECTIVE AND DESIGN: To clarify the effects of dietary supplementation of protocatechuic acid (PCA) and in-depth mechanisms on allergic asthma in ovalbumin (OVA)-induced mice. MATERIALS: Female BALB/c mice were randomly divided into three groups (n = 10 in each group): control group, OVA-induced allergic asthma group, and OVA plus PCA group. TREATMENT: Dietary supplementation of PCA was achieved by adding 50 mg/kg PCA to AIN 93G diet for 25 days. METHODS: Peripheral blood cells, pulmonary inflammatory cell infiltration, the levels of IL-4, IL-5, and IL-13 in bronchoalveolar lavage fluid (BALF), the mRNA levels of Th2-related genes in the lungs, and the protein expressions of the IL-4Rα-STAT6 and the Jagged1/Jagged2-Notch1/Notch2 signaling pathways were measured. RESULTS: Significantly reduced inflammatory cells infiltration and mucosal hypersecretion in the lung tissues, repaired levels of interleukin IL-4, IL-5, and IL-13 in the BALF, and decreased mRNA expression of IL-4, IL-5, and GATA3 were observed in OVA plus PCA group. Moreover, PCA treatment down-regulated the protein levels of IL-4Rα-STAT6 and Jagged1/Jagged2-Notch1/Notch2 signaling pathways. CONCLUSIONS: Dietary supplement of PCA alleviated allergic asthma partly through suppressing the IL-4Rα-STAT6 and Jagged1/Jagged2-Notch1/Notch2 signaling pathways in mice. Our study provided the theoretic basis of PCA used as functional food in preventing allergic asthma.


Subject(s)
Asthma/diet therapy , Dietary Supplements , Hydroxybenzoates/therapeutic use , Allergens , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/genetics , Cytokines/immunology , Female , Functional Food , Jagged-1 Protein/immunology , Jagged-2 Protein/immunology , Leukocyte Count , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Ovalbumin , Receptor, Notch1/immunology , Receptor, Notch2/immunology , Receptors, Cell Surface/immunology , STAT6 Transcription Factor/immunology , Signal Transduction
17.
J Immunol ; 201(9): 2612-2623, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30266770

ABSTRACT

Production of TGF-ß by T cells is key to various aspects of immune homeostasis, with defects in this process causing or aggravating immune-mediated disorders. The molecular mechanisms that lead to TGF-ß generation by T cells remain largely unknown. To address this issue, we take advantage of the fact that intestinal helminths stimulate Th2 cells besides triggering TGF-ß generation by T lymphocytes and regulate immune-mediated disorders. We show that the Th2 cell-inducing transcription factor STAT6 is necessary and sufficient for the expression of TGF-ß propeptide in T cells. STAT6 is also necessary for several helminth-triggered events in mice, such as TGF-ß-dependent suppression of alloreactive inflammation in graft-versus-host disease. Besides STAT6, helminth-induced secretion of active TGF-ß requires cleavage of propeptide by the endopeptidase furin. Thus, for the immune regulatory pathway necessary for TGF-ß production by T cells, our results support a two-step model, composed of STAT6 and furin.


Subject(s)
Furin/immunology , STAT6 Transcription Factor/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/biosynthesis , Animals , Furin/metabolism , Graft vs Host Disease/immunology , Mice , STAT6 Transcription Factor/metabolism , Strongylida Infections/immunology
18.
J Immunol ; 201(10): 2947-2958, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30291166

ABSTRACT

Recently we reported that IL-4 and IL-13 signaling in murine early thymic progenitors (ETPs) expressing the heteroreceptor (HR) comprising IL-4 receptor α (IL-4Rα) and IL-13 receptor α 1 (IL-13Rα1) activate STAT6 and inhibit ETP maturation potential toward T cells. In this study, we asked whether IL-4 and IL-13 signaling through the HR mobilizes other STAT molecules to shape ETP fate decision. The findings indicate that HR+ ETPs undergoing cytokine signaling display increased STAT1, but not STAT3, phosphorylation in addition to STAT6 activation. In parallel, the ETPs had a STAT1-dependent heightened expression of IRF-8, a transcription factor essential for development of CD8α+ dendritic cells (DCs). Interestingly, STAT1 phosphorylation and IRF-8 upregulation, which are independent of STAT6 activation, guided ETP maturation toward myeloid cells with a CD8α+ DC phenotype. Furthermore, these CD8α+ DCs display a thymic resident phenotype, as they did not express SIRPα, a molecule presumed to be involved in cell migration. These findings suggest that IL-4 and IL-13 cytokine-induced HR signaling provides a double-edged sword that simultaneously blocks T cell lineage potential but advances myeloid maturation that could impact T cell selection and central tolerance.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Interleukin-13/metabolism , Interleukin-4/metabolism , Thymocytes/cytology , Animals , Central Tolerance/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-13/immunology , Interleukin-4/immunology , Mice , Mice, Knockout , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Thymocytes/immunology , Thymocytes/metabolism
19.
Int J Mol Sci ; 21(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751111

ABSTRACT

Atopic dermatitis (AD) is an eczematous, pruritic skin disorder with extensive barrier dysfunction and elevated interleukin (IL)-4 and IL-13 signatures. The barrier dysfunction correlates with the downregulation of barrier-related molecules such as filaggrin (FLG), loricrin (LOR), and involucrin (IVL). IL-4 and IL-13 potently inhibit the expression of these molecules by activating signal transducer and activator of transcription (STAT)6 and STAT3. In addition to IL-4 and IL-13, IL-22 and IL-17A are probably involved in the barrier dysfunction by inhibiting the expression of these barrier-related molecules. In contrast, natural or medicinal ligands for aryl hydrocarbon receptor (AHR) are potent upregulators of FLG, LOR, and IVL expression. As IL-4, IL-13, IL-22, and IL-17A are all capable of inducing oxidative stress, antioxidative AHR agonists such as coal tar, glyteer, and tapinarof exert particular therapeutic efficacy for AD. These antioxidative AHR ligands are known to activate an antioxidative transcription factor, nuclear factor E2-related factor 2 (NRF2). This article focuses on the mechanisms by which FLG, LOR, and IVL expression is regulated by IL-4, IL-13, IL-22, and IL-17A. The author also summarizes how AHR and NRF2 dual activators exert their beneficial effects in the treatment of AD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Dermatitis, Atopic/genetics , Membrane Proteins/genetics , NF-E2-Related Factor 2/genetics , Protein Precursors/genetics , Receptors, Aryl Hydrocarbon/genetics , S100 Proteins/genetics , Antioxidants/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/immunology , Coal Tar/therapeutic use , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Filaggrin Proteins , Gene Expression Regulation , Humans , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukins/genetics , Interleukins/immunology , Membrane Proteins/immunology , NF-E2-Related Factor 2/immunology , Oxidative Stress , Protein Precursors/immunology , Receptors, Aryl Hydrocarbon/immunology , Resorcinols/therapeutic use , S100 Proteins/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Signal Transduction , Skin/drug effects , Skin/immunology , Skin/pathology , Stilbenes/therapeutic use , Tars/therapeutic use , Interleukin-22
20.
Molecules ; 25(9)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397290

ABSTRACT

Exposure to particulate matter (PM) has been known to be one of the risk factors to cause allergic asthma, leading to development of respiratory disease. Banhahubak-tang tablet (BHT), a standardized Korean Medicine, is prescribed for neurasthenia, laryngopharyngitis and asthma. In this study, we investigated therapeutic effects of BHT on airway inflammation in ovalbumin (OVA) and PM smaller than 10 µm (PM10)-induced allergic asthma mice. To establish allergic asthma with airway hyper-responsiveness by PM10, BALB/c mice were sensitized and challenged with OVA and PM10, and orally administered BHT. Histological staining was performed to assess airway remodeling. Serum and bronchoalveolar lavage fluid (BALF) was collected for measuring immunoglobulin levels and counting inflammatory cells, respectively. Expression levels of Janus kinase 1 (JAK1)/signal transducer and activator of transcription 6 (STAT6), pro-inflammatory cytokines and type 2 T-helper (Th2)-related cytokines were analyzed in vivo and in vitro models. Histopathological analysis demonstrated that BHT suppressed inflammatory cell infiltration, mucus hypersecretion and collagen deposition in the airway. BHT administration effectively decreased number of inflammatory cells in BALF. BHT reduced total serum Immunoglobulin E (IgE) and Immunoglobulin G (IgG) levels. In addition, BHT significantly inhibited the phosphorylation of JAK1 and STAT6 expressions. Release of pro-inflammatory cytokines and Th2-related cytokines were down-regulated by BHT. In conclusion, BHT mitigated airway inflammation by down-regulating pro-inflammatory and Th2-related cytokines via JAK1/STAT6 signaling. BHT might be a promising herbal medicine for preventing airway inflammation. Moreover, an intervention study among humans is needed to further evaluate the possible beneficial effects of BHT in allergic asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma , Janus Kinase 1/immunology , STAT6 Transcription Factor/immunology , Signal Transduction/drug effects , Animals , Anti-Asthmatic Agents/chemistry , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , Cytokines/immunology , Disease Models, Animal , Female , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Tablets , Th2 Cells/immunology , Th2 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL