Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38889332

ABSTRACT

ILC2s are key players in type 2 immunity and contribute to maintaining homeostasis. ILC2s are also implicated in the development of type 2 inflammation-mediated chronic disorders like asthma. While memory ILC2s have been identified in mouse, it is unknown whether human ILC2s can acquire immunological memory. Here, we demonstrate the persistence of CD45RO, a marker previously linked to inflammatory ILC2s, in resting ILC2s that have undergone prior activation. A high proportion of these cells concurrently reduce the expression of the canonical ILC marker CD127 in a tissue-specific manner. Upon isolation and in vitro stimulation of CD127-CD45RO+ ILC2s, we observed an augmented ability to proliferate and produce cytokines. CD127-CD45RO+ ILC2s are found in both healthy and inflamed tissues and display a gene signature of cell activation. Similarly, mouse memory ILC2s show reduced expression of CD127. Our findings suggest that human ILC2s can acquire innate immune memory and warrant a revision of the current strategies to identify human ILC2s.


Subject(s)
Immunity, Innate , Immunologic Memory , Interleukin-7 Receptor alpha Subunit , Lymphocytes , Humans , Immunologic Memory/immunology , Animals , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocytes/immunology , Mice , Immunity, Innate/immunology , Leukocyte Common Antigens/metabolism , Cytokines/metabolism , Inflammation/immunology , Female , Mice, Inbred C57BL
2.
Immunol Rev ; 323(1): 118-125, 2024 May.
Article in English | MEDLINE | ID: mdl-38506432

ABSTRACT

Group 2 Innate Lymphoid Cells (ILC2s) are innate lymphocytes involved in type 2 immunity. ILC2s are abundant at the barrier tissues and upon allergen exposure, respond to epithelial-derived alarmins by producing type 2 cytokines (e.g., IL-5 and IL-13). Upon activation, some of these activated ILC2s acquire immunological memory and can mount enhanced responses upon further allergen encounters. Here, we review recent findings of the cellular and molecular mechanisms underlying immune memory in ILC2s both in mice and humans and discuss the implications of memory ILC2s in the context of allergic diseases.


Subject(s)
Hypersensitivity , Immunity, Innate , Immunologic Memory , Lymphocytes , Humans , Animals , Lymphocytes/immunology , Lymphocytes/metabolism , Hypersensitivity/immunology , Cytokines/metabolism , Allergens/immunology , Lymphocyte Activation/immunology , Mice
3.
Front Immunol ; 14: 1198310, 2023.
Article in English | MEDLINE | ID: mdl-37809100

ABSTRACT

Introduction: The three groups of helper innate lymphoid cells (ILCs), namely ILC1, ILC2 and ILC3, have been identified by flow cytometry by combinations of cell surface markers. Here, we review various ways ILCs are currently identified, focusing on potential problems and their solutions. The first step to identify all ILCs is to exclude other lymphocytes and myeloid cells by their lineage-specific markers (Lin). However, the Lin cocktail varies in various studies, and the definition of Lin- population containing ILCs is often ambiguous, resulting in contamination of Lin+ cells, particularly T cells. Method: We have designed combinations of cell surface markers to identify ILC populations in various tissues of B6 mice by flow cytometry. To minimize T cell contamination, TCR/CD3ϵ antibodies were used separately from the Lin cocktail. ILCs identified by surface markers are confirmed by the expression of the transcription factors GATA3, RORγt, T-bet and Eomes. Result: ILC1s in the B6 mouse liver are identified by Lin-NKp46+NK1.1+TCR/CD3ϵ-CD49a+CD49b-. However, defining ILC1s in other tissues remains a challenge. ILC2s in the lung are identified by Lin-TCR/CD3ϵ- Thy1+CD127+ST2+ whereas ILC2s in the small intestine and liver are identified by Lin-TCR/CD3ϵ-Thy1+GATA3+RORγt-. ILC3s in B6 mouse spleen, liver, lung and small intestine are identified by Lin-TCR/CD3ϵ- Thy1+CD127+RORγt+. Discussion: The ILC population is heterogeneous and the strategies to identify ILCs have to be designed for each ILC population and tissue. Excluding T cells in all cases is crucial, and a combination of transcription factors GATA3, RORγt, T-bet, and Eomes should be used to identify ILCs. Using CD3ϵ/TCRs in a different fluorochrome not in Lin cocktail minimizes contamination of T cells specifically identify individual ILC populations in various tissues.


Subject(s)
Immunity, Innate , Lymphocytes , Mice , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3 , Flow Cytometry , Transcription Factors , Receptors, Antigen, T-Cell
4.
Front Immunol ; 13: 877005, 2022.
Article in English | MEDLINE | ID: mdl-35572538

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are present in both mouse and human mucosal and non-mucosal tissues and implicated in initiating type 2 inflammation. ILC2s are considered to be tissue resident cells that develop in the perinatal period and persist throughout life with minimal turning over in adulthood. However, recent studies in animal models have shown their ability to circulate between different organs during inflammation and their potential functions in the destined organs, suggesting their roles in mediating multiple type 2 diseases. Here, we review recent findings on ILC2 migration, including migration within, into and out of tissues during inflammation.


Subject(s)
Immunity, Innate , Lymphocytes , Animals , Female , Inflammation , Mice , Pregnancy
5.
Front Immunol ; 12: 679509, 2021.
Article in English | MEDLINE | ID: mdl-34305911

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are tissue resident in the lung and activated by inhaled allergens via epithelial-derived alarmins including IL-33. Activated ILC2s proliferate, produce IL-5 and IL-13, and induce eosinophilic inflammation. Here, we report that intranasal IL-33 or the protease allergen papain administration resulted in increased numbers of ILC2s not only in the lung but also in peripheral blood and liver. Analyses of IL-33 treated parabiosis mice showed that the increase in lung ILC2s was due to proliferation of lung resident ILC2s, whereas the increase in liver ILC2s was due to the migration of activated lung ILC2s. Lung-derived ILC2s induced eosinophilic hepatitis and expression of fibrosis-related genes. Intranasal IL-33 pre-treatment also attenuated concanavalin A-induced acute hepatitis and cirrhosis. These results suggest that activated lung resident ILC2s emigrate from the lung, circulate, settle in the liver and promote type 2 inflammation and attenuate type 1 inflammation.


Subject(s)
Hepatitis/etiology , Hypersensitivity/etiology , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Pneumonia/etiology , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression , Hepatitis/metabolism , Hepatitis/pathology , Hypersensitivity/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Eosinophilia/etiology , Pulmonary Eosinophilia/metabolism , Pulmonary Eosinophilia/pathology
6.
Front Immunol ; 12: 671966, 2021.
Article in English | MEDLINE | ID: mdl-33968080

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) reside in both mucosal and non-mucosal tissues and play critical roles in the first line of defense against parasites and irritants such as allergens. Upon activation by cytokines released from epithelial and stromal cells during tissue damage or stimulation, ILC2s produce copious amounts of IL-5 and IL-13, leading to type 2 inflammation. Over the past 10 years, ILC2 involvement in a variety of human diseases has been unveiled. However, questions remain as to the fate of ILC2s after activation and how that might impact their role in chronic inflammatory diseases such as asthma and fibrosis. Here, we review studies that have revealed novel properties of post-activation ILC2s including the generation of immunological memory, exhausted-like phenotype, transdifferentiation and activation-induced migration.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Animals , Humans
7.
PLoS One ; 16(5): e0252257, 2021.
Article in English | MEDLINE | ID: mdl-34015033

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0214286.].

8.
J Allergy Clin Immunol ; 147(5): 1549-1560, 2021 05.
Article in English | MEDLINE | ID: mdl-33965092

ABSTRACT

Innate lymphoid cells (ILCs) mainly reside at barrier surfaces and regulate tissue homeostasis and immunity. ILCs are divided into 3 groups, group 1 ILCs, group 2 ILCs, and group 3 ILC3, on the basis of their similar effector programs to T cells. The development of ILCs from lymphoid progenitors in adult mouse bone marrow has been studied in detail, and multiple ILC progenitors have been characterized. ILCs are mostly tissue-resident cells that develop in the perinatal period. More recently, ILC progenitors have also been identified in peripheral tissues. In this review, we discuss the stepwise transcription factor-directed differentiation of mouse ILC progenitors into mature ILCs, the critical time windows in ILC development, and the contribution of bone marrow versus tissue ILC progenitors to the pool of mature ILCs in tissues.


Subject(s)
Lymphocytes/cytology , Lymphocytes/immunology , Animals , Core Binding Factor alpha Subunits/immunology , Cytokines/immunology , Epigenesis, Genetic , Humans , Immunity, Innate , Infections/immunology , Inflammation/immunology , Thymus Gland/immunology , Transcription Factors/immunology
9.
Blood Adv ; 4(21): 5362-5372, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33137203

ABSTRACT

Innate lymphoid cells (ILCs) are a recently identified subset of leukocytes that play a central role in pathogen surveillance and resistance, modulation of immune response, and tissue repair. They are remarkably similar to CD4+ T-helper subsets in terms of function and transcription factors required for their development but are distinguished by their lack of antigen-specific receptors. Despite their similarities, the absence of a surface T-cell receptor (TCR) and presence of ILCs and precursors in adult bone marrow has led to speculation that ILCs and T cells develop separately from lineages that branch at the point of precursors within the bone marrow. Considering the common lineage markers and effector cytokine profiles shared between ILCs and T cells, it is surprising that the status of the TCR loci in ILCs was not fully explored at the time of their discovery. Here, we demonstrate that a high proportion of peripheral tissue ILC2s have TCRγ chain gene rearrangements and TCRδ locus deletions. Detailed analyses of these loci show abundant frameshifts and premature stop codons that would encode nonfunctional TCR proteins. Collectively, these data argue that ILC2 can develop from T cells that fail to appropriately rearrange TCR genes, potentially within the thymus.


Subject(s)
Immunity, Innate , Precursor Cells, T-Lymphoid , Leukocytes , Lymphocytes
10.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32573494

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) in mouse lungs are activated by the epithelium-derived alarmin IL-33. Activated ILC2s proliferate and produce IL-5 and IL-13 that drive allergic responses. In neonatal lungs, the occurrence of spontaneous activation of lung ILC2s is dependent on endogenous IL-33. Here, we report that neonatal lung ILC2 activation by endogenous IL-33 has significant effects on ILC2 functions in adulthood. Most neonatal lung ILC2s incorporated 5-bromo-2'-deoxyuridine (BrdU) and persisted into adulthood. BrdU+ ILC2s in adult lungs responded more intensely to IL-33 treatment compared with BrdU- ILC2s. In IL-33-deficient (KO) mice, lung ILC2s develop normally, but they are not activated in the neonatal period. Lung ILC2s in KO mice responded less intensely to IL-33 in adulthood compared with WT ILC2s. While there was no difference in the number of lung ILC2s, there were fewer IL-13+ ILC2s in KO mice compared with those in WT mice. The impaired responsiveness of ILC2s in KO mice was reversed by i.n. administrations of IL-33 in the neonatal period. These results suggest that activation of lung ILC2s by endogenous IL-33 in the neonatal period may "train" ILC2s seeding the lung after birth to become long-lasting resident cells that respond more efficiently to challenges later in life.


Subject(s)
Asthma/genetics , Hypersensitivity/genetics , Immunity, Innate/genetics , Interleukin-33/genetics , Lymphocytes/drug effects , Animals , Animals, Newborn , Asthma/immunology , Asthma/pathology , Cell Proliferation/drug effects , Humans , Hypersensitivity/immunology , Hypersensitivity/pathology , Interleukin-33/pharmacology , Lung/drug effects , Lung/growth & development , Lymphocytes/immunology , Lymphocytes/pathology , Mice , Mice, Knockout , Signal Transduction/drug effects
11.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31816636

ABSTRACT

Lung group 2 innate lymphoid cells (ILC2s) drive allergic inflammation and promote tissue repair. ILC2 development is dependent on the transcription factor retinoic acid receptor-related orphan receptor (RORα), which is also expressed in common ILC progenitors. To elucidate the developmental pathways of lung ILC2s, we generated RORα lineage tracer mice and performed single-cell RNA sequencing, flow cytometry, and functional analyses. In adult mouse lungs, we found an IL-18Rα+ST2- population different from conventional IL-18Rα-ST2+ ILC2s. The former was GATA-3intTcf7EGFP+Kit+, produced few cytokines, and differentiated into multiple ILC lineages in vivo and in vitro. In neonatal mouse lungs, three ILC populations were identified, namely an ILC progenitor population similar to that in adult lungs and two distinct effector ILC2 subsets that differentially produced type 2 cytokines and amphiregulin. Lung ILC progenitors might actively contribute to ILC-poiesis in neonatal and inflamed adult lungs. In addition, neonatal lung ILC2s include distinct proinflammatory and tissue-repairing subsets.


Subject(s)
Immunity, Innate/immunology , Lung/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Stem Cells/immunology , Amphiregulin/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cytokines/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Single-Cell Analysis/methods
12.
J Immunol ; 203(12): 3209-3215, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31676672

ABSTRACT

Innate lymphoid cells (ILCs) are critical for host defense and tissue repair but can also contribute to chronic inflammatory diseases. The transcription factor RORα is required for ILC2 development but is also highly expressed by other ILC subsets where its function remains poorly defined. We previously reported that Rorasg/sg bone marrow chimeric mice (C57BL/6J) were protected from Salmonella-induced intestinal fibrosis due to defective ILC3 responses. In this study, single-cell RNA analysis of ILCs isolated from inflamed tissues indicates that RORα perturbation led to a reduction in ILC3 lineages. Furthermore, residual Rorasg/sg ILC3s have decreased expression of key signature genes, including Rorc and activating cytokine receptors. Collectively, our data suggest that RORα plays a key role in preserving functional ILC3s by modulating their ability to integrate environmental cues to efficiently produce cytokines.


Subject(s)
Enteritis/etiology , Enteritis/metabolism , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Animals , Biomarkers , Chronic Disease , Disease Models, Animal , Enteritis/pathology , Fibrosis , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice
13.
Curr Protoc Immunol ; 125(1): e73, 2019 06.
Article in English | MEDLINE | ID: mdl-30994980

ABSTRACT

Innate lymphoid cells (ILCs) are a heterogeneous family of lymphocytes that populate barrier and non-barrier tissues. ILCs regulate immune responses to pathogens and commensals but also sustain metabolic homeostasis, tissue remodeling after injury and establish dialogue with the nervous system. ILCs rapidly become activated in the absence of adaptive antigen receptors by responding to signaling molecules provided by hematopoietic or non-hematopoietic cells. Here we provide protocols designed for processing the lung, liver, small intestine, bone marrow, mediastinal and mesenteric lymph nodes in order to obtain a purified leukocyte fraction of cells, in which ILC2 enrichment is optimized. In addition, we describe in detail the methodologies used to activate ILC2s and the assays necessary for the detection of their effector cytokines. We highlight the differences in ILC2 characterization within distinct tissues that we have recently identified. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Immunity, Innate , Lymphocytes/immunology , Animals , Bone Marrow/immunology , Intestine, Small/immunology , Liver/immunology , Lung/immunology , Lymph Nodes/immunology , Mice
14.
PLoS One ; 14(3): e0214286, 2019.
Article in English | MEDLINE | ID: mdl-30913260

ABSTRACT

Epidemiological studies have shown sex differences in prevalence of non-allergic asthma. Recent reports demonstrated negative effects of androgen signaling on group 2 innate lymphoid cells (ILC2s), explaining a potential mechanism behind sex bias in asthma prevalence. To further understand sex-related differences in ILC2 functions and ILC2 intrinsic or lung environmental mechanisms behind it, we have investigated the effects of sex and age on lung ILC2 function, the amounts of ILC2-activating cytokines in the lung and gene expression profiles of male and female ILC2s. Flow cytometric analyses of naive male and female mouse lung ILC2s showed no difference in their numbers. However, upon three daily intranasal IL-33 injections, lung ILC2s in postpubertal female mice expanded to a greater degree than male counterpart. In line with in vivo results, purified female mouse lung ILC2s produced more cytokines than male ILC2s upon in vitro stimulation. Gene expression profiles of purified naïve male and female ILC2s differed in 4% of the genes, and gene set enrichment analysis showed that female ILC2s are enriched for gene signatures of memory T cells. We did not observe similar degree of differences between female and male ILC2s after IL-33 stimulation. ILC2-activating cytokines including IL-33, IL-7 and TSLP were more highly expressed in whole lung homogenate samples prepared from naïve post pubertal female mouse lung than male mouse lung. Moreover, the differences in responsiveness of male and female ILC2s to IL-33 were not affected in IL-33-deficient mice. These results suggest that female ILC2s are more readily activated than male ILC2s due to their gene expression at the naïve state, which is potentially influenced by the lung environment.


Subject(s)
Cytokines/metabolism , Lung/metabolism , Lymphocytes/metabolism , Transcriptome , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/analysis , Female , Immunity, Innate/drug effects , Interleukin-33/pharmacology , Isoflurane/pharmacology , Lung/immunology , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Transcriptome/drug effects
15.
Immunol Rev ; 283(1): 41-53, 2018 05.
Article in English | MEDLINE | ID: mdl-29664572

ABSTRACT

Immunological memory, traditionally thought to belong to T and B cells, has now been extended to innate lymphocytes, including NK cells and ILC2s, myeloid cells such as macrophages, also termed "trained immunity" and more recently to epithelial stem cells. In this review, we discuss the mechanisms underlying memory generation on ILC2s and speculate about their potential role in human allergic diseases, such as asthma. Moreover, we examine the relevance of the spontaneous ILC2 activation in the lung during the neonatal period in order to efficiently respond to stimuli later in life. These "training" of neonatal ILC2s may have an impact on the generation of memory ILC2s in the adulthood.


Subject(s)
Immunity, Innate , Immunologic Memory , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Age Factors , Animals , Asthma/etiology , Asthma/metabolism , Cell Differentiation/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Subsets/cytology
16.
Sci Rep ; 8(1): 2924, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29440650

ABSTRACT

Type 2 innate lymphoid cells (ILC2) potentiate immune responses, however, their role in mediating adaptive immunity in cancer has not been assessed. Here, we report that mice genetically lacking ILC2s have significantly increased tumour growth rates and conspicuously higher frequency of circulating tumour cells (CTCs) and resulting metastasis to distal organs. Our data support the model that IL-33 dependent tumour-infiltrating ILC2s are mobilized from the lungs and other tissues through chemoattraction to enter tumours, and subsequently mediate tumour immune-surveillance by cooperating with dendritic cells to promote adaptive cytolytic T cell responses. We conclude that ILC2s play a fundamental, yet hitherto undescribed role in enhancing anti-cancer immunity and controlling tumour metastasis.


Subject(s)
Immunity, Innate , Lymphocytes/immunology , Models, Biological , Neoplasms/immunology , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-33/metabolism , Mice , Neoplasm Metastasis , Neoplasms/metabolism
17.
J Allergy Clin Immunol ; 141(1): 300-310.e11, 2018 01.
Article in English | MEDLINE | ID: mdl-28392332

ABSTRACT

BACKGROUND: Bronchial epithelial barrier leakiness and type 2 innate lymphoid cells (ILC2s) have been separately linked to asthma pathogenesis; however, the influence of ILC2s on the bronchial epithelial barrier has not been investigated previously. OBJECTIVE: We investigated the role of ILC2s in the regulation of bronchial epithelial tight junctions (TJs) and barrier function both in bronchial epithelial cells of asthmatic patients and healthy subjects and general innate lymphoid cell- and ILC2-deficient mice. METHODS: Cocultures of human ILC2s and bronchial epithelial cells were used to determine transepithelial electrical resistance, paracellular flux, and TJ mRNA and protein expressions. The effect of ILC2s on TJs was examined by using a murine model of IL-33-induced airway inflammation in wild-type, recombination-activating gene 2 (Rag2)-/-, Rag2-/-Il2rg-/-, and Rorasg/sg mice undergoing bone marrow transplantation to analyze the in vivo relevance of barrier disruption by ILC2s. RESULTS: ILC2s significantly impaired the epithelial barrier, as demonstrated by reduced transepithelial electrical resistance and increased fluorescein isothiocyanate-dextran permeability in air-liquid interface cultures of human bronchial epithelial cells. This was in parallel to decreased mRNAs and disrupted protein expression of TJ proteins and was restored by neutralization of IL-13. Intranasal administration of recombinant IL-33 to wild-type and Rag2-/- mice lacking T and B cells triggered TJ disruption, whereas Rag2-/-Il2rg-/- and Rorasg/sg mice undergoing bone marrow transplantation that lack ILC2s did not show any barrier leakiness. Direct nasal administration of IL-13 was sufficient to induce deficiency in the TJ barrier in the bronchial epithelium of mice in vivo. CONCLUSION: These data highlight an essential mechanism in asthma pathogenesis by demonstrating that ILC2s are responsible for bronchial epithelial TJ barrier leakiness through IL-13.


Subject(s)
Asthma/immunology , Asthma/metabolism , Immunity, Innate , Interleukin-13/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Tight Junctions/metabolism , Animals , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Interleukin-13/antagonists & inhibitors , Mice , Mice, Transgenic , Mucus/metabolism , Respiratory Mucosa/pathology
18.
Trends Immunol ; 38(6): 423-431, 2017 06.
Article in English | MEDLINE | ID: mdl-28416448

ABSTRACT

Immunological memory has long been described as a property of the adaptive immune system that results in potent responses on exposure to an antigen encountered previously. While this definition appears to exclude cells that do not express antigen receptors, recent studies have shown that innate immune cells, including natural killer (NK) cells, macrophages, and, more recently, group 2 innate lymphoid cells (ILC2s) can record previous activations and respond more vigorously on reactivation. Here we review the similarities and differences between these forms of memory and the underlying mechanisms. Based on these insights, we propose to revise the definition of immunological memory, as the capacity to remember being previously activated and respond more efficiently on reactivation regardless of antigen specificity.


Subject(s)
Immunologic Memory , Killer Cells, Natural/immunology , Lymphocytes/physiology , Macrophages/immunology , Animals , Cell Differentiation , Cytokines/metabolism , Humans , Immunity, Innate/genetics , Lymphocyte Activation , Th2 Cells/immunology , Transcriptome
20.
Immunity ; 45(1): 198-208, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27421705

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) in the lung are stimulated by inhaled allergens. ILC2s do not directly recognize allergens but they are stimulated by cytokines including interleukin (IL)-33 released by damaged epithelium. In response to allergens, lung ILC2s produce T helper 2 cell type cytokines inducing T cell-independent allergic lung inflammation. Here we examined the fate of lung ILC2s upon allergen challenges. ILC2s proliferated and secreted cytokines upon initial stimulation with allergen or IL-33, and this phase was followed by a contraction phase as cytokine production ceased. Some ILC2s persisted long after the resolution of the inflammation as allergen-experienced ILC2s and responded to unrelated allergens more potently than naive ILC2s, mediating severe allergic inflammation. The allergen-experienced ILC2s exhibited a gene expression profile similar to that of memory T cells. The memory-like properties of allergen-experienced ILC2s may explain why asthma patients are often sensitized to multiple allergens.


Subject(s)
Hypersensitivity/immunology , Immunity, Innate , Lymphocytes/immunology , Pneumonia/immunology , Respiratory Mucosa/immunology , Allergens/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Immunologic Memory , Inflammation Mediators/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...