Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 49(1): 144-156, 2019 01.
Article in English | MEDLINE | ID: mdl-29762870

ABSTRACT

Influenza virus infection is an important cause of severe asthma exacerbations, but it remains unclear how a Th1-mediated antiviral response triggers a prototypical Th2 disease. We investigated CD4+ T cells and group 2 innate lymphoid cells (ILC2s) in influenza virus-infected mice. We found that ILC2s accumulated in the lung rapidly after influenza virus infection, but the induction of IL-5 and IL-13 secretion was delayed and concomitant with T cell activation. In an influenza-induced exacerbation of allergic airway inflammation model we noticed an initial reduction of ILC2 numbers and cytokine production in broncho-alveolar lavage compared to chronic house dust mite (HDM)-mediated airway inflammation alone. ILC2s phenotype was characterized by low T1/ST2, ICOS, KLRG1, and CD25 expression, resembling naïve ILC2s. The contribution of ILC2s to type 2 cytokine production in the early stage of the influenza-induced exacerbation was limited. In contrast, T cells showed increased IL-4 and IL-5 production when exposed to both HDM and influenza virus. Upon virus clearance, ILC2s regained an activated T1/ST2high ICOShigh KLRG1high CD25high phenotype paired with cytokine production and were major contributors to the type 2 cytokine milieu. Collectively, our data indicate that both T cells and ILC2s contribute to influenza-induced exacerbation of allergic airway inflammation, but with different kinetics.


Subject(s)
GATA3 Transcription Factor/metabolism , Hypersensitivity/immunology , Inflammation/immunology , Influenza, Human/immunology , Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Respiratory System/immunology , Th2 Cells/immunology , Animals , Antigens, Dermatophagoides/immunology , Cells, Cultured , Cytokines/metabolism , Disease Progression , GATA3 Transcription Factor/genetics , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Pyroglyphidae
3.
J Allergy Clin Immunol ; 142(6): 1793-1807, 2018 12.
Article in English | MEDLINE | ID: mdl-29486229

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are major producers of the cytokines driving allergic asthma, and increased ILC2 numbers have been detected in blood and sputum of asthmatic patients. Asthma susceptibility has a strong genetic component, but the underlying mechanisms and whether asthma genetics affect ILC2 biology remain unclear. OBJECTIVE: We sought to study the ILC2 transcriptome and epigenome during airway inflammation (AI) to couple these to genes and genetic variants associated with asthma pathogenesis. METHODS: Mice harboring a reporter for the key ILC2 transcription factor GATA-3 were subjected to IL-33-driven AI, and ILC2s were isolated from bronchoalveolar lavage fluid and mediastinal lymph nodes. Human ILC2s were purified from peripheral blood and activated in vitro. We used RNA sequencing, genome-wide identification of histone-3 lysine-4 dimethylation-marked chromatin, and computational approaches to study the ILC2 transcriptome and epigenome. RESULTS: Activated ILC2s in mice displayed a tissue-specific gene expression signature that emerged from remarkably similar epigenomes. We identified superenhancers implicated in controlling ILC2 identity and asthma-associated genes. More than 300 asthma-associated genetic polymorphisms identified in genome-wide association studies localized to H3K4Me2+ gene regulatory elements in ILC2s. A refined set of candidate causal asthma-associated variants was uniquely enriched in ILC2, but not TH2 cell, regulatory regions. CONCLUSIONS: ILC2s in AI use a flexible epigenome that couples adaptation to new microenvironments with functional plasticity. Importantly, we reveal strong correlations between gene regulatory mechanisms in ILC2s and the genetic basis of asthma, supporting a pathogenic role for ILC2s in patients with allergic asthma.


Subject(s)
Asthma/genetics , Asthma/immunology , Genetic Predisposition to Disease , Lymphocytes/immunology , Animals , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , Genome , Humans , Immunity, Innate , Mice , Regulatory Sequences, Nucleic Acid , Transcriptome
4.
Front Immunol ; 8: 1684, 2017.
Article in English | MEDLINE | ID: mdl-29250067

ABSTRACT

Group 2 innate lymphoid cells (ILC2) are implicated in allergic asthma as an early innate source of the type 2 cytokines IL-5 and IL-13. However, their induction in house dust mite (HDM)-mediated airway inflammation additionally requires T cell activation. It is currently unknown whether phenotypic differences exist between ILC2s that are activated in a T cell-dependent or T cell-independent fashion. Here, we compared ILC2s in IL-33- and HDM-driven airway inflammation. Using flow cytometry, we found that surface expression levels of various markers frequently used to identify ILC2s were dependent on their mode of activation, highly variable over time, and differed between tissue compartments, including bronchoalveolar lavage (BAL) fluid, lung, draining lymph nodes, and spleen. Whereas in vivo IL-33-activated BAL fluid ILC2s exhibited an almost uniform CD25+CD127+T1/ST2+ICOS+KLRG1+ phenotype, at a comparable time point after HDM exposure BAL fluid ILC2s had a very heterogeneous surface marker phenotype. A major fraction of HDM-activated ILC2s were CD25lowCD127+T1/ST2low ICOSlowKLRG1low, but nevertheless had the capacity to produce large amounts of type 2 cytokines. HDM-activated CD25low ILC2s in BAL fluid and lung rapidly reverted to CD25high ILC2s upon in vivo stimulation with IL-33. Genome-wide transcriptional profiling of BAL ILC2s revealed ~1,600 differentially expressed genes: HDM-stimulated ILC2s specifically expressed genes involved in the regulation of adaptive immunity through B and T cell interactions, whereas IL-33-stimulated ILC2s expressed high levels of proliferation-related and cytokine genes. In both airway inflammation models ILC2s were present in the lung submucosa close to epithelial cells, as identified by confocal microscopy. In chronic HDM-driven airway inflammation ILC2s were also found inside organized cellular infiltrates near T cells. Collectively, our findings show that ILC2s are phenotypically more heterogeneous than previously thought, whereby their surface marker and gene expression profile are highly dynamic.

5.
Sci Rep ; 7(1): 8580, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819261

ABSTRACT

Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.


Subject(s)
Antigen-Presenting Cells/immunology , Leukocytes, Mononuclear/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Ferrets , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macaca fascicularis , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Microscopy, Confocal , Muscle Cells/immunology , Muscle Cells/metabolism , Muscle Cells/virology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vaccinia virus/genetics , Vaccinia virus/physiology
6.
Methods Mol Biol ; 1559: 169-183, 2017.
Article in English | MEDLINE | ID: mdl-28063044

ABSTRACT

Allergic asthma is a chronic inflammatory lung disease mediated by type 2 cytokines produced by T helper 2 (Th2) cells as well as the recently discovered group 2 innate lymphoid cells (ILC2). Due to a lack of unique markers, the accurate phenotypic characterization and quantification of ILC2 requires a comprehensive panel of fluorescently labeled antibodies. The markers that are currently used to characterize ILC2 have not been standardized and often vary between research groups, which poses significant challenges when comparing data. Intranasal administration of the pro-inflammatory cytokine IL-33 in mice is associated with strong, Th2 cell-independent ILC2 activation. ILC2 are also activated in mouse models of allergic asthma based on the physiologically relevant house dust mite (HDM) allergen, which parallel eosinophilic airway inflammation observed in asthma patients. Here, we describe the analysis of ILC2 by flow cytometry in these two commonly used allergic airway inflammation models in the mouse.


Subject(s)
Allergens/administration & dosage , Asthma/immunology , Hypersensitivity/immunology , Immunophenotyping/methods , Interleukin-33/administration & dosage , Lymphocytes/immunology , Administration, Intranasal , Animals , Antibodies/chemistry , Asthma/chemically induced , Asthma/pathology , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Lineage/immunology , Disease Models, Animal , Female , Flow Cytometry/instrumentation , Humans , Hypersensitivity/pathology , Lung/immunology , Lung/pathology , Lymphocyte Activation , Lymphocytes/classification , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Staining and Labeling/methods
7.
J Allergy Clin Immunol ; 140(4): 1079-1089, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28111308

ABSTRACT

BACKGROUND: Allergic asthma is characterized by a TH2 response induced by dendritic cells (DCs) that present inhaled allergen. Although the mechanisms by which they instruct TH2 differentiation are still poorly understood, expression of the Notch ligand Jagged on DCs has been implicated in this process. OBJECTIVE: We sought to establish whether Notch signaling induced by DCs is critical for house dust mite (HDM)-driven allergic airway inflammation (AAI) in vivo. METHODS: The induction of Notch ligand expression on DC subsets by HDM was quantified by using quantitative real-time PCR. We used an HDM-driven asthma mouse model to compare the capacity of Jagged 1 and Jagged 2 single- and double-deficient DCs to induce AAI. In addition, we studied AAI in mice with a T cell-specific deletion of recombination signal-binding protein for immunoglobulin Jκ region (RBPJκ), a downstream effector of Notch signaling. RESULTS: HDM exposure promoted expression of Jagged 1, but not Jagged 2, on DCs. In agreement with published findings, in vitro-differentiated and HDM-pulsed Jagged 1 and Jagged 2 double-deficient DCs lacked the capacity to induce AAI. However, after in vivo intranasal sensitization and challenge with HDM, DC-specific Jagged 1 or Jagged 2 single- or double-deficient mice had eosinophilic airway inflammation and a TH2 cell activation phenotype that was not different from that in control littermates. In contrast, RBPJκ-deficient mice did not experience AAI and airway hyperreactivity. CONCLUSION: Our results show that the Notch signaling pathway in T cells is crucial for the induction of TH2-mediated AAI in an HDM-driven asthma model but that expression of Jagged 1 or Jagged 2 on DCs is not required.


Subject(s)
Dendritic Cells/immunology , Hypersensitivity/immunology , Jagged-1 Protein/metabolism , Jagged-2 Protein/metabolism , Receptors, Notch/metabolism , Th2 Cells/immunology , Animals , Antigens, Dermatophagoides/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Jagged-1 Protein/genetics , Jagged-2 Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae/immunology , Signal Transduction
8.
Eur J Immunol ; 46(6): 1392-403, 2016 06.
Article in English | MEDLINE | ID: mdl-27062360

ABSTRACT

Allergic asthma is a chronic inflammation of the airways mediated by an adaptive type 2 immune response. Upon allergen exposure, group 2 innate lymphoid cells (ILC2s) can be rapidly activated and represent an early innate source of IL-5 and IL-13. Here, we used a house dust mite (HDM)-driven asthma mouse model to study the induction of ILC2s in allergic airway inflammation. In BALF, lungs, and lymph nodes, ILC2 activation is critically dependent on prior sensitization with HDM. Importantly, T cells are required for ILC2 induction, whereby T-cell activation precedes ILC2 induction. During HDM-driven allergic airway inflammation the accumulation of ILC2s in BALF is IL-33 independent, although infiltrating ILC2s produce less cytokines in Il33(-/-) mice. Transfer of in vitro polarized OVA-specific OT-II Th2 cells alone or in combination with Th17 cells followed by OVA and HDM challenge is not sufficient to induce ILC2, despite significant eosinophilic inflammation and T-cell activation. In this asthma model, ILC2s are therefore not an early source of Th2 cytokines, but rather contribute to type 2 inflammation in which Th2 cells play a key role. Taken together, ILC2 induction in HDM-mediated allergic airway inflammation in mice critically depends on activation of T cells.


Subject(s)
Allergens/immunology , Asthma/etiology , Immunity, Innate , Pyroglyphidae/immunology , T-Lymphocyte Subsets/immunology , Animals , Asthma/metabolism , Asthma/pathology , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Immunophenotyping , Inflammation Mediators , Lymphocyte Activation/immunology , Lymphocyte Count , Mice , Mice, Knockout , Phenotype , T-Lymphocyte Subsets/metabolism
9.
Immunology ; 140(3): 281-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23866009

ABSTRACT

Although allergic asthma is a heterogeneous disease, allergen-specific T helper 2 (Th2) cells producing the key cytokines involved in type 2 inflammation, interleukin-4 (IL-4), IL-5 and IL-13, are thought to play a major role in asthma pathogenesis. This model is challenged by the recent discovery of group 2 innate lymphoid cells (ILC2) that represent a critical innate source of type 2 cytokines. These ILC2 are activated by epithelial cell-derived cytokines, including IL-25 and IL-33, which have been implicated in the initiation of asthma. In this review, we will discuss recent studies supporting a significant role for ILC2 in lung inflammation, with special attention to allergen-induced asthma.


Subject(s)
Asthma/immunology , Cytokines/metabolism , Lymphocytes/immunology , Pneumonia/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Immunity, Innate
SELECTION OF CITATIONS
SEARCH DETAIL
...