Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38487999

ABSTRACT

Allergic asthma generally starts during early life and is linked to substantial tissue remodeling and lung dysfunction. Although angiogenesis is a feature of the disrupted airway, the impact of allergic asthma on the pulmonary microcirculation during early life is unknown. Here, using quantitative imaging in precision-cut lung slices (PCLSs), we report that exposure of neonatal mice to house dust mite (HDM) extract disrupts endothelial cell/pericyte interactions in adventitial areas. Central to the blood vessel structure, the loss of pericyte coverage was driven by mast cell (MC) proteases, such as tryptase, that can induce pericyte retraction and loss of the critical adhesion molecule N-cadherin. Furthermore, spatial transcriptomics of pediatric asthmatic endobronchial biopsies suggests intense vascular stress and remodeling linked with increased expression of MC activation pathways in regions enriched in blood vessels. These data provide previously unappreciated insights into the pathophysiology of allergic asthma with potential long-term vascular defects.


Subject(s)
Asthma , Mast Cells , Humans , Child , Animals , Mice , Mast Cells/pathology , Pericytes/metabolism , Endothelial Cells/metabolism , Asthma/pathology , Lung/pathology , Allergens , Pyroglyphidae , Disease Models, Animal
2.
Nature ; 599(7883): 125-130, 2021 11.
Article in English | MEDLINE | ID: mdl-34671159

ABSTRACT

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Subject(s)
Homeostasis , Intestines/immunology , Intestines/physiology , Neuroglia/immunology , Neuroglia/physiology , Regeneration , Adventitia/immunology , Adventitia/parasitology , Animals , Chemokine CXCL10/immunology , Duodenum/immunology , Duodenum/parasitology , Duodenum/pathology , Duodenum/physiology , Female , Gliosis , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Intestines/parasitology , Intestines/pathology , Male , Mice , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Signal Transduction/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Strongylida Infections/pathology
3.
Methods Mol Biol ; 2121: 99-114, 2020.
Article in English | MEDLINE | ID: mdl-32147790

ABSTRACT

ILC2s are a rare innate cell population capable of rapidly producing type 2 cytokines prior to the recruitment and expansion of adaptive type 2 T helper cells. As a result, they are implicated in the pathogenesis of many type-2 immune-mediated diseases, including allergic airway inflammation. Here we describe methods for interrogating and analyzing ILC2 biology in the context of allergic airway inflammation, such as flow cytometric analysis of mouse and human ILC2s as well as live imaging of pulmonary ILC2s.


Subject(s)
Asthma/immunology , Flow Cytometry/methods , Immunity, Innate , Inflammation/immunology , Intravital Microscopy/methods , Lung/cytology , Lymphocyte Subsets/immunology , Animals , Asthma/pathology , Bronchoalveolar Lavage , Cytokines/metabolism , Humans , Inflammation/pathology , Intravital Microscopy/instrumentation , Lung/diagnostic imaging , Lung/immunology , Lung/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Subsets/cytology , Mice
4.
Sci Immunol ; 4(36)2019 06 07.
Article in English | MEDLINE | ID: mdl-31175176

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are enriched in mucosal tissues (e.g., lung) and respond to epithelial cell-derived cytokines initiating type 2 inflammation. During inflammation, ILC2 numbers are increased in the lung. However, the mechanisms controlling ILC2 trafficking and motility within inflamed lungs remain unclear and are crucial for understanding ILC2 function in pulmonary immunity. Using several approaches, including lung intravital microscopy, we demonstrate that pulmonary ILC2s are highly dynamic, exhibit amoeboid-like movement, and aggregate in the lung peribronchial and perivascular spaces. They express distinct chemokine receptors, including CCR8, and actively home to CCL8 deposits located around the airway epithelium. Within lung tissue, ILC2s were particularly motile in extracellular matrix-enriched regions. We show that collagen-I drives ILC2 to markedly change their morphology by remodeling their actin cytoskeleton to promote environmental exploration critical for regulating eosinophilic inflammation. Our study provides previously unappreciated insights into ILC2 migratory patterns during inflammation and highlights the importance of environmental guidance cues in the lung in controlling ILC2 dynamics.


Subject(s)
Lung/immunology , Lymphocytes/immunology , Animals , Cell Movement/drug effects , Collagen/immunology , Eosinophils/immunology , Extracellular Matrix/immunology , Female , Fibronectins/immunology , Humans , Immunity, Innate , Inflammation/immunology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Mice, Inbred BALB C , Mice, Transgenic , Recombinant Proteins/pharmacology
5.
Front Immunol ; 10: 3114, 2019.
Article in English | MEDLINE | ID: mdl-32038635

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are enriched at mucosal sites, including the lung, and play a central role in type 2 immunity and maintaining tissue homeostasis. As a result, since their discovery in 2010, research into ILC2s has increased markedly. Numerous strategies have been used to define ILC2s by flow cytometry, often utilizing different combinations of surface markers despite their expression being variable and context-dependent. In this study, we sought to generate a comprehensive characterization of pulmonary ILC2s, identifying stable and context specific markers from different pulmonary compartments following different airway exposures in different strains of mice. Our analysis revealed that pulmonary ILC2 surface marker phenotype is heterogeneous and is influenced by mouse strain, pulmonary location, in vivo treatment/exposure and ex vivo stimulation. Therefore, we propose that a lineage negative cell expressing CD45 and Gata3 defines an ILC2, and subsequent surface marker expression should be used to describe their phenotype in context-specific scenarios.


Subject(s)
Biomarkers , Immunity, Innate , Immunophenotyping , Lymphocytes/immunology , Lymphocytes/metabolism , Allergens/immunology , Animals , Female , Immunohistochemistry , Interleukin-33/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity
6.
Sci Immunol ; 3(27)2018 09 07.
Article in English | MEDLINE | ID: mdl-30194239

ABSTRACT

Airway hyperresponsiveness (AHR) is a critical feature of wheezing and asthma in children, but the initiating immune mechanisms remain unconfirmed. We demonstrate that both recombinant interleukin-33 (rIL-33) and allergen [house dust mite (HDM) or Alternaria alternata] exposure from day 3 of life resulted in significantly increased pulmonary IL-13+CD4+ T cells, which were indispensable for the development of AHR. In contrast, adult mice had a predominance of pulmonary LinnegCD45+CD90+IL-13+ type 2 innate lymphoid cells (ILC2s) after administration of rIL-33. HDM exposure of neonatal IL-33 knockout (KO) mice still resulted in AHR. However, neonatal CD4creIL-13 KO mice (lacking IL-13+CD4+ T cells) exposed to allergen from day 3 of life were protected from AHR despite persistent pulmonary eosinophilia, elevated IL-33 levels, and IL-13+ ILCs. Moreover, neonatal mice were protected from AHR when inhaled Acinetobacter lwoffii (an environmental bacterial isolate found in cattle farms, which is known to protect from childhood asthma) was administered concurrent with HDM. A. lwoffii blocked the expansion of pulmonary IL-13+CD4+ T cells, whereas IL-13+ ILCs and IL-33 remained elevated. Administration of A. lwoffii mirrored the findings from the CD4creIL-13 KO mice, providing a translational approach for disease protection in early life. These data demonstrate that IL-13+CD4+ T cells, rather than IL-13+ ILCs or IL-33, are critical for inception of allergic AHR in early life.


Subject(s)
Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-13/immunology , Respiratory Hypersensitivity/immunology , Acinetobacter/immunology , Alternaria/immunology , Animals , Animals, Newborn , Female , Interleukin-33/genetics , Male , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Pyroglyphidae/immunology
7.
Cell Host Microbe ; 22(4): 484-493.e5, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29024642

ABSTRACT

Immunity to intestinal helminth infections has been well studied, but the mechanism of helminth killing prior to expulsion remains unclear. Here we identify epithelial-cell-derived phospholipase A2 group 1B (PLA2g1B) as a host-derived endogenous anthelmintic. PLA2g1B is elevated in resistant mice and is responsible for killing tissue-embedded larvae. Despite comparable activities of other essential type-2-dependent immune mechanisms, Pla2g1b-/- mice failed to expel the intestinal helminths Heligmosomoides polygyrus or Nippostrongylus brasiliensis. Expression of Pla2g1b by epithelial cells was dependent upon intestinal microbiota, adaptive immunity, and common-gamma chain-dependent signaling. Notably, Pla2g1b was downregulated in susceptible mice and inhibited by IL-4R-signaling in vitro, uncoupling parasite killing from expulsion mechanisms. Resistance was restored in Pla2g1b-/- mice by treating infective H. polygyrus L3 larvae with PLA2g1B, which reduced larval phospholipid abundance. These findings uncover epithelial-cell-derived Pla2g1b as an essential mediator of helminth killing, highlighting a previously overlooked mechanism of anti-helminth immunity.


Subject(s)
Group IB Phospholipases A2/immunology , Intestinal Mucosa/immunology , Nematospiroides dubius/immunology , Nippostrongylus/immunology , Phospholipids/metabolism , Strongylida Infections/immunology , Adaptive Immunity , Animals , Gastrointestinal Microbiome/immunology , Group IB Phospholipases A2/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Larva/immunology , Mice , Mice, Knockout , Primary Cell Culture
8.
PLoS Pathog ; 13(7): e1006536, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28759611

ABSTRACT

TPL-2 (COT, MAP3K8) kinase activates the MEK1/2-ERK1/2 MAPK signaling pathway in innate immune responses following TLR, TNFR1 and IL-1R stimulation. TPL-2 contributes to type-1/Th17-mediated autoimmunity and control of intracellular pathogens. We recently demonstrated TPL-2 reduces severe airway allergy to house dust mite by negatively regulating type-2 responses. In the present study, we found that TPL-2 deficiency resulted in resistance to Heligmosomoides polygyrus infection, with accelerated worm expulsion, reduced fecal egg burden and reduced worm fitness. Using co-housing experiments, we found resistance to infection in TPL-2 deficient mice (Map3k8-/-) was independent of microbiota alterations in H. polygyrus infected WT and Map3k8-/-mice. Additionally, our data demonstrated immunity to H. polygyrus infection in TPL-2 deficient mice was not due to dysregulated type-2 immune responses. Genome-wide analysis of intestinal tissue from infected TPL-2-deficient mice identified elevated expression of genes involved in chemotaxis and homing of leukocytes and cells, including Ccl24 and alternatively activated genes. Indeed, Map3k8-/-mice had a significant influx of eosinophils, neutrophils, monocytes and Il4GFP+ T cells. Conditional knockout experiments demonstrated that specific deletion of TPL-2 in CD11c+ cells, but not Villin+ epithelial cells, LysM+ myeloid cells or CD4+ T cells, led to accelerated resistance to H. polygyrus. In line with a central role of CD11c+ cells, CD11c+ CD11b+ cells isolated from TPL-2-deficient mice had elevated Ccl24. Finally, Ccl24 neutralization in TPL-2 deficient mice significantly decreased the expression of Arg1, Retnla, Chil3 and Ear11 correlating with a loss of resistance to H. polygyrus. These observations suggest that TPL-2-regulated Ccl24 in CD11c+CD11b+ cells prevents accelerated type-2 mediated immunity to H. polygyrus. Collectively, this study identifies a previously unappreciated role for TPL-2 controlling immune responses to H. polygyrus infection by restricting Ccl24 production.


Subject(s)
Chemokine CCL24/immunology , MAP Kinase Kinase Kinases/immunology , Nematospiroides dubius/immunology , Proto-Oncogene Proteins/immunology , Strongylida Infections/immunology , Animals , Chemokine CCL24/genetics , Female , Humans , Immunity, Innate , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/genetics , Nematospiroides dubius/physiology , Proto-Oncogene Proteins/genetics , Strongylida Infections/enzymology , Strongylida Infections/genetics , Strongylida Infections/parasitology , Th2 Cells/immunology
9.
J Exp Med ; 214(6): 1809-1826, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28507062

ABSTRACT

Immunity to intestinal helminth infections requires the rapid activation of T helper 2 cells (Th2 cells). However, simultaneous expansion of CD4+Foxp3+ regulatory T cells (T reg cells) impedes protective responses, resulting in chronic infections. The ratio between T reg and effector T cells can therefore determine the outcome of infection. The redifferentiation of T reg cells into Th cells has been identified in hyperinflammatory diseases. In this study, we asked whether ex-T reg Th2 cells develop and contribute to type-2 immunity. Using multigene reporter and fate-reporter systems, we demonstrate that a significant proportion of Th2 cells derive from Foxp3+ cells after Heligmosomoides polygyrus infection and airway allergy. Ex-Foxp3 Th2 cells exhibit characteristic Th2 effector functions and provide immunity to H. polygyrus Through selective deletion of Il4ra on Foxp3+ cells, we further demonstrate IL-4 is required for the development of ex-Foxp3 Th2 cells. Collectively, our findings indicate that converting T reg cells into Th2 cells could concomitantly enhance Th2 cells and limit T reg cell-mediated suppression.


Subject(s)
Forkhead Transcription Factors/metabolism , Immunity , Interleukin-4/metabolism , Intestines/immunology , Intestines/parasitology , Nematospiroides dubius/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Polarity , Gene Expression Profiling , Immunity/genetics , Mice, Inbred C57BL , Receptors, Interleukin-4/metabolism , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Lymphocytes, Regulatory/immunology
10.
PLoS Pathog ; 12(8): e1005783, 2016 08.
Article in English | MEDLINE | ID: mdl-27487182

ABSTRACT

Persistent TH2 cytokine responses following chronic helminth infections can often lead to the development of tissue pathology and fibrotic scarring. Despite a good understanding of the cellular mechanisms involved in fibrogenesis, there are very few therapeutic options available, highlighting a significant medical need and gap in our understanding of the molecular mechanisms of TH2-mediated immunopathology. In this study, we found that the Map3 kinase, TPL-2 (Map3k8; Cot) regulated TH2-mediated intestinal, hepatic and pulmonary immunopathology following Schistosoma mansoni infection or S. mansoni egg injection. Elevated inflammation, TH2 cell responses and exacerbated fibrosis in Map3k8-/-mice was observed in mice with myeloid cell-specific (LysM) deletion of Map3k8, but not CD4 cell-specific deletion of Map3k8, indicating that TPL-2 regulated myeloid cell function to limit TH2-mediated immunopathology. Transcriptional and metabolic assays of Map3k8-/-M2 macrophages identified that TPL-2 was required for lipolysis, M2 macrophage activation and the expression of a variety of genes involved in immuno-regulatory and pro-fibrotic pathways. Taken together this study identified that TPL-2 regulated TH2-mediated inflammation by supporting lipolysis and M2 macrophage activation, preventing TH2 cell expansion and downstream immunopathology and fibrosis.


Subject(s)
Cell Differentiation/immunology , Lipolysis/immunology , MAP Kinase Kinase Kinases/immunology , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Fibrosis , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipolysis/genetics , MAP Kinase Kinase Kinases/genetics , Macrophages/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/pathology , Th2 Cells/pathology
11.
PLoS Pathog ; 11(7): e1004994, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26147567

ABSTRACT

Parasitic helminths establish chronic infections in mammalian hosts. Helminth/Plasmodium co-infections occur frequently in endemic areas. However, it is unclear whether Plasmodium infections compromise anti-helminth immunity, contributing to the chronicity of infection. Immunity to Plasmodium or helminths requires divergent CD4+ T cell-driven responses, dominated by IFNγ or IL-4, respectively. Recent literature has indicated that Th cells, including Th2 cells, have phenotypic plasticity with the ability to produce non-lineage associated cytokines. Whether such plasticity occurs during co-infection is unclear. In this study, we observed reduced anti-helminth Th2 cell responses and compromised anti-helminth immunity during Heligmosomoides polygyrus and Plasmodium chabaudi co-infection. Using newly established triple cytokine reporter mice (Il4gfpIfngyfpIl17aFP635), we demonstrated that Il4gfp+ Th2 cells purified from in vitro cultures or isolated ex vivo from helminth-infected mice up-regulated IFNγ following adoptive transfer into Rag1-/- mice infected with P. chabaudi. Functionally, Th2 cells that up-regulated IFNγ were transcriptionally re-wired and protected recipient mice from high parasitemia. Mechanistically, TCR stimulation and responsiveness to IL-12 and IFNγ, but not type I IFN, was required for optimal IFNγ production by Th2 cells. Finally, blockade of IL-12 and IFNγ during co-infection partially preserved anti-helminth Th2 responses. In summary, this study demonstrates that Th2 cells retain substantial plasticity with the ability to produce IFNγ during Plasmodium infection. Consequently, co-infection with Plasmodium spp. may contribute to the chronicity of helminth infection by reducing anti-helminth Th2 cells and converting them into IFNγ-secreting cells.


Subject(s)
Coinfection/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Malaria/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nematospiroides dubius/immunology , Plasmodium chabaudi/immunology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...