Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Discov Immunol ; 2(1): kyad009, 2023.
Article in English | MEDLINE | ID: mdl-37545765

ABSTRACT

The lung is a dynamic mucosal surface constantly exposed to a variety of immunological challenges including harmless environmental antigens, pollutants, and potentially invasive microorganisms. Dysregulation of the immune system at this crucial site is associated with a range of chronic inflammatory conditions including asthma and Chronic Pulmonary Obstructive Disease (COPD). However, due to its relative inaccessibility, our fundamental understanding of the human lung immune compartment is limited. To address this, we performed flow cytometric immune phenotyping of human lung tissue and matched blood samples that were isolated from 115 donors undergoing lung tissue resection. We provide detailed characterization of the lung mononuclear phagocyte and T cell compartments, demonstrating clear phenotypic differences between lung tissue cells and those in peripheral circulation. Additionally, we show that CD103 expression demarcates pulmonary T cells that have undergone recent TCR and IL-7R signalling. Unexpectedly, we discovered that the immune landscape from asthmatic or COPD donors was broadly comparable to controls. Our data provide a much-needed expansion of our understanding of the pulmonary immune compartment in both health and disease.

3.
Front Immunol ; 13: 906338, 2022.
Article in English | MEDLINE | ID: mdl-35958580

ABSTRACT

Schistosomiasis is a disease of global significance, with severity and pathology directly related to how the host responds to infection. The immunological narrative of schistosomiasis has been constructed through decades of study, with researchers often focussing on isolated time points, cell types and tissue sites of interest. However, the field currently lacks a comprehensive and up-to-date understanding of the immune trajectory of schistosomiasis over infection and across multiple tissue sites. We have defined schistosome-elicited immune responses at several distinct stages of the parasite lifecycle, in three tissue sites affected by infection: the liver, spleen, and mesenteric lymph nodes. Additionally, by performing RNA-seq on the livers of schistosome infected mice, we have generated novel transcriptomic insight into the development of schistosome-associated liver pathology and fibrosis across the breadth of infection. Through depletion of CD11c+ cells during peak stages of schistosome-driven inflammation, we have revealed a critical role for CD11c+ cells in the co-ordination and regulation of Th2 inflammation during infection. Our data provide an updated and high-resolution account of how host immune responses evolve over the course of murine schistosomiasis, underscoring the significance of CD11c+ cells in dictating host immunopathology against this important helminth infection.


Subject(s)
Schistosomiasis mansoni , Schistosomiasis , Animals , CD11c Antigen , Immunity , Inflammation , Mice , Schistosoma mansoni
4.
J Am Soc Mass Spectrom ; 33(4): 649-659, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35262356

ABSTRACT

Microbes exert influence across the microbiome-gut-brain axis through neurotransmitter production, induction of host immunomodulators, or the release or induction of other microbial or host molecules. Here, we used mass spectrometry imaging (MSI), a label-free imaging tool, to map molecular changes in the gut and brain in germ-free, antibiotic-treated and control mice. We determined spatial distribution and relative quantification of neurotransmitters and their precursors in response to the microbiome. Using untargeted MSI, we detected a significant change in the levels of four identified small molecules in the brains of germ-free animals compared to controls. However, antibiotic treatment induced no significant changes in these same metabolites in the brain after 1 week of treatment. This work exemplifies the utility of MSI as a tool for the study of known and discovery of novel, mediators of microbiome-gut-brain axis communication.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Brain/metabolism , Brain-Gut Axis , Gastrointestinal Microbiome/physiology , Mass Spectrometry/methods , Mice
5.
Immunohorizons ; 5(8): 721-732, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34462311

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are potent producers of type I IFN (IFN-I) during viral infection and respond to IFN-I in a positive feedback loop that promotes their function. IFN-I shapes dendritic cell responses during helminth infection, impacting their ability to support Th2 responses. However, the role of pDCs in type 2 inflammation is unclear. Previous studies have shown that pDCs are dispensable for hepatic or splenic Th2 responses during the early stages of murine infection with the trematode Schistosoma mansoni at the onset of parasite egg laying. However, during S. mansoni infection, an ongoing Th2 response against mature parasite eggs is required to protect the liver and intestine from acute damage and how pDCs participate in immune responses to eggs and adult worms in various tissues beyond acute infection remains unclear. We now show that pDCs are required for optimal Th2 cytokine production in response to S. mansoni eggs in the intestinal-draining mesenteric lymph nodes throughout infection and for egg-specific IFN-γ at later time points of infection. Further, pDC depletion at chronic stages of infection led to increased hepatic and splenic pathology as well as abrogated Th2 cell cytokine production and activation in the liver. In vitro, mesenteric lymph node pDCs supported Th2 cell responses from infection-experienced CD4+ T cells, a process dependent on pDC IFN-I responsiveness, yet independent of Ag. Together, these data highlight a previously unappreciated role for pDCs and IFN-I in maintaining and reinforcing type 2 immunity in the lymph nodes and inflamed tissue during helminth infection.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/parasitology , Cytokines/metabolism , Dendritic Cells/parasitology , Female , Flow Cytometry/methods , Host-Parasite Interactions/immunology , Lymphocyte Count , Mice, Inbred C57BL , Mice, Knockout , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/parasitology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/parasitology
6.
Front Immunol ; 11: 592325, 2020.
Article in English | MEDLINE | ID: mdl-33193437

ABSTRACT

The balance of type 1 and type 2 immune responses plays a crucial role in anti-helminth immunity and can either support chronic infection or drive type 2 mediated expulsion of the parasite. Helminth antigens and secreted molecules directly influence this balance and induce a favorable immunological environment for the parasite's survival. However, less is known if the site of infection also influences the balance of type 1 and type 2 immunity. Here, we report that tissue-specific immune responses are mounted against helminth antigens, which elicited strong IL-4 responses when injected into the skin, while the same antigen, delivered into the intestinal subserosa, induced increased IFN-γ and reduced Th2 responses. Immune responses in individual mesenteric lymph nodes that drain defined regions of the intestine furthermore displayed a site-specific pattern of type 1 and type 2 immunity after Schistosoma mansoni or Heligmosomoides polygyrus infection. S. mansoni egg-specific Th2 responses were detectable in all mesenteric lymph nodes but Th1 responses were only present in those draining the colon, while H. polygyrus infection elicited mixed Th1 and Th2 responses in the lymph nodes associated with the site of infection. Similar site-specific type 1 and type 2 immune responses were observed in the draining lymph nodes after the controlled delivery of S. mansoni eggs into different segments of the small and large intestine using microsurgical techniques. Different subsets of intestinal dendritic cells were hereby responsible for the uptake and priming of Th1 and Th2 responses against helminth antigens. Migratory CD11b+CD103- and especially CD11b+CD103+ DC2s transported S. mansoni egg antigens to the draining lymph nodes to induce Th1 and Th2 responses, while CD103+ DC1s induced only IFN-γ responses. In contrast, H. polygyrus antigens were predominantly transported by CD11b+CD103- DC2s and CD103+ DC1s and all DC subsets induced similar Th1 but weaker Th2 responses, compared to S. mansoni egg antigens. The development of adaptive anti-helminth immune responses is therefore influenced by the antigen itself, the uptake and priming characteristics of antigen-positive dendritic cell subsets and the site of infection, which shape the level of Th1 and Th2 responses in order to create a favorable immunological environment for the parasite.


Subject(s)
Antigens, Helminth/immunology , Host-Parasite Interactions/immunology , Lymph Nodes/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Biomarkers , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Host-Parasite Interactions/genetics , Immunization , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Count , Mesentery , Mice , Mice, Knockout , Schistosomiasis mansoni/parasitology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
7.
Front Immunol ; 11: 183, 2020.
Article in English | MEDLINE | ID: mdl-32117307

ABSTRACT

Methyl-CpG-binding domain-2 (Mbd2) acts as an epigenetic regulator of gene expression, by linking DNA methylation to repressive chromatin structure. Although Mbd2 is widely expressed in gastrointestinal immune cells and is implicated in regulating intestinal cancer, anti-helminth responses and colonic inflammation, the Mbd2-expressing cell types that control these responses are incompletely defined. Indeed, epigenetic control of gene expression in cells that regulate intestinal immunity is generally poorly understood, even though such mechanisms may explain the inability of standard genetic approaches to pinpoint the causes of conditions like inflammatory bowel disease. In this study we demonstrate a vital role for Mbd2 in regulating murine colonic inflammation. Mbd2-/- mice displayed dramatically worse pathology than wild type controls during dextran sulfate sodium (DSS) induced colitis, with increased inflammatory (IL-1ß+) monocytes. Profiling of mRNA from innate immune and epithelial cell (EC) populations suggested that Mbd2 suppresses inflammation and pathology via control of innate-epithelial cell crosstalk and T cell recruitment. Consequently, restriction of Mbd2 deficiency to CD11c+ dendritic cells and macrophages, or to ECs, resulted in increased DSS colitis severity. Our identification of this dual role for Mbd2 in regulating the inflammatory capacity of both CD11c+ cells and ECs highlights how epigenetic control mechanisms may limit intestinal inflammatory responses.


Subject(s)
Colitis/etiology , Colon/immunology , DNA-Binding Proteins/physiology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Animals , CD11 Antigens/analysis , Colitis/immunology , Disease Susceptibility , Female , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Transcriptome
8.
Sci Adv ; 6(11): eaax6328, 2020 03.
Article in English | MEDLINE | ID: mdl-32195337

ABSTRACT

Alterations to the gut microbiome are associated with various neurological diseases, yet evidence of causality and identity of microbiome-derived compounds that mediate gut-brain axis interaction remain elusive. Here, we identify two previously unknown bacterial metabolites 3-methyl-4-(trimethylammonio)butanoate and 4-(trimethylammonio)pentanoate, structural analogs of carnitine that are present in both gut and brain of specific pathogen-free mice but absent in germ-free mice. We demonstrate that these compounds are produced by anaerobic commensal bacteria from the family Lachnospiraceae (Clostridiales) family, colocalize with carnitine in brain white matter, and inhibit carnitine-mediated fatty acid oxidation in a murine cell culture model of central nervous system white matter. This is the first description of direct molecular inter-kingdom exchange between gut prokaryotes and mammalian brain cells, leading to inhibition of brain cell function.


Subject(s)
Carnitine , Clostridiales/metabolism , Gastrointestinal Microbiome , Intestinal Mucosa , White Matter/metabolism , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice
9.
Nat Commun ; 10(1): 2344, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138806

ABSTRACT

Infection by soil transmitted parasitic helminths, such as Trichuris spp, are ubiquitous in humans and animals but the mechanisms determining persistence of chronic infections are poorly understood. Here we show that p43, the single most abundant protein in T. muris excretions/secretions, is non-immunogenic during infection and has an unusual sequence and structure containing subdomain homology to thrombospondin type 1 and interleukin (IL)-13 receptor (R) α2. Binding of p43 to IL-13, the key effector cytokine responsible for T. muris expulsion, inhibits IL-13 function both in vitro and in vivo. Tethering of p43 to matrix proteoglycans presents a bound source of p43 to facilitate interaction with IL-13, which may underpin chronic intestinal infection. Our results suggest that exploiting the biology of p43 may open up new approaches to modulating IL-13 function and control of Trichuris infections.


Subject(s)
Helminth Proteins/metabolism , Interleukin-13/metabolism , Intestinal Diseases, Parasitic/metabolism , Proteoglycans/metabolism , Trichuris/metabolism , Animals , Extracellular Matrix/metabolism , Helminth Proteins/immunology , Interleukin-13/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Intestinal Diseases, Parasitic/immunology , Mice , Sequence Homology, Amino Acid , Thrombospondin 1/metabolism , Trichuriasis
10.
Nat Immunol ; 20(5): 571-580, 2019 05.
Article in English | MEDLINE | ID: mdl-30936493

ABSTRACT

Fine control of macrophage activation is needed to prevent inflammatory disease, particularly at barrier sites such as the lungs. However, the dominant mechanisms that regulate the activation of pulmonary macrophages during inflammation are poorly understood. We found that alveolar macrophages (AlvMs) were much less able to respond to the canonical type 2 cytokine IL-4, which underpins allergic disease and parasitic worm infections, than macrophages from lung tissue or the peritoneal cavity. We found that the hyporesponsiveness of AlvMs to IL-4 depended upon the lung environment but was independent of the host microbiota or the lung extracellular matrix components surfactant protein D (SP-D) and mucin 5b (Muc5b). AlvMs showed severely dysregulated metabolism relative to that of cavity macrophages. After removal from the lungs, AlvMs regained responsiveness to IL-4 in a glycolysis-dependent manner. Thus, impaired glycolysis in the pulmonary niche regulates AlvM responsiveness during type 2 inflammation.


Subject(s)
Inflammation/immunology , Lung/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Animals , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Larva/immunology , Larva/physiology , Lung/metabolism , Lung/pathology , Macrophage Activation/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/parasitology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucin-5B/genetics , Mucin-5B/immunology , Mucin-5B/metabolism , Nippostrongylus/immunology , Nippostrongylus/physiology , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Strongylida Infections/genetics , Strongylida Infections/immunology , Strongylida Infections/parasitology
11.
Front Immunol ; 9: 2764, 2018.
Article in English | MEDLINE | ID: mdl-30542349

ABSTRACT

Background: Macrophages are pivotal in coordinating a range of important processes in the intestines, including controlling intracellular infections and limiting damaging inflammation against the microbiota. However, it is not clear how gut macrophages, relative to recruited blood monocytes and other myeloid cells, contribute to the intestinal inflammatory milieu, nor how macrophages and their monocyte precursors mediate recruitment of other immune cells to the inflamed intestine. Methods: Myeloid cell populations isolated from colonic inflammatory bowel disease (IBD) or murine dextran sulphate sodium (DSS) induced colitis were assessed using flow cytometry and compared to healthy controls. In addition, mRNA expression profiles in human and murine colon samples, and in macrophages and monocytes from healthy and inflamed murine colons, were analysed by quantitative PCR (qPCR) and mRNA microarray. Results: We show that the monocyte:macrophage balance is disrupted in colon inflammation to favour recruitment of CD14+HLA-DRInt cells in humans, and Ly6CHi monocytes in mice. In addition, we identify that murine blood monocytes receive systemic signals enabling increased release of IL-1ß prior to egress from the blood into the colon. Further, once within the colon and relative to other myeloid cells, monocytes represent the dominant local source of both IL-1ß and TNF. Finally, our data reveal that, independent of inflammation, murine colon macrophages act as a major source of Ccl7 and Ccl8 chemokines that trigger further recruitment of their pro-inflammatory monocyte precursors. Conclusions: Our work suggests that strategies targeting macrophage-mediated monocyte recruitment may represent a promising approach for limiting the chronic inflammation that characterises IBD.


Subject(s)
Colitis/immunology , Colon/immunology , Macrophage Activation/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Chemokine CCL7/immunology , Chemokine CCL8/immunology , Dextran Sulfate/immunology , Female , Humans , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factors/immunology
12.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28716804

ABSTRACT

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Th2 Cells/immunology , Allergens/immunology , Animals , Mice , Mice, Knockout , Pyroglyphidae/immunology , Receptor, Interferon alpha-beta/deficiency , Schistosoma mansoni/immunology
13.
Nat Commun ; 6: 6920, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25908537

ABSTRACT

Dendritic cells (DCs) direct CD4(+) T-cell differentiation into diverse helper (Th) subsets that are required for protection against varied infections. However, the mechanisms used by DCs to promote Th2 responses, which are important both for immunity to helminth infection and in allergic disease, are currently poorly understood. We demonstrate a key role for the protein methyl-CpG-binding domain-2 (Mbd2), which links DNA methylation to repressive chromatin structure, in regulating expression of a range of genes that are associated with optimal DC activation and function. In the absence of Mbd2, DCs display reduced phenotypic activation and a markedly impaired capacity to initiate Th2 immunity against helminths or allergens. These data identify an epigenetic mechanism that is central to the activation of CD4(+) T-cell responses by DCs, particularly in Th2 settings, and reveal methyl-CpG-binding proteins and the genes under their control as possible therapeutic targets for type-2 inflammation.


Subject(s)
DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Gene Expression Regulation/genetics , RNA, Messenger/metabolism , Th2 Cells/immunology , Allergens , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Polarity , Chromatin Immunoprecipitation , DNA Methylation , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Epigenesis, Genetic , Flow Cytometry , Hypersensitivity/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Pyroglyphidae/immunology , Reverse Transcriptase Polymerase Chain Reaction , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology
14.
Int Immunol ; 18(1): 69-78, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16291653

ABSTRACT

B cells act as efficient antigen-presenting cells if they acquire antigen via membrane-bound Ig [termed the B cell receptor (BCR)]. Ligation of the BCR leads to antigen internalization, processing and presentation to CD4+ T cells in association with MHC class II molecules. Ligation of the BCR also leads to the generation of activation signals. One short-term consequence of this is the up-regulation of co-stimulatory molecule expression by the B cell, allowing full T cell activation. Other antigen receptors expressed by B cells can also mediate efficient antigen presentation to CD4+ T cells. Ligating one such receptor, complement receptor 2 (CR2), has also been described to induce co-stimulatory molecule expression. If correct, this may have serious consequences for ensuring the specificity of the resultant B cell response. We have therefore investigated the effects of ligating both the BCR and CR2 independently of each other, as well as with reagents to cross-link the two receptors, in order to clarify these findings. In contrast to the effects seen upon BCR ligation, we find no evidence for co-stimulatory molecule up-regulation following CR2 ligation. As antigen presentation in the absence of co-stimulation may lead to the induction of tolerogenic or regulatory signals being delivered to T cell populations, these findings imply that the role of CR2 in B cell-mediated antigen presentation is different from that of the BCR.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Receptors, Complement 3d/immunology , Spleen/immunology , Animals , B-Lymphocytes/cytology , Cells, Cultured , Mice , Mice, Inbred BALB C , Signal Transduction/immunology , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...