Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Nature ; 628(8009): 854-862, 2024 Apr.
Article En | MEDLINE | ID: mdl-38570678

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Intestinal Mucosa , Mucous Membrane , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Immune Tolerance/immunology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Integrin alpha Chains/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mucous Membrane/cytology , Mucous Membrane/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Single-Cell Gene Expression Analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , Transcriptome
2.
Mucosal Immunol ; 17(1): 111-123, 2024 Feb.
Article En | MEDLINE | ID: mdl-37995912

The relationship between gastrointestinal tract infection, the host immune response, and the clinical outcome of disease is not well understood in COVID-19. We sought to understand the effect of intestinal immune responses to SARS-CoV-2 on patient outcomes including the magnitude of systemic antibody induction. Combining two prospective cohort studies, International Severe Acute Respiratory and emerging Infections Consortium Comprehensive Clinical Characterisations Collaboration (ISARIC4C) and Integrated Network for Surveillance, Trials and Investigations into COVID-19 Transmission (INSTINCT), we acquired samples from 88 COVID-19 cases representing the full spectrum of disease severity and analysed viral RNA and host gut cytokine responses in the context of clinical and virological outcome measures. There was no correlation between the upper respiratory tract and faecal viral loads. Using hierarchical clustering, we identified a group of fecal cytokines including Interleukin-17A, Granulocyte macrophage colony-stimulating factor, Tumor necrosis factorα, Interleukin-23, and S100A8, that were transiently elevated in mild cases and also correlated with the magnitude of systemic anti-Spike-receptor-binding domain antibody induction. Receiver operating characteristic curve analysis showed that expression of these gut cytokines at study enrolment in hospitalised COVID-19 cases was associated negatively with overall clinical severity implicating a protective role in COVID-19. This suggests that a productive intestinal immune response may be beneficial in the response to a respiratory pathogen and a biomarker of a successful barrier response.


COVID-19 , Humans , Cytokines/metabolism , SARS-CoV-2 , Prospective Studies , Feces , Antibodies, Viral
3.
Nat Commun ; 13(1): 7472, 2022 12 03.
Article En | MEDLINE | ID: mdl-36463279

Interactions with commensal microbes shape host immunity on multiple levels and play a pivotal role in human health and disease. Tissue-dwelling, antigen-specific T cells are poised to respond to local insults, making their phenotype important in the relationship between host and microbes. Here we show that MHC-II restricted, commensal-reactive T cells in the colon of both humans and mice acquire transcriptional and functional characteristics associated with innate-like T cells. This cell population is abundant and conserved in the human and murine colon and endowed with polyfunctional effector properties spanning classic Th1- and Th17-cytokines, cytotoxic molecules, and regulators of epithelial homeostasis. T cells with this phenotype are increased in ulcerative colitis patients, and their presence aggravates pathology in dextran sodium sulphate-treated mice, pointing towards a pathogenic role in colitis. Our findings add to the expanding spectrum of innate-like immune cells positioned at the frontline of intestinal immune surveillance, capable of acting as sentinels of microbes and the local cytokine milieu.


Coleoptera , Colitis , Humans , Mice , Animals , Lymphocyte Count , Immunologic Surveillance , Colitis/chemically induced , Cytokines
4.
Immunity ; 55(10): 1751-1753, 2022 10 11.
Article En | MEDLINE | ID: mdl-36223721

Unconventional T cells (UTCs) play important roles in protecting barrier tissues. In this issue of Immunity, Ataide and colleagues show an additional function for these cells: populating downstream lymph nodes and skewing their cytokine profiles. This process allows tissues to adjust the function of draining lymph nodes through imprinting UTC cytokine production, subsequently influencing the immune responses that are triggered.


Lymph Nodes , T-Lymphocytes , Cytokines
5.
Immunity ; 55(4): 718-733.e8, 2022 04 12.
Article En | MEDLINE | ID: mdl-35349789

Resident memory B (BRM) cells develop and persist in the lungs of influenza-infected mice and humans; however, their contribution to recall responses has not been defined. Here, we used two-photon microscopy to visualize BRM cells within the lungs of influenza -virus immune and reinfected mice. Prior to re-exposure, BRM cells were sparsely scattered throughout the tissue, displaying limited motility. Within 24 h of rechallenge, these cells increased their migratory capacity, localized to infected sites, and subsequently differentiated into plasma cells. Alveolar macrophages mediated this process, in part by inducing expression of chemokines CXCL9 and CXCL10 from infiltrating inflammatory cells. This led to the recruitment of chemokine receptor CXCR3-expressing BRM cells to infected regions and increased local antibody concentrations. Our study uncovers spatiotemporal mechanisms that regulate lung BRM cell reactivation and demonstrates their capacity to rapidly deliver antibodies in a highly localized manner to sites of viral replication.


Influenza, Human , Orthomyxoviridae Infections , Orthomyxoviridae , Animals , Antibodies , Humans , Immunologic Memory , Memory B Cells , Mice
6.
Oxf Open Immunol ; 2(1): iqab002, 2021.
Article En | MEDLINE | ID: mdl-33585820

COVID-19 was initially characterized as a disease primarily of the lungs, but it is becoming increasingly clear that the SARS-CoV2 virus is able to infect many organs and cause a broad pathological response. The primary infection site is likely to be a mucosal surface, mainly the lungs or the intestine, where epithelial cells can be infected with virus. Although it is clear that virus within the lungs can cause severe pathology, driven by an exaggerated immune response, infection within the intestine generally seems to cause minor or no symptoms. In this review, we compare the disease processes between the lungs and gastrointestinal tract, and what might drive these different responses. As the microbiome is a key part of mucosal barrier sites, we also consider the effect that microbial species may play on infection and the subsequent immune responses. Because of difficulties obtaining tissue samples, there are currently few studies focused on the local mucosal response rather than the systemic response, but understanding the local immune response will become increasingly important for understanding the mechanisms of disease in order to develop better treatments.

7.
J Exp Med ; 215(8): 1987-1998, 2018 08 06.
Article En | MEDLINE | ID: mdl-29980582

Psoriasis is a complex inflammatory skin disease affecting ∼3% of the population worldwide. Although type I interferons (IFN-I) are thought to be involved in its pathogenesis, the details of this relationship remain elusive. Here we show that in a murine model of imiquimod-driven psoriatic skin inflammation, Foxp3+ regulatory T cells (T reg cells) control inflammation severity by restraining IFN-I. Depletion of T reg cells induces IFN-I and IFN-stimulated gene expression, and leads to accumulation of CD8+ T cells in lesional skin. Mononuclear phagocytes (MNPs) were the source of IFN-I, and their depletion reversed the effect of T reg cell depletion. Blockade of IFN-I signaling abolished CD8+ T cell infiltration and excess inflammation in the skin of T reg cell-depleted mice. Depletion of CD8+ T cells attenuated pathology, confirming their role as critical effector cells downstream of IFN-I. Our results describe an unexpected role for T reg cells in restraint of an MNP-IFN-I-driven CD8+ T cell response during psoriasiform skin inflammation. These findings highlight a pathway with potential relevance for the treatment of early-stage disease.


CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Inflammation/immunology , Interferon Type I/metabolism , Psoriasis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Mice, Inbred C57BL , Phagocytes/metabolism , Severity of Illness Index , Skin/pathology
8.
Nature ; 544(7648): 105-109, 2017 04 06.
Article En | MEDLINE | ID: mdl-28329764

Platelets are critical for haemostasis, thrombosis, and inflammatory responses, but the events that lead to mature platelet production remain incompletely understood. The bone marrow has been proposed to be a major site of platelet production, although there is indirect evidence that the lungs might also contribute to platelet biogenesis. Here, by directly imaging the lung microcirculation in mice, we show that a large number of megakaryocytes circulate through the lungs, where they dynamically release platelets. Megakaryocytes that release platelets in the lungs originate from extrapulmonary sites such as the bone marrow; we observed large megakaryocytes migrating out of the bone marrow space. The contribution of the lungs to platelet biogenesis is substantial, accounting for approximately 50% of total platelet production or 10 million platelets per hour. Furthermore, we identified populations of mature and immature megakaryocytes along with haematopoietic progenitors in the extravascular spaces of the lungs. Under conditions of thrombocytopenia and relative stem cell deficiency in the bone marrow, these progenitors can migrate out of the lungs, repopulate the bone marrow, completely reconstitute blood platelet counts, and contribute to multiple haematopoietic lineages. These results identify the lungs as a primary site of terminal platelet production and an organ with considerable haematopoietic potential.


Blood Platelets/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Lung/blood supply , Lung/cytology , Animals , Bone Marrow , Cell Lineage , Female , Lung/anatomy & histology , Male , Megakaryocytes/cytology , Mice , Microcirculation , Platelet Count , Thrombocytopenia/pathology
9.
Elife ; 5: e10066, 2016 Jan 18.
Article En | MEDLINE | ID: mdl-26780670

Innate lymphoid cells (ILCs) contribute to host defence and tissue repair but can induce immunopathology. Recent work has revealed tissue-specific roles for ILCs; however, the question of how a small population has large effects on immune homeostasis remains unclear. We identify two mechanisms that ILC3s utilise to exert their effects within intestinal tissue. ILC-driven colitis depends on production of granulocyte macrophage-colony stimulating factor (GM-CSF), which recruits and maintains intestinal inflammatory monocytes. ILCs present in the intestine also enter and exit cryptopatches in a highly dynamic process. During colitis, ILC3s mobilize from cryptopatches, a process that can be inhibited by blocking GM-CSF, and mobilization precedes inflammatory foci elsewhere in the tissue. Together these data identify the IL-23R/GM-CSF axis within ILC3 as a key control point in the accumulation of innate effector cells in the intestine and in the spatio-temporal dynamics of ILCs in the intestinal inflammatory response.


Colitis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Intestines/immunology , Lymphocytes/immunology , Humans , Interleukin-23 Subunit p19/metabolism
10.
Trends Immunol ; 35(8): 379-86, 2014 Aug.
Article En | MEDLINE | ID: mdl-24974157

The lung is a complex structure that is interdigitated with immune cells. Understanding the 4D process of normal and defective lung function and immunity has been a centuries-old problem. Challenges intrinsic to the lung have limited adequate microscopic evaluation of its cellular dynamics in real time, until recently. Because of emerging technologies, we now recognize alveolar-to-airway transport of inhaled antigen. We understand the nature of neutrophil entry during lung injury and are learning more about cellular interactions during inflammatory states. Insights are also accumulating in lung development and the metastatic niche of the lung. Here we assess the developing technology of lung imaging, its merits for studies of pathophysiology and areas where further advances are needed.


Lung Neoplasms/immunology , Lung/immunology , Animals , Cell Communication , Humans , Imaging, Three-Dimensional , Inflammation/immunology , Lung/pathology , Organogenesis
11.
Immunity ; 40(3): 414-24, 2014 Mar 20.
Article En | MEDLINE | ID: mdl-24631157

Chitin, a polysaccharide constituent of many allergens and parasites, initiates innate type 2 lung inflammation through incompletely defined pathways. We show that inhaled chitin induced expression of three epithelial cytokines, interleukin-25 (IL-25), IL-33, and thymic stromal lymphopoietin (TSLP), which nonredundantly activated resident innate lymphoid type 2 cells (ILC2s) to express IL-5 and IL-13 necessary for accumulation of eosinophils and alternatively activated macrophages (AAMs). In the absence of all three epithelial cytokines, ILC2s normally populated the lung but failed to increase IL-5 and IL-13. Although eosinophils and AAMs were attenuated, neutrophil influx remained normal without these epithelial cytokines. Genetic ablation of ILC2s, however, enhanced IL-1ß, TNFα, and IL-23 expression, increased activation of IL-17A-producing γδ T cells, and prolonged neutrophil influx. Thus, chitin elicited patterns of innate cytokines that targeted distinct populations of resident lymphoid cells, revealing divergent but interacting pathways underlying the tissue accumulation of specific types of inflammatory myeloid cells.


Chitin/immunology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Chitin/administration & dosage , Cytokines/biosynthesis , Cytokines/genetics , Eosinophils/immunology , Eosinophils/metabolism , Gene Expression Regulation/drug effects , Inflammation/genetics , Lung/immunology , Lung/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout
12.
Nature ; 502(7470): 245-8, 2013 Oct 10.
Article En | MEDLINE | ID: mdl-24037376

Eosinophils are specialized myeloid cells associated with allergy and helminth infections. Blood eosinophils demonstrate circadian cycling, as described over 80 years ago, and are abundant in the healthy gastrointestinal tract. Although a cytokine, interleukin (IL)-5, and chemokines such as eotaxins mediate eosinophil development and survival, and tissue recruitment, respectively, the processes underlying the basal regulation of these signals remain unknown. Here we show that serum IL-5 levels are maintained by long-lived type 2 innate lymphoid cells (ILC2) resident in peripheral tissues. ILC2 cells secrete IL-5 constitutively and are induced to co-express IL-13 during type 2 inflammation, resulting in localized eotaxin production and eosinophil accumulation. In the small intestine where eosinophils and eotaxin are constitutive, ILC2 cells co-express IL-5 and IL-13; this co-expression is enhanced after caloric intake. The circadian synchronizer vasoactive intestinal peptide also stimulates ILC2 cells through the VPAC2 receptor to release IL-5, linking eosinophil levels with metabolic cycling. Tissue ILC2 cells regulate basal eosinophilopoiesis and tissue eosinophil accumulation through constitutive and stimulated cytokine expression, and this dissociated regulation can be tuned by nutrient intake and central circadian rhythms.


Eosinophils/metabolism , Homeostasis , Lymphocytes/metabolism , Animals , Cells, Cultured , Circadian Rhythm , Collagen/metabolism , Eosinophils/immunology , Eosinophils/parasitology , Female , Gene Expression Regulation , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-5/blood , Interleukin-5/genetics , Interleukin-5/metabolism , Lung/immunology , Lung/metabolism , Lung/parasitology , Lymphocytes/immunology , Lymphocytes/parasitology , Male , Mice , Mice, Inbred C57BL , Nippostrongylus/physiology , Strongylida Infections/immunology
13.
J Exp Med ; 209(6): 1183-99, 2012 Jun 04.
Article En | MEDLINE | ID: mdl-22585735

Asthma pathogenesis is focused around conducting airways. The reasons for this focus have been unclear because it has not been possible to track the sites and timing of antigen uptake or subsequent antigen presentation to effector T cells. In this study, we use two-photon microscopy of the lung parenchyma and note accumulation of CD11b(+) dendritic cells (DCs) around the airway after allergen challenge but very limited access of these airway-adjacent DCs to the contents of the airspace. In contrast, we observed prevalent transepithelial uptake of particulate antigens by alveolar DCs. These distinct sites are temporally linked, as early antigen uptake in alveoli gives rise to DC and antigen retention in the airway-adjacent region. Antigen-specific T cells also accumulate in the airway-adjacent region after allergen challenge and are activated by the accumulated DCs. Thus, we propose that later airway hyperreactivity results from selective retention of allergen-presenting DCs and antigen-specific T cells in airway-adjacent interaction zones, not from variation in the abilities of individual DCs to survey the lung.


Antigens/metabolism , Dendritic Cells/immunology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , T-Lymphocytes/immunology , Allergens/immunology , Animals , Antigen Presentation/immunology , Asthma/immunology , Asthma/pathology , Bronchial Hyperreactivity/immunology , CD11c Antigen/genetics , In Vitro Techniques , Lung/cytology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy/methods
14.
Curr Protoc Cytom ; Chapter 12: 12.28.1-12.28.12, 2012 Apr.
Article En | MEDLINE | ID: mdl-22470155

Live imaging is critical to determining the dynamics and spatial interactions of cells within the tissue environment. In the lung, this has proven to be difficult due to the motion incurred by ventilation and cardiac contractions. In this chapter, we report protocols for imaging ex vivo live lung slices and the intact mouse lung.


Diagnostic Imaging/methods , Lung/anatomy & histology , Animals , Cell Movement , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Instillation, Drug , Lung/cytology , Lung/surgery , Mice , Microscopy , Staining and Labeling
15.
Nat Methods ; 8(1): 91-6, 2011 Jan.
Article En | MEDLINE | ID: mdl-21151136

Real-time imaging of cellular and subcellular dynamics in vascularized organs requires image resolution and image registration to be simultaneously optimized without perturbing normal physiology. This problem is particularly pronounced in the lung, in which cells may transit at speeds >1 mm s(-1) and in which normal respiration results in large-scale tissue movements that prevent image registration. Here we report video-rate, two-photon imaging of a physiologically intact preparation of the mouse lung that is stabilizing and nondisruptive. Using our method, we obtained evidence for differential trapping of T cells and neutrophils in mouse pulmonary capillaries, and observed neutrophil mobilization and dynamic vascular leak in response to stretch and inflammatory models of lung injury in mice. The system permits physiological measurement of motility rates of >1 mm s(-1), observation of detailed cellular morphology and could be applied in the future to other organs and tissues while maintaining intact physiology.


Imaging, Three-Dimensional/methods , Immunologic Surveillance/immunology , Lung/cytology , Lung/immunology , Movement , Animals , Lung/blood supply , Mice , Microscopy, Fluorescence
...