Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.653
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 259-288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38277692

ABSTRACT

Gastrointestinal nematode (GIN) infection has applied significant evolutionary pressure to the mammalian immune system and remains a global economic and human health burden. Upon infection, type 2 immune sentinels activate a common antihelminth response that mobilizes and remodels the intestinal tissue for effector function; however, there is growing appreciation of the impact GIN infection also has on the distal tissue immune state. Indeed, this effect is observed even in tissues through which GINs never transit. This review highlights how GIN infection modulates systemic immunity through (a) induction of host resistance and tolerance responses, (b) secretion of immunomodulatory products, and (c) interaction with the intestinal microbiome. It also discusses the direct consequences that changes to distal tissue immunity can have for concurrent and subsequent infection, chronic noncommunicable diseases, and vaccination efficacy.


Subject(s)
Gastrointestinal Microbiome , Nematoda , Nematode Infections , Animals , Humans , Nematode Infections/immunology , Nematoda/immunology , Nematoda/physiology , Gastrointestinal Microbiome/immunology , Immunomodulation , Host-Parasite Interactions/immunology , Intestinal Diseases, Parasitic/immunology , Immune Tolerance , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology
2.
Annu Rev Immunol ; 40: 499-523, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35471839

ABSTRACT

The bladder is a major component of the urinary tract, an organ system that expels metabolic waste and excess water, which necessitates proximity to the external environment and its pathogens. It also houses a commensal microbiome. Therefore, its tissue immunity must resist pathogen invasion while maintaining tolerance to commensals. Bacterial infection of the bladder is common, with half of women globally experiencing one or more episodes of cystitis in their lifetime. Despite this, our knowledge of bladder immunity, particularly in humans, is incomplete. Here we consider the current view of tissue immunity in the bladder, with a focus on defense against infection. The urothelium has robust immune functionality, and its defensive capabilities are supported by resident immune cells, including macrophages, dendritic cells, natural killer cells, and γδ T cells. We discuss each in turn and consider why adaptive immune responses are often ineffective in preventing recurrent infection, as well as areas of priority for future research.


Subject(s)
Bacterial Infections , Urinary Bladder , Animals , Female , Humans , Immune Tolerance , Immunity, Innate , Macrophages , Urinary Bladder/microbiology
3.
Annu Rev Immunol ; 38: 23-48, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340570

ABSTRACT

The gastrointestinal tract harbors numerous commensal bacteria, referred to as the microbiota, that benefit host health by digesting dietary components and eliminating pathogens. The intestinal microbiota maintains epithelial barrier integrity and shapes the mucosal immune system, balancing host defense and oral tolerance with microbial metabolites, components, and attachment to host cells. To avoid aberrant immune responses, epithelial cells segregate the intestinal microbiota from immune cells by constructing chemical and physical barriers, leading to the establishment of host-commensal mutualism. Furthermore, intestinal immune cells participate in the maintenance of a healthy microbiota community and reinforce epithelial barrier functions. Perturbations of the microbiota composition are commonly observed in patients with autoimmune diseases and chronic inflammatory disorders. An understanding of the intimate interactions between the intestinal microbiota, epithelial cells, and immune cells that are crucial for the maintenance of intestinal homeostasis might promote advances in diagnostic and therapeutic approaches for various diseases.


Subject(s)
Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Animals , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism
4.
Annu Rev Immunol ; 38: 203-228, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31986071

ABSTRACT

Mucosal-associated invariant T (MAIT) cells have been attracting increasing attention over the last few years as a potent unconventional T cell subset. Three factors largely account for this emerging interest. Firstly, these cells are abundant in humans, both in circulation and especially in some tissues such as the liver. Secondly is the discovery of a ligand that has uncovered their microbial targets, and also allowed for the development of tools to accurately track the cells in both humans and mice. Finally, it appears that the cells not only have a diverse range of functions but also are sensitive to a range of inflammatory triggers that can enhance or even bypass T cell receptor-mediated signals-substantially broadening their likely impact in health and disease. In this review we discuss how MAIT cells display antimicrobial, homeostatic, and amplifier roles in vivo, and how this may lead to protection and potentially pathology.


Subject(s)
Disease Susceptibility , Homeostasis , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Animals , Biomarkers , Host-Pathogen Interactions , Humans , Immunity, Mucosal , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
Annu Rev Immunol ; 35: 501-532, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28226227

ABSTRACT

Respiratory syncytial virus (RSV) is an exceptional mucosal pathogen. It specializes in infection of the ciliated respiratory epithelium, causing disease of variable severity with little or no direct systemic effects. It infects virtually all children by the age of three years and then repeatedly infects throughout life; this it does despite relatively slight variations in antigenicity, apparently by inducing selective immunological amnesia. Inappropriate or dysregulated responses to RSV can be pathogenic, causing disease-enhancing inflammation that contributes to short- and long-term effects. In addition, RSV's importance as a largely unrecognized pathogen of debilitated older people is increasingly evident. Vaccines that induce nonpathogenic protective immunity may soon be available, and it is possible that different vaccines will be optimal for infants; older children; young to middle-age adults (including pregnant women); and elderly persons. At the dawn of RSV vaccination, it is timely to review what is known (and unknown) about immune responses to this fascinating virus.


Subject(s)
Respiratory Mucosa/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Viral Vaccines/immunology , Adult , Aged , Animals , Child , Humans , Immune Evasion , Immunomodulation , Respiratory Mucosa/virology
6.
Cell ; 187(19): 5393-5412.e30, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39121857

ABSTRACT

Negative psychological states impact immunity by altering the gut microbiome. However, the relationship between brain states and microbiome composition remains unclear. We show that Brunner's glands in the duodenum couple stress-sensitive brain circuits to bacterial homeostasis. Brunner's glands mediated the enrichment of gut Lactobacillus species in response to vagus nerve stimulation. Cell-specific ablation of the glands markedly suppressed Lactobacilli counts and heightened vulnerability to infection. In the forebrain, we mapped a vagally mediated, polysynaptic circuit connecting the central nucleus of the amygdala to Brunner's glands. Chronic stress suppressed central amygdala activity and phenocopied the effects of gland lesions. Conversely, excitation of either the central amygdala or parasympathetic vagal neurons activated Brunner's glands and reversed the effects of stress on the gut microbiome and immunity. The findings revealed a tractable brain-body mechanism linking psychological states to host defense.


Subject(s)
Duodenum , Gastrointestinal Microbiome , Stress, Psychological , Vagus Nerve , Animals , Mice , Duodenum/microbiology , Vagus Nerve/physiology , Male , Mice, Inbred C57BL , Amygdala/physiology , Lactobacillus/physiology , Neurons/metabolism
7.
Cell ; 186(21): 4632-4651.e23, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37776858

ABSTRACT

The dynamics of immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infants and young children by analyzing blood samples and weekly nasal swabs collected before, during, and after infection with Omicron and non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, showed no sign of decay for up to 300 days. Infants mounted a robust mucosal immune response characterized by inflammatory cytokines, interferon (IFN) α, and T helper (Th) 17 and neutrophil markers (interleukin [IL]-17, IL-8, and CXCL1). The immune response in blood was characterized by upregulation of activation markers on innate cells, no inflammatory cytokines, but several chemokines and IFNα. The latter correlated with viral load and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell multi-omics. Together, these data provide a snapshot of immunity to infection during the initial weeks and months of life.


Subject(s)
COVID-19 , SARS-CoV-2 , Adult , Child , Infant , Humans , Child, Preschool , SARS-CoV-2/metabolism , Multiomics , Cytokines/metabolism , Interferon-alpha , Immunity, Mucosal
8.
Annu Rev Immunol ; 33: 201-25, 2015.
Article in English | MEDLINE | ID: mdl-25533702

ABSTRACT

Helminth parasites are a highly successful group of pathogens that challenge the immune system in a manner distinct from rapidly replicating infectious agents. Of this group, roundworms (nematodes) that dwell in the intestines of humans and other animals are prevalent worldwide. Currently, more than one billion people are infected by at least one species, often for extended periods of time. Thus, host-protective immunity is rarely complete. The reasons for this are complex, but laboratory investigation of tractable model systems in which protective immunity is effective has provided a mechanistic understanding of resistance that is characterized almost universally by a type 2/T helper 2 response. Greater understanding of the mechanisms of susceptibility has also provided the basis for defining host immunoregulation and parasite-evasion strategies, helping place in context the changing patterns of immunological disease observed worldwide.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Host-Pathogen Interactions/immunology , Adaptive Immunity , Animals , Antigens, Helminth/immunology , Disease Resistance , Disease Susceptibility , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/parasitology , Gastrointestinal Microbiome , Humans , Immunity, Innate , Nematoda/immunology , Nematode Infections/immunology , Nematode Infections/microbiology , Nematode Infections/parasitology
9.
Cell ; 185(19): 3501-3519.e20, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36041436

ABSTRACT

How intestinal microbes regulate metabolic syndrome is incompletely understood. We show that intestinal microbiota protects against development of obesity, metabolic syndrome, and pre-diabetic phenotypes by inducing commensal-specific Th17 cells. High-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection. Microbiota-induced Th17 cells afforded protection by regulating lipid absorption across intestinal epithelium in an IL-17-dependent manner. Diet-induced loss of protective Th17 cells was mediated by the presence of sugar. Eliminating sugar from high-fat diets protected mice from obesity and metabolic syndrome in a manner dependent on commensal-specific Th17 cells. Sugar and ILC3 promoted outgrowth of Faecalibaculum rodentium that displaced Th17-inducing microbiota. These results define dietary and microbiota factors posing risk for metabolic syndrome. They also define a microbiota-dependent mechanism for immuno-pathogenicity of dietary sugar and highlight an elaborate interaction between diet, microbiota, and intestinal immunity in regulation of metabolic disorders.


Subject(s)
Metabolic Syndrome , Microbiota , Animals , Diet, High-Fat , Dietary Sugars , Interleukin-17 , Intestinal Mucosa , Lipids , Mice , Mice, Inbred C57BL , Obesity , Th17 Cells
10.
Cell ; 185(1): 113-130.e15, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34921774

ABSTRACT

mRNA-1273 vaccine efficacy against SARS-CoV-2 Delta wanes over time; however, there are limited data on the impact of durability of immune responses on protection. Here, we immunized rhesus macaques and assessed immune responses over 1 year in blood and upper and lower airways. Serum neutralizing titers to Delta were 280 and 34 reciprocal ID50 at weeks 6 (peak) and 48 (challenge), respectively. Antibody-binding titers also decreased in bronchoalveolar lavage (BAL). Four days after Delta challenge, the virus was unculturable in BAL, and subgenomic RNA declined by ∼3-log10 compared with control animals. In nasal swabs, sgRNA was reduced by 1-log10, and the virus remained culturable. Anamnestic antibodies (590-fold increased titer) but not T cell responses were detected in BAL by day 4 post-challenge. mRNA-1273-mediated protection in the lungs is durable but delayed and potentially dependent on anamnestic antibody responses. Rapid and sustained protection in upper and lower airways may eventually require a boost.

11.
Cell ; 185(5): 896-915.e19, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35180381

ABSTRACT

The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Immunity, Mucosal , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , COVID-19/virology , COVID-19 Vaccines/immunology , Cytokines/blood , Genetic Vectors/genetics , Genetic Vectors/immunology , Genetic Vectors/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutralization Tests , Nucleocapsid/genetics , Nucleocapsid/immunology , Nucleocapsid/metabolism , Pan troglodytes , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Cell ; 184(15): 4090-4104.e15, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34129837

ABSTRACT

The oral mucosa remains an understudied barrier tissue. This is a site of rich exposure to antigens and commensals, and a tissue susceptible to one of the most prevalent human inflammatory diseases, periodontitis. To aid in understanding tissue-specific pathophysiology, we compile a single-cell transcriptome atlas of human oral mucosa in healthy individuals and patients with periodontitis. We uncover the complex cellular landscape of oral mucosal tissues and identify epithelial and stromal cell populations with inflammatory signatures that promote antimicrobial defenses and neutrophil recruitment. Our findings link exaggerated stromal cell responsiveness with enhanced neutrophil and leukocyte infiltration in periodontitis. Our work provides a resource characterizing the role of tissue stroma in regulating mucosal tissue homeostasis and disease pathogenesis.


Subject(s)
Immunity, Mucosal , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Neutrophils/cytology , Adult , Epithelial Cells/cytology , Gene Expression Regulation , Genetic Predisposition to Disease , Gingiva/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Microbiota , Myeloid Cells/cytology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Single-Cell Analysis , Stromal Cells/cytology , T-Lymphocytes/cytology
13.
Cell ; 184(12): 3178-3191.e18, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34022140

ABSTRACT

Gasdermin B (GSDMB) belongs to a large family of pore-forming cytolysins that execute inflammatory cell death programs. While genetic studies have linked GSDMB polymorphisms to human disease, its function in the immunological response to pathogens remains poorly understood. Here, we report a dynamic host-pathogen conflict between GSDMB and the IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri. We show that IpaH7.8 ubiquitinates and targets GSDMB for 26S proteasome destruction. This virulence strategy protects Shigella from the bacteriocidic activity of natural killer cells by suppressing granzyme-A-mediated activation of GSDMB. In contrast to the canonical function of most gasdermin family members, GSDMB does not inhibit Shigella by lysing host cells. Rather, it exhibits direct microbiocidal activity through recognition of phospholipids found on Gram-negative bacterial membranes. These findings place GSDMB as a central executioner of intracellular bacterial killing and reveal a mechanism employed by pathogens to counteract this host defense system.


Subject(s)
Biomarkers, Tumor/metabolism , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Neoplasm Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Shigella flexneri/physiology , Ubiquitination , Animals , Bacterial Proteins/metabolism , Cardiolipins/metabolism , Cell Line , Cell Membrane/metabolism , Female , Granzymes/metabolism , Humans , Lipid A/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Substrate Specificity
14.
Cell ; 183(1): 169-184.e13, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931734

ABSTRACT

The coronavirus disease 2019 pandemic has made deployment of an effective vaccine a global health priority. We evaluated the protective activity of a chimpanzee adenovirus-vectored vaccine encoding a prefusion stabilized spike protein (ChAd-SARS-CoV-2-S) in challenge studies with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mice expressing the human angiotensin-converting enzyme 2 receptor. Intramuscular dosing of ChAd-SARS-CoV-2-S induces robust systemic humoral and cell-mediated immune responses and protects against lung infection, inflammation, and pathology but does not confer sterilizing immunity, as evidenced by detection of viral RNA and induction of anti-nucleoprotein antibodies after SARS-CoV-2 challenge. In contrast, a single intranasal dose of ChAd-SARS-CoV-2-S induces high levels of neutralizing antibodies, promotes systemic and mucosal immunoglobulin A (IgA) and T cell responses, and almost entirely prevents SARS-CoV-2 infection in both the upper and lower respiratory tracts. Intranasal administration of ChAd-SARS-CoV-2-S is a candidate for preventing SARS-CoV-2 infection and transmission and curtailing pandemic spread.


Subject(s)
Coronavirus Infections/immunology , Immunogenicity, Vaccine , Pneumonia, Viral/immunology , Viral Vaccines/immunology , Adenoviridae/genetics , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 , COVID-19 Vaccines , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Female , HEK293 Cells , Humans , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Pandemics , Pneumonia, Viral/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells , Viral Vaccines/administration & dosage
15.
Cell ; 180(4): 749-763.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32059780

ABSTRACT

Immune responses in diverse tissue sites are critical for protective immunity and homeostasis. Here, we investigate how tissue localization regulates the development and function of human natural killer (NK) cells, innate lymphocytes important for anti-viral and tumor immunity. Integrating high-dimensional analysis of NK cells from blood, lymphoid organs, and mucosal tissue sites from 60 individuals, we identify tissue-specific patterns of NK cell subset distribution, maturation, and function maintained across age and between individuals. Mature and terminally differentiated NK cells with enhanced effector function predominate in blood, bone marrow, spleen, and lungs and exhibit shared transcriptional programs across sites. By contrast, precursor and immature NK cells with reduced effector capacity populate lymph nodes and intestines and exhibit tissue-resident signatures and site-specific adaptations. Together, our results reveal anatomic control of NK cell development and maintenance as tissue-resident populations, whereas mature, terminally differentiated subsets mediate immunosurveillance through diverse peripheral sites. VIDEO ABSTRACT.


Subject(s)
Aging/immunology , Killer Cells, Natural/cytology , Lymphopoiesis , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Child , Female , Humans , Immunity, Innate , Intestinal Mucosa/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Lung/cytology , Lymph Nodes/cytology , Male , Middle Aged , Spleen/cytology
16.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442401

ABSTRACT

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Subject(s)
B-Lymphocytes/physiology , Immunity, Mucosal , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Fasting , Gene Expression Regulation , Glycolysis , Immunoglobulin A/metabolism , Male , Mice , Mice, Inbred BALB C , Nutritional Status , Ovalbumin/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/pathology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
17.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38744292

ABSTRACT

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Subject(s)
Acetylcholine , Intestinal Mucosa , Animals , Acetylcholine/metabolism , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Choline O-Acetyltransferase/metabolism , Interleukin-13/metabolism , Interleukin-13/immunology , Mice, Knockout , Mice, Inbred C57BL , Helminthiasis/immunology , Helminthiasis/parasitology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Immunity, Innate , Nematospiroides dubius/immunology , Tuft Cells
18.
Cell ; 175(5): 1307-1320.e22, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30392957

ABSTRACT

In the small intestine, a niche of accessory cell types supports the generation of mature epithelial cell types from intestinal stem cells (ISCs). It is unclear, however, if and how immune cells in the niche affect ISC fate or the balance between self-renewal and differentiation. Here, we use single-cell RNA sequencing (scRNA-seq) to identify MHC class II (MHCII) machinery enrichment in two subsets of Lgr5+ ISCs. We show that MHCII+ Lgr5+ ISCs are non-conventional antigen-presenting cells in co-cultures with CD4+ T helper (Th) cells. Stimulation of intestinal organoids with key Th cytokines affects Lgr5+ ISC renewal and differentiation in opposing ways: pro-inflammatory signals promote differentiation, while regulatory cells and cytokines reduce it. In vivo genetic perturbation of Th cells or MHCII expression on Lgr5+ ISCs impacts epithelial cell differentiation and IEC fate during infection. These interactions between Th cells and Lgr5+ ISCs, thus, orchestrate tissue-wide responses to external signals.


Subject(s)
Cell Differentiation , Cell Self Renewal , Interleukin-10/metabolism , Stem Cells/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cytokines/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Histocompatibility Antigens Class II/metabolism , Immune System/metabolism , Intestines/cytology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Salmonella enterica/pathogenicity , Stem Cells/metabolism , T-Lymphocytes, Helper-Inducer/cytology
19.
Immunity ; 56(8): 1894-1909.e5, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37421943

ABSTRACT

Infancy and childhood are critical life stages for generating immune memory to protect against pathogens; however, the timing, location, and pathways for memory development in humans remain elusive. Here, we investigated T cells in mucosal sites, lymphoid tissues, and blood from 96 pediatric donors aged 0-10 years using phenotypic, functional, and transcriptomic profiling. Our results revealed that memory T cells preferentially localized in the intestines and lungs during infancy and accumulated more rapidly in mucosal sites compared with blood and lymphoid organs, consistent with site-specific antigen exposure. Early life mucosal memory T cells exhibit distinct functional capacities and stem-like transcriptional profiles. In later childhood, they progressively adopt proinflammatory functions and tissue-resident signatures, coincident with increased T cell receptor (TCR) clonal expansion in mucosal and lymphoid sites. Together, our findings identify staged development of memory T cells targeted to tissues during the formative years, informing how we might promote and monitor immunity in children.


Subject(s)
Lymphoid Tissue , Memory T Cells , Child , Humans , Infant , CD8-Positive T-Lymphocytes , Immunologic Memory , Lymphoid Tissue/metabolism , Mucous Membrane , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Infant, Newborn , Child, Preschool
20.
Immunity ; 56(4): 797-812.e4, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36801011

ABSTRACT

The aryl-hydrocarbon receptor (AHR) is a ligand-activated transcription factor that buoys intestinal immune responses. AHR induces its own negative regulator, the AHR repressor (AHRR). Here, we show that AHRR is vital to sustaining intestinal intraepithelial lymphocytes (IELs). AHRR deficiency reduced IEL representation in a cell-intrinsic fashion. Single-cell RNA sequencing revealed an oxidative stress profile in Ahrr-/- IELs. AHRR deficiency unleashed AHR-induced expression of CYP1A1, a monooxygenase that generates reactive oxygen species, increasing redox imbalance, lipid peroxidation, and ferroptosis in Ahrr-/- IELs. Dietary supplementation with selenium or vitamin E to restore redox homeostasis rescued Ahrr-/- IELs. Loss of IELs in Ahrr-/- mice caused susceptibility to Clostridium difficile infection and dextran sodium-sulfate-induced colitis. Inflamed tissue of inflammatory bowel disease patients showed reduced Ahrr expression that may contribute to disease. We conclude that AHR signaling must be tightly regulated to prevent oxidative stress and ferroptosis of IELs and to preserve intestinal immune responses.


Subject(s)
Ferroptosis , Intraepithelial Lymphocytes , Animals , Mice , Intraepithelial Lymphocytes/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Oxidative Stress , Hydrocarbons
SELECTION OF CITATIONS
SEARCH DETAIL