Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Annu Rev Immunol ; 42(1): 489-519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941607

ABSTRACT

Recent advances have contributed to a mechanistic understanding of neuroimmune interactions in the intestine and revealed an essential role of this cross talk for gut homeostasis and modulation of inflammatory and infectious intestinal diseases. In this review, we describe the innervation of the intestine by intrinsic and extrinsic neurons and then focus on the bidirectional communication between neurons and immune cells. First, we highlight the contribution of neuronal subtypes to the development of colitis and discuss the different immune and epithelial cell types that are regulated by neurons via the release of neuropeptides and neurotransmitters. Next, we review the role of intestinal inflammation in the development of visceral hypersensitivity and summarize how inflammatory mediators induce peripheral and central sensitization of gut-innervating sensory neurons. Finally, we outline the importance of immune cells and gut microbiota for the survival and function of different neuronal populations at homeostasis and during bacterial and helminth infection.


Subject(s)
Neuroimmunomodulation , Humans , Animals , Intestines/immunology , Homeostasis , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/metabolism , Neurons/immunology , Neuropeptides/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism
2.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31813624

ABSTRACT

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Nociceptors/physiology , Animals , Epithelium/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Peyer's Patches/innervation , Peyer's Patches/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
3.
Immunity ; 55(9): 1663-1679.e6, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36070768

ABSTRACT

Interleukin-23 receptor plays a critical role in inducing inflammation and autoimmunity. Here, we report that Th1-like cells differentiated in vitro with IL-12 + IL-21 showed similar IL-23R expression to that of pathogenic Th17 cells using eGFP reporter mice. Fate mapping established that these cells did not transition through a Th17 cell state prior to becoming Th1-like cells, and we observed their emergence in vivo in the T cell adoptive transfer colitis model. Using IL-23R-deficient Th1-like cells, we demonstrated that IL-23R was required for the development of a highly colitogenic phenotype. Single-cell RNA sequencing analysis of intestinal T cells identified IL-23R-dependent genes in Th1-like cells that differed from those expressed in Th17 cells. The perturbation of one of these regulators (CD160) in Th1-like cells inhibited the induction of colitis. We thus uncouple IL-23R as a purely Th17 cell-specific factor and implicate IL-23R signaling as a pathogenic driver in Th1-like cells inducing tissue inflammation.


Subject(s)
Colitis , Receptors, Interleukin , Animals , Inflammation/metabolism , Interleukin-23/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Th1 Cells , Th17 Cells
4.
Immunity ; 51(6): 1102-1118.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31757673

ABSTRACT

Young children are more susceptible to developing allergic asthma than adults. As neural innervation of the peripheral tissue continues to develop after birth, neurons may modulate tissue inflammation in an age-related manner. Here we showed that sympathetic nerves underwent a dopaminergic-to-adrenergic transition during post-natal development of the lung in mice and humans. Dopamine signaled through a specific dopamine receptor (DRD4) to promote T helper 2 (Th2) cell differentiation. The dopamine-DRD4 pathway acted synergistically with the cytokine IL-4 by upregulating IL-2-STAT5 signaling and reducing inhibitory histone trimethylation at Th2 gene loci. In murine models of allergen exposure, the dopamine-DRD4 pathway augmented Th2 inflammation in the lungs of young mice. However, this pathway operated marginally after sympathetic nerves became adrenergic in the adult lung. Taken together, the communication between dopaminergic nerves and CD4+ T cells provides an age-related mechanism underlying the susceptibility to allergic inflammation in the early lung.


Subject(s)
Adrenergic Neurons/cytology , Asthma/pathology , Dopamine/metabolism , Dopaminergic Neurons/cytology , Lung/pathology , Th2 Cells/immunology , Adolescent , Adult , Age Factors , Aged , Animals , Asthma/immunology , Cells, Cultured , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Interleukin-2/metabolism , Interleukin-4/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurogenesis/physiology , Receptors, Dopamine D4/metabolism , STAT5 Transcription Factor/metabolism , Sympathetic Nervous System/cytology
5.
Immunity ; 51(4): 696-708.e9, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618654

ABSTRACT

Signaling abnormalities in immune responses in the small intestine can trigger chronic type 2 inflammation involving interaction of multiple immune cell types. To systematically characterize this response, we analyzed 58,067 immune cells from the mouse small intestine by single-cell RNA sequencing (scRNA-seq) at steady state and after induction of a type 2 inflammatory reaction to ovalbumin (OVA). Computational analysis revealed broad shifts in both cell-type composition and cell programs in response to the inflammation, especially in group 2 innate lymphoid cells (ILC2s). Inflammation induced the expression of exon 5 of Calca, which encodes the alpha-calcitonin gene-related peptide (α-CGRP), in intestinal KLRG1+ ILC2s. α-CGRP antagonized KLRG1+ ILC2s proliferation but promoted IL-5 expression. Genetic perturbation of α-CGRP increased the proportion of intestinal KLRG1+ ILC2s. Our work highlights a model where α-CGRP-mediated neuronal signaling is critical for suppressing ILC2 expansion and maintaining homeostasis of the type 2 immune machinery.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Inflammation/immunology , Intestines/immunology , Lymphocytes/immunology , Neuropeptides/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Cells, Cultured , Computational Biology , Immunity, Innate , Interleukin-5/genetics , Interleukin-5/metabolism , Lectins, C-Type/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neuropeptides/genetics , Receptors, Immunologic/metabolism , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Th2 Cells/immunology , Transcriptome , Up-Regulation
6.
Immunity ; 51(4): 709-723.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31604686

ABSTRACT

Neuroimmune interactions have emerged as critical modulators of allergic inflammation, and type 2 innate lymphoid cells (ILC2s) are an important cell type for mediating these interactions. Here, we show that ILC2s expressed both the neuropeptide calcitonin gene-related peptide (CGRP) and its receptor. CGRP potently inhibited alarmin-driven type 2 cytokine production and proliferation by lung ILC2s both in vitro and in vivo. CGRP induced marked changes in ILC2 expression programs in vivo and in vitro, attenuating alarmin-driven proliferative and effector responses. A distinct subset of ILCs scored highly for a CGRP-specific gene signature after in vivo alarmin stimulation, suggesting CGRP regulated this response. Finally, we observed increased ILC2 proliferation and type 2 cytokine production as well as exaggerated responses to alarmins in mice lacking the CGRP receptor. Together, these data indicate that endogenous CGRP is a critical negative regulator of ILC2 responses in vivo.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Lymphocytes/physiology , Neuropeptides/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Alarmins/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Cell Proliferation , Cells, Cultured , Feedback, Physiological , Immunity, Innate , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Neuropeptides/genetics , Receptors, Calcitonin Gene-Related Peptide/genetics , Signal Transduction , Th2 Cells/immunology
7.
Nature ; 549(7672): 351-356, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28902842

ABSTRACT

Type 2 innate lymphoid cells (ILC2s) both contribute to mucosal homeostasis and initiate pathologic inflammation in allergic asthma. However, the signals that direct ILC2s to promote homeostasis versus inflammation are unclear. To identify such molecular cues, we profiled mouse lung-resident ILCs using single-cell RNA sequencing at steady state and after in vivo stimulation with the alarmin cytokines IL-25 and IL-33. ILC2s were transcriptionally heterogeneous after activation, with subpopulations distinguished by expression of proliferative, homeostatic and effector genes. The neuropeptide receptor Nmur1 was preferentially expressed by ILC2s at steady state and after IL-25 stimulation. Neuromedin U (NMU), the ligand of NMUR1, activated ILC2s in vitro, and in vivo co-administration of NMU with IL-25 strongly amplified allergic inflammation. Loss of NMU-NMUR1 signalling reduced ILC2 frequency and effector function, and altered transcriptional programs following allergen challenge in vivo. Thus, NMUR1 signalling promotes inflammatory ILC2 responses, highlighting the importance of neuro-immune crosstalk in allergic inflammation at mucosal surfaces.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/pathology , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Animals , Female , Gene Expression Regulation , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukin-33/immunology , Ligands , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Neurotransmitter/biosynthesis , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , Transcription, Genetic
9.
Immunol Rev ; 286(1): 53-73, 2018 11.
Article in English | MEDLINE | ID: mdl-30294962

ABSTRACT

Type 2 immunity against pathogens is tightly regulated to ensure appropriate inflammatory responses that clear infection and prevent excessive tissue damage. Recent research has shown that type 2 innate lymphoid cells (ILC2s) contribute to steady-state tissue integrity and exert tissue-specific functions. However, upon exposure to inflammatory stimuli, they also initiate and amplify type 2 inflammation by inducing mucus production, eosinophilia, and Th2 differentiation. In this review, we discuss the regulation of ILC2 activation by transcription factors and metabolic pathways, as well as by extrinsic signals such as cytokines, lipid mediators, hormones, and neuropeptides. We also review recent discoveries about ILC2 plasticity and heterogeneity in different tissues, as revealed partly through single-cell RNA sequencing of transcriptional responses to various stimuli. Understanding the tissue-specific pathways that regulate ILC2 diversity and function is a critical step in the development of potential therapies for allergic diseases.


Subject(s)
Inflammation/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Cytokines/metabolism , High-Throughput Nucleotide Sequencing , Humans , Immunity, Innate , Lymphocyte Activation , Single-Cell Analysis
10.
Trends Neurosci ; 45(4): 251-253, 2022 04.
Article in English | MEDLINE | ID: mdl-34973845

ABSTRACT

In the gut, coordinated cell interactions regulate tissue repair and immunity. How enteric glial cells (EGCs) mediate these processes remained elusive. In a recent paper, Progatzky et al. demonstrate that EGCs interact with immune and mesothelial cells under homeostasis and helminth infection, revealing an indispensable role of an interferon-γ (IFNγ)-EGC-CXCL10 axis in tissue repair.


Subject(s)
Enteric Nervous System , Neuroglia , Cell Communication , Homeostasis , Humans , Neuroglia/physiology
11.
Nat Med ; 24(10): 1625-1626, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30013196

ABSTRACT

In the version of this article initially published, the line graph showing TNF-α levels in Fig. 2d was inadvertently duplicated. A graph of IL-6 levels should be shown in place of the duplication.These results were also incorrectly described in the main text, which originally stated: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokines TNF-α and IL-6, and the chemokine CXCL-1 (Fig. 2d)". This should instead read: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokine TNF-α and the chemokine CXCL-1 (Fig. 2d)".In the supplementary information initially posted online, incorrect bar graphs were presented in Supplementary Fig. 1b (VG, TRPV1+ data, top panel) and Supplementary Fig. 4b (DRG, CGRP+ data, middle panel).

12.
Nat Med ; 24(4): 417-426, 2018 05.
Article in English | MEDLINE | ID: mdl-29505031

ABSTRACT

Lung-innervating nociceptor sensory neurons detect noxious or harmful stimuli and consequently protect organisms by mediating coughing, pain, and bronchoconstriction. However, the role of sensory neurons in pulmonary host defense is unclear. Here, we found that TRPV1+ nociceptors suppressed protective immunity against lethal Staphylococcus aureus pneumonia. Targeted TRPV1+-neuron ablation increased survival, cytokine induction, and lung bacterial clearance. Nociceptors suppressed the recruitment and surveillance of neutrophils, and altered lung γδ T cell numbers, which are necessary for immunity. Vagal ganglia TRPV1+ afferents mediated immunosuppression through release of the neuropeptide calcitonin gene-related peptide (CGRP). Targeting neuroimmunological signaling may be an effective approach to treat lung infections and bacterial pneumonia.


Subject(s)
Bacterial Infections/immunology , Neutrophils/metabolism , Nociceptors/metabolism , Pneumonia/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Sensory Receptor Cells/metabolism , T-Lymphocytes/immunology , Animals , Bacterial Infections/microbiology , Calcitonin Gene-Related Peptide/metabolism , Cytokines/metabolism , Female , Host-Pathogen Interactions/immunology , Male , Mice, Inbred C57BL , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/physiology , TRPV Cation Channels/metabolism , Vagus Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL