Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Immunol ; 23(4): 619-631, 2022 04.
Article in English | MEDLINE | ID: mdl-35332328

ABSTRACT

Innate lymphocytes encompass a diverse array of phenotypic identities with specialized functions. DNA methylation and hydroxymethylation are essential for epigenetic fidelity and fate commitment. The landscapes of these modifications are unknown in innate lymphocytes. Here, we characterized the whole-genome distribution of methyl-CpG and 5-hydroxymethylcytosine (5hmC) in mouse innate lymphoid cell 3 (ILC3), ILC2 and natural killer (NK) cells. We identified differentially methylated regions (DMRs) and differentially hydroxymethylated regions (DHMRs) between ILC and NK cell subsets and correlated them with transcriptional signatures. We associated lineage-determining transcription factors (LDTFs) with demethylation and demonstrated unique patterns of DNA methylation/hydroxymethylation in relationship to open chromatin regions (OCRs), histone modifications and TF-binding sites. We further identified an association between hydroxymethylation and NK cell superenhancers (SEs). Using mice lacking the DNA hydroxymethylase TET2, we showed the requirement for TET2 in optimal production of hallmark cytokines by ILC3s and interleukin-17A (IL-17A) by inflammatory ILC2s. These findings provide a powerful resource for studying innate lymphocyte epigenetic regulation and decode the regulatory logic governing their identity.


Subject(s)
DNA Methylation , Immunity, Innate , Animals , Chromatin/genetics , Epigenesis, Genetic , Immunity, Innate/genetics , Killer Cells, Natural , Lymphocytes , Mice
2.
Cell ; 172(3): 534-548.e19, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29275861

ABSTRACT

Many tumors produce platelet-derived growth factor (PDGF)-DD, which promotes cellular proliferation, epithelial-mesenchymal transition, stromal reaction, and angiogenesis through autocrine and paracrine PDGFRß signaling. By screening a secretome library, we found that the human immunoreceptor NKp44, encoded by NCR2 and expressed on natural killer (NK) cells and innate lymphoid cells, recognizes PDGF-DD. PDGF-DD engagement of NKp44 triggered NK cell secretion of interferon gamma (IFN)-γ and tumor necrosis factor alpha (TNF-α) that induced tumor cell growth arrest. A distinctive transcriptional signature of PDGF-DD-induced cytokines and the downregulation of tumor cell-cycle genes correlated with NCR2 expression and greater survival in glioblastoma. NKp44 expression in mouse NK cells controlled the dissemination of tumors expressing PDGF-DD more effectively than control mice, an effect enhanced by blockade of the inhibitory receptor CD96 or CpG-oligonucleotide treatment. Thus, while cancer cell production of PDGF-DD supports tumor growth and stromal reaction, it concomitantly activates innate immune responses to tumor expansion.


Subject(s)
Brain Neoplasms/immunology , Cell Cycle Checkpoints , Glioblastoma/immunology , Killer Cells, Natural/immunology , Platelet-Derived Growth Factor/metabolism , Animals , Brain Neoplasms/pathology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Glioblastoma/pathology , Humans , Immunity, Innate , Interferon-gamma/metabolism , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Nat Immunol ; 20(8): 980-991, 2019 08.
Article in English | MEDLINE | ID: mdl-31209406

ABSTRACT

Innate lymphoid cells (ILCs) are tissue-resident lymphocytes categorized on the basis of their core regulatory programs and the expression of signature cytokines. Human ILC3s that produce the cytokine interleukin-22 convert into ILC1-like cells that produce interferon-γ in vitro, but whether this conversion occurs in vivo remains unclear. In the present study we found that ILC3s and ILC1s in human tonsils represented the ends of a spectrum that included additional discrete subsets. RNA velocity analysis identified an intermediate ILC3-ILC1 cluster, which had strong directionality toward ILC1s. In humanized mice, the acquisition of ILC1 features by ILC3s showed tissue dependency. Chromatin studies indicated that the transcription factors Aiolos and T-bet cooperated to repress regulatory elements active in ILC3s. A transitional ILC3-ILC1 population was also detected in the human intestine. We conclude that ILC3s undergo conversion into ILC1-like cells in human tissues in vivo, and that tissue factors and Aiolos were required for this process.


Subject(s)
Immunity, Innate/immunology , Interferon-gamma/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Lymphocytes/immunology , Palatine Tonsil/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Child , Child, Preschool , Humans , Ikaros Transcription Factor/metabolism , Intestinal Mucosa/cytology , Lymphocytes/classification , Lymphocytes/cytology , Mice , T-Box Domain Proteins/metabolism , Interleukin-22
5.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759002

ABSTRACT

Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-ß family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-ß family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-ß signaling mediated by the cytokine receptor TGFßR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-ß. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-ß signaling in NK cells.


Subject(s)
Killer Cells, Natural/cytology , Lymphopoiesis/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/immunology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/immunology , Animals , Case-Control Studies , Cell Differentiation , Gene Expression Profiling , Humans , Immunity, Innate/immunology , Immunoblotting , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocytes/cytology , Melanoma, Experimental/immunology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Smad4 Protein/immunology
6.
Immunity ; 52(4): 650-667.e10, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294406

ABSTRACT

Appropriate balance of T helper 17 (Th17) and regulatory T (Treg) cells maintains immune tolerance and host defense. Disruption of Th17-Treg cell balance is implicated in a number of immune-mediated diseases, many of which display dysregulation of the insulin-like growth factor (IGF) system. Here, we show that, among effector T cell subsets, Th17 and Treg cells selectively expressed multiple components of the IGF system. Signaling through IGF receptor (IGF1R) activated the protein kinase B-mammalian target of rapamycin (AKT-mTOR) pathway, increased aerobic glycolysis, favored Th17 cell differentiation over that of Treg cells, and promoted a heightened pro-inflammatory gene expression signature. Group 3 innate lymphoid cells (ILC3s), but not ILC1s or ILC2s, were similarly responsive to IGF signaling. Mice with deficiency of IGF1R targeted to T cells failed to fully develop disease in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Thus, the IGF system represents a previously unappreciated pathway by which type 3 immunity is modulated and immune-mediated pathogenesis controlled.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, IGF Type 1/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , Th17 Cells/immunology , Animals , Cell Communication , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Immune Tolerance , Immunity, Innate , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Receptor, IGF Type 1/genetics , Signal Transduction , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/genetics , Th17 Cells/pathology
7.
Nat Immunol ; 17(7): 783-9, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27328008

ABSTRACT

Innate lymphoid cells (ILCs) are a family of innate immune cells that have diverse functions during homeostasis and disease. Subsets of ILCs have phenotypes that mirror those of polarized helper T cell subsets in their expression of core transcription factors and effector cytokines. Given the similarities between these two classes of lymphocytes, it is important to understand which functions of ILCs are specialized and which are redundant with those of T cells. Here we discuss genetic mouse models that have been used to delineate the contributions of ILCs versus those of T cells and review the current understanding of the specialized in vivo functions of ILCs.


Subject(s)
Immunity, Innate , Inflammation/immunology , Lymphocytes/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Animals, Genetically Modified , Cytokines/metabolism , Disease Models, Animal , Homeostasis , Humans , Mice
8.
Nat Immunol ; 16(2): 153-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25501629

ABSTRACT

Fetal lymphoid tissue inducer (LTi) cells are required for lymph node and Peyer's patch (PP) organogenesis, but where these specialized group 3 innate lymphoid cells (ILC3s) develop remains unclear. Here, we identify extrahepatic arginase-1(+) Id2(+) fetal ILC precursors that express a transitional developmental phenotype (ftILCPs) and differentiate into ILC1s, ILC2s and ILC3s in vitro. These cells populate the intestine by embryonic day (E) 13.5 and, before PP organogenesis (E14.5-15), are broadly dispersed in the proximal gut, correlating with regions where PPs first develop. At E16.5, after PP development begins, ftILCPs accumulate at PP anlagen in a lymphotoxin-α-dependent manner. Thus, ftILCPs reside in the intestine during PP development, where they aggregate at PP anlagen after stromal cell activation and become a localized source of ILC populations.


Subject(s)
Cell Differentiation , Immunity, Innate , Intestine, Small/cytology , Intestine, Small/embryology , Lymphoid Tissue/cytology , Lymphoid Tissue/embryology , Animals , Arginase/metabolism , Cells, Cultured , Fetus/cytology , Fetus/immunology , Flow Cytometry , Immunohistochemistry , Intestine, Small/immunology , Lymphoid Tissue/immunology , Mice
9.
Immunity ; 48(6): 1208-1219.e4, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29858011

ABSTRACT

While signals that activate group 3 innate lymphoid cells (ILC3s) have been described, the factors that negatively regulate these cells are less well understood. Here we found that the tumor necrosis factor (TNF) superfamily member receptor activator of nuclear factor κB ligand (RANKL) suppressed ILC3 activity in the intestine. Deletion of RANKL in ILC3s and T cells increased C-C motif chemokine receptor 6 (CCR6)+ ILC3 abundance and enhanced production of interleukin-17A (IL-17A) and IL-22 in response to IL-23 and during infection with the enteric murine pathogen Citrobacter rodentium. Additionally, CCR6+ ILC3s produced higher amounts of the master transcriptional regulator RORγt at steady state in the absence of RANKL. RANKL-mediated suppression was independent of T cells, and instead occurred via interactions between CCR6+ ILC3s that expressed both RANKL and its receptor, RANK. Thus, RANK-RANKL interactions between ILC3s regulate ILC3 abundance and activation, suggesting that cell clustering may control ILC3 activity.


Subject(s)
Immunity, Innate/immunology , Lymphocyte Subsets/immunology , RANK Ligand/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Lymphocyte Subsets/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , RANK Ligand/metabolism , Receptors, CCR6/immunology
10.
Immunity ; 44(5): 1127-39, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27156386

ABSTRACT

The signals guiding differentiation of innate lymphoid cells (ILCs) within tissues are not well understood. Salivary gland (SG) ILCs as well as liver and intestinal intraepithelial ILC1 have markers that denote tissue residency and transforming growth factor-ß (TGF-ß) imprinting. We deleted Tgfbr2 in cells expressing the ILC and NK marker NKp46 and found that SG ILCs were reduced in number. They lost distinct tissue markers, such as CD49a, and the effector molecules TRAIL and CD73. Expression of the transcription factor Eomes, which promotes NK cell differentiation, was elevated. Conversely, Eomes deletion in NKp46(+) cells enhanced TGF-ß-imprinting of SG ILCs. Thus, TGF-ß induces SG ILC differentiation by suppressing Eomes. TGF-ß acted through a JNK-dependent, Smad4-independent pathway. Transcriptome analysis demonstrated that SG ILCs had characteristic of both NK cells and ILC1. Finally, TGF-ß imprinting of SG ILCs was synchronized with SG development, highlighting the impact of tissue microenvironment on ILC development.


Subject(s)
Cell Differentiation , Killer Cells, Natural/physiology , Lymphocytes/physiology , Salivary Glands/immunology , Transforming Growth Factor beta/metabolism , Animals , Antigens, Ly/metabolism , Cellular Microenvironment , Gene Expression Profiling , Immunity, Innate , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad4 Protein/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 119(45): e2214900119, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36279426

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are RORγT+ lymphocytes that are predominately enriched in mucosal tissues and produce IL-22 and IL-17A. They are the innate counterparts of Th17 cells. While Th17 lymphocytes utilize unique metabolic pathways in their differentiation program, it is unknown whether ILC3s make similar metabolic adaptations. We employed single-cell RNA sequencing and metabolomic profiling of intestinal ILC subsets to identify an enrichment of polyamine biosynthesis in ILC3s, converging on the rate-limiting enzyme ornithine decarboxylase (ODC1). In vitro and in vivo studies demonstrated that exogenous supplementation with the polyamine putrescine or its biosynthetic substrate, ornithine, enhanced ILC3 production of IL-22. Conditional deletion of ODC1 in ILC3s impaired mouse antibacterial defense against Citrobacter rodentium infection, which was associated with a decrease in anti-microbial peptide production by the intestinal epithelium. Furthermore, in a model of anti-CD40 colitis, deficiency of ODC1 in ILC3s markedly reduced the production of IL-22 and severity of inflammatory colitis. We conclude that ILC3-intrinsic polyamine biosynthesis facilitates efficient defense against enteric pathogens as well as exacerbates autoimmune colitis, thus representing an attractive target to modulate ILC3 function in intestinal disease.


Subject(s)
Colitis , Enterobacteriaceae Infections , Mice , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3 , Interleukin-17 , Ornithine Decarboxylase/genetics , Immunity, Innate , Putrescine , Colitis/genetics , Enterobacteriaceae Infections/genetics , Th17 Cells/metabolism , Ornithine , Anti-Bacterial Agents , Interleukin-22
12.
Proc Natl Acad Sci U S A ; 119(23): e2204557119, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35653568

ABSTRACT

C-type lectin domain family 4, member a4 (Clec4a4) is a C-type lectin inhibitory receptor specific for glycans thought to be exclusively expressed on murine CD8α− conventional dendritic cells. Using newly generated Clec4a4-mCherry knock-in mice, we identify a subset of Clec4a4-expressing eosinophils uniquely localized in the small intestine lamina propria. Clec4a4+ eosinophils evinced an immunomodulatory signature, whereas Clec4a4− eosinophils manifested a proinflammatory profile. Clec4a4+ eosinophils expressed high levels of aryl hydrocarbon receptor (Ahr), which drove the expression of Clec4a4 as well as other immunomodulatory features, such as PD-L1. The abundance of Clec4a4+ eosinophils was dependent on dietary AHR ligands, increased with aging, and declined in inflammatory conditions. Mice lacking AHR in eosinophils expanded innate lymphoid cells of type 2 and cleared Nippostrongylus brasiliensis infection more effectively than did wild-type mice. These results highlight the heterogeneity of eosinophils in response to tissue cues and identify a unique AHR-dependent subset of eosinophils in the small intestine with an immunomodulatory profile.


Subject(s)
Eosinophils , Receptors, Aryl Hydrocarbon , Receptors, Cell Surface , Eosinophilia/therapy , Food Hypersensitivity/therapy , Immunomodulation , Intestine, Small , Leukocyte Count , Ligands , Receptors, Aryl Hydrocarbon/genetics
13.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34083442

ABSTRACT

Lymphoid tissue inducer (LTi)-like cells are tissue resident innate lymphocytes that rapidly secrete cytokines that promote gut epithelial integrity and protect against extracellular bacterial infections.Here, we report that the retention of LTi-like cells in conventional solitary intestinal lymphoid tissue (SILT) is essential for controlling LTi-like cell function and is maintained by expression of the chemokine receptor CXCR5. Deletion of Cxcr5 functionally unleashed LTi-like cells in a cell intrinsic manner, leading to uncontrolled IL-17 and IL-22 production. The elevated production of IL-22 in Cxcr5-deficient mice improved gut barrier integrity and protected mice during infection with the opportunistic pathogen Clostridium difficile Interestingly, Cxcr5-/- mice developed LTi-like cell aggregates that were displaced from their typical niche at the intestinal crypt, and LTi-like cell hyperresponsiveness was associated with the local formation of this unconventional SILT. Thus, LTi-like cell positioning within mucosa controls their activity via niche-specific signals that temper cytokine production during homeostasis.


Subject(s)
Immunity, Innate , Interleukin-17/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, CXCR5/immunology , Animals , Gene Deletion , Interleukin-17/genetics , Interleukins/genetics , Intestinal Mucosa/cytology , Lymphocytes/cytology , Mice , Mice, Knockout , Receptors, CXCR5/genetics , Interleukin-22
14.
Nat Immunol ; 13(1): 58-66, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138715

ABSTRACT

Interleukin 4 (IL-4) and IL-13 are critical for responses to parasitic helminthes. We used genetically engineered reporter mice to assess the temporal and spatial production of these cytokines in vivo. In lymph nodes, IL-4, but not IL-13, was made by follicular helper T cells (T(FH) cells). In contrast, tissue type 2 helper T cells (T(H)2 cells) produced both cytokines. There was also divergent production of IL-4 and IL-13 among cells of the innate immune system, whereby basophils produced IL-4, whereas innate helper type 2 cells (Ih2 cells) produced IL-13. IL-13 production by T(H)2 and Ih2 cells was dependent on the transcription factor GATA-3, which was present in large amounts in these cells, and in contrast to the small amount of GATA-3 in T(FH) cells and basophils. The distinct localization and cellular expression of IL-4 and IL-13 explains their unique roles during allergic immunity.


Subject(s)
Hypersensitivity/immunology , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Animals , Basophils/immunology , GATA3 Transcription Factor/metabolism , Gene Expression , Hypersensitivity/genetics , Immunity, Innate , Interleukin-13/genetics , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Transport , STAT6 Transcription Factor/metabolism , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Th2 Cells/immunology
15.
Nat Immunol ; 12(6): 527-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21552267

ABSTRACT

Contributions by basophils to allergic and helminth immunity remain incompletely defined. Using sensitive interleukin 4 (Il4) reporter alleles, we demonstrate here that basophil IL-4 production occurs by a CD4(+) T cell-dependent process restricted to the peripheral tissues affected. We genetically marked and achieved specific deletion of basophils and found that basophils did not mediate T helper type 2 (T(H)2) priming in vivo. Two-photon imaging confirmed that basophils did not interact with antigen-specific T cells in lymph nodes but engaged in prolonged serial interactions with T cells in lung tissues. Although targeted deletion of IL-4 and IL-13 in either CD4(+) T cells or basophils had a minimal effect on worm clearance, deletion from both lineages demonstrated a nonredundant role for basophil cytokines in primary helminth immunity.


Subject(s)
Basophils/immunology , Interleukin-4/immunology , Lung/immunology , Strongylida Infections/immunology , Animals , Basophils/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Helminthiasis, Animal/immunology , Helminthiasis, Animal/metabolism , Helminthiasis, Animal/parasitology , Host-Parasite Interactions/immunology , Humans , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Liver/immunology , Liver/metabolism , Liver/parasitology , Lung/metabolism , Lung/parasitology , Lung Diseases, Parasitic/immunology , Lung Diseases, Parasitic/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Nippostrongylus/immunology , Nippostrongylus/physiology , Schistosoma mansoni/immunology , Schistosoma mansoni/physiology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
16.
Trends Immunol ; 41(8): 643-645, 2020 08.
Article in English | MEDLINE | ID: mdl-32605803

ABSTRACT

A recent article published in Immunity by Naoko Satoh-Takayama et al. examines interactions between group 2 innate lymphocytes and gastric microbes that enhance IgA production.


Subject(s)
Immunity, Innate , Lymphocytes , Antibody Formation , Bacteria , Humans , Immunoglobulin A , Stomach
17.
Immunity ; 37(4): 735-46, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23021952

ABSTRACT

After an immune response, the expanded population of antigen-specific CD4(+) T cells contract to steady state levels. We have found that the contraction is neither cell-autonomous nor mediated by competition for generic trophic factors, but regulated by relatively rare subsets of neighboring CD4(+) T cells not necessarily of a conventional regulatory T cell lineage. These regulators, referred to as deletors, specifically limit the frequency of particular antigen-specific T cells even though they are not reactive to the same agonist as their targets. Instead, an isolated deletor could outcompete the target for recognition of a shared, nonstimulatory endogenous peptide-MHC ligand. This mechanism was sufficient to prevent even agonist-driven autoimmune disease in a lymphopenic environment. Such a targeted regulation of homeostasis within narrow colonies of T cells with related TCR specificities for subthreshold ligands might help to prevent the loss of unrelated TCRs during multiple responses, preserving the valuable diversity of the repertoire.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , T-Cell Antigen Receptor Specificity , Animals , Autoimmunity , Cell Survival , Cells, Cultured , Ligands , Lymphopenia/immunology , Mice , Receptors, Antigen, T-Cell/immunology
18.
Eur J Immunol ; 43(8): 2010-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630120

ABSTRACT

Traumatic brain injury (TBI) elicits innate inflammatory responses that can lead to secondary brain injury. To better understand the mechanisms involved in TBI-induced inflammation, we examined the nature of macrophages responding to TBI in mice. In this model, brain macrophages were increased >20-fold the day after injury and >77-fold 4 days after injury in the ipsilateral hemisphere compared with sham controls. TBI macrophage subsets were identified by using a reporter mouse strain (YARG) that expresses eYFP from an internal ribosome entry site (IRES) inserted at the 3' end of the gene for arginase-1 (Arg1), a hallmark of alternatively activated (M2) macrophages. One day after TBI, 21 ± 1.5% of ipsilateral brain macrophages expressed relatively high levels of Arg1 as detected by yellow fluorescent protein, and this subpopulation declined thereafter. Arg1(+) cells localized with macrophages near the TBI lesion. Gene expression analysis of sorted Arg1(+) and Arg1(-) brain macrophages revealed that both populations had profiles that included features of conventional M2 macrophages and classically activated (M1) macrophages. The Arg1(+) cells differed from Arg1(-) cells in multiple aspects, most notably in their chemokine repertoires. Thus, the macrophage response to TBI initially involves heterogeneous polarization toward at least two major subsets.


Subject(s)
Arginase/metabolism , Brain Injuries/immunology , Brain/immunology , Macrophage Activation/immunology , Macrophages/immunology , Animals , Arginase/genetics , Bacterial Proteins/genetics , Cell Movement , Chemokines/biosynthesis , Gene Expression Profiling , Inflammation/immunology , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ribosomes/genetics , Ribosomes/metabolism
19.
Sci Immunol ; 9(95): eadi5374, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758808

ABSTRACT

The gut microbiota and tumor-associated macrophages (TAMs) affect tumor responses to anti-programmed cell death protein 1 (PD-1) immune checkpoint blockade. Reprogramming TAM by either blocking or deleting the macrophage receptor triggering receptor on myeloid cells 2 (TREM2) attenuates tumor growth, and lack of functional TREM2 enhances tumor elimination by anti-PD-1. Here, we found that anti-PD-1 treatment combined with TREM2 deficiency in mice induces proinflammatory programs in intestinal macrophages and a concomitant expansion of Ruminococcus gnavus in the gut microbiota. Gavage of wild-type mice with R. gnavus enhanced anti-PD-1-mediated tumor elimination, recapitulating the effect occurring in the absence of TREM2. A proinflammatory intestinal environment coincided with expansion, increased circulation, and migration of TNF-producing CD4+ T cells to the tumor bed. Thus, TREM2 remotely controls anti-PD-1 immune checkpoint blockade through modulation of the intestinal immune environment and microbiota, with R. gnavus emerging as a potential probiotic agent for increasing responsiveness to anti-PD-1.


Subject(s)
Gastrointestinal Microbiome , Immunotherapy , Macrophages , Membrane Glycoproteins , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Receptors, Immunologic , Animals , Receptors, Immunologic/immunology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Mice , Gastrointestinal Microbiome/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Macrophages/immunology , Immune Checkpoint Inhibitors/pharmacology , Mice, Knockout , Female , Intestines/immunology
20.
Immunohorizons ; 5(4): 193-209, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33906960

ABSTRACT

Necrotizing enterocolitis (NEC) causes significant morbidity and mortality in premature infants; therefore, the identification of therapeutic and preventative strategies against NEC remains a high priority. The ligand-dependent transcription factor aryl hydrocarbon receptor (AhR) is well known to contribute to the regulation of intestinal microbial communities and amelioration of intestinal inflammation. However, the role of AhR signaling in NEC is unclear. Experimental NEC was induced in 4-d-old wild-type mice or mice lacking AhR expression in the intestinal epithelial cells or AhR expression in CD11c+ cells (AhRΔCD11c) by subjecting animals to twice daily hypoxic stress and gavage feeding with formula supplemented with LPS and enteric bacteria. During NEC, compared with wild-type mice treated with vehicle, littermates treated with an AhR proligand, indole-3-carbinol, had reduced expression of Il1b and Marco, a scavenger receptor that mediates dendritic cell activation and the recognition and clearance of bacterial pathogens by macrophages. Furthermore, indole-3-carbinol treatment led to the downregulation of genes involved in cytokine and chemokine, as revealed by pathway enrichment analysis. AhR expression in the intestinal epithelial cells and their cre-negative mouse littermates were similarly susceptible to experimental NEC, whereas AhRΔCD11c mice with NEC exhibited heightened inflammatory responses compared with their cre-negative mouse littermates. In seeking to determine the mechanisms involved in this increased inflammatory response, we identified the Tim-4- monocyte-dependent subset of macrophages as increased in AhRΔCD11c mice compared with their cre-negative littermates. Taken together, these findings demonstrate the potential for AhR ligands as a novel immunotherapeutic approach to the management of this devastating disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Enterocolitis, Necrotizing/drug therapy , Indoles/pharmacology , Intestinal Mucosa/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/pathology , Humans , Indoles/therapeutic use , Interleukin-1beta/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL