Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Immunity ; 54(6): 1320-1337.e4, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33945787

ABSTRACT

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1s) are heterogenous innate lymphocytes broadly defined in mice as Lin-NK1.1+NKp46+ cells that express the transcription factor T-BET and produce interferon-γ. The ILC1 definition primarily stems from studies on liver and small intestinal populations. However, NK1.1+NKp46+ cells in the salivary glands, uterus, adipose, and other tissues exhibit nonuniform programs that differ from those of liver or intestinal ILC1s or NK cells. Here, we performed single-cell RNA sequencing on murine NK1.1+NKp46+ cells from blood, spleen, various tissues, and solid tumors. We identified gene expression programs of tissue-specific ILC1s, tissue-specific NK cells, and non-tissue-specific populations in blood, spleen, and other tissues largely corresponding to circulating cells. Moreover, we found that circulating NK cell programs were reshaped in tumor-bearing mice. Core programs of circulating and tumor NK cells paralleled conserved human NK cells signatures, advancing our understanding of the human NK-ILC1 spectrum.


Subject(s)
Immunity, Innate/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Animals , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Neoplasms/immunology , Single-Cell Analysis/methods , Transcription Factors/immunology
2.
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
3.
Immunity ; 51(3): 479-490.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31402259

ABSTRACT

Natural killer (NK) cells are cytotoxic type 1 innate lymphoid cells (ILCs) that defend against viruses and mediate anti-tumor responses, yet mechanisms controlling their development and function remain incompletely understood. We hypothesized that the abundantly expressed microRNA-142 (miR-142) is a critical regulator of type 1 ILC biology. Interleukin-15 (IL-15) signaling induced miR-142 expression, whereas global and ILC-specific miR-142-deficient mice exhibited a cell-intrinsic loss of NK cells. Death of NK cells resulted from diminished IL-15 receptor signaling within miR-142-deficient mice, likely via reduced suppressor of cytokine signaling-1 (Socs1) regulation by miR-142-5p. ILCs persisting in Mir142-/- mice demonstrated increased expression of the miR-142-3p target αV integrin, which supported their survival. Global miR-142-deficient mice exhibited an expansion of ILC1-like cells concurrent with increased transforming growth factor-ß (TGF-ß) signaling. Further, miR-142-deficient mice had reduced NK-cell-dependent function and increased susceptibility to murine cytomegalovirus (MCMV) infection. Thus, miR-142 critically integrates environmental cues for proper type 1 ILC homeostasis and defense against viral infection.


Subject(s)
Homeostasis/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , MicroRNAs/immunology , Animals , Cell Line , Female , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , NIH 3T3 Cells , Receptors, Interleukin-15/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Transforming Growth Factor beta/immunology
4.
Nat Immunol ; 16(3): 306-17, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25621825

ABSTRACT

The recognized diversity of innate lymphoid cells (ILCs) is rapidly expanding. Three ILC classes have emerged, ILC1, ILC2 and ILC3, with ILC1 and ILC3 including several subsets. The classification of some subsets is unclear, and it remains controversial whether natural killer (NK) cells and ILC1 cells are distinct cell types. To address these issues, we analyzed gene expression in ILCs and NK cells from mouse small intestine, spleen and liver, as part of the Immunological Genome Project. The results showed unique gene-expression patterns for some ILCs and overlapping patterns for ILC1 cells and NK cells, whereas other ILC subsets remained indistinguishable. We identified a transcriptional program shared by small intestine ILCs and a core ILC signature. We revealed and discuss transcripts that suggest previously unknown functions and developmental paths for ILCs.


Subject(s)
Immunity, Innate/genetics , Immunity, Innate/immunology , Lymphocytes/physiology , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Animals , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Lymphocytes/immunology , Male , Mice , Mice, Inbred C57BL
5.
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
6.
J Immunol ; 192(10): 4487-91, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24740507

ABSTRACT

Nfil3 is viewed as an obligate transcription factor for NK cell development. However, mouse CMV (MCMV) infection recently was shown to bypass the requirement for Nfil3 by inducing the appearance of NK cells that express the MCMV-specific receptor Ly49H. Thus, signals transmitted by Ly49H and proinflammatory cytokines are sufficient to promote NK cell differentiation in the absence of Nfil3. In this study, we report that salivary gland (SG) NK cells develop in an Nfil3-independent fashion in the steady-state in the absence of MCMV or any infection. Moreover, we show that SG NK cells have an integrin profile reminiscent of tissue-resident lymphocytes and express TRAIL for killing target cells. These results demonstrate that SG NK cells, although related to conventional NK cells, are a distinct subset of innate lymphoid cells that deviates from the conventional developmental pathway, perhaps under the influence of tissue-specific factors.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Killer Cells, Natural/immunology , Salivary Glands/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , Organ Specificity/genetics , Organ Specificity/immunology , Salivary Glands/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
7.
J Exp Med ; 221(3)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38197946

ABSTRACT

Innate lymphoid cells (ILCs) are a heterogeneous population of lymphocytes that coordinate early immune responses and maintain tissue homeostasis. Type 1 innate immune responses are mediated by natural killer (NK) cells and group 1 ILCs (ILC1s). Despite their shared features, NK cells and ILC1s display profound differences among various tissue microenvironments. Here, we identify the inositol polyphosphatase INPP4B as a hallmark feature of tissue-resident ILC1s and intratumoral NK cells using an scRNA-seq atlas of tissue-associated and circulating NK/ILC1s. Conditional deletion of Inpp4b in ILC1s and NK cells reveals that it is necessary for the homeostasis of tissue-resident ILC1s but not circulating NK cells at steady-state. Inpp4b-deficient cells display increased rates of apoptosis and reduced activation of the prosurvival molecule AKT. Furthermore, expression of Inpp4b by NK/ILC1s is necessary for their presence in the intratumoral environment, and lack of Inpp4b impairs antitumor immunity. These findings highlight INPP4B as a novel regulator of tissue residency and antitumor function in ILC1s and NK cells.


Subject(s)
Immunity, Innate , Proto-Oncogene Proteins c-akt , Killer Cells, Natural , Homeostasis
8.
Cancers (Basel) ; 13(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546248

ABSTRACT

Natural Killer cells belong to group 1 innate lymphoid cells, which also includes ILC1s. NK/ILC1s are highly heterogeneous cell types showing distinct phenotypes across tissues and conditions. NK cells have long been described as innate lymphocytes able to directly and rapidly kill tumor cells without antigen-restriction. Different mechanisms were shown to modulate NK cell activation and tumor resistance, mainly based on cytokine stimulation and receptor-ligand interactions, and several strategies have been developed to target NK cells in tumor immunotherapy to promote NK cell function and overcome tumor evasion. The characterization of ILC1 distinct phenotype and function and the specific role in tumors still needs further investigation and will be essential to better understand the impact of innate lymphoid cells in tumors. Here, we review key aspects of NK cell biology that are relevant in tumor immune surveillance, emphasizing the most recent findings in the field. We describe the novel therapeutical strategies that have been developed in tumor immunotherapy targeting NK cells, and we summarize some recent findings related to NK cell/ILC1 transition in tumor models.

9.
Curr Protoc ; 1(3): e77, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33740294

ABSTRACT

Recent findings position tuft cells as key mediators of intestinal immunity through their production of the cytokine interleukin (IL)-25 and activation of group 2 innate lymphoid cells (ILC2s). Though tuft cells are found in numerous epithelial tissues, their phenotype and function have been best characterized in the small intestine, where robust in vivo techniques have enabled the dissection of their cellular function, ontogeny, and key signaling pathways. We describe methods for the identification, quantification, and manipulation of tuft cells, focusing on analysis of ILC2s as a readout of tuft cell function. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry Alternate Protocol: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry in the context of type 2 inflammation Basic Protocol 2: Ex vivo analysis of small intestinal tuft cells by imaging of intestinal Swiss roll Basic Protocol 3: Tuft-ILC2 circuit activation by oral gavage of adult Nippostrongylus brasiliensis worms Basic Protocol 4: Circuit activation by colonization with Tritrichomonas spp. Basic Protocol 5: Circuit activation by treatment with succinate in drinking water Basic Protocol 6: Circuit activation by treatment with recombinant IL-25.


Subject(s)
Immunity, Innate , Tritrichomonas , Animals , Intestine, Small , Lymphocytes , Nippostrongylus
10.
Front Immunol ; 10: 1423, 2019.
Article in English | MEDLINE | ID: mdl-31312200

ABSTRACT

The gastrointestinal tract hosts the largest collection of commensal microbes in the body. Infections at this site can cause significant perturbations in the microbiota, known as dysbiosis, that facilitate the expansion of pathobionts, and can elicit inappropriate immune responses that impair the intestinal barrier function. Dysbiosis typically occurs during intestinal infection with Toxoplasma gondii. Host resistance to T. gondii depends on a potent Th1 response. In addition, a Th17 response is also elicited. How Th17 cells contribute to the host response to T. gondii remains unclear. Here we show that class I-restricted T cell-associated molecule (CRTAM) expression on T cells is required for an optimal IL-17 production during T. gondii infection. Moreover, that the lack of IL-17, results in increased immunopathology caused by an impaired antimicrobial peptide production and bacterial translocation from the intestinal lumen to the mesenteric lymph nodes and spleen.


Subject(s)
Dysbiosis/parasitology , Gastrointestinal Microbiome/immunology , Immunoglobulins/metabolism , Interleukin-17/metabolism , Th17 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cell Differentiation/genetics , Dysbiosis/immunology , Female , Immunoglobulins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Toxoplasmosis, Animal/parasitology , alpha-Defensins/metabolism , beta-Defensins/metabolism
11.
Science ; 357(6353): 806-810, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28775213

ABSTRACT

The small intestine contains CD4+CD8αα+ double-positive intraepithelial lymphocytes (DP IELs), which originate from intestinal CD4+ T cells through down-regulation of the transcription factor Thpok and have regulatory functions. DP IELs are absent in germ-free mice, which suggests that their differentiation depends on microbial factors. We found that DP IEL numbers in mice varied in different vivaria, correlating with the presence of Lactobacillus reuteri This species induced DP IELs in germ-free mice and conventionally-raised mice lacking these cells. L. reuteri did not shape the DP-IEL-TCR (TCR, T cell receptor) repertoire but generated indole derivatives of tryptophan that activated the aryl-hydrocarbon receptor in CD4+ T cells, allowing Thpok down-regulation and differentiation into DP IELs. Thus, L. reuteri, together with a tryptophan-rich diet, can reprogram intraepithelial CD4+ T cells into immunoregulatory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Limosilactobacillus reuteri/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Germ-Free Life , Indoles/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Tryptophan/metabolism
12.
Immunol Lett ; 179: 19-24, 2016 11.
Article in English | MEDLINE | ID: mdl-27394699

ABSTRACT

Innate lymphoid cells (ILCs) are a heterogeneous population of cells with diverse roles in immune responses. Three major groups of ILCs have been defined on the basis of similarity in their production of signature cytokines, developmental requirements, and phenotypic markers. Group 1 ILCs produce IFN-γ, express the T-box transcription factors (TF) T-bet and/or Eomesodermin (Eomes), group 2 ILCs secrete IL-5 and IL-13 and express the TF GATA-3, while group 3 ILCs produce IL-22 and IL-17 and express the TF RORgt. In this review, we will briefly overview each group in terms of phenotype, function and development and then focus more extensively on group 1 ILCs, expanding on their emerging diversity, their disparate functions and the differences between NK cells and ILC1.


Subject(s)
Cell Plasticity/immunology , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Plasticity/genetics , Cytokines/metabolism , Disease Susceptibility , Gene Expression Regulation, Developmental , Humans , Interferon-gamma/biosynthesis , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/cytology , Organ Specificity , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Curr Protoc ; 1(7): e205, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34242484
14.
Curr Opin Immunol ; 32: 71-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25615701

ABSTRACT

Here, we illustrate the complexity of ILC subsets and discuss novel functions, focusing on emerging mechanisms of crosstalk with other immune cells and the microbiota. Furthermore, we highlight recent insights into the development of ILCs, including the common pathways they share as well as points of divergence between ILC groups and subsets.


Subject(s)
Cell Differentiation , Immunity, Innate , Lymphocyte Subsets/cytology , Lymphocyte Subsets/physiology , Animals , Humans , Lymphoid Progenitor Cells/cytology , Phenotype
15.
J Exp Med ; 211(4): 623-33, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24687959

ABSTRACT

Retention of lymphocytes in the intestinal mucosa requires specialized chemokine receptors and adhesion molecules. We find that both CD4(+)CD8(+) and CD4(+) T cells in the intestinal epithelium, as well as CD8(+) T cells in the intestinal mucosa and mesenteric lymph nodes, express the cell adhesion molecule class I-restricted T cell-associated molecule (Crtam) upon activation, whereas the ligand of Crtam, cell adhesion molecule 1 (Cadm1), is expressed on gut CD103(+)DCs. Lack of Crtam-Cadm1 interactions in Crtam(-/-) and Cadm1(-/-) mice results in loss of CD4(+)CD8(+) T cells, which arise from mucosal CD4(+) T cells that acquire a CD8 lineage expression profile. After acute oral infection with Toxoplasma gondii, both WT and Crtam(-/-) mice mounted a robust TH1 response, but markedly fewer TH17 cells were present in the intestinal mucosa of Crtam(-/-) mice. The almost exclusive TH1 response in Crtam(-/-) mice resulted in more efficient control of intestinal T. gondii infection. Thus, Crtam-Cadm1 interactions have a major impact on the residency and maintenance of CD4(+)CD8(+) T cells in the gut mucosa in the steady state. During pathogenic infection, Crtam-Cadm1 interactions regulate the dynamic equilibrium between newly formed CD4(+) T cells and their retention in the gut, thereby shaping representation of disparate CD4(+) T cell subsets and the overall quality of the CD4(+) T cell response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology , Immunoglobulins/metabolism , Th17 Cells/immunology , Toxoplasma/physiology , Animals , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Polarity , Cytokines/biosynthesis , Dendritic Cells/metabolism , Gastrointestinal Tract/pathology , Immunoglobulins/deficiency , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Ligands , Lymphocyte Count , Mice , Mice, Inbred C57BL , Neutralization Tests , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL