Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cell ; 180(6): 1280-1280.e1, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200803

ABSTRACT

NK cells are broadly distributed innate lymphoid cells (ILCs) encompassing distinct populations based on CD11b and CD27 expression in mice or CD56 intensity in humans. Involved in anti-viral and anti-tumor immunity thanks to their cytokines and chemokines secretion as well as their cytotoxic capabilities, NK cells have emerged as a promising therapeutic target in several solid tumors and hematological malignancies. To view this Snapshot, open or download the PDF.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Animals , Cytokines/metabolism , Humans , Immunity, Innate , Immunotherapy, Active/methods , Mice , Neoplasms/immunology
2.
Cell ; 176(4): 716-728.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712871

ABSTRACT

Sensory axons degenerate following separation from their cell body, but partial injury to peripheral nerves may leave the integrity of damaged axons preserved. We show that an endogenous ligand for the natural killer (NK) cell receptor NKG2D, Retinoic Acid Early 1 (RAE1), is re-expressed in adult dorsal root ganglion neurons following peripheral nerve injury, triggering selective degeneration of injured axons. Infiltration of cytotoxic NK cells into the sciatic nerve by extravasation occurs within 3 days following crush injury. Using a combination of genetic cell ablation and cytokine-antibody complex stimulation, we show that NK cell function correlates with loss of sensation due to degeneration of injured afferents and reduced incidence of post-injury hypersensitivity. This neuro-immune mechanism of selective NK cell-mediated degeneration of damaged but intact sensory axons complements Wallerian degeneration and suggests the therapeutic potential of modulating NK cell function to resolve painful neuropathy through the clearance of partially damaged nerves.


Subject(s)
Killer Cells, Natural/physiology , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Peripheral Nerve Injuries/metabolism , Animals , Axons , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nerve Regeneration , Neurons/cytology , Neurons, Afferent/immunology , Neurons, Afferent/metabolism , Nuclear Matrix-Associated Proteins/physiology , Nucleocytoplasmic Transport Proteins/physiology , Pain , Peripheral Nerve Injuries/immunology , Peripheral Nervous System Diseases , Sciatic Nerve , Sensory Receptor Cells/metabolism
3.
Nat Immunol ; 19(9): 954-962, 2018 09.
Article in English | MEDLINE | ID: mdl-30127438

ABSTRACT

Controlling the balance between immunity and immunopathology is crucial for host resistance to pathogens. After infection, activation of the hypothalamic-pituitary-adrenal (HPA) axis leads to the production of glucocorticoids. However, the pleiotropic effects of these steroid hormones make it difficult to delineate their precise role(s) in vivo. Here we found that the regulation of natural killer (NK) cell function by the glucocorticoid receptor (GR) was required for host survival after infection with mouse cytomegalovirus (MCMV). Mechanistically, endogenous glucocorticoids produced shortly after infection induced selective and tissue-specific expression of the checkpoint receptor PD-1 on NK cells. This glucocorticoid-PD-1 pathway limited production of the cytokine IFN-γ by spleen NK cells, which prevented immunopathology. Notably, this regulation did not compromise viral clearance. Thus, the fine tuning of NK cell functions by the HPA axis preserved tissue integrity without impairing pathogen elimination, which reveals a novel aspect of neuroimmune regulation.


Subject(s)
Glucocorticoids/metabolism , Herpesviridae Infections/immunology , Killer Cells, Natural/physiology , Muromegalovirus/physiology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cells, Cultured , Female , Hypothalamo-Hypophyseal System , Immunity, Innate , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Organ Specificity , Pituitary-Adrenal System , Receptors, Glucocorticoid/genetics , Signal Transduction , Viral Load
4.
Nat Immunol ; 17(2): 179-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595889

ABSTRACT

Intestinal T cells and group 3 innate lymphoid cells (ILC3 cells) control the composition of the microbiota and gut immune responses. Within the gut, ILC3 subsets coexist that either express or lack the natural cytoxicity receptor (NCR) NKp46. We identified here the transcriptional signature associated with the transcription factor T-bet-dependent differentiation of NCR(-) ILC3 cells into NCR(+) ILC3 cells. Contrary to the prevailing view, we found by conditional deletion of the key ILC3 genes Stat3, Il22, Tbx21 and Mcl1 that NCR(+) ILC3 cells were redundant for the control of mouse colonic infection with Citrobacter rodentium in the presence of T cells. However, NCR(+) ILC3 cells were essential for cecal homeostasis. Our data show that interplay between intestinal ILC3 cells and adaptive lymphocytes results in robust complementary failsafe mechanisms that ensure gut homeostasis.


Subject(s)
Immunity, Innate , Interleukins/biosynthesis , Lymphocytes/immunology , Lymphocytes/metabolism , Animals , Citrobacter rodentium/immunology , Cluster Analysis , Disease Models, Animal , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/mortality , Enterobacteriaceae Infections/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Homeostasis , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/deficiency , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Signal Transduction , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcriptome , Interleukin-22
5.
Immunity ; 49(5): 971-986.e5, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413361

ABSTRACT

Natural killer (NK) cells are innate lymphoid cells (ILCs) involved in antimicrobial and antitumoral responses. Several NK cell subsets have been reported in humans and mice, but their heterogeneity across organs and species remains poorly characterized. We assessed the diversity of human and mouse NK cells by single-cell RNA sequencing on thousands of individual cells isolated from spleen and blood. Unbiased transcriptional clustering revealed two distinct signatures differentiating between splenic and blood NK cells. This analysis at single-cell resolution identified three subpopulations in mouse spleen and four in human spleen, and two subsets each in mouse and human blood. A comparison of transcriptomic profiles within and between species highlighted the similarity of the two major subsets, NK1 and NK2, across organs and species. This unbiased approach provides insight into the biology of NK cells and establishes a rationale for the translation of mouse studies to human physiology and disease.


Subject(s)
Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Transcriptome , Animals , Biomarkers , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunity, Innate , Immunophenotyping , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Mice , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , Single-Cell Analysis
6.
Nature ; 594(7861): 94-99, 2021 06.
Article in English | MEDLINE | ID: mdl-34012116

ABSTRACT

Inflammation is a defence response to tissue damage that requires tight regulation in order to prevent impaired healing. Tissue-resident macrophages have a key role in tissue repair1, but the precise molecular mechanisms that regulate the balance between inflammatory and pro-repair macrophage responses during healing remain poorly understood. Here we demonstrate a major role for sensory neurons in promoting the tissue-repair function of macrophages. In a sunburn-like model of skin damage in mice, the conditional ablation of sensory neurons expressing the Gαi-interacting protein (GINIP) results in defective tissue regeneration and in dermal fibrosis. Elucidation of the underlying molecular mechanisms revealed a crucial role for the neuropeptide TAFA4, which is produced in the skin by C-low threshold mechanoreceptors-a subset of GINIP+ neurons. TAFA4 modulates the inflammatory profile of macrophages directly in vitro. In vivo studies in Tafa4-deficient mice revealed that TAFA4 promotes the production of IL-10 by dermal macrophages after UV-induced skin damage. This TAFA4-IL-10 axis also ensures the survival and maintenance of IL-10+TIM4+ dermal macrophages, reducing skin inflammation and promoting tissue regeneration. These results reveal a neuroimmune regulatory pathway driven by the neuropeptide TAFA4 that promotes the anti-inflammatory functions of macrophages and prevents fibrosis after tissue damage, and could lead to new therapeutic perspectives for inflammatory diseases.


Subject(s)
Cytokines/metabolism , Macrophages/metabolism , Regeneration , Sensory Receptor Cells/metabolism , Wound Healing , Animals , Cell Survival , Cytokines/deficiency , Disease Models, Animal , Female , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Macrophages/radiation effects , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/radiation effects , Skin/pathology , Skin/radiation effects , Sunburn/complications , Sunburn/etiology , Sunburn/metabolism , Sunburn/pathology , Ultraviolet Rays/adverse effects
7.
Immunity ; 46(3): 340-342, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329698

ABSTRACT

Brain ischemia induces profound systemic immunosuppression, leading to infectious complications. In this issue of Immunity, Liu et al. (2017) demonstrate that distinct neuroendocrine pathways differentially inhibit natural killer (NK) cell responses in the central nervous system and the periphery after cerebral infarction.


Subject(s)
Brain , Killer Cells, Natural/immunology , Humans
8.
Brain Behav Immun ; 119: 750-766, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710336

ABSTRACT

Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.


Subject(s)
Chronic Pain , Dysbiosis , Ganglia, Spinal , Gastrointestinal Microbiome , Mice, Knockout , Myosin Type I , Animals , Female , Male , Mice , Chronic Pain/metabolism , Chronic Pain/microbiology , Dysbiosis/metabolism , Ganglia, Spinal/metabolism , Gastrointestinal Microbiome/physiology , Macrophages/metabolism , Mice, Inbred C57BL , Myosin Type I/metabolism
9.
J Allergy Clin Immunol ; 147(1): 349-360, 2021 01.
Article in English | MEDLINE | ID: mdl-32417134

ABSTRACT

BACKGROUND: Programmed cell death protein 1 (PD-1)-immune checkpoint blockade has provided significant clinical efficacy across various types of cancer by unleashing both T and natural killer (NK) cell-mediated antitumor responses. However, resistance to immunotherapy occurs for many patients, rendering the identification of the mechanisms that control PD-1 expression extremely important to increase the response to the therapy. OBJECTIVE: We sought to identify the stimuli and the molecular mechanisms that induce the de novo PD-1 expression on human NK cells in the tumor setting. METHODS: NK cells freshly isolated from peripheral blood of healthy donors were stimulated with different combinations of molecules, and PD-1 expression was studied at the mRNA and protein levels. Moreover, ex vivo analysis of tumor microenvironment and NK cell phenotype was performed. RESULTS: Glucocorticoids are indispensable for PD-1 induction on human NK cells, in cooperation with a combination of cytokines that are abundant at the tumor site. Mechanistically, glucocorticoids together with IL-12, IL-15, and IL-18 not only upregulate PDCD1 transcription, but also activate a previously unrecognized transcriptional program leading to enhanced mRNA translation and resulting in an increased PD-1 amount in NK cells. CONCLUSIONS: These results provide evidence of a novel immune suppressive mechanism of glucocorticoids involving the transcriptional and translational control of an important immune checkpoint.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Glucocorticoids/immunology , Interleukin-15/immunology , Interleukin-18/immunology , Interleukin-2/immunology , Killer Cells, Natural/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology , A549 Cells , Humans , K562 Cells
10.
Immunol Rev ; 286(1): 120-136, 2018 11.
Article in English | MEDLINE | ID: mdl-30294960

ABSTRACT

The activities of the immune system in repairing tissue injury and combating pathogens were long thought to be independent of the nervous system. However, a major regulatory role of immunomodulatory molecules released locally or systemically by the neuroendocrine system has recently emerged. A number of observations and discoveries support indeed the notion of the nervous system as an immunoregulatory system involved in immune responses. Innate lymphoid cells (ILCs), including natural killer (NK) cells and tissue-resident ILCs, form a family of effector cells present in organs and mucosal barriers. ILCs are involved in the maintenance of tissue integrity and homeostasis. They can also secrete effector cytokines rapidly, and this ability enables them to play early roles in the immune response. ILCs are activated by multiple pathways including epithelial and myeloid cell-derived cytokines. Their functions are also regulated by mediators produced by the nervous system. In particular, the peripheral nervous system, through neurotransmitters and neuropeptides, works in parallel with the hypothalamic-pituitary-adrenal and gonadal axis to modulate inflammatory events and maintain homeostasis. We summarize here recent findings concerning the regulation of ILC activities by neuroendocrine mediators in homeostatic and inflammatory conditions.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Neurotransmitter Agents/immunology , Pituitary-Adrenal System/physiology , Animals , Homeostasis , Humans , Immunity, Innate , Lymphocytes , Neuroimmunomodulation , Neurosecretory Systems
12.
Nat Immunol ; 9(5): 503-10, 2008 May.
Article in English | MEDLINE | ID: mdl-18425107

ABSTRACT

Natural killer (NK) cells are effector lymphocytes of the innate immune system that control several types of tumors and microbial infections by limiting their spread and subsequent tissue damage. Recent research highlights the fact that NK cells are also regulatory cells engaged in reciprocal interactions with dendritic cells, macrophages, T cells and endothelial cells. NK cells can thus limit or exacerbate immune responses. Although NK cells might appear to be redundant in several conditions of immune challenge in humans, NK cell manipulation seems to hold promise in efforts to improve hematopoietic and solid organ transplantation, promote antitumor immunotherapy and control inflammatory and autoimmune disorders.


Subject(s)
Killer Cells, Natural/immunology , Animals , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Host vs Graft Reaction/immunology , Humans , Immune Tolerance , Immunotherapy, Adoptive , Inflammation/immunology , Macrophages/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Virus Diseases/immunology
13.
Circ Res ; 122(1): 47-57, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29046274

ABSTRACT

RATIONALE: Chronic inflammation is central in the development of atherosclerosis. Both innate and adaptive immunities are involved. Although several studies have evaluated the functions of natural killer (NK) cells in experimental animal models of atherosclerosis, it is not yet clear whether NK cells behave as protective or proatherogenic effectors. One of the main caveats of previous studies was the lack of specificity in targeting loss or gain of function of NK cells. OBJECTIVES: We used 2 selective genetic approaches to investigate the role of NK cells in atherosclerosis: (1) Ncr1iCre/+R26lsl-DTA/+ mice in which NK cells were depleted and (2) Noé mice in which NK cells are hyperresponsive. METHODS AND RESULTS: No difference in atherosclerotic lesion size was found in Ldlr-/- (low-density lipoprotein receptor null) mice transplanted with bone marrow (BM) cells from Ncr1iCreR26Rlsl-DTA , Noé, or wild-type mice. Also, no difference was observed in plaque composition in terms of collagen content, macrophage infiltration, or the immune profile, although Noé chimera had more IFN (interferon)-γ-producing NK cells, compared with wild-type mice. Then, we investigated the NK-cell selectivity of anti-asialoganglioside M1 antiserum, which was previously used to conclude the proatherogenicity of NK cells. Anti-asialoganglioside M1 treatment decreased atherosclerosis in both Ldlr-/- mice transplanted with Ncr1iCreR26Rlsl-DTA or wild-type bone marrow, indicating that its antiatherogenic effects are unrelated to NK-cell depletion, but to CD8+ T and NKT cells. Finally, to determine whether NK cells could contribute to the disease in conditions of pathological NK-cell overactivation, we treated irradiated Ldlr-/- mice reconstituted with either wild-type or Ncr1iCreR26Rlsl-DTA bone marrow with the viral mimic polyinosinic:polycytidylic acid and found a significant reduction of plaque size in NK-cell-deficient chimeric mice. CONCLUSIONS: Our findings, using state-of-the-art mouse models, demonstrate that NK cells have no direct effect on the natural development of hypercholesterolemia-induced atherosclerosis, but may play a role when an additional systemic NK-cell overactivation occurs.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/metabolism , Gene Deletion , Killer Cells, Natural/metabolism , Animals , Atherosclerosis/immunology , Cells, Cultured , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
EMBO J ; 33(12): 1295-303, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24674969

ABSTRACT

Immune memory has traditionally been the domain of the adaptive immune system, present only in antigen-specific T and B cells. The purpose of this review is to summarize the evidence for immunological memory in lower organisms (which are not thought to possess adaptive immunity) and within specific cell subsets of the innate immune system. A special focus will be given to recent findings in both mouse and humans for specificity and memory in natural killer (NK) cells, which have resided under the umbrella of innate immunity for decades. The surprising longevity and enhanced responses of previously primed NK cells will be discussed in the context of several immunization settings.


Subject(s)
Biological Evolution , Immunity, Innate/immunology , Immunologic Memory/immunology , Invertebrates/immunology , Killer Cells, Natural/immunology , Models, Neurological , Animals , Humans , Mice , Species Specificity
15.
Blood ; 123(5): 678-86, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24326534

ABSTRACT

Natural killer (NK) cells mediate antilymphoma activity by spontaneous cytotoxicity and antibody-dependent cell-mediated cytotoxicity (ADCC) when triggered by rituximab, an anti-CD20 monoclonal antibody (mAb) used to treat patients with B-cell lymphomas. The balance of inhibitory and activating signals determines the magnitude of the efficacy of NK cells by spontaneous cytotoxicity. Here, using a killer-cell immunoglobulin-like receptor (KIR) transgenic murine model, we show that blockade of the interface of inhibitory KIRs with major histocompatibility complex (MHC) class I antigens on lymphoma cells by anti-KIR antibodies prevents a tolerogenic interaction and augments NK-cell spontaneous cytotoxicity. In combination with anti-CD20 mAbs, anti-KIR treatment induces enhanced NK-cell-mediated, rituximab-dependent cytotoxicity against lymphoma in vitro and in vivo in KIR transgenic and syngeneic murine lymphoma models. These results support a therapeutic strategy of combination rituximab and KIR blockade through lirilumab, illustrating the potential efficacy of combining a tumor-targeting therapy with an NK-cell agonist, thus stimulating the postrituximab antilymphoma immune response.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Killer Cells, Natural/immunology , Lymphoma/therapy , Receptors, KIR/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line , Female , Histocompatibility Antigens Class I/immunology , Humans , Lymphoma/immunology , Male , Mice , Rituximab
16.
Blood ; 122(3): 394-404, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23687088

ABSTRACT

B7-H6, a member of the B7 family of immunoreceptors, is as a cell-surface ligand for the NKp30-activating receptor expressed on natural killer cells. B7-H6 is not detected in normal human tissues at steady state but is expressed on tumor cells. However, whether B7-H6 can be expressed in other conditions remains unknown. We analyzed here the pathways that lead to the expression of B7-H6 in nontransformed cells. In vitro, B7-H6 was induced at the surface of CD14(+)CD16(+) proinflammatory monocytes and neutrophils upon stimulation by ligands of Toll-like receptors or proinflammatory cytokines such as interleukin-1ß and tumor necrosis factor α. In these conditions, a soluble form of B7-H6 (sB7-H6) was also produced by activated monocytes and neutrophils. In vivo, B7-H6 was expressed on circulating proinflammatory CD14(+)CD16(+) monocytes in a group of patients in sepsis conditions, and was linked to an increased mortality. sB7-H6 was selectively detected in the sera of patients with gram-negative sepsis and was associated with membrane vesicles that co-sedimented with the exosomal fraction. These findings reveal that B7-H6 is not only implicated in tumor immunosurveillance but also participates in the inflammatory response in infectious conditions.


Subject(s)
B7 Antigens/metabolism , Inflammation/immunology , Natural Cytotoxicity Triggering Receptor 3/metabolism , Antibodies, Monoclonal/metabolism , B7 Antigens/blood , B7 Antigens/genetics , B7 Antigens/immunology , Cell Membrane/metabolism , Cells, Cultured , Humans , Inflammation/pathology , Ligands , Monocytes/metabolism , Neutrophils/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sepsis/immunology , Sepsis/pathology , Solubility
17.
Immunol Cell Biol ; 92(3): 221-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366519

ABSTRACT

Natural killer (NK) cells are innate lymphoid cells (ILCs) that participate to the clearance of pathogen-infected cells and tumour cells. NK cells and subsets of ILCs express the natural cytotoxicity receptors (NCRs) NKp46, NKp44 and NKp30 at their surface. NCRs have been shown to recognize a broad spectrum of ligands ranging from viral-, parasite- and bacterial-derived ligands to cellular ligands; however, the full identification of NCR ligands remains to be performed and will undoubtedly contribute to a better understanding of NK cell and ILC biology.


Subject(s)
Ligands , Receptors, Natural Cytotoxicity Triggering/metabolism , Animals , Bacteria/metabolism , Humans , Models, Immunological , Parasites/metabolism , Viruses/metabolism
18.
Proc Natl Acad Sci U S A ; 108(45): 18324-9, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22021440

ABSTRACT

NKp46 is a cell surface receptor expressed on natural killer (NK) cells, on a minute subset of T cells, and on a population of innate lymphoid cells that produce IL-22 and express the transcription factor retinoid-related orphan receptor (ROR)-γt, referred to as NK cell receptor (NKR)(+)ROR-γt(+) cells. Here we describe Nkp46(iCre) knock-in mice in which the gene encoding the improved Cre (iCre) recombinase was inserted into the Nkp46 locus. This mouse was used to noninvasively trace cells expressing NKp46 in vivo. Fate mapping experiments demonstrated the stable expression of NKp46 on NK cells and allowed a reappraisal of the sequential steps of NK cell maturation. NKp46 genetic tracing also showed that gut NKR(+)ROR-γt(+) and NK cells represent two distinct lineages. In addition, the genetic heterogeneity of liver NK cells was evidenced. Finally, Nkp46(iCre) mice also represent a unique mouse model of conditional mutagenesis specifically in NKp46(+) cells, paving the way for further developments in the biology of NKp46(+) NK, T, and NKR(+)ROR-γt(+) cells.


Subject(s)
Antigens, Ly/metabolism , Lymphoid Tissue/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, Ly/genetics , Cell Differentiation , Cell Lineage , Intestines/cytology , Liver/cytology , Lymphoid Tissue/cytology , Mice , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics
19.
J Exp Med ; 204(4): 853-63, 2007 Apr 16.
Article in English | MEDLINE | ID: mdl-17420270

ABSTRACT

Mouse cytomegalovirus (MCMV) susceptibility often results from defects of natural killer (NK) cell function. Here we describe Jinx, an N-ethyl-N-nitrosourea-induced MCMV susceptibility mutation that permits unchecked proliferation of the virus, causing death. In Jinx homozygotes, activated NK cells and cytotoxic T lymphocytes (CTLs) fail to degranulate, although they retain the ability to produce cytokines, and cytokine levels are markedly elevated in the blood of infected mutant mice. Jinx was mapped to mouse chromosome 11 on a total of 246 meioses and confined to a 4.60-million basepair critical region encompassing 122 annotated genes. The phenotype was ascribed to the creation of a novel donor splice site in Unc13d, the mouse orthologue of human MUNC13-4, in which mutations cause type 3 familial hemophagocytic lymphohistiocytosis (FHL3), a fatal disease marked by massive hepatosplenomegaly, anemia, and thrombocytopenia. Jinx mice do not spontaneously develop clinical features of hemophagocytic lymphohistiocytosis (HLH), but do so when infected with lymphocytic choriomeningitis virus, exhibiting hyperactivation of CTLs and antigen-presenting cells, and inadequate restriction of viral proliferation. In contrast, neither Listeria monocytogenes nor MCMV induces the syndrome. In mice, the HLH phenotype is conditional, which suggests the existence of a specific infectious trigger of FHL3 in humans.


Subject(s)
Disease Models, Animal , Genetic Predisposition to Disease , Herpesviridae Infections/metabolism , Lymphohistiocytosis, Hemophagocytic/metabolism , Lymphohistiocytosis, Hemophagocytic/pathology , Membrane Proteins/metabolism , Muromegalovirus/physiology , Animals , Antigen-Presenting Cells/metabolism , Apoptosis , Cloning, Molecular , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Interferon-gamma/biosynthesis , Lymphohistiocytosis, Hemophagocytic/classification , Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Proteins/genetics , Mice , Mutation/genetics , Phenotype
20.
Blood ; 117(10): 2874-82, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21239699

ABSTRACT

Natural killer (NK) cells are innate immune cells that express members of the leukocyte ß2 integrin family in humans and mice. These CD11/CD18 heterodimers play critical roles in leukocyte trafficking, immune synapse formation, and costimulation. The cell-surface expression of one of these integrins, CD11b/CD18, is also recognized as a major marker of mouse NK-cell maturation, but its function on NK cells has been largely ignored. Using N-ethyl-N-nitrosourea (ENU) mutagenesis, we generated a mouse carrying an A → T transverse mutation in the Itgb2 gene, resulting in a mutation that prevented the cell-surface expression of CD18 and its associated CD11a, CD11b, and CD11c proteins. We show that ß2 integrin-deficient NK cells have a hyporesponsive phenotype in vitro, and present an alteration of their in vivo developmental program characterized by a selective accumulation of c-kit(+) cells. NK-cell missing-self recognition was partially altered in vivo, whereas the early immune response to mouse cytomegalovirus (MCMV) infection occurred normally in CD18-deficient mice. Therefore, ß2 integrins are required for optimal NK-cell maturation, but this deficiency is partial and can be bypassed during MCMV infection, highlighting the robustness of antiviral protective responses.


Subject(s)
CD18 Antigens/immunology , CD18 Antigens/metabolism , Cell Differentiation/immunology , Killer Cells, Natural/immunology , Animals , Cell Separation , Flow Cytometry , Herpesviridae Infections/immunology , Killer Cells, Natural/cytology , Mice , Muromegalovirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL