Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 18(10): 1117-1127, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28805812

ABSTRACT

The transcription factor T-bet has been associated with increased susceptibility to systemic and organ-specific autoimmunity, but the mechanism by which T-bet expression promotes neuroinflammation remains unknown. In this study, we demonstrate a cardinal role of T-bet-dependent NKp46+ innate lymphoid cells (ILCs) in the initiation of CD4+ TH17-mediated neuroinflammation. Loss of T-bet specifically in NKp46+ ILCs profoundly impaired the ability of myelin-reactive TH17 cells to invade central nervous system (CNS) tissue and protected the mice from autoimmunity. T-bet-dependent NKp46+ ILCs localized in the meninges and acted as chief coordinators of meningeal inflammation by inducing the expression of proinflammatory cytokines, chemokines and matrix metalloproteinases, which together facilitated T cell entry into CNS parenchyma. Our findings uncover a detrimental role of T-bet-dependent NKp46+ ILCs in the development of CNS autoimmune disease.


Subject(s)
Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Cell Movement/genetics , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression , Immunophenotyping , Mice , Mice, Knockout , Myelin Sheath/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , T-Box Domain Proteins , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Nat Immunol ; 17(2): 169-78, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595886

ABSTRACT

The transcription factor GATA-3 is indispensable for the development of all innate lymphoid cells (ILCs) that express the interleukin 7 receptor α-chain (IL-7Rα). However, the function of low GATA-3 expression in committed group 3 ILCs (ILC3 cells) has not been identified. We found that GATA-3 regulated the homeostasis of ILC3 cells by controlling IL-7Rα expression. In addition, GATA-3 served a critical function in the development of the NKp46(+) ILC3 subset by regulating the balance between the transcription factors T-bet and RORγt. Among NKp46(+) ILC3 cells, although GATA-3 positively regulated genes specific to the NKp46(+) ILC3 subset, it negatively regulated genes specific to lymphoid tissue-inducer (LTi) or LTi-like ILC3 cells. Furthermore, GATA-3 was required for IL-22 production in both ILC3 subsets. Thus, despite its low expression, GATA-3 was critical for the homeostasis, development and function of ILC3 subsets.


Subject(s)
Cell Differentiation , GATA3 Transcription Factor/metabolism , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cluster Analysis , GATA3 Transcription Factor/deficiency , GATA3 Transcription Factor/genetics , Gene Expression Profiling , Gene Expression Regulation , Homeostasis , Immunity, Innate/genetics , Immunophenotyping , Interleukins/biosynthesis , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Protein Binding , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , T-Box Domain Proteins/metabolism , Interleukin-22
3.
Nat Immunol ; 16(4): 376-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25729921

ABSTRACT

An important cause of obesity-induced insulin resistance is chronic systemic inflammation originating in visceral adipose tissue (VAT). VAT inflammation is associated with the accumulation of proinflammatory macrophages in adipose tissue, but the immunological signals that trigger their accumulation remain unknown. We found that a phenotypically distinct population of tissue-resident natural killer (NK) cells represented a crucial link between obesity-induced adipose stress and VAT inflammation. Obesity drove the upregulation of ligands of the NK cell-activating receptor NCR1 on adipocytes; this stimulated NK cell proliferation and interferon-γ (IFN-γ) production, which in turn triggered the differentiation of proinflammatory macrophages and promoted insulin resistance. Deficiency of NK cells, NCR1 or IFN-γ prevented the accumulation of proinflammatory macrophages in VAT and greatly ameliorated insulin sensitivity. Thus NK cells are key regulators of macrophage polarization and insulin resistance in response to obesity-induced adipocyte stress.


Subject(s)
Adipocytes/immunology , Insulin Resistance/immunology , Intra-Abdominal Fat/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Obesity/immunology , Adipocytes/pathology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Cell Differentiation , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Insulin/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Intra-Abdominal Fat/pathology , Killer Cells, Natural/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Obesity/genetics , Obesity/pathology , Signal Transduction
4.
Dig Dis Sci ; 69(9): 3276-3289, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39020183

ABSTRACT

OBJECTIVE: NK cells play a vital role in tumor immune resistance. Various factors affect NK cell activity. While NK cell dysfunction has been observed in numerous malignancies, the underlying mechanisms in gastric cancer remain unclear. METHOD: Flow cytometry was used to identify the phenotypic distribution and expression of activated receptors on NK cells. ELISA was used to determine the expression of cytokines. We examined the expression of NK cell-related genes and explored their association with survival and prognosis. Additionally, we conducted PCR detection of miR-552-5p expression levels in plasma exosomes of patients and investigated its correlation with phenotypic distribution and activated receptors. We used flow cytometry and ELISA to verify the role of miR-552-5p in NK cell dysfunction. Furthermore, we investigated the potential role of PD-1/PD-L1 in regulating NK cell dysfunction in patients' cells. RESULTS: We observed a significant decrease in the percentage of NKG2D and NKp30 and IFN-γ and TNF-α in patients than in healthy volunteers. Patients with low levels of CD56, CD16, NKG2D, and NKP46 exhibited poorer survival prognoses. Moreover, increased expression levels of plasma exosomal miR-552-5p in patients were negatively associated with NK cell phenotypic distribution and activated receptor expression. MiR-552-5p downregulated the secretion of perforin, granzyme, and IFN-γ as well as the expression of NKp30, NKp46, and NKG2D. Additionally, it suppressed the cytotoxicity of NK cells. The inhibitory effect of miR-552-5p, on NK cell function was reversed when anti-PD-L1 antibodies were used. CONCLUSION: Exosomal miR-552-5p targets the PD-1/PD-L1 axis, leading to impaired NK cell function.


Subject(s)
B7-H1 Antigen , Exosomes , Killer Cells, Natural , MicroRNAs , Programmed Cell Death 1 Receptor , Stomach Neoplasms , Aged , Female , Humans , Male , Middle Aged , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Case-Control Studies , CD56 Antigen/metabolism , Exosomes/metabolism , Exosomes/genetics , Exosomes/immunology , Gene Expression Regulation, Neoplastic , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 3/genetics , Natural Cytotoxicity Triggering Receptor 3/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Receptors, IgG/genetics , Receptors, IgG/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649222

ABSTRACT

Natural killer (NK) cells are innate effectors armed with cytotoxic and cytokine-secreting capacities whose spontaneous antitumor activity is key to numerous immunotherapeutic strategies. However, current mouse models fail to mirror the extensive immune system variation that exists in the human population which may impact on NK cell-based therapies. We performed a comprehensive profiling of NK cells in the Collaborative Cross (CC), a collection of novel recombinant inbred mouse strains whose genetic diversity matches that of humans, thereby providing a unique and highly diverse small animal model for the study of immune variation. We demonstrate that NK cells from CC strains displayed a breadth of phenotypic and functional variation reminiscent of that reported for humans with regards to cell numbers, key marker expression, and functional capacities. We took advantage of the vast genetic diversity of the CC and identified nine genomic loci through quantitative trait locus mapping driving these phenotypic variations. SNP haplotype patterns and variant effect analyses identified candidate genes associated with lung NK cell numbers, frequencies of CD94+ NK cells, and expression levels of NKp46. Thus, we demonstrate that the CC represents an outstanding resource to study NK cell diversity and its regulation by host genetics.


Subject(s)
Antigens, Ly , Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily D , Natural Cytotoxicity Triggering Receptor 1 , Polymorphism, Single Nucleotide , Quantitative Trait Loci/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Crosses, Genetic , Mice , Mice, Inbred Strains , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology
6.
Nat Immunol ; 12(4): 320-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21336274

ABSTRACT

Lymphoid cells that express the nuclear hormone receptor RORγt are involved in containment of the large intestinal microbiota and defense against pathogens through the production of interleukin 17 (IL-17) and IL-22. They include adaptive IL-17-producing helper T cells (T(H)17 cells), as well as innate lymphoid cells (ILCs) such as lymphoid tissue-inducer (LTi) cells and IL-22-producing NKp46+ cells. Here we show that in contrast to T(H)17 cells, both types of RORγt+ ILCs constitutively produced most of the intestinal IL-22 and that the symbiotic microbiota repressed this function through epithelial expression of IL-25. This function was greater in the absence of adaptive immunity and was fully restored and required after epithelial damage, which demonstrates a central role for RORγt+ ILCs in intestinal homeostasis. Our data identify a finely tuned equilibrium among intestinal symbionts, adaptive immunity and RORγt+ ILCs.


Subject(s)
Intestines/immunology , Lymphoid Tissue/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Signal Transduction/immunology , Adaptive Immunity/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Female , Flow Cytometry , Homeostasis/immunology , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Symbiosis/immunology , Time Factors , Interleukin-22
7.
J Immunol ; 206(3): 465-470, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33443057

ABSTRACT

Heterogeneity among naive adaptive lymphocytes determines their individual functions and fate decisions during an immune response. NK cells are innate lymphocytes capable of generating "adaptive" responses during infectious challenges. However, the factors that govern various NK cell functions are not fully understood. In this study, we use a reporter mouse model to permanently "time stamp" NK cells and type 1 innate lymphoid cells (ILC1s) to characterize the dynamics of their homeostatic turnover. We found that the homeostatic turnover of tissue-resident ILC1s is much slower than that of circulating NK cells. NK cell homeostatic turnover is further accelerated without the transcription factor Eomes. Finally, heterogeneity in NK cell age diversifies NK cell function, with "older" NK cells exhibiting more potent IFN-γ production to activating stimuli and more robust adaptive responses during CMV infection. These results provide insight into how the functional response of an NK cell varies over its lifespan.


Subject(s)
Antigens, Ly/metabolism , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Animals , Antigens, Ly/genetics , Cell Differentiation , Cell Self Renewal , Cellular Senescence/immunology , Cytokines/metabolism , Homeostasis , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/immunology
8.
PLoS Pathog ; 16(2): e1008279, 2020 02.
Article in English | MEDLINE | ID: mdl-32023327

ABSTRACT

IFN-γ is an enigmatic cytokine that shows direct anti-viral effects, confers upregulation of MHC-II and other components relevant for antigen presentation, and that adjusts the composition and balance of complex cytokine responses. It is produced during immune responses by innate as well as adaptive immune cells and can critically affect the course and outcome of infectious diseases, autoimmunity, and cancer. To selectively analyze the function of innate immune cell-derived IFN-γ, we generated conditional IFN-γOFF mice, in which endogenous IFN-γ expression is disrupted by a loxP flanked gene trap cassette inserted into the first intron of the IFN-γ gene. IFN-γOFF mice were intercrossed with Ncr1-Cre or CD4-Cre mice that express Cre mainly in NK cells (IFN-γNcr1-ON mice) or T cells (IFN-γCD4-ON mice), respectively. Rosa26RFP reporter mice intercrossed with Ncr1-Cre mice showed selective RFP expression in more than 80% of the NK cells, while upon intercrossing with CD4-Cre mice abundant RFP expression was detected in T cells, but also to a minor extent in other immune cell subsets. Previous studies showed that IFN-γ expression is needed to promote survival of vaccinia virus (VACV) infection. Interestingly, during VACV infection of wild type and IFN-γCD4-ON mice two waves of serum IFN-γ were induced that peaked on day 1 and day 3/4 after infection. Similarly, VACV infected IFN-γNcr1-ON mice mounted two waves of IFN-γ responses, of which the first one was moderately and the second one profoundly reduced when compared with WT mice. Furthermore, IFN-γNcr1-ON as well as IFN-γCD4-ON mice survived VACV infection, whereas IFN-γOFF mice did not. As expected, ex vivo analysis of splenocytes derived from VACV infected IFN-γNcr1-ON mice showed IFN-γ expression in NK cells, but not T cells, whereas IFN-γOFF mice showed IFN-γ expression neither in NK cells nor T cells. VACV infected IFN-γNcr1-ON mice mounted normal cytokine responses, restored neutrophil accumulation, and showed normal myeloid cell distribution in blood and spleen. Additionally, in these mice normal MHC-II expression was detected on peripheral macrophages, whereas IFN-γOFF mice did not show MHC-II expression on such cells. In conclusion, upon VACV infection Ncr1 positive cells including NK cells mount two waves of early IFN-γ responses that are sufficient to promote the induction of protective anti-viral immunity.


Subject(s)
Antigens, Ly/immunology , Gene Expression Regulation/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , Antigens, Ly/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Interferon-gamma/genetics , Killer Cells, Natural/pathology , Mice , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vaccinia/genetics , Vaccinia/pathology , Vaccinia virus/genetics
9.
Nat Immunol ; 11(2): 121-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20023661

ABSTRACT

The mechanism of action of natural killer (NK) cells in type 1 diabetes is still unknown. Here we show that the activating receptor NKp46 recognizes mouse and human ligands on pancreatic beta cells. NK cells appeared in the pancreas when insulitis progressed to type 1 diabetes, and NKp46 engagement by beta cells led to degranulation of NK cells. NKp46-deficient mice had less development of type 1 diabetes induced by injection of a low dose of streptozotocin. Injection of soluble NKp46 proteins into nonobese diabetic mice during the early phase of insulitis and the prediabetic stage prevented the development of type 1 diabetes. Our findings demonstrate that NKp46 is essential for the development of type 1 diabetes and highlight potential new therapeutic modalities for this disease.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Antigens, Ly/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Degranulation/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred NOD , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism
10.
Immunity ; 38(2): 322-35, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23352232

ABSTRACT

Dendritic cells (DCs) are crucial for mounting allergic airway inflammation, but it is unclear which subset of DCs performs this task. By using CD64 and MAR-1 staining, we reliably separated CD11b(+) monocyte-derived DCs (moDCs) from conventional DCs (cDCs) and studied antigen uptake, migration, and presentation assays of lung and lymph node (LN) DCs in response to inhaled house dust mite (HDM). Mainly CD11b(+) cDCs but not CD103(+) cDCs induced T helper 2 (Th2) cell immunity in HDM-specific T cells in vitro and asthma in vivo. Studies in Flt3l(-/-) mice, lacking all cDCs, revealed that moDCs were also sufficient to induce Th2 cell-mediated immunity but only when high-dose HDM was given. The main function of moDCs was the production of proinflammatory chemokines and allergen presentation in the lung during challenge. Thus, we have identified migratory CD11b(+) cDCs as the principal subset inducing Th2 cell-mediated immunity in the LN, whereas moDCs orchestrate allergic inflammation in the lung.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Asthma/immunology , Dendritic Cells/immunology , Immunity, Cellular , Pyroglyphidae/immunology , Th2 Cells/immunology , Administration, Inhalation , Adoptive Transfer , Allergens/isolation & purification , Animals , Antigens, Dermatophagoides/administration & dosage , Antigens, Dermatophagoides/isolation & purification , Antigens, Ly/genetics , Antigens, Ly/immunology , Asthma/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Movement , Cell Proliferation , Dendritic Cells/transplantation , Gene Expression , Inflammation/immunology , Inflammation/pathology , Lung/immunology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/transplantation , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Organ Specificity , Receptors, IgG/genetics , Receptors, IgG/immunology
11.
Eur J Immunol ; 50(5): 656-665, 2020 05.
Article in English | MEDLINE | ID: mdl-32027754

ABSTRACT

Natural Killer (NK) cell responses are regulated by a variety of different surface receptors. While we can determine the overall positive or negative effect of a given receptor on NK cell functions, investigating NK cell regulation in a quantitative way is challenging. To quantitatively investigate individual receptors for their effect on NK cell activation, we chose to functionalize latex beads that have approximately the same size as lymphocytes with defined amounts of specific antibodies directed against distinct activating receptors. This enabled us to investigate NK cell reactivity in a defined, clean, and controllable system. Only CD16 and NKp30 could activate the degranulation of resting human NK cells. CD16, NKG2D, NKp30, NKp44, and NKp46 were able to activate cultured NK cells. NK cell activation resulted in the induction of polyfunctional cells that degranulated and produced IFN-γ and MIP-1ß. Interestingly, polyfunctional NK cells were only induced by triggering ITAM-coupled receptors. NKp44 showed a very sensitive response pattern, where a small increase in receptor stimulation caused maximal NK cell activity. In contrast, stimulation of 2B4 induced very little NK cell degranulation, while providing sufficient signal for NK cell adhesion. Our data demonstrate that activating receptors differ in their effectiveness to stimulate NK cells.


Subject(s)
Antibodies/pharmacology , Gene Expression Regulation/drug effects , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Antibodies/chemistry , Cell Adhesion/drug effects , Cell Degranulation/drug effects , Cells, Cultured , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Microspheres , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Natural Cytotoxicity Triggering Receptor 2/genetics , Natural Cytotoxicity Triggering Receptor 2/immunology , Natural Cytotoxicity Triggering Receptor 3/genetics , Natural Cytotoxicity Triggering Receptor 3/immunology , Protein Binding , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction
12.
PLoS Pathog ; 15(6): e1007797, 2019 06.
Article in English | MEDLINE | ID: mdl-31220194

ABSTRACT

During viral infection, tight regulation of CD8+ T-cell functions determines the outcome of the disease. Recently, others and we determined that the natural killer (NK) cells kill hyperproliferative CD8+ T cells in the context of viral infection, but molecules that are involved in shaping the regulatory capability of NK cells remain virtually unknown. Here we used mice lacking the Fc-receptor common gamma chain (FcRγ, FcεRIγ, Fcer1g-/- mice) to determine the role of Fc-receptor and NK-receptor signaling in the process of CD8+ T-cell regulation. We found that the lack of FcRγ on NK cells limits their ability to restrain virus-specific CD8+ T cells and that the lack of FcRγ in Fcer1g-/- mice leads to enhanced CD8+ T-cell responses and rapid control of the chronic docile strain of the lymphocytic choriomeningitis virus (LCMV). Mechanistically, FcRγ stabilized the expression of NKp46 but not that of other killer cell-activating receptors on NK cells. Although FcRγ did not influence the development or activation of NK cell during LCMV infection, it specifically limited their ability to modulate CD8+ T-cell functions. In conclusion, we determined that FcRγ plays an important role in regulating CD8+ T-cell functions during chronic LCMV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Fc/immunology , Acute Disease , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , CD8-Positive T-Lymphocytes/pathology , Chronic Disease , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Receptors, Fc/genetics
13.
FASEB J ; 34(3): 4462-4481, 2020 03.
Article in English | MEDLINE | ID: mdl-31989715

ABSTRACT

Myeloid progenitor cells have generally been considered the predominant source of myeloid cells under steady-state conditions. Here we show that NK cells contributed to a myeloid cell lineage pool in naïve and tumor-bearing mice. Using fate tracing of NKp46+ cells, we found that myeloid cells could be derived from NK cells. Notably, among mature CD11b+ CD27+ NK cells, c-Kit+ CD24+ NK cells were capable of differentiating into a range of myeloid lineages in vitro and produced neutrophils and monocytes in vivo. The differentiation was completely inhibited by NK-stimulating cytokines. In addition to the potential for differentiation into myeloid cells, c-Kit+ CD24+ NK cells retained NK cell phenotypes and effector functions. Mechanistically, GATA-2 was necessary for the differentiation of c-Kit+ CD24+ NK cells. Therefore, we discovered that GATA-2-dependent differentiation of c-Kit+ CD24+ NK cells contributes to myeloid cell development and identified a novel pathway for myeloid lineage commitment under physiological conditions.


Subject(s)
Cell Proliferation/physiology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/physiology , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
14.
PLoS Biol ; 16(4): e2004867, 2018 04.
Article in English | MEDLINE | ID: mdl-29702643

ABSTRACT

NKp46, a natural killer (NK) cell-activating receptor, is involved in NK cell cytotoxicity against virus-infected cells or tumor cells. However, the role of NKp46 in other NKp46+ non-NK innate lymphoid cell (ILC) populations has not yet been characterized. Here, an NKp46 deficiency model of natural cytotoxicity receptor 1 (Ncr1)gfp/gfp and Ncr1gfp/+ mice, i.e., homozygous and heterozygous knockout (KO), was used to explore the role of NKp46 in regulating the development of the NKp46+ ILCs. Surprisingly, our studies demonstrated that homozygous NKp46 deficiency resulted in a nearly complete depletion of the ILC1 subset (ILC1) of group 1 ILCs, and heterozygote KO decreased the number of cells in the ILC1 subset. Moreover, transplantation studies confirmed that ILC1 development depends on NKp46 and that the dependency is cell intrinsic. Interestingly, however, the cell depletion specifically occurred in the ILC1 subset but not in the other ILCs, including ILC2s, ILC3s, and NK cells. Thus, our studies reveal that NKp46 selectively participates in the regulation of ILC1 development.


Subject(s)
Antigens, Ly/genetics , Cell Lineage/immunology , Gene Expression Regulation, Developmental/immunology , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , Animals , Antigens, Ly/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Lineage/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Heterozygote , Homozygote , Immunity, Innate , Immunophenotyping , Integrin alpha2/genetics , Integrin alpha2/immunology , Killer Cells, Natural/cytology , Liver/cytology , Liver/immunology , Mice , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/deficiency , Natural Cytotoxicity Triggering Receptor 1/immunology , Spleen/cytology , Spleen/immunology , TNF-Related Apoptosis-Inducing Ligand/deficiency , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
15.
J Immunol ; 203(7): 1981-1988, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31444264

ABSTRACT

NK cells can recognize target cells such as virus-infected and tumor cells through integration of activation and inhibitory receptors. Recognition by NK cells can lead to direct lysis of the target cell and production of the signature cytokine IFN-γ. However, it is unclear whether stimulation through activation receptors alone is sufficient for IFN-γ production. In this study, we show that NK activation receptor engagement requires additional signals for optimal IFN-γ production, which could be provided by IFN-ß or IL-12. Stimulation of murine NK cells with soluble Abs directed against NK1.1, Ly49H, Ly49D, or NKp46 required additional stimulation with cytokines, indicating that a range of activation receptors with distinct adaptor molecules require additional stimulation for IFN-γ production. The requirement for multiple signals extends to stimulation with primary m157-transgenic target cells, which triggers the activation receptor Ly49H, suggesting that NK cells do require multiple signals for IFN-γ production in the context of target cell recognition. Using quantitative PCR and RNA flow cytometry, we found that cytokines, not activating ligands, act on NK cells to express Ifng transcripts. Ly49H engagement is required for IFN-γ translational initiation. Results using inhibitors suggest that the proteasome-ubiquitin-IKK-TPL2-MNK1 axis was required during activation receptor engagement. Thus, this study indicates that activation receptor-dependent IFN-γ production is regulated on the transcriptional and translational levels.


Subject(s)
Interferon-gamma/immunology , Killer Cells, Natural/immunology , Proteasome Endopeptidase Complex/immunology , Protein Biosynthesis/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Interferon-gamma/genetics , Killer Cells, Natural/cytology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , NK Cell Lectin-Like Receptor Subfamily B/genetics , NK Cell Lectin-Like Receptor Subfamily B/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Proteasome Endopeptidase Complex/genetics , Signal Transduction/genetics
16.
J Immunol ; 200(6): 1982-1987, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29440507

ABSTRACT

B6.SJL-Ptprca Pepcb /Boy (CD45.1) mice have been used in hundreds of congenic competitive transplants, with the presumption that they differ from C57BL/6 mice only at the CD45 locus. In this study, we describe a point mutation in the natural cytotoxicity receptor 1 (Ncr1) locus fortuitously identified in the CD45.1 strain. This point mutation was mapped at the 40th nucleotide of the Ncr1 locus causing a single amino acid mutation from cysteine to arginine at position 14 from the start codon, resulting in loss of NCR1 expression. We found that these mice were more resistant to CMV due to a hyper innate IFN-γ response in the absence of NCR1. In contrast, loss of NCR1 increased susceptibility to influenza virus, a result that is consistent with the role of NCR1 in the recognition of influenza Ag, hemagglutinin. This work sheds light on potential confounding experimental interpretation when this congenic strain is used as a tool for tracking lymphocyte development.


Subject(s)
Antigens, Ly/genetics , Leukocyte Common Antigens/genetics , Natural Cytotoxicity Triggering Receptor 1/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Point Mutation/genetics , Animals , Hemagglutinins/immunology , Immunity, Innate , Interferon-gamma/immunology , Mice , Mice, Congenic , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics
17.
J Immunol ; 201(9): 2551-2556, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30275046

ABSTRACT

NK cells accumulate in adult murine and human uteri during decidualization induced physiologically, pathologically, or experimentally. Adoptive transfer studies indicate that uterine NK (uNK) cells arise from circulating progenitors. However, virgin uteri contain few circulating NK1.1+CD49a- conventional NK cells, whereas NK1.1+CD49a+ tissue-resident NK (trNK) cells are abundant. In this study, we employed a novel, immune-competent NK cell-specific reporter mouse to track accumulation of uNK cells during unmanipulated pregnancies. We identified conventional NK and trNK cells accumulating in both decidua basalis and myometrium. Only trNK cells showed evidence of proliferation. In parabiosis studies using experimentally induced deciduomata, the accumulated uNK cells were proliferating trNK cells; migrating NK cells made no contribution. Together, these data suggest proliferating trNK cells are the source of uNK cells during endometrial decidualization.


Subject(s)
Cell Movement/immunology , Cell Proliferation/physiology , Decidua/cytology , Killer Cells, Natural/immunology , Pregnancy, Animal , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Decidua/immunology , Female , Green Fluorescent Proteins/genetics , Killer Cells, Natural/cytology , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Parabiosis , Pregnancy
18.
Eur J Immunol ; 48(7): 1137-1152, 2018 07.
Article in English | MEDLINE | ID: mdl-29624673

ABSTRACT

The bone marrow hosts NK cells whose distribution, motility and response to systemic immune challenge are poorly understood. At steady state, two-photon microscopy of the bone marrow in Ncr1gfp/+ mice captured motile NK cells interacting with dendritic cells. NK cells expressed markers and effector molecules of mature cells. Following poly (I:C) injection, RNA-Seq of NK cells revealed three phases of transcription featuring immune response genes followed by posttranscriptional processes and proliferation. Functionally, poly (I:C) promoted upregulation of granzyme B, enhanced cytotoxicity in vitro and in vivo, and, in the same individual cells, triggered proliferation. Two-photon imaging revealed that the proportion of sinusoidal NK cells decreased, while at the same time parenchymal NK cells accelerated, swelled and divided within the bone marrow. MVA viremia induced similar responses. Our findings demonstrate that the bone marrow is patrolled by mature NK cells that rapidly proliferate in response to systemic viral challenge while maintaining their effector functions.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Viremia/immunology , Animals , Antigens, Ly/genetics , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic , Granzymes/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Poly I-C/immunology , Virus Activation
19.
Nature ; 494(7436): 261-5, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23334414

ABSTRACT

At mucosal surfaces, the immune system should not initiate inflammatory immune responses to the plethora of antigens constantly present in the environment, but should remain poised to unleash a potent assault on intestinal pathogens. The transcriptional programs and regulatory factors required for immune cells to switch from homeostatic (often tissue-protective) function to potent antimicrobial immunity are poorly defined. Mucosal retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) innate lymphoid cells (ILCs) are emerging as an important innate lymphocyte population required for immunity to intestinal infections. Various subsets of RORγt(+) ILCs have been described but the transcriptional programs controlling their specification and fate remain largely unknown. Here we provide evidence that the transcription factor T-bet determines the fate of a distinct lineage of CCR6(-)RORγt(+) ILCs. Postnatally emerging CCR6(-)RORγt(+) ILCs upregulated T-bet and this was controlled by cues from the commensal microbiota and interleukin-23 (IL-23). In contrast, CCR6(+)RORγt(+) ILCs, which arise earlier during ontogeny, did not express T-bet. T-bet instructed the expression of T-bet target genes such as interferon-γ (IFN-γ) and of the natural cytotoxicity receptor NKp46. Mice genetically lacking T-bet showed normal development of CCR6(-)RORγt(+) ILCs, but they could not differentiate into NKp46-expressing RORγt(+) ILCs (that is, IL-22-producing natural killer (NK-22) cells) and failed to produce IFN-γ. The production of IFN-γ by T-bet-expressing CCR6(-)RORγt(+) ILCs was essential for the release of mucus-forming glycoproteins required to protect the epithelial barrier during Salmonella enterica infection. Salmonella infection also causes severe enterocolitis that is at least partly driven by IFN-γ. Mice deficient for T-bet or depleted of ILCs developed only mild enterocolitis. Thus, graded expression of T-bet in CCR6(-)RORγt(+) ILCs facilitates the differentiation of IFN-γ-producing CCR6(-)RORγt(+) ILCs required to protect the epithelial barrier against Salmonella infections. Co-expression of T-bet and RORγt, which is also found in subsets of IL-17-producing T-helper (T(H)17) cells, may be an evolutionarily conserved transcriptional program that originally developed as part of the innate defence against infections but that also confers an increased risk of immune-mediated pathology.


Subject(s)
Cell Lineage , Immunity, Innate/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, CCR6/deficiency , T-Box Domain Proteins/metabolism , Animals , Antigens, Ly/genetics , Cell Differentiation , Cells, Cultured , Enterocolitis/immunology , Enterocolitis/metabolism , Enterocolitis/pathology , Epithelium/immunology , Epithelium/metabolism , Epithelium/microbiology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-23/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucus/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Receptors, CCR6/metabolism , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity
20.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L977-L990, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30234375

ABSTRACT

Natural killer (NK) cells are cytotoxic innate lymphoid cells with an established role in the regulation of vascular structure in pregnancy and cancer. Impaired NK cell function has been identified in patients with pulmonary arterial hypertension (PAH), a disease of obstructive vascular remodeling in the lungs, as well as in multiple rodent models of disease. However, the precise contribution of NK cell impairment to the initiation and progression of PAH remains unknown. Here, we report the development of spontaneous pulmonary hypertension in two independent genetic models of NK cell dysfunction, including Nfil3-/- mice, which are deficient in NK cells due to the absence of the NFIL3 transcription factor, and Ncr1-Gfp mice, which lack the NK activating receptor NKp46. Mouse models of NK insufficiency exhibited increased right ventricular systolic pressure and muscularization of the pulmonary arteries in the absence of elevated left ventricular end-diastolic pressure, indicating that the development of pulmonary hypertension was not secondary to left heart dysfunction. In cases of severe NK cell impairment or loss, a subset of mice failed to develop pulmonary hypertension and instead exhibited reduced systemic blood pressure, demonstrating an extension of vascular abnormalities beyond the pulmonary circulation into the systemic vasculature. In both mouse models, the development of PAH was linked to elevated interleukin-23 production, whereas systemic hypotension in Ncr1-Gfp mice was accompanied by a loss of angiopoietin-2. Together, these results support an important role for NK cells in the regulation of pulmonary and systemic vascular function and the pathogenesis of PAH.


Subject(s)
Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Killer Cells, Natural/pathology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Disease Models, Animal , Endothelial Cells/pathology , Humans , Lung/pathology , Mice , Natural Cytotoxicity Triggering Receptor 1/genetics , Pulmonary Artery/pathology , Vascular Remodeling/genetics
SELECTION OF CITATIONS
SEARCH DETAIL