Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Immunol ; 16(11): 1124-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414766

ABSTRACT

Subsets of innate lymphoid cells (ILCs) reside in the mucosa and regulate immune responses to external pathogens. While ILCs can be phenotypically classified into ILC1, ILC2 and ILC3 subsets, the transcriptional control of commitment to each ILC lineage is incompletely understood. Here we report that the transcription factor Runx3 was essential for the normal development of ILC1 and ILC3 cells but not of ILC2 cells. Runx3 controlled the survival of ILC1 cells but not of ILC3 cells. Runx3 was required for expression of the transcription factor RORγt and its downstream target, the transcription factor AHR, in ILC3 cells. The absence of Runx3 in ILCs exacerbated infection with Citrobacter rodentium. Therefore, our data establish Runx3 as a key transcription factor in the lineage-specific differentiation of ILC1 and ILC3 cells.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Animals , Antigens, Ly/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/immunology , Cell Lineage/immunology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Core Binding Factor Alpha 3 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Enterobacteriaceae Infections/etiology , Enterobacteriaceae Infections/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocyte Subsets/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
2.
Nature ; 584(7822): 624-629, 2020 08.
Article in English | MEDLINE | ID: mdl-32788723

ABSTRACT

Conventional type 1 dendritic cells (cDC1)1 are thought to perform antigen cross-presentation, which is required to prime CD8+ T cells2,3, whereas cDC2 are specialized for priming CD4+ T cells4,5. CD4+ T cells are also considered to help CD8+ T cell responses through a variety of mechanisms6-11, including a process whereby CD4+ T cells 'license' cDC1 for CD8+ T cell priming12. However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4+ and CD8+ T cells. As expected, tumour rejection required cDC1 and CD8+ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4+ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4+ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4+ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8+ T cell priming, but also for initial CD4+ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4+ and CD8+ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Neoplasms/immunology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/cytology , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Histocompatibility Antigens Class II/immunology , Mice , Signal Transduction
3.
Nat Immunol ; 12(4): 335-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21399639

ABSTRACT

NF-κB is a major gene regulator in immune responses, and ribosomal protein S3 (RPS3) is an NF-κB subunit that directs specific gene transcription. However, it is unknown how nuclear translocation of RPS3 is regulated. Here we report that phosphorylation of RPS3 Ser209 by the kinase IKKß was crucial for nuclear localization of RPS3 in response to activating stimuli. Moreover, virulence protein NleH1 of the foodborne pathogen Escherichia coli strain O157:H7 specifically inhibited phosphorylation of RPS3 Ser209 and blocked RPS3 function, thereby promoting bacterial colonization and diarrhea but resulting in less mortality in a gnotobiotic piglet-infection model. Thus, the IKKß-dependent modification of a specific amino acid in RPS3 promoted specific NF-κB functions that underlie the molecular pathogenetic mechanisms of E. coli O157:H7.


Subject(s)
Escherichia coli Proteins/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Ribosomal Proteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/virology , Escherichia coli O157/genetics , Escherichia coli O157/metabolism , Escherichia coli O157/physiology , Escherichia coli Proteins/genetics , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , I-kappa B Kinase/genetics , Immunoblotting , Jurkat Cells , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , RNA Interference , Ribosomal Proteins/genetics , Sequence Homology, Amino Acid , Serine/genetics , Serine/metabolism , Swine
4.
Mol Cell ; 52(4): 517-28, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24184211

ABSTRACT

Metazoan gene expression is often regulated after the recruitment of RNA polymerase II (Pol II) to promoters, through the controlled release of promoter-proximally paused Pol II into productive RNA synthesis. Despite the prevalence of paused Pol II, very little is known about the dynamics of these early elongation complexes or the fate of the short transcription start site-associated (tss) RNAs they produce. Here, we demonstrate that paused elongation complexes can be remarkably stable, with half-lives exceeding 15 min at genes with inefficient pause release. Promoter-proximal termination by Pol II is infrequent, and released tssRNAs are targeted for rapid degradation. Further, we provide evidence that the predominant tssRNA species observed are nascent RNAs held within early elongation complexes. We propose that stable pausing of polymerase provides a temporal window of opportunity for recruitment of factors to modulate gene expression and that the nascent tssRNA represents an appealing target for these interactions.


Subject(s)
Drosophila Proteins/genetics , RNA Polymerase II/physiology , RNA, Small Cytoplasmic/metabolism , Animals , Base Sequence , Cell Line , Chromatin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Exosome Multienzyme Ribonuclease Complex/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , RNA Stability , Signal Transduction , Transcription Elongation, Genetic
5.
Proc Natl Acad Sci U S A ; 114(40): E8440-E8447, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923946

ABSTRACT

Natural killer (NK) cells express MHC class I (MHC-I)-specific receptors, such as Ly49A, that inhibit killing of cells expressing self-MHC-I. Self-MHC-I also "licenses" NK cells to become responsive to activating stimuli and regulates the surface level of NK-cell inhibitory receptors. However, the mechanisms of action resulting from these interactions of the Ly49s with their MHC-I ligands, particularly in vivo, have been controversial. Definitive studies could be derived from mice with targeted mutations in inhibitory Ly49s, but there are inherent challenges in specifically altering a single gene within a multigene family. Herein, we generated a knock-in mouse with a targeted mutation in the immunoreceptor tyrosine-based inhibitory motif (ITIM) of Ly49A that abolished the inhibitory function of Ly49A in cytotoxicity assays. This mutant Ly49A caused a licensing defect in NK cells, but the surface expression of Ly49A was unaltered. Moreover, NK cells that expressed this mutant Ly49A exhibited an altered inhibitory receptor repertoire. These results demonstrate that Ly49A ITIM signaling is critical for NK-cell effector inhibition, licensing, and receptor repertoire development.


Subject(s)
Cytotoxicity, Immunologic/immunology , Genes, MHC Class I/immunology , Immunoreceptor Tyrosine-Based Inhibition Motif , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/physiology , Receptors, NK Cell Lectin-Like/metabolism , Animals , Cells, Cultured , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Receptors, NK Cell Lectin-Like/genetics , Tyrosine/metabolism
6.
bioRxiv ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38895234

ABSTRACT

Natural killer (NK) cells recognize target cells through germline-encoded activation and inhibitory receptors enabling effective immunity against viruses and cancer. The Ly49 receptor family in the mouse and killer immunoglobin-like receptor family in humans play a central role in NK cell immunity through recognition of MHC class I and related molecules. Functionally, these receptor families are involved in licensing and rejection of MHC-I-deficient cells through missing-self. The Ly49 family is highly polymorphic, making it challenging to detail the contributions of individual Ly49 receptors to NK cell function. Herein, we showed mice lacking expression of all Ly49s were unable to reject missing-self target cells in vivo, were defective in NK cell licensing, and displayed lower KLRG1 on the surface of NK cells. Expression of Ly49A alone on a H-2Dd background restored missing-self target cell rejection, NK cell licensing, and NK cell KLRG1 expression. Thus, a single inhibitory Ly49 receptor is sufficient to license NK cells and mediate missing-self in vivo.

7.
Crit Care Explor ; 5(10): e0990, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37868029

ABSTRACT

BACKGROUND: We report the case of a patient with aplastic anemia and pancytopenia on immune-suppressive therapy who developed invasive pulmonary infection with mucormycosis and was treated with immune adjuvant therapy. CASE SUMMARY: Given the patient's profound lymphopenia and progressive invasive mucor despite dual antifungal drug therapy, interleukin (IL)-7, a cytokine that induces lymphocyte activation and proliferation, was instituted and resulted in normalization of absolute lymphocyte counts and was temporally associated with clearance of fungal pathogens and resolution of clinical symptoms. CONCLUSION: Patients with life-threatening fungal infections are frequently immune suppressed and immune adjuvant therapies should be considered in patients who are not responding to antifungal drugs and source control. Well-designed, double-blind, placebo-controlled trials are needed to advance the field. Although a number of immune adjuvants may be beneficial in fungal sepsis, IL-7 is a particularly attractive immune adjuvant because of its broad immunologic effects on key immunologic pathways that mediate enhanced antifungal immune system activity.

8.
Elife ; 112022 10 03.
Article in English | MEDLINE | ID: mdl-36190189

ABSTRACT

BTB domain And CNC Homolog 2 (Bach2) is a transcription repressor that actively participates in T and B lymphocyte development, but it is unknown if Bach2 is also involved in the development of innate immune cells, such as natural killer (NK) cells. Here, we followed the expression of Bach2 during murine NK cell development, finding that it peaked in immature CD27+CD11b+ cells and decreased upon further maturation. Bach2 showed an organ and tissue-specific expression pattern in NK cells. Bach2 expression positively correlated with the expression of transcription factor TCF1 and negatively correlated with genes encoding NK effector molecules and those involved in the cell cycle. Lack of Bach2 expression caused changes in chromatin accessibility of corresponding genes. In the end, Bach2 deficiency resulted in increased proportions of terminally differentiated NK cells with increased production of granzymes and cytokines. NK cell-mediated control of tumor metastasis was also augmented in the absence of Bach2. Therefore, Bach2 is a key checkpoint protein regulating NK terminal maturation.


Subject(s)
BTB-POZ Domain , Basic-Leucine Zipper Transcription Factors , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin , Cytokines/genetics , Granzymes , Killer Cells, Natural , Mice , Transcription Factors/genetics
9.
Cell Rep ; 32(4): 107969, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726632

ABSTRACT

Major histocompatibility complex class I (MHC-I)-restricted immune responses are largely attributed to cytotoxic T lymphocytes (CTLs). However, natural killer (NK) cells, as predicted by the missing-self hypothesis, have opposing requirements for MHC-I, suggesting that they may also demonstrate MHC-I-restricted effects. In mice, the Ly49 inhibitory receptors prevent NK cell killing of missing-self targets in effector responses, and they have a proposed second function in licensing or educating NK cells via self-MHC-I in vivo. Here we show MHC-I-restricted control of murine cytomegalovirus (MCMV) infection in vivo that is NK cell dependent. Using mice lacking specific Ly49 receptors, we show that control of MCMV requires inhibitory Ly49 receptors and an inhibitory signaling motif and the capacity for MCMV to downregulate MHC-I. Taken together, these data provide definitive evidence that the inhibitory receptors are required for missing-self rejection and are relevant to MHC-I-restricted NK cell control of a viral infection in vivo.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Antigens, Ly , Cytomegalovirus Infections/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , Muromegalovirus/pathogenicity , NK Cell Lectin-Like Receptor Subfamily A/immunology , Receptors, Natural Killer Cell , Virus Diseases
10.
J Exp Med ; 216(1): 99-116, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30559128

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that are thought to kill cells that down-regulate MHC class I (MHC-I) through "missing-self" recognition. NK cells from B2m-/- mice that lack surface MHC-I, however, are not autoreactive as predicted by the missing-self hypothesis. As a result, it is unclear if MHC-I down-regulation in vivo induces NK cell reactivity or tolerance to missing-self. Here, we generated a floxed B2m mouse to acutely down-regulate MHC-I in vivo in a host that normally expresses MHC-I. Global down-regulation of MHC-I induced NK cell hyporesponsiveness and tolerance to missing-self without overt missing-self reactivity. In contrast, down-regulation of MHC-I on a small fraction of hematopoietic cells triggered missing-self reactivity. Surprisingly, down-regulation of MHC-I only on CD4+ T cells predominately induced tolerance to missing-self without resetting NK cell responsiveness. In this setting, inflammation triggered substantial missing-self reactivity. These results show that MHC-I down-regulation can induce either NK cell tolerance or killing in vivo and that inflammation promotes missing-self reactivity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Down-Regulation/immunology , Immune Tolerance , Killer Cells, Natural/immunology , beta 2-Microglobulin/immunology , Animals , Female , Male , Mice , Mice, Knockout , beta 2-Microglobulin/genetics
11.
J Exp Med ; 216(1): 117-132, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30559127

ABSTRACT

Tissue-resident memory CD8+ T cells (TRMs) confer rapid protection and immunity against viral infections. Many viruses have evolved mechanisms to inhibit MHCI presentation in order to evade CD8+ T cells, suggesting that these mechanisms may also apply to TRM-mediated protection. However, the effects of viral MHCI inhibition on the function and generation of TRMs is unclear. Herein, we demonstrate that viral MHCI inhibition reduces the abundance of CD4+ and CD8+ TRMs, but its effects on the local microenvironment compensate to promote antigen-specific CD8+ TRM formation. Unexpectedly, local cognate antigen enhances CD8+ TRM development even in the context of viral MHCI inhibition and CD8+ T cell evasion, strongly suggesting a role for in situ cross-presentation in local antigen-driven TRM differentiation. However, local cognate antigen is not required for CD8+ TRM maintenance. We also show that viral MHCI inhibition efficiently evades CD8+ TRM effector functions. These findings indicate that viral evasion of MHCI antigen presentation has consequences on the development and response of antiviral TRMs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Evasion , Immunologic Memory , Virus Diseases/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chlorocebus aethiops , Dogs , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic , Vero Cells , Virus Diseases/genetics , Virus Diseases/pathology
12.
Elife ; 82019 08 08.
Article in English | MEDLINE | ID: mdl-31393266

ABSTRACT

Innate lymphoid cells (ILCs) were originally classified based on their cytokine profiles, placing natural killer (NK) cells and ILC1s together, but recent studies support their separation into different lineages at steady-state. However, tumors may induce NK cell conversion into ILC1-like cells that are limited to the tumor microenvironment and whether this conversion occurs beyond this environment remains unknown. Here, we describe Toxoplasma gondii infection converts NK cells into ILC1-like cells that are distinct from both steady-state NK cells and ILC1s in uninfected mice. These cells were Eomes-dependent, indicating that NK cells can give rise to Eomes- Tbet-dependent ILC1-like cells that circulate widely and persist independent of ongoing infection. Moreover, these changes appear permanent, as supported by epigenetic analyses. Thus, these studies markedly expand current concepts of NK cells, ILCs, and their potential conversion.


Subject(s)
Cell Transdifferentiation , Killer Cells, Natural/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Disease Models, Animal , Mice , T-Box Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL