Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(3): 112135, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36840944

ABSTRACT

Micronutrient deficiency is a major cause of disease throughout the world. Yet, how perturbations influence the immune-microbiome interface remains poorly understood. Here, we report that loss of dietary tryptophan (Trp) reshapes intestinal microbial communities, including the depletion of probiotic L. reuteri, drives transcriptional changes to immune response genes in the intestinal ileum, and reshapes the regulatory T cell (Treg) compartment. Dietary Trp deficiency promotes expansion of RORγt+ Treg cells and the loss of Gata3+ Tregs in a microbiota-dependent manner. In the absence of dietary Trp, provision of the AhR ligand indole-3-carbinol is sufficient to restore the Treg compartment. Together, these data show that dietary Trp deficiency perturbs the interaction between the host and its bacterial symbionts to regulate Treg homeostasis via the deprivation of bacterially derived Trp metabolites. Our findings highlight an essential role for immune-microbiome crosstalk as a key homeostatic regulator during nutrient deficiency.


Subject(s)
Microbiota , T-Lymphocytes, Regulatory , Tryptophan/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Homeostasis , Receptors, Aryl Hydrocarbon/genetics
2.
Sci Immunol ; 7(69): eabn8041, 2022 03 25.
Article in English | MEDLINE | ID: mdl-35333545

ABSTRACT

Targeting the potent immunosuppressive properties of FOXP3+ regulatory T cells (Tregs) has substantial therapeutic potential for treating autoimmune and inflammatory diseases. Yet, the molecular mechanisms controlling Treg homeostasis, particularly during inflammation, remain unclear. We report that caspase-8 is a central regulator of Treg homeostasis in a context-specific manner that is decisive during immune responses. In mouse genetic models, targeting caspase-8 in Tregs led to accumulation of effector Tregs resistant to apoptotic cell death. Conversely, inflammation induced the MLKL-dependent necroptosis of caspase-8-deficient lymphoid and tissue Tregs, which enhanced immunity to a variety of chronic infections to promote clearance of viral or parasitic pathogens. However, improved immunity came at the risk of lethal inflammation in overwhelming infections. Caspase-8 inhibition using a clinical-stage compound revealed that human Tregs have heightened sensitivity to necroptosis compared with conventional T cells. These findings reveal a fundamental mechanism in Tregs that could be targeted to manipulate the balance between immune tolerance versus response for therapeutic benefit.


Subject(s)
Caspase 8/metabolism , Immune Tolerance , T-Lymphocytes, Regulatory , Animals , Homeostasis , Inflammation/metabolism , Mice
3.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32160524

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Subject(s)
Immunity, Cellular , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-33/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Tryptophan Hydroxylase/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Disease Susceptibility , Gene Expression Regulation/immunology , Immunity, Innate , Immunity, Mucosal , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-33/genetics , Larva/growth & development , Larva/immunology , Larva/pathogenicity , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/growth & development , Nippostrongylus/pathogenicity , Primary Cell Culture , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology , Strongylida Infections/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/parasitology , Tryptophan Hydroxylase/genetics
4.
Immunity ; 49(2): 201-203, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30134197

ABSTRACT

Intestinal Treg cells suppress colitis; yet the mechanisms behind the intricate pathways involved in this process remain largely unknown. In this issue of Immunity,Bauché et al. (2018) show that Treg cells engage MHCII on CX3CR1+ macrophages via LAG3. This indirectly reduces IL-22 mediated colonic inflammation.


Subject(s)
Colitis , T-Lymphocytes, Regulatory , CX3C Chemokine Receptor 1 , Humans , Interleukin-23 , Intestines , Macrophages
5.
J Autoimmun ; 91: 73-82, 2018 07.
Article in English | MEDLINE | ID: mdl-29724515

ABSTRACT

Regulatory T (Treg) cells maintain immunological tolerance in steady-state and after immune challenge. Activated Treg cells can undergo further differentiation into an effector state that highly express genes critical for Treg cell function, including ICOS, TIGIT and IL-10, although how this process is controlled is poorly understood. Effector Treg cells also specifically express the transcriptional regulator Blimp-1 whose expression overlaps with many of the canonical markers associated with effector Treg cells, although not all ICOS+TIGIT+ Treg cells express Blimp-1 or IL-10. In this study, we addressed the role of Blimp-1 in effector Treg cell function. Mice lacking Blimp-1 specifically in Treg cells mature normally, but succumb to a multi-organ inflammatory disease later in life. Blimp-1 is not required for Treg cell differentiation, with mutant mice having increased numbers of effector Treg cells, but regulated a suite of genes involved in cell signaling, communication and survival, as well as being essential for the expression of the immune modulatory cytokine IL-10. Thus, Blimp-1 is a marker of effector Treg cells in all contexts examined and is required for the full functionality of these cells during aging.


Subject(s)
Aging/immunology , Inflammation/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immune Tolerance , Inflammation/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/genetics , Signal Transduction
6.
Cell ; 173(3): 554-567, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677509

ABSTRACT

The essential roles played by the immune system in the discrimination between self- versus non/altered-self and its integral role in promoting host defense against invading microbes and tumors have been extensively studied for many years. In these contexts, significant advances have been made in defining the molecular and cellular networks that orchestrate cell-cell communication to mediate host defense and pathogen expulsion. Notably, recent studies indicate that in addition to these classical immune functions, cells of the innate and adaptive immune system also sense complex tissue- and environment-derived signals, including those from the nervous system and the diet. In turn these responses regulate physiologic processes in multiple tissues throughout the body, including nervous system function, metabolic state, thermogenesis, and tissue repair. In this review we propose an integrated view of how the mammalian immune system senses and interacts with other complex organ systems to maintain tissue and whole-body homeostasis.


Subject(s)
Energy Metabolism , Immune System/physiology , Immunity, Innate/physiology , Adaptive Immunity , Animals , Cell Communication , Diet , Homeostasis , Host-Pathogen Interactions , Humans , Inflammation , Neurons/physiology , Regeneration , Sympathetic Nervous System , Vasoactive Intestinal Peptide/chemistry
7.
Science ; 359(6379): 1056-1061, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29496881

ABSTRACT

The type 2 inflammatory response is induced by various environmental and infectious stimuli. Although recent studies identified group 2 innate lymphoid cells (ILC2s) as potent sources of type 2 cytokines, the molecular pathways controlling ILC2 responses are incompletely defined. Here we demonstrate that murine ILC2s express the ß2-adrenergic receptor (ß2AR) and colocalize with adrenergic neurons in the intestine. ß2AR deficiency resulted in exaggerated ILC2 responses and type 2 inflammation in intestinal and lung tissues. Conversely, ß2AR agonist treatment was associated with impaired ILC2 responses and reduced inflammation in vivo. Mechanistically, we demonstrate that the ß2AR pathway is a cell-intrinsic negative regulator of ILC2 responses through inhibition of cell proliferation and effector function. Collectively, these data provide the first evidence of a neuronal-derived regulatory circuit that limits ILC2-dependent type 2 inflammation.


Subject(s)
Adaptive Immunity , Adrenergic Neurons/immunology , Immunity, Innate , Lymphocytes/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Humans , Inflammation/immunology , Intestines/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Nerve Net/immunology , Receptors, Adrenergic, beta-2/genetics , Signal Transduction
8.
Nature ; 549(7671): 282-286, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28869965

ABSTRACT

The type 2 cytokines interleukin (IL)-4, IL-5, IL-9 and IL-13 have important roles in stimulating innate and adaptive immune responses that are required for resistance to helminth infection, promotion of allergic inflammation, metabolic homeostasis and tissue repair. Group 2 innate lymphoid cells (ILC2s) produce type 2 cytokines, and although advances have been made in understanding the cytokine milieu that promotes ILC2 responses, how ILC2 responses are regulated by other stimuli remains poorly understood. Here we demonstrate that ILC2s in the mouse gastrointestinal tract co-localize with cholinergic neurons that express the neuropeptide neuromedin U (NMU). In contrast to other haematopoietic cells, ILC2s selectively express the NMU receptor 1 (NMUR1). In vitro stimulation of ILC2s with NMU induced rapid cell activation, proliferation, and secretion of the type 2 cytokines IL-5, IL-9 and IL-13 that was dependent on cell-intrinsic expression of NMUR1 and Gαq protein. In vivo administration of NMU triggered potent type 2 cytokine responses characterized by ILC2 activation, proliferation and eosinophil recruitment that was associated with accelerated expulsion of the gastrointestinal nematode Nippostrongylus brasiliensis or induction of lung inflammation. Conversely, worm burden was higher in Nmur1-/- mice than in control mice. Furthermore, use of gene-deficient mice and adoptive cell transfer experiments revealed that ILC2s were necessary and sufficient to mount NMU-elicited type 2 cytokine responses. Together, these data indicate that the NMU-NMUR1 neuronal signalling circuit provides a selective mechanism through which the enteric nervous system and innate immune system integrate to promote rapid type 2 cytokine responses that can induce anti-microbial, inflammatory and tissue-protective type 2 responses at mucosal sites.


Subject(s)
Cytokines/immunology , Immunity, Innate , Inflammation/immunology , Lymphocytes/immunology , Neuropeptides/metabolism , Adoptive Transfer , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cytokines/metabolism , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/immunology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/innervation , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/pathology , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice , Neuropeptides/pharmacology , Nippostrongylus/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Receptors, Neurotransmitter/deficiency , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Signal Transduction/drug effects
9.
Sci Signal ; 9(426): ra46, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27141930

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are composed of subsets that are either positive or negative for the natural cytotoxicity receptor (NCR) NKp46 (encoded by Ncr1). ILC3s are located at mucosal sites, such as in the intestine and lung, where they are exposed to billions of commensal microbes and potentially harmful pathogens. Together with T cells, the various ILC3 subsets maintain the balance between homeostasis and immune activation. Through genetic mapping, we identified a previously uncharacterized subset of NCR(-) ILC3s in mice that transiently express Ncr1, demonstrating previously undescribed heterogeneity within the ILC3 population. In addition, we showed that sustained Notch signaling was required for the maintenance of the NCR(+) phenotype and that the cytokine transforming growth factor-ß (TGF-ß) impaired the development of NCR(+) ILC3s. Thus, the plasticity of ILC3s is regulated by the balance between the opposing effects of Notch and TGF-ß signaling, maintaining homeostasis in the face of continual challenges.


Subject(s)
Lymphocytes/cytology , Receptor, Notch1/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cytokines/metabolism , Female , Immunity, Innate , Intestinal Mucosa/metabolism , Ligands , Lung/metabolism , Male , Mice , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/metabolism , Signal Transduction , T-Lymphocytes/cytology , Transcription, Genetic
10.
Immunity ; 44(1): 103-115, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26795246

ABSTRACT

The inhibitor of DNA binding 2 (Id2) is essential for natural killer (NK) cell development with its canonical role being to antagonize E-protein function and alternate lineage fate. Here we have identified a key role for Id2 in regulating interleukin-15 (IL-15) receptor signaling and homeostasis of NK cells by repressing multiple E-protein target genes including Socs3. Id2 deletion in mature NK cells was incompatible with their homeostasis due to impaired IL-15 receptor signaling and metabolic function and this could be rescued by strong IL-15 receptor stimulation or genetic ablation of Socs3. During NK cell maturation, we observed an inverse correlation between E-protein target genes and Id2. These results shift the current paradigm on the role of ID2, indicating that it is required not only to antagonize E-proteins during NK cell commitment, but constantly required to titrate E-protein activity to regulate NK cell fitness and responsiveness to IL-15.


Subject(s)
Cell Differentiation/immunology , Inhibitor of Differentiation Protein 2/immunology , Interleukin-15/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Animals , Cell Lineage/immunology , Cells, Cultured , Female , Flow Cytometry , Male , Mice , Mice, Mutant Strains , Receptors, Interleukin-15/immunology , Receptors, Interleukin-15/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/immunology , Transcription Factors/metabolism
11.
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
12.
J Exp Med ; 211(9): 1733-40, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25092873

ABSTRACT

Innate lymphoid cell (ILC) populations protect against infection and are essential for lymphoid tissue formation and tissue remodeling after damage. Nfil3 is implicated in the function of adaptive immune lineages and NK cell development, but it is not yet known if Nfil3 regulates other innate lymphoid lineages. Here, we identify that Nfil3 is essential for the development of Peyer's patches and ILC2 and ILC3 subsets. Loss of Nfil3 selectively reduced Peyer's patch formation and was accompanied by impaired recruitment and distribution of lymphocytes within the patches. ILC subsets exhibited high Nfil3 expression and genetic deletion of Nfil3 severely compromised the development of all subsets. Subsequently, Nfil3(-/-) mice were highly susceptible to disease when challenged with inflammatory or infectious agents. Thus, we demonstrate that Nfil3 is a key regulator of the development of ILC subsets essential for immune protection in the lung and gut.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Immunity, Innate , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Gene Expression , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Killer Cells, Natural/immunology , Lung/cytology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/cytology , Peyer's Patches/immunology , Transplantation Chimera/genetics , Transplantation Chimera/immunology
13.
Nat Commun ; 5: 4539, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25119382

ABSTRACT

The cytokine IL-15 is required for natural killer (NK) cell homeostasis; however, the intrinsic mechanism governing this requirement remains unexplored. Here we identify the absolute requirement for myeloid cell leukaemia sequence-1 (Mcl1) in the sustained survival of NK cells in vivo. Mcl1 is highly expressed in NK cells and regulated by IL-15 in a dose-dependent manner via STAT5 phosphorylation and subsequent binding to the 3'-UTR of Mcl1. Specific deletion of Mcl1 in NK cells results in the absolute loss of NK cells from all tissues owing to a failure to antagonize pro-apoptotic proteins in the outer mitochondrial membrane. This NK lymphopenia results in mice succumbing to multiorgan melanoma metastases, being permissive to allogeneic transplantation and being resistant to toxic shock following polymicrobial sepsis challenge. These results clearly demonstrate a non-redundant pathway linking IL-15 to Mcl1 in the maintenance of NK cells and innate immune responses in vivo.


Subject(s)
Gene Deletion , Immune System Diseases/genetics , Immunity, Innate/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cytokines/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Immune System Diseases/physiopathology , Immunity, Innate/physiology , Interleukin-15/pharmacology , Interleukin-15/physiology , Killer Cells, Natural/drug effects , Lymphopenia/genetics , Lymphopenia/pathology , Lymphopenia/physiopathology , Male , Mice , Mice, Inbred C57BL , STAT5 Transcription Factor/physiology , Signal Transduction/physiology
14.
J Immunol ; 191(8): 4383-91, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24038093

ABSTRACT

Innate lymphocyte populations play a central role in conferring protective immunity at the mucosal frontier. In this study, we demonstrate that T cell factor 1 (TCF-1; encoded by Tcf7), a transcription factor also important for NK and T cell differentiation, is expressed by multiple innate lymphoid cell (ILC) subsets, including GATA3(+) nuocytes (ILC2) and NKp46(+) ILCs (ILC3), which confer protection against lung and intestinal inflammation. TCF-1 was intrinsically required for the differentiation of both ILC2 and NKp46(+) ILC3. Loss of TCF-1 expression impaired the capacity of these ILC subsets to produce IL-5, IL-13, and IL-22 and resulted in crippled responses to intestinal infection with Citrobacter rodentium. Furthermore, a reduction in T-bet expression required for Notch-2-dependent development of NKp46(+) ILC3 showed a dose-dependent reduction in TCF-1 expression. Collectively, our findings demonstrate an essential requirement for TCF-1 in ILC2 differentiation and reveal a link among Tcf7, Notch, and Tbx21 in NKp46(+) ILC3 development.


Subject(s)
Intestines/immunology , Killer Cells, Natural/metabolism , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, Ly/metabolism , Cell Differentiation/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 1-alpha , Inflammation/immunology , Inflammation/microbiology , Interleukin-13/biosynthesis , Interleukin-5/biosynthesis , Interleukins/biosynthesis , Intestines/microbiology , Lymphocyte Activation , Mice , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptor, Notch2/metabolism , T Cell Transcription Factor 1/genetics , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/immunology , Interleukin-22
15.
Nat Immunol ; 14(4): 389-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455676

ABSTRACT

NKp46+ innate lymphoid cells (ILCs) serve important roles in regulating the intestinal microbiota and defense against pathogens. Whether NKp46+ ILCs arise directly from lymphoid tissue-inducer (LTi) cells or represent a separate lineage remains controversial. We report here that the transcription factor T-bet (encoded by Tbx21) was essential for the development of NKp46+ ILCs but not of LTi cells or nuocytes. Deficiency in interleukin 22 (IL-22)-producing NKp46+ ILCs resulted in greater susceptibility of Tbx21-/- mice to intestinal infection. Haploinsufficient T-bet expression resulted in lower expression of the signaling molecule Notch, and Notch signaling was necessary for the transition of LTi cells into NKp46+ ILCs. Furthermore, NKp46+ ILCs differentiated solely from the CD4- LTi population, not the CD4+ LTi population. Our results pinpoint the regulation of Notch signaling by T-bet as a distinct molecular pathway that guides the development of NKp46+ ILCs.


Subject(s)
Antigens, Ly/metabolism , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Receptors, Notch/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Lymphocyte Subsets/cytology , Mice , Mice, Knockout , T-Box Domain Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...