Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Front Immunol ; 12: 760999, 2021.
Article in English | MEDLINE | ID: mdl-34804049

ABSTRACT

The loss of inhibitor of differentiation-2 (ID2) could lead to the development of colitis in mice, supplementation with exogenous ID2 protein might be a potential strategy to ameliorate colitis. In this study, the effects of ID2 protein supplementation on Dextran sodium sulfate (DSS)-induced colitis were investigated. Firstly, we confirmed that the expression of ID2 was reduced in the colon tissues of DSS-induced colitis mice and patients with ulcerative colitis (UC). Then, we constructed a recombinant plasmid containing the human Id2 gene and expressed it in Escherichia coli (E. coli) successfully. After purification and identification, purified hID2 could ameliorate DSS-induced colitis efficiently in mice by improving disease symptoms, decreasing the levels of proinflammatory cytokines in colon tissues, maintaining the integrity of intestinal barrier and reducing the infiltration of neutrophils and macrophages in the colon. Further study showed that hID2 could be endocytosed efficiently by neutrophils and macrophages, and hID2 lost its protection function against colitis when neutrophils were depleted with an anti-Gr-1 antibody. hID2 decreased the mRNA levels of IL-6, IL-1ß and TNF-α in lipopolysaccharides (LPS)-stimulated neutrophils and efficiently inhibited the activation of NF-κB signalling pathway in neutrophils. Interestingly, hID2 showed a synergistic role in inhibition of NF-κB activation with pyrrolidine dithiocarbamic acid (PDTC), an inhibitor of NF-κB activation. Therefore, this study demonstrated the potential use of hID2 to treat UC, and hID2 protein might be a promising anti-inflammatory agent that targets the NF-κB signalling pathway in neutrophils.


Subject(s)
Colitis, Ulcerative/immunology , Inhibitor of Differentiation Protein 2/immunology , Animals , Caco-2 Cells , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colon/immunology , Colon/pathology , Cytokines/immunology , Dextran Sulfate , Feces/microbiology , Humans , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/therapeutic use , Male , Mice, Inbred C57BL , NF-kappa B/immunology , Neutrophils/immunology , Recombinant Proteins/therapeutic use
2.
Immunity ; 52(5): 808-824.e7, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433949

ABSTRACT

Tissue-resident memory CD8+ T cells (Trm) provide host protection through continuous surveillance of non-lymphoid tissues. Using single-cell RNA-sequencing (scRNA-seq) and genetic reporter mice, we identified discrete lineages of intestinal antigen-specific CD8+ T cells, including a Blimp1hiId3lo tissue-resident effector cell population most prominent in the early phase of acute viral and bacterial infections and a molecularly distinct Blimp1loId3hi tissue-resident memory population that subsequently accumulated at later infection time points. These Trm populations exhibited distinct cytokine production, secondary memory potential, and transcriptional programs including differential roles for transcriptional regulators Blimp1, T-bet, Id2, and Id3 in supporting and maintaining intestinal Trm. Extending our analysis to malignant tissue, we also identified discrete populations of effector-like and memory-like CD8+ T cell populations with tissue-resident gene-expression signatures that shared features of terminally exhausted and progenitor-exhausted T cells, respectively. Our findings provide insight into the development and functional heterogeneity of Trm cells, which has implications for enhancing vaccination and immunotherapy approaches.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Immunotherapy/methods , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Inhibitor of Differentiation Proteins/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism
3.
J Immunol ; 202(8): 2296-2306, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30814306

ABSTRACT

NK cells are innate-like lymphocytes that eliminate virally infected and cancerous cells, but the mechanisms that control NK cell development and cytotoxicity are incompletely understood. We identified roles for sclerostin domain-containing-1 (Sostdc1) in NK cell development and function. Sostdc1-knockout (Sostdc1 -/-) mice display a progressive accumulation of transitional NK cells (tNKs) (CD27+CD11b+) with age, indicating a partial developmental block. The NK cell Ly49 repertoire in Sostdc1 -/- mice is also changed. Lower frequencies of Sostdc1 -/- splenic tNKs express inhibitory Ly49G2 receptors, but higher frequencies express activating Ly49H and Ly49D receptors. However, the frequencies of Ly49I+, G2+, H+, and D+ populations were universally decreased at the most mature (CD27-CD11b+) stage. We hypothesized that the Ly49 repertoire in Sostdc1 -/- mice would correlate with NK killing ability and observed that Sostdc1-/- NK cells are hyporesponsive against MHC class I-deficient cell targets in vitro and in vivo, despite higher CD107a surface levels and similar IFN-γ expression to controls. Consistent with Sostdc1's known role in Wnt signaling regulation, Tcf7 and Lef1 levels were higher in Sostdc1 -/- NK cells. Expression of the NK development gene Id2 was decreased in Sostdc1-/- immature NK and tNK cells, but Eomes and Tbx21 expression was unaffected. Reciprocal bone marrow transplant experiments showed that Sostdc1 regulates NK cell maturation and expression of Ly49 receptors in a cell-extrinsic fashion from both nonhematopoietic and hematopoietic sources. Taken together, these data support a role for Sostdc1 in the regulation of NK cell maturation and cytotoxicity, and identify potential NK cell niches.


Subject(s)
Bone Morphogenetic Proteins/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Wnt Signaling Pathway/immunology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/cytology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/immunology , Mice , Mice, Knockout , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Wnt Signaling Pathway/genetics
4.
Mol Immunol ; 115: 56-63, 2019 11.
Article in English | MEDLINE | ID: mdl-30144957

ABSTRACT

NK cells are cytotoxic lymphocytes with a key role in limiting tumour metastases. In mice, the NK cell lineage continually expresses high levels of the Inhibitor of DNA-binding 2 (Id2) protein and loss of Id2 is incongruous with their survival due to aberrant E-protein target gene activity. Using novel Id2 and E-protein antibodies that detect both mouse and human proteins, we have extensively characterised Id2 and E-protein expression in murine and human NK cells. We detected clear expression of E2 A and HEB, and to a lesser extent E2-2 in murine NK cells. In contrast HEB appears to be the major E-protein expressed in human NK cells, with minor E2-2 expression and surprisingly, no E2 A detected in primary NK cells nor human NK cell lines. These novel antibodies are also functional in immunofluorescence and immunoprecipitation. Mass spectrometry analysis of Id2 immuno-precipitated from murine NK cells revealed a number of novel associated proteins including several members of the SWI/SNF-related matrix-associated actin-dependent regulator chromatin (SMARC) and Mediator complex (MED) families. Taken together, these data highlight the utility of novel Id2 and E-protein antibodies and caution against mouse models for understanding Id2/E-protein biology in NK cells given their clearly disparate expression patternbetween species.


Subject(s)
Antibodies/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/immunology , Transcription Factor 4/immunology , Animals , Cell Line , Cell Lineage/immunology , Chromatin/immunology , Humans , Mice , Mice, Inbred C57BL
5.
Eur J Immunol ; 49(3): 476-489, 2019 03.
Article in English | MEDLINE | ID: mdl-30578645

ABSTRACT

Antigen-specific Th1 cells could be a passage to the infection sites during infection to execute effector functions, such as help CD8+ T cells to localize in these sites by secretion of anti-viral cytokines-IFN-γ or direct cytotoxicity of antigen-bearing cells. However, the molecular components that modulate Th1 cell differentiation and function in response to viral infection remain incompletely understood. Here, we reported that both inhibitor of DNA binding 3(Id3) protein and inhibitor of DNA binding 2(Id2) protein promoted Th1 cell differentiation. Depletion of Id3 or Id2 led to severe defect of Th1 cell differentiation during influenza virus infection. Whereas depletion of both Id3 and Id2 in CD4+ T cells restrained Th1 cell differentiation to a greater extent, indicating that Id3 and Id2 nonredundantly regulate Th1 cell differentiation. Moreover, deletion of E-proteins, the antagonists of Id proteins, greatly enhanced Th1 cell differentiation. Mechanistic study indicated that E-proteins suppressed Th1 cell differentiation by directly binding to the regulatory elements of Th1 cell master regulator T-bet and regulate T-bet expression. Thus, our findings identified Id-protein's importance for Th1 cells and clarified the nonredundant role of Id3 and Id2 in regulating Th1 cell differentiation, providing novel insight that Id3-Id2-E protein axis are essential for Th1 cell polarization.


Subject(s)
Cell Differentiation/immunology , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Proteins/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Th1 Cells/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Regulatory Elements, Transcriptional/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Th1 Cells/virology
6.
Nat Immunol ; 19(12): 1427-1440, 2018 12.
Article in English | MEDLINE | ID: mdl-30374131

ABSTRACT

Multipotent progenitor cells confirm their T cell-lineage identity in the CD4-CD8- double-negative (DN) pro-T cell DN2 stages, when expression of the essential transcription factor Bcl11b begins. In vivo and in vitro stage-specific deletions globally identified Bcl11b-controlled target genes in pro-T cells. Proteomics analysis revealed that Bcl11b associated with multiple cofactors and that its direct action was needed to recruit those cofactors to selective target sites. Regions near functionally regulated target genes showed enrichment for those sites of Bcl11b-dependent recruitment of cofactors, and deletion of individual cofactors relieved the repression of many genes normally repressed by Bcl11b. Runx1 collaborated with Bcl11b most frequently for both activation and repression. In parallel, Bcl11b indirectly regulated a subset of target genes by a gene network circuit via the transcription inhibitor Id2 (encoded by Id2) and transcription factor PLZF (encoded by Zbtb16); Id2 and Zbtb16 were directly repressed by Bcl11b, and Id2 and PLZF controlled distinct alternative programs. Thus, our study defines the molecular basis of direct and indirect Bcl11b actions that promote T cell identity and block alternative potentials.


Subject(s)
Lymphopoiesis/immunology , Precursor Cells, T-Lymphoid/immunology , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Repressor Proteins/immunology , Tumor Suppressor Proteins/immunology , Animals , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology , Promyelocytic Leukemia Zinc Finger Protein/immunology
7.
Sci Immunol ; 3(22)2018 04 27.
Article in English | MEDLINE | ID: mdl-29703840

ABSTRACT

All innate lymphoid cells (ILCs) require the small helix-loop-helix transcription factor ID2, but the functions of ID2 are not well understood in these cells. We show that mature natural killer (NK) cells, the prototypic ILCs, developed in mice lacking ID2 but remained as precursor CD27+CD11b- cells that failed to differentiate into CD27-CD11b+ cytotoxic effectors. We show that ID2 limited chromatin accessibility at E protein binding sites near naïve T lymphocyte-associated genes including multiple chemokine receptors, cytokine receptors, and signaling molecules and altered the NK cell response to inflammatory cytokines. In the absence of ID2, CD27+CD11b- NK cells expressed ID3, a helix-loop-helix protein associated with naïve T cells, and they transitioned from a CD8 memory precursor-like to a naïve-like chromatin accessibility state. We demonstrate that ID3 was required for the development of ID2-deficient NK cells, indicating that completely unfettered E protein function is incompatible with NK cell development. These data solidify the roles of ID2 and ID3 as mediators of effector and naïve gene programs, respectively, and revealed a critical role for ID2 in promoting a chromatin state and transcriptional program in CD27+CD11b- NK cells that supports cytotoxic effector differentiation and cytokine responses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/immunology , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/immunology , Chromatin/metabolism , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism
8.
J Immunol ; 198(8): 3136-3148, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28258199

ABSTRACT

Inhibitor of DNA binding (Id) proteins, including Id1-4, are transcriptional regulators involved in promoting cell proliferation and survival in various cell types. Although upregulation of Id proteins is associated with a broad spectrum of tumors, recent studies have identified that Id3 plays a tumor-suppressor role in the development of Burkitt's lymphoma in humans and hepatosplenic T cell lymphomas in mice. In this article, we report rapid lymphoma development in Id2/Id3 double-knockout mice that is caused by unchecked expansion of invariant NKT (iNKT) cells or a unique subset of innate-like CD1d-independent T cells. These populations began to expand in neonatal mice and, upon malignant transformation, resulted in mortality between 3 and 11 mo of age. The malignant cells also gave rise to lymphomas upon transfer to Rag-deficient and wild-type hosts, reaffirming their inherent tumorigenic potential. Microarray analysis revealed a significantly modified program in these neonatal iNKT cells that ultimately led to their malignant transformation. The lymphoma cells demonstrated chromosome instability along with upregulation of several signaling pathways, including the cytokine-cytokine receptor interaction pathway, which can promote their expansion and migration. Dysregulation of genes with reported driver mutations and the NF-κB pathway were found to be shared between Id2/Id3 double-knockout lymphomas and human NKT tumors. Our work identifies a distinct premalignant state and multiple tumorigenic pathways caused by loss of function of Id2 and Id3. Thus, conditional deletion of Id2 and Id3 in developing T cells establishes a unique animal model for iNKT and relevant innate-like lymphomas.


Subject(s)
Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Proteins/immunology , Lymphoma/immunology , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Cell Separation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Flow Cytometry , Humans , Lymphoma/pathology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
9.
Protein Cell ; 8(4): 242-254, 2017 04.
Article in English | MEDLINE | ID: mdl-28108952

ABSTRACT

Research on innate lymphoid cells (ILC) has recently been a fast paced topic of immunological research. As ILCs are able to produce signature Th cytokine, ILCs have garnered considerable attention and have been described to represent the innate counterpart of the CD4+ T helper (Th) cells. The development and function of ILCs are precisely regulated by a network of crucial transcription factors, which are also involved in the development or differentiation of conventional natural killer (cNK) cells and T cells. In this review, we will summarize the key transcriptional regulators and their functions through each phases of ILC development. With the phase of ILC lineage commitment, we will focus in particular on the roles of the transcription regulators Id2 and GATA-3, which in collaboration with other transcriptional factors, are critically involved in the generation of ILC fate determined progenitors. Once an ILC lineage has been established, several other transcription factors are required for the specification and functional regulation of distinct mature ILC subsets. Thus, a comprehensive understanding of the interactions and regulatory mechanisms mediated by these transcription factors will help us to further understand how ILCs exert their helper-like functions and bridge the innate and adaptive immunity.


Subject(s)
GATA3 Transcription Factor/immunology , Immunity, Innate/physiology , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Humans
10.
Sci Signal ; 9(447): ra94, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27678219

ABSTRACT

Despite the potent ability of dendritic cells (DCs) to stimulate lymphocyte responses and host immunity, granulocyte-macrophage colony-stimulating factor-derived DCs (GM-DCs) used as antitumor vaccines have demonstrated relatively modest success in cancer immunotherapy. We found that injecting GM-DCs into melanoma tumors in mice, or culturing GM-DCs with melanoma-secreted cytokines or melanoma-conditioned medium, rapidly suppressed DC-intrinsic expression of the gene encoding inhibitor of differentiation 2 (ID2), a transcriptional regulator. Melanoma-associated cytokines repressed Id2 transcription in murine DCs through the activation of signal transducer and activator of transcription 3 (STAT3). Enforced expression of ID2 in GM-DCs (ID2-GM-DCs) suppressed their production of the proinflammatory cytokine tumor necrosis factor-α (TNF-α). Vaccination with ID2-GM-DCs slowed the progression of melanoma tumors and enhanced animal survival, which was associated with an increased abundance of tumor-infiltrating interferon-γ-positive CD4(+) effector and CD8(+) cytotoxic T cells and a decreased number of tumor-infiltrating regulatory CD4(+) T cells. The efficacy of the ID2-GM-DC vaccine was improved by combinatorial treatment with a blocking antibody to programmed cell death protein-1 (PD-1), a current immunotherapy that overcomes suppressive immune checkpoint signaling. Collectively, our data reveal a previously unrecognized STAT3-mediated immunosuppressive mechanism in DCs and indicate that DC-intrinsic ID2 promotes tumor immunity by modulating tumor-associated CD4(+) T cell responses. Thus, inhibiting STAT3 or overexpressing ID2 selectively in DCs may improve the efficiency of DC vaccines in cancer therapy.


Subject(s)
Dendritic Cells/immunology , Immunity, Cellular , Inhibitor of Differentiation Protein 2/immunology , Melanoma/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/pathology , Inhibitor of Differentiation Protein 2/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , STAT3 Transcription Factor/genetics , Signal Transduction/genetics
11.
Immunity ; 45(3): 610-625, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27612641

ABSTRACT

The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.


Subject(s)
Celiac Disease/immunology , Interleukin-15/immunology , Intestines/immunology , Lymphoma/immunology , T-Lymphocyte Subsets/immunology , Animals , CD3 Complex/immunology , Cell Differentiation/immunology , Cells, Cultured , Granzymes/immunology , Humans , Inhibitor of Differentiation Protein 2/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Receptor, Notch1/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology
12.
J Exp Med ; 213(6): 897-911, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27185854

ABSTRACT

Plasmacytoid dendritic cells (DCs [pDCs]) develop from pre-pDCs, whereas two lineages of conventional DCs (cDCs; cDC1s and cDC2s) develop from lineage-committed pre-cDCs. Several transcription factors (TFs) have been implicated in regulating the development of pDCs (E2-2 and Id2) and cDC1s (Irf8, Id2, and Batf3); however, those required for the early commitment of pre-cDCs toward the cDC2 lineage are unknown. Here, we identify the TF zinc finger E box-binding homeobox 2 (Zeb2) to play a crucial role in regulating DC development. Zeb2 was expressed from the pre-pDC and pre-cDC stage onward and highly expressed in mature pDCs and cDC2s. Mice conditionally lacking Zeb2 in CD11c(+) cells had a cell-intrinsic reduction in pDCs and cDC2s, coupled with an increase in cDC1s. Conversely, mice in which CD11c(+) cells overexpressed Zeb2 displayed a reduction in cDC1s. This was accompanied by altered expression of Id2, which was up-regulated in cDC2s and pDCs from conditional knockout mice. Zeb2 chromatin immunoprecipitation analysis revealed Id2 to be a direct target of Zeb2. Thus, we conclude that Zeb2 regulates commitment to both the cDC2 and pDC lineages through repression of Id2.


Subject(s)
Dendritic Cells/immunology , Homeodomain Proteins/immunology , Inhibitor of Differentiation Protein 2/immunology , Plasma Cells/immunology , Repressor Proteins/immunology , Up-Regulation/immunology , Animals , Dendritic Cells/cytology , Homeodomain Proteins/genetics , Inhibitor of Differentiation Protein 2/genetics , Mice , Mice, Knockout , Plasma Cells/cytology , Repressor Proteins/genetics , Zinc Finger E-box Binding Homeobox 2
13.
J Exp Med ; 213(5): 687-96, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27069114

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are a subset of ILCs that play a protective role in the response to helminth infection, but they also contribute to allergic lung inflammation. Here, we report that the deletion of the ETS1 transcription factor in lymphoid cells resulted in a loss of ILC2s in the bone marrow and lymph nodes and that ETS1 promotes the fitness of the common progenitor of all ILCs. ETS1-deficient ILC2 progenitors failed to up-regulate messenger RNA for the E protein transcription factor inhibitor ID2, a critical factor for ILCs, and these cells were unable to expand in cytokine-driven in vitro cultures. In vivo, ETS1 was required for the IL-33-induced accumulation of lung ILC2s and for the production of the T helper type 2 cytokines IL-5 and IL-13. IL-25 also failed to elicit an expansion of inflammatory ILC2s when these cells lacked ETS1. Our data reveal ETS1 as a critical regulator of ILC2 expansion and cytokine production and implicate ETS1 in the regulation of Id2 at the inception of ILC2 development.


Subject(s)
Cytokines/immunology , Immunity, Innate , Inhibitor of Differentiation Protein 2/immunology , Lymphocytes/immunology , Proto-Oncogene Protein c-ets-1/immunology , Animals , Cytokines/genetics , Inhibitor of Differentiation Protein 2/genetics , Mice , Mice, Knockout , Proto-Oncogene Protein c-ets-1/genetics
14.
Cell Rep ; 14(6): 1500-1516, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26832410

ABSTRACT

T and innate lymphoid cells (ILCs) share some aspects of their developmental programs. However, although Notch signaling is strictly required for T cell development, it is dispensable for fetal ILC development. Constitutive activation of Notch signaling, at the common lymphoid progenitor stage, drives T cell development and abrogates ILC development by preventing Id2 expression. By combining single-cell transcriptomics and clonal culture strategies, we characterize two heterogeneous α4ß7-expressing lymphoid progenitor compartments. αLP1 (Flt3(+)) still retains T cell potential and comprises the global ILC progenitor, while αLP2 (Flt3(-)) consists of ILC precursors that are primed toward the different ILC lineages. Only a subset of αLP2 precursors is sensitive to Notch signaling required for their proliferation. Our study identifies, in a refined manner, the diversity of transitional stages of ILC development, their transcriptional signatures, and their differential dependence on Notch signaling.


Subject(s)
B-Lymphocyte Subsets/immunology , Gene Expression Regulation, Developmental , Immunity, Innate , Signal Transduction , T-Lymphocyte Subsets/immunology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation , Cell Lineage/immunology , Cell Proliferation , Fetus , Gene Expression Profiling , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Integrins/genetics , Integrins/immunology , Mice , Mice, Transgenic , Receptors, Notch/genetics , Receptors, Notch/immunology , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocyte Subsets/cytology , Transcription, Genetic , Transcriptome , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/immunology
15.
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
16.
PLoS One ; 10(6): e0129237, 2015.
Article in English | MEDLINE | ID: mdl-26079374

ABSTRACT

Tumor vaccines have held much promise, but to date have demonstrated little clinical success. This lack of success is conceivably due to poor tumor antigen presentation combined with immuno-suppressive mechanisms exploited by the tumor itself. Knock down of Inhibitor of differentiation protein 2 (Id2-kd) in mouse neuroblastoma whole tumor cells rendered these cells immunogenic. Id2-kd neuroblastoma (Neuro2a) cells (Id2-kd N2a) failed to grow in most immune competent mice and these mice subsequently developed immunity against further wild-type Neuro2a tumor cell challenge. Id2-kd N2a cells grew aggressively in immune-compromised hosts, thereby establishing the immunogenicity of these cells. Therapeutic vaccination with Id2-kd N2a cells alone suppressed tumor growth even in established neuroblastoma tumors and when used in combination with immune checkpoint blockade eradicated large established tumors. Mechanistically, immune cell depletion studies demonstrated that while CD8+ T cells are critical for antitumor immunity, CD4+ T cells are also required to induce a sustained long-lasting helper effect. An increase in number of CD8+ T-cells and enhanced production of interferon gamma (IFNγ) was observed in tumor antigen stimulated splenocytes of vaccinated mice. More importantly, a massive influx of cytotoxic CD8+ T-cells infiltrated the shrinking tumor following combined immunotherapy. These findings show that down regulation of Id2 induced tumor cell immunity and in combination with checkpoint blockade produced a novel, potent, T-cell mediated tumor vaccine strategy.


Subject(s)
Cancer Vaccines/immunology , Inhibitor of Differentiation Protein 2/immunology , Lymphocyte Depletion/methods , Neuroblastoma/immunology , RNA Interference/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Disease-Free Survival , Female , Humans , Immunity/immunology , Immunotherapy/methods , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred Strains , Mice, Nude , Mice, SCID , Neuroblastoma/pathology , Neuroblastoma/therapy , RNAi Therapeutics/methods
17.
Immunity ; 42(4): 731-43, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902484

ABSTRACT

Microbiota-mediated effects on the host immune response facilitate colonization resistance against pathogens. However, it is unclear whether and how the host immune response can regulate the microbiota to mediate colonization resistance. ID2, an essential transcriptional regulator for the development of innate lymphoid cell (ILC) progenitors, remains highly expressed in differentiated ILCs with unknown function. Using conditionally deficient mice in which ID2 is deleted from differentiated ILC3s, we observed that these mutant mice exhibited greatly impaired gut colonization resistance against Citrobacter rodentium. Utilizing gnotobiotic hosts, we showed that the ID2-dependent early colonization resistance was mediated by interleukin-22 (IL-22) regulation of the microbiota. In addition to regulating development, ID2 maintained homeostasis of ILC3s and controlled IL-22 production through an aryl hydrocarbon receptor (AhR) and IL-23 receptor pathway. Thus, ILC3s can mediate immune surveillance, which constantly maintains a proper microbiota, to facilitate early colonization resistance through an ID2-dependent regulation of IL-22.


Subject(s)
Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/pathology , Inhibitor of Differentiation Protein 2/immunology , Interleukins/immunology , Lymphocytes/pathology , Receptors, Aryl Hydrocarbon/immunology , Animals , Cell Differentiation , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Gene Expression Regulation , Germ-Free Life/immunology , Homeostasis/immunology , Immunity, Innate , Inhibitor of Differentiation Protein 2/deficiency , Inhibitor of Differentiation Protein 2/genetics , Interleukins/genetics , Lymphocytes/immunology , Lymphocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction , Interleukin-22
18.
Nat Immunol ; 15(8): 767-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973820

ABSTRACT

Regulatory T (Treg) cells suppress the development of inflammatory disease, but our knowledge of transcriptional regulators that control this function remains incomplete. Here we show that expression of Id2 and Id3 in Treg cells was required to suppress development of fatal inflammatory disease. We found that T cell antigen receptor (TCR)-driven signaling initially decreased the abundance of Id3, which led to the activation of a follicular regulatory T (TFR) cell-specific transcription signature. However, sustained lower abundance of Id2 and Id3 interfered with proper development of TFR cells. Depletion of Id2 and Id3 expression in Treg cells resulted in compromised maintenance and localization of the Treg cell population. Thus, Id2 and Id3 enforce TFR cell checkpoints and control the maintenance and homing of Treg cells.


Subject(s)
Inflammation/immunology , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Cell Proliferation , Female , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Inflammation/genetics , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/biosynthesis , Inhibitor of Differentiation Proteins/genetics , Interleukin-10/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, CXCR5/biosynthesis , Sequence Analysis, RNA
19.
J Immunol ; 192(5): 2227-36, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24470501

ABSTRACT

Disease outcome is known to be influenced by defined subsets of invariant NKT (iNKT) cells residing in distinct locations within peripheral tissue. However, the factors governing the development of these unique iNKT sublineages during thymic development are unknown. In this study we explored the mechanism by which E protein transcription factors and their negative regulators, the Id proteins, control the development of iNKT sublineages after positive selection. We found that E proteins directly bound the promyelocytic leukemia zinc finger (PLZF) promoter and were required for expression of this lineage-defining transcription factor and for the maturation and expansion of thymic iNKT cells. Moreover, expression of the negative regulators of E proteins, Id2 and Id3, defined distinct iNKT cell sublineages. Id3 was expressed in PLZF(high) NKT2 cells and loss of Id3 allowed for increased thymic iNKT cell expansion and abundance of the PLZF(+) NKT2 sublineage. Id2 was expressed in T-BET(+) NKT1 cells, and both Id proteins were required for the formation of this sublineage. Thus, we provide insight into E and Id protein regulation of iNKT cell proliferation and differentiation to specific sublineages during development in the thymus.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Cell Differentiation/physiology , Cell Proliferation , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Proteins/immunology , Natural Killer T-Cells/immunology , Thymus Gland/immunology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/biosynthesis , Inhibitor of Differentiation Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Mice , Mice, Transgenic , Natural Killer T-Cells/cytology , Promyelocytic Leukemia Zinc Finger Protein , Thymus Gland/cytology , Thymus Gland/metabolism
20.
Adv Exp Med Biol ; 785: 9-26, 2013.
Article in English | MEDLINE | ID: mdl-23456833

ABSTRACT

The family of innate lymphoid cells (ILCs) comprises of natural killer (NK) cells, Rorγt-dependent ILCs (lymphoid tissue inducer (LTi) cells, ILC22, and ILC17), and type 2 ILCs. Apart from a common requirement for inhibitor of DNA binding 2 (Id2) expression and common γ-chain (γc) signaling, the differentiation of ILC populations is regulated by distinct transcription factors. ILCs play fundamental roles in processes such as cytotoxicity, lymphoid organogenesis, intestinal homeostasis, immunity against infections, and wound healing. However, the dysregulation of ILCs has been implicated in autoimmune and inflammatory diseases. Here, we will review the recent advances in ILC development and their roles in immunity and disease, with a primary focus on type 2 ILCs.


Subject(s)
Helminthiasis/immunology , Immunity, Innate , Inhibitor of Differentiation Protein 2/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Animals , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic , Gene Expression/immunology , Helminthiasis/parasitology , Helminths/immunology , Humans , Immunophenotyping , Inhibitor of Differentiation Protein 2/genetics , Lymphocytes/classification , Lymphocytes/parasitology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...