Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 6(63): eabg6895, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34516781

ABSTRACT

CD8+ T cell responses to pulmonary challenges are primed by lung migratory dendritic cells (mDCs), which capture antigens in the lungs and migrate to the lung-draining mediastinal lymph node (med-LN) to activate T cells. The lungs and the spleen are not connected by the lymphatic vasculature. Thus, the current paradigm suggests that, in response to respiratory virus infections that are restricted to the respiratory tract, priming of T cell responses by lung mDCs takes place entirely in the med-LN. Our results challenge this "LN-centric" paradigm by demonstrating that, during influenza virus infection, lung mDCs egress the med-LN and traffic to the spleen, where they prime influenza-specific CD8+ T cells. CD8+ T cells primed in the spleen are transcriptionally distinct and have enhanced ability to differentiate into long-lived memory cells compared with med-LN­primed counterparts. Thus, our data identify a lung mDC trafficking pathway that connects the lungs with the spleen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Spleen/immunology , Animals , Cell Movement/immunology , Dendritic Cells/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, T-Lymphoid/immunology
2.
Immunol Res ; 69(4): 323-333, 2021 08.
Article in English | MEDLINE | ID: mdl-34037945

ABSTRACT

Autoreactive T cell is one of the leading causes of immunological tolerance defects in the chronic inflammatory lesions of rheumatoid arthritis (RA). There have been several extracellular signals and intracellular pathways reported in regulating this process but largely remain unknown yet. In this study, we explored the roles of intestinal Wnt/ß-catenin on disease severity during collagen-induced arthritis model (CIA), an animal model of RA. We first testified the activity pattern Wnt/ß-catenin shifted by intragastric administration of LiCl and DKK-1 in the intestine by real-time PCR and WB analysis. The arthritis scores showing the disease severity in the DKK-1 group was significantly ameliorated compared with the control group at the late stage of the disease, while in the LiCl group, the scores were significantly elevated which was consistent with pathology score analysis of H&E staining. Next, ELISA was performed and showed that TNF-α and IL-17 in the LiCl group were significantly higher than that of the control group. IL-10 in the DKK-1 group was significantly higher than that in the LiCl-1 group and control group, P < 0.05. Flow cytometry of spleen T cells differentiation ratio showed that: Th1 from the DKK-1 and LiCl groups and Th17 from the LiCl group was significantly different from that of the blank model group, P < 0.05. Finally, we explored the effects of intestinal Wnt/ß-catenin on T cell differentiation regulator ROR-γt and TCF1 and found that both transcription factors were up-regulated in the LiCl group. Together, these data suggested the pro-information role of Wnt/ß-catenin pathway from the intestine in the CIA mouse, implying its use as a potential therapeutic target for the treatment of inflammatory diseases such as RA.


Subject(s)
Arthritis, Experimental/immunology , Intestines/immunology , Wnt Signaling Pathway , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cytokines/immunology , Gene Expression , Hepatocyte Nuclear Factor 1-alpha/immunology , Intercellular Signaling Peptides and Proteins/administration & dosage , Joints/pathology , Lithium Chloride/administration & dosage , Male , Mice, Inbred DBA , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Severity of Illness Index , T-Lymphocytes/immunology , Tarsal Bones/pathology
3.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33372138

ABSTRACT

Precise regulation of coinhibitory receptors is essential for maintaining immune tolerance without interfering with protective immunity, yet the mechanism underlying such a balanced act remains poorly understood. In response to protein immunization, T follicular helper (TFH) cells lacking Tcf1 and Lef1 transcription factors were phenotypically normal but failed to promote germinal center formation and antibody production. Transcriptomic profiling revealed that Tcf1/Lef1-deficient TFH cells aberrantly up-regulated CTLA4 and LAG3 expression, and treatment with anti-CTLA4 alone or combined with anti-LAG3 substantially rectified B-cell help defects by Tcf1/Lef1-deficient TFH cells. Mechanistically, Tcf1 and Lef1 restrain chromatin accessibility at the Ctla4 and Lag3 loci. Groucho/Tle corepressors, which are known to cooperate with Tcf/Lef factors, were essential for TFH cell expansion but dispensable for repressing coinhibitory receptors. In contrast, mutating key amino acids in histone deacetylase (HDAC) domain in Tcf1 resulted in CTLA4 derepression in TFH cells. These findings demonstrate that Tcf1-instrinsic HDAC activity is necessary for preventing excessive CTLA4 induction in protein immunization-elicited TFH cells and hence guarding their B-cell help function.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , T Follicular Helper Cells/immunology , Animals , Antigens, CD , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/metabolism , Cell Differentiation/immunology , Female , Germinal Center/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Immune Tolerance , Lymphoid Enhancer-Binding Factor 1/immunology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-6 , T Follicular Helper Cells/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Lymphocyte Activation Gene 3 Protein
4.
PLoS Pathog ; 16(6): e1008555, 2020 06.
Article in English | MEDLINE | ID: mdl-32579593

ABSTRACT

Exhaustion is a dysfunctional state of cytotoxic CD8+ T cells (CTL) observed in chronic infection and cancer. Current in vivo models of CTL exhaustion using chronic viral infections or cancer yield very few exhausted CTL, limiting the analysis that can be done on these cells. Establishing an in vitro system that rapidly induces CTL exhaustion would therefore greatly facilitate the study of this phenotype, identify the truly exhaustion-associated changes and allow the testing of novel approaches to reverse or prevent exhaustion. Here we show that repeat stimulation of purified TCR transgenic OT-I CTL with their specific peptide induces all the functional (reduced cytokine production and polyfunctionality, decreased in vivo expansion capacity) and phenotypic (increased inhibitory receptors expression and transcription factor changes) characteristics of exhaustion. Importantly, in vitro exhausted cells shared the transcriptomic characteristics of the gold standard of exhaustion, CTL from LCMV cl13 infections. Gene expression of both in vitro and in vivo exhausted CTL was distinct from T cell anergy. Using this system, we show that Tcf7 promoter DNA methylation contributes to TCF1 downregulation in exhausted CTL. Thus this novel in vitro system can be used to identify genes and signaling pathways involved in exhaustion and will facilitate the screening of reagents that prevent/reverse CTL exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA Methylation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Promoter Regions, Genetic/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology
5.
Immunity ; 52(5): 825-841.e8, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32396847

ABSTRACT

CD8+ T cell exhaustion is a major barrier to current anti-cancer immunotherapies. Despite this, the developmental biology of exhausted CD8+ T cells (Tex) remains poorly defined, restraining improvement of strategies aimed at "re-invigorating" Tex cells. Here, we defined a four-cell-stage developmental framework for Tex cells. Two TCF1+ progenitor subsets were identified, one tissue restricted and quiescent and one more blood accessible, that gradually lost TCF1 as it divided and converted to a third intermediate Tex subset. This intermediate subset re-engaged some effector biology and increased upon PD-L1 blockade but ultimately converted into a fourth, terminally exhausted subset. By using transcriptional and epigenetic analyses, we identified the control mechanisms underlying subset transitions and defined a key interplay between TCF1, T-bet, and Tox in the process. These data reveal a four-stage developmental hierarchy for Tex cells and define the molecular, transcriptional, and epigenetic mechanisms that could provide opportunities to improve cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epigenesis, Genetic/immunology , Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Transcription, Genetic/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Epigenesis, Genetic/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/therapy , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocyte Subsets/metabolism , Transcription, Genetic/genetics
7.
Nat Immunol ; 20(9): 1150-1160, 2019 09.
Article in English | MEDLINE | ID: mdl-31358996

ABSTRACT

Innate lymphoid cells (ILCs) play important functions in immunity and tissue homeostasis, but their development is poorly understood. Through the use of single-cell approaches, we examined the transcriptional and functional heterogeneity of ILC progenitors, and studied the precursor-product relationships that link the subsets identified. This analysis identified two successive stages of ILC development within T cell factor 1-positive (TCF-1+) early innate lymphoid progenitors (EILPs), which we named 'specified EILPs' and 'committed EILPs'. Specified EILPs generated dendritic cells, whereas this potential was greatly decreased in committed EILPs. TCF-1 was dispensable for the generation of specified EILPs, but required for the generation of committed EILPs. TCF-1 used a pre-existing regulatory landscape established in upstream lymphoid precursors to bind chromatin in EILPs. Our results provide insight into the mechanisms by which TCF-1 promotes developmental progression of ILC precursors, while constraining their dendritic cell lineage potential and enforcing commitment to ILC fate.


Subject(s)
Cell Lineage/immunology , Dendritic Cells/cytology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lymphoid Progenitor Cells/cytology , T-Lymphocytes/cytology , Animals , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice , Mice, Inbred C57BL , Transcription, Genetic/genetics
8.
J Immunol ; 203(4): 801-806, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31300510

ABSTRACT

Differentiation of T follicular helper (TFH) cells is regulated by a complex transcriptional network, with mutually antagonistic Bcl6-Blimp1 as a core regulatory axis. It is well established that Tcf1 acts upstream of Bcl6 for its optimal induction to program TFH cell differentiation. In this study, we show that whereas genetic ablation of Tcf1 in mice greatly diminished TFH cells in response to viral infection, compound deletion of Blimp1 with Tcf1 restored TFH cell frequency, numbers, and generation of germinal center B cells. Aberrant upregulation of T-bet and Id2 in Tcf1-deficient TFH cells was also largely rectified by ablating Blimp1. Tcf1 chromatin immunoprecipitation sequencing in TFH cells identified two strong Tcf1 binding sites in the Blimp1 gene at a 24-kb upstream and an intron-3 element. Deletion of the intron-3 element, but not the 24-kb upstream element, compromised production of TFH cells. Our data demonstrate that Tcf1-mediated Blimp1 repression is functionally critical for safeguarding TFH cell differentiation.


Subject(s)
Cell Differentiation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocytes, Helper-Inducer/cytology , Virus Diseases/immunology , Animals , Gene Expression Regulation/immunology , Germinal Center/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Positive Regulatory Domain I-Binding Factor 1/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
9.
J Immunol ; 203(2): 323-327, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31175159

ABSTRACT

The differentiation of memory CD8+ T cells is critical to the long-term cellular immunity. The transcription factor BCL6 has been reportedly important for the generation and maintenance of memory CD8+ T cells; however, using the newly established BCL6 conditional knockout mouse model, we demonstrate that BCL6 is dispensable for the maintenance of established memory CD8+ T cell pool, although BCL6 is still required for the generation of CD8+ memory precursors upon acute viral infection. In addition, BCL6 promotes the expression of TCF-1 via directly binding to the Tcf7 (gene symbol for TCF-1) allele in CD8+ memory precursors and forced expression of TCF-1 restores the generation of BCL6-deficient memory precursors. Thus, our findings clarify that BCL6 is dispensable for the maintenance of memory CD8+ T cells, but functions as an important upstream of TCF-1 to regulate the generation of memory precursors in acute viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Transcription Factors/genetics , Virus Diseases/genetics , Acute Disease , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Immunologic Memory/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-6/immunology , Transcription Factors/immunology , Virus Diseases/immunology
10.
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
11.
Nat Immunol ; 19(12): 1366-1378, 2018 12.
Article in English | MEDLINE | ID: mdl-30420627

ABSTRACT

Thymocyte development requires a complex orchestration of multiple transcription factors. Ablating either TCF-1 or HEB in CD4+CD8+ thymocytes elicits similar developmental outcomes including increased proliferation, decreased survival, and fewer late Tcra rearrangements. Here, we provide a mechanistic explanation for these similarities by showing that TCF-1 and HEB share ~7,000 DNA-binding sites genome wide and promote chromatin accessibility. The binding of both TCF-1 and HEB was required at these shared sites for epigenetic and transcriptional gene regulation. Binding of TCF-1 and HEB to their conserved motifs in the enhancer regions of genes associated with T cell differentiation promoted their expression. Binding to sites lacking conserved motifs in the promoter regions of cell-cycle-associated genes limited proliferation. TCF-1 displaced nucleosomes, allowing for chromatin accessibility. Importantly, TCF-1 inhibited Notch signaling and consequently protected HEB from Notch-mediated proteasomal degradation. Thus, TCF-1 shifts nucleosomes and safeguards HEB, thereby enabling their cooperation in establishing the epigenetic and transcription profiles of CD4+CD8+ thymocytes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Gene Expression Regulation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lymphopoiesis/immunology , Thymocytes/immunology , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Blood Adv ; 2(14): 1685-1690, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30021780

ABSTRACT

Expression of the transcription factor T-cell factor 1 (TCF1) identifies antigen-experienced murine CD8+ T cells that retain potential for lymphoid recirculation and the ability to self-renew while producing more differentiated effector cells. We found that CD8+ T cells in the blood of both healthy and chronically infected humans expressed TCF1 at 3 distinct levels: high (TCF1-hi), intermediate (TCF1-int), and low (TCF1-lo). TCF1-hi cells could be found within both the naive and memory compartments and were characterized by relative quiescence and lack of immediate effector function. A substantial fraction of TCF1-int cells were found among memory cells, and TCF1-int cells exhibited robust immediate effector functions. TCF1-lo cells were most enriched in effector memory cells that expressed the senescence marker CD57. Following reactivation, TCF1-hi cells gave rise to TCF1-lo descendants while self-renewing the TCF1-hi progenitor. By contrast, reactivation of TCF1-lo cells produced more TCF1-lo cells without evidence of de-differentiating into TCF1-hi cells. Flow cytometric analyses of TCF1 expression from patient specimens may become a useful biomarker for adaptive immune function in response to vaccination, infection, autoimmunity, and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immunologic Memory , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Female , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Male
13.
Biochem Med (Zagreb) ; 28(2): 020703, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29666556

ABSTRACT

INTRODUCTION: Maturity onset diabetes of the young due to HNF1A mutations (HNF1A-MODY) is the most frequent form of monogenic diabetes in adults. It is often misdiagnosed as type 1 or type 2 diabetes, but establishing genetic diagnosis is important, as treatment differs from the common types of diabetes. HNF1A-MODY has not been investigated in Croatia before due to limited access to genetic testing. In this study we aimed to describe the characteristics of young adults diagnosed with diabetes before the age of 45 years, who have rare HNF1A allele variants, and estimate the prevalence of HNF1A-MODY in Croatia. MATERIALS AND METHODS: We recruited 477 C-peptide positive and beta cell antibody negative subjects through the Croatian Diabetes Registry. HNF1A was sequenced for all participants and systematic assessment of the variants found was performed. The prevalence of HNF1A-MODY was calculated in the study group and results extrapolated to estimate the proportion of diabetic individuals with HNF1A-MODY in Croatia and the population prevalence. RESULTS: Our study identified 13 individuals harbouring rare HNF1A allelic variants. After systematic assessment, 8 were assigned a diagnosis of HNF1A-MODY. Two individuals were able to discontinue insulin treatment following the diagnosis. We estimated that HNF1A-MODY in Croatia has a prevalence of 66 (95% CI 61 - 72) cases per million. CONCLUSIONS: The estimated prevalence of HNF1A-MODY in Croatia is similar to that reported in other European countries. Finding cases lead to important treatment changes for patients. This strongly supports the introduction of diagnostic genetic testing for monogenic diabetes in Croatia.


Subject(s)
C-Peptide/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Hepatocyte Nuclear Factor 1-alpha/genetics , Mutation , Registries , Adolescent , Adult , Aged , Alleles , Autoantibodies/blood , Biomarkers/blood , Croatia/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Female , Gene Expression , Gene Frequency , Genetic Testing , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Male , Middle Aged , Prevalence , Sequence Analysis, DNA
14.
J Immunol ; 200(10): 3397-3406, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29632143

ABSTRACT

T cell factor 1 (TCF-1) is expressed in both developing and mature T cells and has been shown to restrain mature T cell-mediated Th17 responses by inhibiting IL-17 expression. However, it is not clear when TCF-1 is required in vivo to restrain the magnitude of peripheral Th17 responses and what the molecular mechanisms responsible for TCF-1-regulated IL-17 gene expression are. In this study, we showed that conditional deletion of TCF-1 at the early but not later CD4+CD8+ double-positive stage in mice enhanced Th17 differentiation and aggravated experimental autoimmune encephalomyelitis, which correlates with abnormally high IL-17 expression. Expression of TCF-1 in TCF-1-deficient thymocytes but not TCF-1-deficient Th17 cells inhibited IL-17 expression. TCF-1 binds to IL-17 promoter regions, and deletion of two TCF-1 binding sites relieves TCF-1-mediated inhibition of IL-17 promoter activity. Lastly, wild-type TCF-1, but not a TCF-1 mutant that has no intrinsic histone deacetylase activity, was able to inhibit IL-17 expression in TCF-1 deficient mouse thymocytes. Thus, our study demonstrates the requirement of TCF-1 in vivo at stages earlier than double-positive cells to restrain peripheral Th17 immunity by directly binding and inhibiting IL-17 promoter in its intrinsic histone deacetylase-dependent manner.


Subject(s)
Gene Expression/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Immunity/genetics , Immunity/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression/genetics , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Mice , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Thymocytes/immunology
15.
J Exp Med ; 215(2): 575-594, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29282254

ABSTRACT

Upon infection with an intracellular pathogen, cytotoxic CD8+ T cells develop diverse differentiation states characterized by function, localization, longevity, and the capacity for self-renewal. The program of differentiation is determined, in part, by FOXO1, a transcription factor known to integrate extrinsic input in order to specify survival, DNA repair, self-renewal, and proliferation. At issue is whether the state of T cell differentiation is specified by initial conditions of activation or is actively maintained. To study the spectrum of T cell differentiation, we have analyzed an infection with mouse cytomegalovirus, a persistent-latent virus that elicits different cytotoxic T cell responses characterized as acute resolving or inflationary. Our results show that FOXO1 is continuously required for all the phenotypic characteristics of memory-effector T cells such that with acute inactivation of the gene encoding FOXO1, T cells revert to a short-lived effector phenotype, exhibit reduced viability, and manifest characteristics of anergy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Anergy , Forkhead Box Protein O1/immunology , Immunologic Memory , Adoptive Transfer , Animals , Antigens, Viral , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Survival/immunology , Forkhead Box Protein O1/deficiency , Forkhead Box Protein O1/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Lectins, C-Type , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Receptors, Immunologic/immunology
16.
Immunity ; 47(6): 1129-1141.e5, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246443

ABSTRACT

During chronic stimulation, CD8+ T cells acquire an exhausted phenotype characterized by expression of inhibitory receptors, down-modulation of effector function, and metabolic impairments. T cell exhaustion protects from excessive immunopathology but limits clearance of virus-infected or tumor cells. We transcriptionally profiled antigen-specific T cells from mice infected with lymphocytic choriomeningitis virus strains that cause acute or chronic disease. T cell exhaustion during chronic infection was driven by high amounts of T cell receptor (TCR)-induced transcription factors IRF4, BATF, and NFATc1. These regulators promoted expression of inhibitory receptors, including PD-1, and mediated impaired cellular metabolism. Furthermore, they repressed the expression of TCF1, a transcription factor required for memory T cell differentiation. Reducing IRF4 expression restored the functional and metabolic properties of antigen-specific T cells and promoted memory-like T cell development. These findings indicate that IRF4 functions as a central node in a TCR-responsive transcriptional circuit that establishes and sustains T cell exhaustion during chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon Regulatory Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Gene Expression Regulation , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 114(42): E8865-E8874, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973925

ABSTRACT

The factors and steps controlling postinfection CD8+ T cell terminal effector versus memory differentiation are incompletely understood. Whereas we found that naive TCF7 (alias "Tcf-1") expression is FOXO1 independent, early postinfection we report bimodal, FOXO1-dependent expression of the memory-essential transcription factor TCF7 in pathogen-specific CD8+ T cells. We determined the early postinfection TCF7high population is marked by low TIM3 expression and bears memory signature hallmarks before the appearance of established memory precursor marker CD127 (IL-7R). These cells exhibit diminished TBET, GZMB, mTOR signaling, and cell cycle progression. Day 5 postinfection, TCF7high cells express higher memory-associated BCL2 and EOMES, as well as increased accumulation potential and capacity to differentiate into memory phenotype cells. TCF7 retroviral transduction opposes GZMB expression and the formation of KLRG1pos phenotype cells, demonstrating an active role for TCF7 in extinguishing the effector program and forestalling terminal differentiation. Past the peak of the cellular immune response, we report a gradient of FOXO1 and TCF7 expression, which functions to oppose TBET and orchestrate a continuum of effector-to-memory phenotypes.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Forkhead Box Protein O1/metabolism , Immunologic Memory/physiology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Forkhead Box Protein O1/genetics , Granzymes/genetics , Granzymes/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Lectins, C-Type , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, Immunologic/metabolism
18.
Cell Rep ; 20(3): 613-626, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723565

ABSTRACT

The transcription factor Tcf1 is essential for the development of natural killer (NK) cells. However, its precise role has not been clarified. Our combined analysis of Tcf1-deficient and transgenic mice indicated that Tcf1 guides NK cells through three stages of development. Tcf1 expression directed bone marrow progenitors toward the NK cell lineage and ensured the survival of NK-committed cells, and its downregulation was needed for terminal maturation. Impaired survival of NK-committed cells was due to excessive expression of granzyme B (GzmB) and other granzyme family members, which induced NK cell self-destruction during maturation and following activation with cytokines or target cells. Mechanistically, Tcf1 binding reduced the activity of a Gzmb-associated regulatory element, and this accounted for the reduced Gzmb expression in Tcf1-expressing NK cells. These data identify an unexpected requirement to limit the expression of cytotoxic effector molecules for the normal expansion and function of NK cells.


Subject(s)
Gene Expression Regulation, Enzymologic/immunology , Granzymes/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Killer Cells, Natural/immunology , Animals , Granzymes/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice , Mice, Knockout
19.
Nat Commun ; 8: 15050, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28466857

ABSTRACT

Differentiation and fate of virus-specific CD8+ T cells after cessation of chronic antigen stimulation is unclear. Here we show that a TCF1+CD127+PD1+ hepatitis C virus (HCV)-specific CD8+ T-cell subset exists in chronically infected patients with phenotypic features of T-cell exhaustion and memory, both before and after treatment with direct acting antiviral (DAA) agents. This subset is maintained during, and for a long duration after, HCV elimination. After antigen re-challenge the less differentiated TCF1+CD127+PD1+ population expands, which is accompanied by emergence of terminally exhausted TCF1-CD127-PD1hi HCV-specific CD8+ T cells. These results suggest the TCF1+CD127+PD1+ HCV-specific CD8+ T-cell subset has memory-like characteristics, including antigen-independent survival and recall proliferation. We thus provide evidence for the establishment of memory-like virus-specific CD8+ T cells in a clinically relevant setting of chronic viral infection and we uncover their fate after cessation of chronic antigen stimulation, implicating a potential strategy for antiviral immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Adult , Aged , Antiviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Female , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Young Adult
20.
Cell Rep ; 17(2): 436-447, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705792

ABSTRACT

Innate lymphoid cells (ILCs) are enriched at mucosal surfaces, where they provide immune surveillance. All ILC subsets develop from a common progenitor that gives rise to pre-committed progenitors for each of the ILC lineages. Currently, the temporal control of gene expression that guides the emergence of these progenitors is poorly understood. We used global transcriptional mapping to analyze gene expression in different ILC progenitors. We identified PD-1 to be specifically expressed in PLZF+ ILCp and revealed that the timing and order of expression of the transcription factors NFIL3, ID2, and TCF-1 was critical. Importantly, induction of ILC lineage commitment required only transient expression of NFIL3 prior to ID2 and TCF-1 expression. These findings highlight the importance of the temporal program that permits commitment of progenitors to the ILC lineage, and they expand our understanding of the core transcriptional program by identifying potential regulators of ILC development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunity, Innate/immunology , Lymphocytes/immunology , Programmed Cell Death 1 Receptor/genetics , Animals , Basic-Leucine Zipper Transcription Factors/immunology , Bone Marrow Cells/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-alpha/immunology , Immunity, Innate/genetics , Killer Cells, Natural/immunology , Mice , Programmed Cell Death 1 Receptor/immunology , Transcription Factors/genetics , Transcription Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL