Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 287
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636132

ABSTRACT

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Subject(s)
Amino Acids/metabolism , CD8-Positive T-Lymphocytes/cytology , Immunologic Memory , Signal Transduction , Amino Acid Transport Systems/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Cycle , Cell Differentiation , Disease Models, Animal , Female , Gene Knock-In Techniques , Lymphocytic Choriomeningitis/immunology , Male , Mice , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology
2.
Nat Immunol ; 22(3): 370-380, 2021 03.
Article in English | MEDLINE | ID: mdl-33574619

ABSTRACT

During chronic infection and cancer, a self-renewing CD8+ T cell subset maintains long-term immunity and is critical to the effectiveness of immunotherapy. These stem-like CD8+ T cells diverge from other CD8+ subsets early after chronic viral infection. However, pathways guarding stem-like CD8+ T cells against terminal exhaustion remain unclear. Here, we show that the gene encoding transcriptional repressor BACH2 is transcriptionally and epigenetically active in stem-like CD8+ T cells but not terminally exhausted cells early after infection. BACH2 overexpression enforced stem-like cell fate, whereas BACH2 deficiency impaired stem-like CD8+ T cell differentiation. Single-cell transcriptomic and epigenomic approaches revealed that BACH2 established the transcriptional and epigenetic programs of stem-like CD8+ T cells. In addition, BACH2 suppressed the molecular program driving terminal exhaustion through transcriptional repression and epigenetic silencing. Thus, our study reveals a new pathway that enforces commitment to stem-like CD8+ lineage and prevents an alternative terminally exhausted cell fate.


Subject(s)
Arenaviridae Infections/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Epigenesis, Genetic , Precursor Cells, T-Lymphoid/metabolism , Transcription, Genetic , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Lineage , Cells, Cultured , Chronic Disease , Disease Models, Animal , Host-Pathogen Interactions , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/virology , Signal Transduction
3.
Nat Immunol ; 22(4): 434-448, 2021 04.
Article in English | MEDLINE | ID: mdl-33649580

ABSTRACT

T cells dynamically interact with multiple, distinct cellular subsets to determine effector and memory differentiation. Here, we developed a platform to quantify cell location in three dimensions to determine the spatial requirements that direct T cell fate. After viral infection, we demonstrated that CD8+ effector T cell differentiation is associated with positioning at the lymph node periphery. This was instructed by CXCR3 signaling since, in its absence, T cells are confined to the lymph node center and alternatively differentiate into stem-like memory cell precursors. By mapping the cellular sources of CXCR3 ligands, we demonstrated that CXCL9 and CXCL10 are expressed by spatially distinct dendritic and stromal cell subsets. Unlike effector cells, retention of stem-like memory precursors in the paracortex is associated with CCR7 expression. Finally, we demonstrated that T cell location can be tuned, through deficiency in CXCL10 or type I interferon signaling, to promote effector or stem-like memory fates.


Subject(s)
Arenaviridae Infections/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Immunologic Memory , Lymph Nodes/metabolism , Precursor Cells, T-Lymphoid/metabolism , Receptors, CXCR3/metabolism , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Lineage , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL9/genetics , Chemotaxis, Leukocyte , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Interferon Type I/metabolism , Ligands , Lymph Nodes/immunology , Lymph Nodes/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/virology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptors, CCR7/metabolism , Receptors, CXCR3/genetics , Signal Transduction , Stem Cell Niche , Stromal Cells/immunology , Stromal Cells/metabolism
4.
Nat Immunol ; 21(10): 1256-1266, 2020 10.
Article in English | MEDLINE | ID: mdl-32839610

ABSTRACT

CD8+ T cells responding to chronic infections or tumors acquire an 'exhausted' state associated with elevated expression of inhibitory receptors, including PD-1, and impaired cytokine production. Exhausted T cells are continuously replenished by T cells with precursor characteristics that self-renew and depend on the transcription factor TCF1; however, their developmental requirements are poorly understood. In the present study, we demonstrate that high antigen load promoted the differentiation of precursor T cells, which acquired hallmarks of exhaustion within days of infection, whereas early effector cells retained polyfunctional features. Early precursor T cells showed epigenetic imprinting characteristic of T cell receptor-dependent transcription factor binding and were restricted to the generation of cells displaying exhaustion characteristics. Transcription factors BACH2 and BATF were key regulators with opposing functions in the generation of early precursor T cells. Overall, we demonstrate that exhaustion manifests first in TCF1+ precursor T cells and is propagated subsequently to the pool of antigen-specific T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Precursor Cells, T-Lymphoid/immunology , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Chronic Disease , Clonal Anergy , Epigenesis, Genetic , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity
5.
Nat Immunol ; 21(9): 1010-1021, 2020 09.
Article in English | MEDLINE | ID: mdl-32661362

ABSTRACT

Robust CD8+ T cell memory is essential for long-term protective immunity but is often compromised in cancer, where T cell exhaustion leads to loss of memory precursors. Immunotherapy via checkpoint blockade may not effectively reverse this defect, potentially underlying disease relapse. Here we report that mice with a CD8+ T cell-restricted neuropilin-1 (NRP1) deletion exhibited substantially enhanced protection from tumor rechallenge and sensitivity to anti-PD1 immunotherapy, despite unchanged primary tumor growth. Mechanistically, NRP1 cell-intrinsically limited the self-renewal of the CD44+PD1+TCF1+TIM3- progenitor exhausted T cells, which was associated with their reduced ability to induce c-Jun/AP-1 expression on T cell receptor restimulation, a mechanism that may contribute to terminal T cell exhaustion at the cost of memory differentiation in wild-type tumor-bearing hosts. These data indicate that blockade of NRP1, a unique 'immune memory checkpoint', may promote the development of long-lived tumor-specific Tmem that are essential for durable antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Proteins/metabolism , Melanoma, Experimental/immunology , Neuropilin-1/metabolism , Precursor Cells, T-Lymphoid/immunology , Animals , Cell Line, Tumor , Humans , Immune Checkpoint Proteins/genetics , Immune Tolerance , Immunity , Immunologic Memory , Mice , Mice, Knockout , Neuropilin-1/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction
6.
Nat Immunol ; 20(11): 1456-1468, 2019 11.
Article in English | MEDLINE | ID: mdl-31636466

ABSTRACT

T cell specification and commitment require Notch signaling. Although the requirement for Notch signaling during intrathymic T cell development is known, it is still unclear whether the onset of T cell priming can occur in a prethymic niche and whether RBPJ-dependent Notch signaling has a role during this event. Here, we established an Rbpj-inducible system that allowed temporal and tissue-specific control of the responsiveness to Notch in all hematopoietic cells. Using this system, we found that Notch signaling was required before the early T cell progenitor stage in the thymus. Lymphoid-primed multipotent progenitors in the bone marrow underwent Notch signaling with Rbpj induction, which inhibited development towards the myeloid lineage in thymus-seeding progenitors. Thus, our results indicated that the onset of T cell differentiation occurred in a prethymic setting, and that Notch played an important role during this event.


Subject(s)
Cell Differentiation/immunology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, Notch/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cell Lineage/immunology , Cell Separation , Female , Flow Cytometry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Mice , Mice, Transgenic , Primary Cell Culture , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
7.
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
8.
Nat Immunol ; 18(2): 173-183, 2017 02.
Article in English | MEDLINE | ID: mdl-27992401

ABSTRACT

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Regulatory/physiology , Transcriptional Activation/immunology , Animals , Autoimmunity , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Immune Tolerance , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Precursor Cells, T-Lymphoid/physiology
9.
Nat Immunol ; 18(1): 45-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27869820

ABSTRACT

TET proteins oxidize 5-methylcytosine in DNA to 5-hydroxymethylcytosine and other oxidation products. We found that simultaneous deletion of Tet2 and Tet3 in mouse CD4+CD8+ double-positive thymocytes resulted in dysregulated development and proliferation of invariant natural killer T cells (iNKT cells). Tet2-Tet3 double-knockout (DKO) iNKT cells displayed pronounced skewing toward the NKT17 lineage, with increased DNA methylation and impaired expression of genes encoding the key lineage-specifying factors T-bet and ThPOK. Transfer of purified Tet2-Tet3 DKO iNKT cells into immunocompetent recipient mice resulted in an uncontrolled expansion that was dependent on the nonclassical major histocompatibility complex (MHC) protein CD1d, which presents lipid antigens to iNKT cells. Our data indicate that TET proteins regulate iNKT cell fate by ensuring their proper development and maturation and by suppressing aberrant proliferation mediated by the T cell antigen receptor (TCR).


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Natural Killer T-Cells/physiology , Precursor Cells, T-Lymphoid/physiology , Proto-Oncogene Proteins/metabolism , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Nat Immunol ; 18(7): 771-779, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530714

ABSTRACT

TCRαß+CD4-CD8α+CD8ß- intestinal intraepithelial lymphocytes (CD8αα IELs) are an abundant population of thymus-derived T cells that protect the gut barrier surface. We sought to better define the thymic IEL precursor (IELp) through analysis of its maturation, localization and emigration. We defined two precursor populations among TCRß+CD4-CD8- thymocytes by dependence on the kinase TAK1 and rigorous lineage-exclusion criteria. Those IELp populations included a nascent PD-1+ population and a T-bet+ population that accumulated with age. Both gave rise to intestinal CD8αα IELs after adoptive transfer. The PD-1+ IELp population included more strongly self-reactive clones and was largely restricted by classical major histocompatibility complex (MHC) molecules. Those cells localized to the cortex and efficiently emigrated in a manner dependent on the receptor S1PR1. The T-bet+ IELp population localized to the medulla, included cells restricted by non-classical MHC molecules and expressed the receptor NK1.1, the integrin CD103 and the chemokine receptor CXCR3. The two IELp populations further differed in their use of the T cell antigen receptor (TCR) α-chain variable region (Vα) and ß-chain variable region (Vß). These data provide a foundation for understanding the biology of CD8αα IELs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Intestinal Mucosa/immunology , Precursor Cells, T-Lymphoid/immunology , Thymocytes/immunology , Adaptive Immunity/immunology , Adoptive Transfer , Animals , Antigens, CD , Antigens, Ly/immunology , CD8 Antigens/immunology , Cell Lineage , Cell Movement/immunology , Flow Cytometry , Fluorescent Antibody Technique , Histocompatibility Antigens/immunology , Immunity, Mucosal/immunology , Integrin alpha Chains , Intestinal Mucosa/cytology , Lymphocytes , Mice , NK Cell Lectin-Like Receptor Subfamily B/immunology , Phenotype , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, CXCR3 , Receptors, Lysosphingolipid/immunology , Sphingosine-1-Phosphate Receptors , T-Box Domain Proteins/immunology , Thymocytes/cytology , Thymus Gland/cytology
11.
Nat Immunol ; 17(6): 704-11, 2016 06.
Article in English | MEDLINE | ID: mdl-27064374

ABSTRACT

The asymmetric partitioning of fate-determining proteins has been shown to contribute to the generation of CD8(+) effector and memory T cell precursors. Here we demonstrate the asymmetric partitioning of mTORC1 activity after the activation of naive CD8(+) T cells. This results in the generation of two daughter T cells, one of which shows increased mTORC1 activity, increased glycolytic activity and increased expression of effector molecules. The other daughter T cell has relatively low mTORC1 activity and increased lipid metabolism, expresses increased amounts of anti-apoptotic molecules and subsequently displays enhanced long-term survival. Mechanistically, we demonstrate a link between T cell antigen receptor (TCR)-induced asymmetric expression of amino acid transporters and RagC-mediated translocation of mTOR to the lysosomes. Overall, our data provide important insight into how mTORC1-mediated metabolic reprogramming affects the fate decisions of T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Precursor Cells, T-Lymphoid/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Female , Glycolysis , Immunologic Memory , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
12.
Nat Immunol ; 17(3): 304-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829766

ABSTRACT

The role of anergy, an acquired state of T cell functional unresponsiveness, in natural peripheral tolerance remains unclear. In this study, we found that anergy was selectively induced in fetal antigen-specific maternal CD4(+) T cells during pregnancy. A naturally occurring subpopulation of anergic polyclonal CD4(+) T cells, enriched for self antigen-specific T cell antigen receptors, was also present in healthy hosts. Neuropilin-1 expression in anergic conventional CD4(+) T cells was associated with hypomethylation of genes related to thymic regulatory T cells (Treg cells), and this correlated with their ability to differentiate into Foxp3(+) Treg cells that suppressed immunopathology. Thus, our data suggest that not only is anergy induction important in preventing autoimmunity but also it generates the precursors for peripheral Treg cell differentiation.


Subject(s)
Autoimmunity/immunology , Cell Differentiation/immunology , Clonal Anergy/immunology , Histocompatibility, Maternal-Fetal/immunology , Peripheral Tolerance/immunology , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Arthritis, Experimental/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Genes, T-Cell Receptor alpha , Immunoblotting , Male , Mice , Mice, Knockout , Neuropilin-1/metabolism , Pregnancy , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance , Thymocytes/immunology
13.
Nat Immunol ; 17(10): 1206-1215, 2016 10.
Article in English | MEDLINE | ID: mdl-27548434

ABSTRACT

Thymic epithelial cell differentiation, growth and function depend on the expression of the transcription factor Foxn1; however, its target genes have never been physically identified. Using static and inducible genetic model systems and chromatin studies, we developed a genome-wide map of direct Foxn1 target genes for postnatal thymic epithelia and defined the Foxn1 binding motif. We determined the function of Foxn1 in these cells and found that, in addition to the transcriptional control of genes involved in the attraction and lineage commitment of T cell precursors, Foxn1 regulates the expression of genes involved in antigen processing and thymocyte selection. Thus, critical events in thymic lympho-stromal cross-talk and T cell selection are indispensably choreographed by Foxn1.


Subject(s)
Epithelial Cells/physiology , Forkhead Transcription Factors/metabolism , Precursor Cells, T-Lymphoid/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Antigen Presentation/genetics , Cell Communication , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Clonal Selection, Antigen-Mediated/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genome/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic
14.
Immunity ; 51(5): 930-948.e6, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31604687

ABSTRACT

Generation of the first T lymphocytes in the human embryo involves the emergence, migration, and thymus seeding of lymphoid progenitors together with concomitant thymus organogenesis, which is the initial step to establish the entire adaptive immune system. However, the cellular and molecular programs regulating this process remain unclear. We constructed a single-cell transcriptional landscape of human early T lymphopoiesis by using cells from multiple hemogenic and hematopoietic sites spanning embryonic and fetal stages. Among heterogenous early thymic progenitors, one subtype shared common features with a subset of lymphoid progenitors in fetal liver that are known as thymus-seeding progenitors. Unbiased bioinformatics analysis identified a distinct type of pre-thymic lymphoid progenitors in the aorta-gonad-mesonephros (AGM) region. In parallel, we investigated thymic epithelial cell development and potential cell-cell interactions during thymus organogenesis. Together, our data provide insights into human early T lymphopoiesis that prospectively direct T lymphocyte regeneration, which might lead to development of clinical applications.


Subject(s)
Cell Differentiation/genetics , Lymphopoiesis/genetics , Organogenesis/genetics , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/embryology , Biomarkers , Cell Differentiation/immunology , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Lymphopoiesis/immunology , Signal Detection, Psychological , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Transcriptome
15.
Nature ; 609(7926): 354-360, 2022 09.
Article in English | MEDLINE | ID: mdl-35978192

ABSTRACT

CD8+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2-is maintained by precursors of exhausted T (TPEX) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6. Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-myb , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunotherapy , L-Selectin/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Viruses/immunology
16.
Nat Immunol ; 16(6): 628-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939024

ABSTRACT

Most T lymphocytes, including regulatory T cells (Treg cells), differentiate in the thymus. The age-dependent involution of this organ leads to decreasing production of T cells. Here we found that the output of new Treg cells from the thymus decreased substantially more than that of conventional T cells. Peripheral mouse and human Treg cells recirculated back to the thymus, where they constituted a large proportion of the pool of Treg cells and displayed an activated and differentiated phenotype. In the thymus, the recirculating cells exerted their regulatory function by inhibiting interleukin 2 (IL-2)-dependent de novo differentiation of Treg cells. Thus, Treg cell development is controlled by a negative feedback loop in which mature progeny cells return to the thymus and restrain development of precursors of Treg cells.


Subject(s)
Precursor Cells, T-Lymphoid/physiology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/immunology , Aging/immunology , Animals , Blood Circulation , Cell Differentiation/genetics , Cells, Cultured , Child , Feedback, Physiological , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Nat Immunol ; 14(11): 1190-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056746

ABSTRACT

Although intergenic long noncoding RNAs (lincRNAs) have been linked to gene regulation in various tissues, little is known about lincRNA transcriptomes in the T cell lineages. Here we identified 1,524 lincRNA clusters in 42 T cell samples, from early T cell progenitors to terminally differentiated helper T cell subsets. Our analysis revealed highly dynamic and cell-specific expression patterns for lincRNAs during T cell differentiation. These lincRNAs were located in genomic regions enriched for genes that encode proteins with immunoregulatory functions. Many were bound and regulated by the key transcription factors T-bet, GATA-3, STAT4 and STAT6. We found that the lincRNA LincR-Ccr2-5'AS, together with GATA-3, was an essential component of a regulatory circuit in gene expression specific to the TH2 subset of helper T cells and was important for the migration of TH2 cells.


Subject(s)
Gene Expression Regulation/immunology , Precursor Cells, T-Lymphoid/metabolism , RNA, Long Noncoding/genetics , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Animals , Cell Differentiation , Cell Movement , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Genetic Loci , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Protein Binding , RNA, Long Noncoding/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Transcriptome/immunology
18.
Blood ; 142(3): 274-289, 2023 07 20.
Article in English | MEDLINE | ID: mdl-36989489

ABSTRACT

Interleukin-7 (IL-7) supports the growth and chemoresistance of T-cell acute lymphoblastic leukemia (T-ALL), particularly the early T-cell precursor subtype (ETP-ALL), which frequently has activating mutations of IL-7 signaling. Signal transducer and activator of transcription (STAT5) is an attractive therapeutic target because it is almost universally activated in ETP-ALL, even in the absence of mutations of upstream activators such as the IL-7 receptor (IL-7R), Janus kinase, and Fms-like tyrosine kinase 3 (FLT3). To examine the role of activated STAT5 in ETP-ALL, we have used a Lmo2-transgenic (Lmo2Tg) mouse model in which we can monitor chemoresistant preleukemia stem cells (pre-LSCs) and leukemia stem cells (LSCs) that drive T-ALL development and relapse following chemotherapy. Using IL-7R-deficient Lmo2Tg mice, we show that IL-7 signaling was not required for the formation of pre-LSCs but essential for their expansion and clonal evolution into LSCs to generate T-ALL. Activated STAT5B was sufficient for the development of T-ALL in IL-7R-deficient Lmo2Tg mice, indicating that inhibition of STAT5 is required to block the supportive signals provided by IL-7. To further understand the role of activated STAT5 in LSCs of ETP-ALL, we developed a new transgenic mouse that enables T-cell specific and doxycycline-inducible expression of the constitutively activated STAT5B1∗6 mutant. Expression of STAT5B1∗6 in T cells had no effect alone but promoted expansion and chemoresistance of LSCs in Lmo2Tg mice. Pharmacologic inhibition of STAT5 with pimozide-induced differentiation and loss of LSCs, while enhancing response to chemotherapy. Furthermore, pimozide significantly reduced leukemia burden in vivo and overcame chemoresistance of patient-derived ETP-ALL xenografts. Overall, our results demonstrate that STAT5 is an attractive therapeutic target for eradicating LSCs in ETP-ALL.


Subject(s)
Precursor Cells, T-Lymphoid , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Mice , Animals , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Interleukin-7/genetics , Interleukin-7/metabolism , Pimozide/therapeutic use , Mice, Transgenic
19.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33479171

ABSTRACT

Runt domain-related (Runx) transcription factors are essential for early T cell development in mice from uncommitted to committed stages. Single and double Runx knockouts via Cas9 show that target genes responding to Runx activity are not solely controlled by the dominant factor, Runx1. Instead, Runx1 and Runx3 are coexpressed in single cells; bind to highly overlapping genomic sites; and have redundant, collaborative functions regulating genes pivotal for T cell development. Despite stable combined expression levels across pro-T cell development, Runx1 and Runx3 preferentially activate and repress genes that change expression dynamically during lineage commitment, mostly activating T-lineage genes and repressing multipotent progenitor genes. Furthermore, most Runx target genes are sensitive to Runx perturbation only at one stage and often respond to Runx more for expression transitions than for maintenance. Contributing to this highly stage-dependent gene regulation function, Runx1 and Runx3 extensively shift their binding sites during commitment. Functionally distinct Runx occupancy sites associated with stage-specific activation or repression are also distinguished by different patterns of partner factor cobinding. Finally, Runx occupancies change coordinately at numerous clustered sites around positively or negatively regulated targets during commitment. This multisite binding behavior may contribute to a developmental "ratchet" mechanism making commitment irreversible.


Subject(s)
Cell Lineage/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes/immunology , Transcriptome , Animals , Cell Differentiation , Cell Lineage/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Core Binding Factor Alpha 3 Subunit/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Male , Mice , Precursor Cells, T-Lymphoid/cytology , Primary Cell Culture , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Lymphocytes/classification , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
20.
Cancer ; 129(1): 49-59, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36281717

ABSTRACT

BACKGROUND: Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) is a distinct subtype of T-ALL with a unique immunophenotype and high treatment failure rate. The molecular genetic abnormalities and their prognostic impact in ETP-ALL patients are poorly understood. METHODS: The authors performed systematic analyses of the clinicopathologic features with an emphasis on molecular genetic aspects of 32 patients with ETP-ALL. RESULTS: The median age was 43 years (range, 16-71). The blasts were positive for cytoplasmic CD3 and CD7 and negative for CD1a and CD8. Other markers expressed included CD34 (88%), CD33 (72%), CD117 (68%), CD13 (58%), CD5 (partial, 56%), CD2 (38%), CD10 (25%), CD56 (partial, 19%), and CD4 (6%). Cytogenetic analyses revealed a diploid karyotype in 10 patients, simple (1-2) abnormalities in 10 patients, and complex karyotype in 10 patients. Next-generation sequencing for 21 patients demonstrated that all had gene mutations (median, four mutations per patient). The most frequently mutated genes were WT1 (38%), NOTCH1 (29%), NRAS (29%), PHF6 (25%), TP53 (24%), ASXL1 (19%), FLT3 (19%), and IKZF1 (19%). All patients except one received multi-agent chemotherapy, and 22 patients underwent allogeneic stem cell transplantation. Thrombocytopenia, an abnormal karyotype, and TP53 mutation were associated with markedly shortened overall survival. Stem cell transplantation significantly improved overall survival. CONCLUSIONS: Patients with ETP-ALL often have high mutation burden with increased genomic instability. TP53 mutation was the only molecular prognostic marker and was associated with complex karyotype and greater than or equal to five mutations. These patients may benefit from stem cell transplantation, and recurrent gene mutations may be novel therapeutic markers.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cells, T-Lymphoid , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Immunophenotyping , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL