Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.893
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 37: 457-495, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30676822

ABSTRACT

Exhausted CD8 T (Tex) cells are a distinct cell lineage that arise during chronic infections and cancers in animal models and humans. Tex cells are characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression, metabolic dysregulation, poor memory recall and homeostatic self-renewal, and distinct transcriptional and epigenetic programs. The ability to reinvigorate Tex cells through inhibitory receptor blockade, such as αPD-1, highlights the therapeutic potential of targeting this population. Emerging insights into the mechanisms of exhaustion are informing immunotherapies for cancer and chronic infections. However, like other immune cells, Tex cells are heterogeneous and include progenitor and terminal subsets with unique characteristics and responses to checkpoint blockade. Here, we review our current understanding of Tex cell biology, including the developmental paths, transcriptional and epigenetic features, and cell intrinsic and extrinsic factors contributing to exhaustion and how this knowledge may inform therapeutic targeting of Tex cells in chronic infections, autoimmunity, and cancer.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/metabolism , Immunotherapy/methods , Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/physiology , Virus Diseases/immunology , Animals , Cellular Senescence , Chronic Disease , Clonal Anergy , Epigenesis, Genetic , Humans , Neoplasms/therapy , Virus Diseases/therapy
2.
Annu Rev Immunol ; 37: 145-171, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30526160

ABSTRACT

Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimeric antigen receptor (CAR) T cells were recently approved by the US Food and Drug Administration and are poised to enter the practice of medicine for leukemia and lymphoma, demonstrating that engineered immune cells can serve as a powerful new class of cancer therapeutics. The emergence of synthetic biology approaches for cellular engineering provides a broadly expanded set of tools for programming immune cells for enhanced function. Advances in T cell engineering, genetic editing, the selection of optimal lymphocytes, and cell manufacturing have the potential to broaden T cell-based therapies and foster new applications beyond oncology, in infectious diseases, organ transplantation, and autoimmunity.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy, Adoptive/trends , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/physiology , Animals , Genetic Engineering , Humans , Neoplasms/immunology , T-Lymphocytes/transplantation , United States , United States Food and Drug Administration
3.
Annu Rev Immunol ; 35: 85-118, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28226225

ABSTRACT

Intrathymic T cell development is a complex process that depends upon continuous guidance from thymus stromal cell microenvironments. The thymic epithelium within the thymic stroma comprises highly specialized cells with a high degree of anatomic, phenotypic, and functional heterogeneity. These properties are collectively required to bias thymocyte development toward production of self-tolerant and functionally competent T cells. The importance of thymic epithelial cells (TECs) is evidenced by clear links between their dysfunction and multiple diseases where autoimmunity and immunodeficiency are major components. Consequently, TECs are an attractive target for cell therapies to restore effective immune system function. The pathways and molecular regulators that control TEC development are becoming clearer, as are their influences on particular stages of T cell development. Here, we review both historical and the most recent advances in our understanding of the cellular and molecular mechanisms controlling TEC development, function, dysfunction, and regeneration.


Subject(s)
Epithelial Cells/metabolism , T-Lymphocytes/physiology , Thymus Gland/pathology , Animals , Autoimmunity , Cell Differentiation , Epithelial Cells/immunology , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Thymus Gland/immunology , Transcription Factors/metabolism , AIRE Protein
4.
Cell ; 174(3): 672-687.e27, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053426

ABSTRACT

TCR-signaling strength generally correlates with peptide-MHC binding affinity; however, exceptions exist. We find high-affinity, yet non-stimulatory, interactions occur with high frequency in the human T cell repertoire. Here, we studied human TCRs that are refractory to activation by pMHC ligands despite robust binding. Analysis of 3D affinity, 2D dwell time, and crystal structures of stimulatory versus non-stimulatory TCR-pMHC interactions failed to account for their different signaling outcomes. Using yeast pMHC display, we identified peptide agonists of a formerly non-responsive TCR. Single-molecule force measurements demonstrated the emergence of catch bonds in the activating TCR-pMHC interactions, correlating with exclusion of CD45 from the TCR-APC contact site. Molecular dynamics simulations of TCR-pMHC disengagement distinguished agonist from non-agonist ligands based on the acquisition of catch bonds within the TCR-pMHC interface. The isolation of catch bonds as a parameter mediating the coupling of TCR binding and signaling has important implications for TCR and antigen engineering for immunotherapy.


Subject(s)
Histocompatibility Antigens Class I/physiology , Lymphocyte Activation/physiology , Adult , Female , Humans , Kinetics , Ligands , Major Histocompatibility Complex/physiology , Male , Middle Aged , Molecular Dynamics Simulation , Oligopeptides , Peptides , Protein Binding/physiology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Single Molecule Imaging , T-Lymphocytes/physiology
5.
Nat Immunol ; 21(10): 1280-1292, 2020 10.
Article in English | MEDLINE | ID: mdl-32719521

ABSTRACT

The development of TCRαß and TCRγδ T cells comprises a step-wise process in which regulatory events control differentiation and lineage outcome. To clarify these mechanisms, we employed RNA-sequencing, ATAC-sequencing and ChIPmentation on well-defined thymocyte subsets that represent the continuum of human T cell development. The chromatin accessibility dynamics show clear stage specificity and reveal that human T cell-lineage commitment is marked by GATA3- and BCL11B-dependent closing of PU.1 sites. A temporary increase in H3K27me3 without open chromatin modifications is unique for ß-selection, whereas emerging γδ T cells, which originate from common precursors of ß-selected cells, show large chromatin accessibility changes due to strong T cell receptor (TCR) signaling. Furthermore, we unravel distinct chromatin landscapes between CD4+ and CD8+ αß-lineage cells that support their effector functions and reveal gene-specific mechanisms that define mature T cells. This resource provides a framework for studying gene regulatory mechanisms that drive normal and malignant human T cell development.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Cell Differentiation , Cell Lineage , Cells, Cultured , Chromatin/metabolism , Clonal Selection, Antigen-Mediated , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Lymphocyte Activation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, RNA , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Nat Immunol ; 20(12): 1584-1593, 2019 12.
Article in English | MEDLINE | ID: mdl-31745336

ABSTRACT

The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.


Subject(s)
Immunotherapy/trends , Interleukin-7/metabolism , Neoplasms/immunology , Receptors, Interleukin-7/metabolism , T-Lymphocytes/physiology , Animals , Cell Differentiation , Cell Survival , Homeostasis , Humans , Interleukin-7/immunology , Receptors, Interleukin-7/immunology
7.
Nat Immunol ; 20(10): 1381-1392, 2019 10.
Article in English | MEDLINE | ID: mdl-31451788

ABSTRACT

Proliferation is tightly regulated during T cell development, and is limited to immature CD4-CD8- thymocytes. The major proliferative event is initiated at the 'ß-selection' stage following successful rearrangement of Tcrß, and is triggered by and dependent on concurrent signaling by Notch and the pre-T cell receptor (TCR); however, it is unclear how these signals cooperate to promote cell proliferation. Here, we found that ß-selection-associated proliferation required the combined activity of two Skp-cullin-F-box (SCF) ubiquitin ligase complexes that included as substrate recognition subunits the F-box proteins Fbxl1 or Fbxl12. Both SCF complexes targeted the cyclin-dependent kinase inhibitor Cdkn1b for polyubiquitination and proteasomal degradation. We found that Notch signals induced the transcription of Fbxl1, whereas pre-TCR signals induced the transcription of Fbxl12. Thus, concurrent Notch and pre-TCR signaling induced the expression of two genes, Fbxl1 and Fbxl12, whose products functioned identically but additively to promote degradation of Cdkn1b, cell cycle progression, and proliferation of ß-selected thymocytes.


Subject(s)
F-Box Proteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Notch/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Animals , Cell Differentiation , Cell Proliferation , Clonal Selection, Antigen-Mediated , Cyclin-Dependent Kinase Inhibitor p27/metabolism , F-Box Proteins/genetics , Gene Expression Regulation , Genes, T-Cell Receptor beta , Mice , Mice, Inbred C57BL , Receptor Cross-Talk , Signal Transduction
8.
Nat Immunol ; 20(10): 1322-1334, 2019 10.
Article in English | MEDLINE | ID: mdl-31427773

ABSTRACT

We report a new immunodeficiency disorder in mice caused by a viable hypomorphic mutation of Snrnp40, an essential gene encoding a subunit of the U5 small nuclear ribonucleoprotein (snRNP) complex of the spliceosome. Snrnp40 is ubiquitous but strongly expressed in lymphoid tissue. Homozygous mutant mice showed hypersusceptibility to infection by murine cytomegalovirus and multiple defects of lymphoid development, stability and function. Cell-intrinsic defects of hematopoietic stem cell differentiation also affected homozygous mutants. SNRNP40 deficiency in primary hematopoietic stem cells or T cells or the EL4 cell line increased the frequency of splicing errors, mostly intron retention, in several hundred messenger RNAs. Altered expression of proteins associated with immune cell function was also observed in Snrnp40-mutant cells. The immunological consequences of SNRNP40 deficiency presumably result from cumulative, moderate effects on processing of many different mRNA molecules and secondary reductions in the expression of critical immune proteins, yielding a syndromic immune disorder.


Subject(s)
Hematopoietic Stem Cells/physiology , Herpesviridae Infections/immunology , Immunologic Deficiency Syndromes/immunology , Muromegalovirus/physiology , Ribonucleoprotein, U5 Small Nuclear/metabolism , Spliceosomes/metabolism , T-Lymphocytes/physiology , Alleles , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Susceptibility , Herpesviridae Infections/genetics , Immunologic Deficiency Syndromes/genetics , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , RNA Splicing , Ribonucleoprotein, U5 Small Nuclear/genetics
9.
Nat Immunol ; 18(3): 263-273, 2017 03.
Article in English | MEDLINE | ID: mdl-28135252

ABSTRACT

Aire is a transcription factor that controls T cell tolerance by inducing the expression of a large repertoire of genes specifically in thymic stromal cells. It interacts with scores of protein partners of diverse functional classes. We found that Aire and some of its partners, notably those implicated in the DNA-damage response, preferentially localized to and activated long chromatin stretches that were overloaded with transcriptional regulators, known as super-enhancers. We also identified topoisomerase 1 as a cardinal Aire partner that colocalized on super-enhancers and was required for the interaction of Aire with all of its other associates. We propose a model that entails looping of super-enhancers to efficiently deliver Aire-containing complexes to local and distal transcriptional start sites.


Subject(s)
Chromatin Assembly and Disassembly , DNA Topoisomerases, Type I/metabolism , Enhancer Elements, Genetic/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Transcription Factors/metabolism , Transcriptional Activation , Animals , Autoimmunity , DNA Damage/genetics , DNA Repair/genetics , DNA Topoisomerases, Type I/genetics , Epigenesis, Genetic , Gene Regulatory Networks , HEK293 Cells , Humans , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Transcription Factors/genetics , AIRE Protein
10.
Nat Immunol ; 18(2): 161-172, 2017 02.
Article in English | MEDLINE | ID: mdl-27941786

ABSTRACT

Aire is a transcriptional regulator that induces promiscuous expression of thousands of genes encoding tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs). While the target genes of Aire are well characterized, the transcriptional programs that regulate its own expression have remained elusive. Here we comprehensively analyzed both cis-acting and trans-acting regulatory mechanisms and found that the Aire locus was insulated by the global chromatin organizer CTCF and was hypermethylated in cells and tissues that did not express Aire. In mTECs, however, Aire expression was facilitated by concurrent eviction of CTCF, specific demethylation of exon 2 and the proximal promoter, and the coordinated action of several transcription activators, including Irf4, Irf8, Tbx21, Tcf7 and Ctcfl, which acted on mTEC-specific accessible regions in the Aire locus.


Subject(s)
Epithelial Cells/immunology , Gene Regulatory Networks , T-Lymphocytes/physiology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Antigen Presentation/genetics , Autoantigens/metabolism , CCCTC-Binding Factor , Cell Differentiation , Cells, Cultured , Clonal Selection, Antigen-Mediated , DNA Methylation , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Thymus Gland/cytology , Transcription Factors/genetics , AIRE Protein
11.
Mol Cell ; 81(17): 3637-3649.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34478654

ABSTRACT

The off-target activity of the CRISPR-associated nuclease Cas9 is a potential concern for therapeutic genome editing applications. Although high-fidelity Cas9 variants have been engineered, they exhibit varying efficiencies and have residual off-target effects, limiting their applicability. Here, we show that CRISPR hybrid RNA-DNA (chRDNA) guides provide an effective approach to increase Cas9 specificity while preserving on-target editing activity. Across multiple genomic targets in primary human T cells, we show that 2'-deoxynucleotide (dnt) positioning affects guide activity and specificity in a target-dependent manner and that this can be used to engineer chRDNA guides with substantially reduced off-target effects. Crystal structures of DNA-bound Cas9-chRDNA complexes reveal distorted guide-target duplex geometry and allosteric modulation of Cas9 conformation. These structural effects increase specificity by perturbing DNA hybridization and modulating Cas9 activation kinetics to disfavor binding and cleavage of off-target substrates. Overall, these results pave the way for utilizing customized chRDNAs in clinical applications.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/physiology , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/physiology , DNA/genetics , Endonucleases/genetics , Gene Editing/methods , Genetic Techniques , Genome/genetics , Genomics/methods , Humans , Leukocytes, Mononuclear/metabolism , Molecular Conformation , RNA, Guide, Kinetoplastida/genetics , Structure-Activity Relationship , T-Lymphocytes/physiology
12.
Nat Immunol ; 17(9): 1075-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27400149

ABSTRACT

Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Minor Histocompatibility Antigens/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/physiology , Animals , Cells, Cultured , Homeostasis , Ion Transport , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptors, Lymphocyte Homing/genetics , Solute Carrier Family 12, Member 2/metabolism , WNK Lysine-Deficient Protein Kinase 1
13.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27695000

ABSTRACT

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lymphoid Progenitor Cells/physiology , Myeloid Progenitor Cells/physiology , Receptors, Notch/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Fetus , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
14.
Nat Immunol ; 17(7): 844-50, 2016 07.
Article in English | MEDLINE | ID: mdl-27213689

ABSTRACT

Most adaptive immune responses require the activation of specific T cells through the T cell antigen receptor (TCR)-CD3 complex. Here we show that cholesterol sulfate (CS), a naturally occurring analog of cholesterol, inhibits CD3 ITAM phosphorylation, a crucial first step in T cell activation. In biochemical studies, CS disrupted TCR multimers, apparently by displacing cholesterol, which is known to bind TCRß. Moreover, CS-deficient mice showed heightened sensitivity to a self-antigen, whereas increasing CS content by intrathymic injection inhibited thymic selection, indicating that this molecule is an intrinsic regulator of thymocyte development. These results reveal a regulatory role for CS in TCR signaling and thymic selection, highlighting the importance of the membrane microenvironment in modulating cell surface receptor activation.


Subject(s)
Cell Membrane/metabolism , Cholesterol Esters/metabolism , Cholesterol/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Autoimmunity/genetics , Cells, Cultured , Cholesterol/analogs & derivatives , Clonal Selection, Antigen-Mediated , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Multimerization/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , Sulfotransferases/genetics
15.
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
16.
Nat Immunol ; 17(8): 956-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27376470

ABSTRACT

During T cell development, multipotent progenitors relinquish competence for other fates and commit to the T cell lineage by turning on Bcl11b, which encodes a transcription factor. To clarify lineage commitment mechanisms, we followed developing T cells at the single-cell level using Bcl11b knock-in fluorescent reporter mice. Notch signaling and Notch-activated transcription factors collaborate to activate Bcl11b expression irrespectively of Notch-dependent proliferation. These inputs work via three distinct, asynchronous mechanisms: an early locus 'poising' function dependent on TCF-1 and GATA-3, a stochastic-permissivity function dependent on Notch signaling, and a separate amplitude-control function dependent on Runx1, a factor already present in multipotent progenitors. Despite their necessity for Bcl11b expression, these inputs act in a stage-specific manner, providing a multitiered mechanism for developmental gene regulation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphopoiesis/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Tracking , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , GATA3 Transcription Factor/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Signal Transduction , Single-Cell Analysis , Tumor Suppressor Proteins/genetics
17.
Nat Immunol ; 17(6): 721-727, 2016 06.
Article in English | MEDLINE | ID: mdl-27043412

ABSTRACT

The mouse thymus produces discrete γδ T cell subsets that make either interferon-γ (IFN-γ) or interleukin 17 (IL-17), but the role of the T cell antigen receptor (TCR) in this developmental process remains controversial. Here we show that Cd3g(+/-) Cd3d(+/-) (CD3 double-haploinsufficient (CD3DH)) mice have reduced TCR expression and signaling strength on γδ T cells. CD3DH mice had normal numbers and phenotypes of αß thymocyte subsets, but impaired differentiation of fetal Vγ6(+) (but not Vγ4(+)) IL-17-producing γδ T cells and a marked depletion of IFN-γ-producing CD122(+) NK1.1(+) γδ T cells throughout ontogeny. Adult CD3DH mice showed reduced peripheral IFN-γ(+) γδ T cells and were resistant to experimental cerebral malaria. Thus, TCR signal strength within specific thymic developmental windows is a major determinant of the generation of proinflammatory γδ T cell subsets and their impact on pathophysiology.


Subject(s)
Cell Differentiation , Inflammation/immunology , Malaria, Cerebral/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/physiology , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Antigens, Ly/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction
18.
Nat Immunol ; 17(6): 712-20, 2016 06.
Article in English | MEDLINE | ID: mdl-27111141

ABSTRACT

Sustained glucose and glutamine transport are essential for activated T lymphocytes to support ATP and macromolecule biosynthesis. We found that glutamine and glucose also fuel an indispensable dynamic regulation of intracellular protein O-GlcNAcylation at key stages of T cell development, transformation and differentiation. Glucose and glutamine are precursors of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), a substrate for cellular glycosyltransferases. Immune-activated T cells contained higher concentrations of UDP-GlcNAc and increased intracellular protein O-GlcNAcylation controlled by the enzyme O-linked-ß-N-acetylglucosamine (O-GlcNAc) glycosyltransferase as compared with naive cells. We identified Notch, the T cell antigen receptor and c-Myc as key controllers of T cell protein O-GlcNAcylation via regulation of glucose and glutamine transport. Loss of O-GlcNAc transferase blocked T cell progenitor renewal, malignant transformation and peripheral T cell clonal expansion. Nutrient-dependent signaling pathways regulated by O-GlcNAc glycosyltransferase are thus fundamental for T cell biology.


Subject(s)
Glucose/metabolism , Glutamine/metabolism , N-Acetylglucosaminyltransferases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Uridine Diphosphate N-Acetylglucosamine/metabolism , Animals , Cell Proliferation/genetics , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Clone Cells , Female , Lymphocyte Activation/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Proto-Oncogene Proteins c-myc/genetics , Receptors, Notch/metabolism
19.
Nat Immunol ; 16(6): 618-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939025

ABSTRACT

A20 is an anti-inflammatory protein linked to multiple human diseases; however, the mechanisms by which A20 prevents inflammatory disease are incompletely defined. We found that A20-deficient T cells and fibroblasts were susceptible to caspase-independent and kinase RIPK3-dependent necroptosis. Global deficiency in RIPK3 significantly restored the survival of A20-deficient mice. A20-deficient cells exhibited exaggerated formation of RIPK1-RIPK3 complexes. RIPK3 underwent physiological ubiquitination at Lys5 (K5), and this ubiquitination event supported the formation of RIPK1-RIPK3 complexes. Both the ubiquitination of RIPK3 and formation of the RIPK1-RIPK3 complex required the catalytic cysteine of A20's deubiquitinating motif. Our studies link A20 and the ubiquitination of RIPK3 to necroptotic cell death and suggest additional mechanisms by which A20 might prevent inflammatory disease.


Subject(s)
Cysteine Endopeptidases/metabolism , Fibroblasts/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/physiology , Animals , Apoptosis/genetics , Catalytic Domain/genetics , Cysteine Endopeptidases/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Necrosis/genetics , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/genetics , Ubiquitins/metabolism
20.
Immunity ; 48(2): 227-242.e8, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466755

ABSTRACT

How chromatin reorganization coordinates differentiation and lineage commitment from hematopoietic stem and progenitor cells (HSPCs) to mature immune cells has not been well understood. Here, we carried out an integrative analysis of chromatin accessibility, topologically associating domains, AB compartments, and gene expression from HSPCs to CD4+CD8+ T cells. We found that abrupt genome-wide changes at all three levels of chromatin organization occur during the transition from double-negative stage 2 (DN2) to DN3, accompanying the T lineage commitment. The transcription factor BCL11B, a critical regulator of T cell commitment, is associated with increased chromatin interaction, and Bcl11b deletion compromised chromatin interaction at its target genes. We propose that these large-scale and concerted changes in chromatin organization present an energy barrier to prevent the cell from reversing its fate to earlier stages or redirecting to alternatives and thus lock the cell fate into the T lineages.


Subject(s)
Cell Lineage , Cell Nucleus/physiology , Chromatin/physiology , T-Lymphocytes/physiology , Animals , Cell Differentiation , Humans , Repressor Proteins/physiology , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL