Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
Nat Immunol ; 20(1): 73-85, 2019 01.
Article in English | MEDLINE | ID: mdl-30538336

ABSTRACT

γδ T cells that produce the cytokine IL-17 (Tγδ17 cells) are innate-like mediators of immunity that undergo effector programming in the thymus. While regulators of Tγδ17 specialization restricted to various Vγ subsets are known, a commitment factor essential to all Tγδ17 cells has remained undefined. In this study, we identified the transcription factor c-Maf as a universal regulator of Tγδ17 cell differentiation and maintenance. Maf deficiency caused an absolute lineage block at the immature CD24+CD45RBlo γδ thymocyte stage, which revealed a critical checkpoint in the acquisition of effector functions. Here, c-Maf enforced Tγδ17 cell identity by promoting chromatin accessibility and expression of key type 17 program genes, notably Rorc and Blk, while antagonizing the transcription factor TCF1, which promotes interferon-γ-producing γδ T cells (Tγδ1 cells). Furthermore, γδ T cell antigen receptor (γδTCR) signal strength tuned c-Maf expression, which indicates that c-Maf is a core node that connects γδTCR signals to Tγδ17 cell transcriptional programming.


Subject(s)
Interleukin-17/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th17 Cells/physiology , Thymocytes/physiology , Animals , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Immunity, Innate , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Proto-Oncogene Proteins c-maf/genetics , Signal Transduction , src-Family Kinases/genetics
4.
Nat Immunol ; 18(3): 274-282, 2017 03.
Article in English | MEDLINE | ID: mdl-28135253

ABSTRACT

Although invariant Vα14+ natural killer T cells (NKT cells) are thought to be generated from CD4+CD8+ double-positive (DP) thymocytes, the developmental origin of CD4-CD8- double-negative (DN) NKT cells still remains unresolved. Here we provide definitive genetic evidence obtained, through studies of mice with DP-stage-specific ablation of expression of the gene encoding the recombinase component RAG-2 (Rag2) and by a fate-mapping approach, that supports the proposal of the existence of an alternative developmental pathway through which a fraction of DN NKT cells with strong T-helper-type-1 (TH1)-biased and cytotoxic characteristics develop from late DN-stage thymocytes, bypassing the DP stage. These findings provide new insight into understanding of the development of NKT cells and propose a role for timing of expression of the invariant T cell antigen receptor in determining the functional properties of NKT cells.


Subject(s)
Natural Killer T-Cells/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/physiology , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , DNA-Binding Proteins/genetics , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/immunology
5.
Nat Immunol ; 16(5): 517-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25848867

ABSTRACT

Lethal-7 (let-7) microRNAs (miRNAs) are the most abundant miRNAs in the genome, but their role in developing thymocytes is unclear. We found that let-7 miRNAs targeted Zbtb16 mRNA, which encodes the lineage-specific transcription factor PLZF, to post-transcriptionally regulate PLZF expression and thereby the effector functions of natural killer T cells (NKT cells). Dynamic upregulation of let-7 miRNAs during the development of NKT thymocytes downregulated PLZF expression and directed their terminal differentiation into interferon-γ (IFN-γ)-producing NKT1 cells. Without upregulation of let-7 miRNAs, NKT thymocytes maintained high PLZF expression and terminally differentiated into interleukin 4 (IL-4)-producing NKT2 cells or IL-17-producing NKT17 cells. Upregulation of let-7 miRNAs in developing NKT thymocytes was signaled by IL-15, vitamin D and retinoic acid. Such targeting of a lineage-specific transcription factor by miRNA represents a previously unknown level of developmental regulation in the thymus.


Subject(s)
Cytokines/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Natural Killer T-Cells/physiology , Thymocytes/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cytotoxicity, Immunologic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Promyelocytic Leukemia Zinc Finger Protein , Protein Binding , RNA Processing, Post-Transcriptional , Tretinoin/metabolism , Up-Regulation , Vitamin D/metabolism
6.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34323272

ABSTRACT

During positive selection at the transition from CD4+CD8+ double-positive (DP) to single-positive (SP) thymocyte, TCR signalling results in appropriate MHC restriction and signals for survival and progression. We show that the pioneer transcription factors Foxa1 and Foxa2 are required to regulate RNA splicing during positive selection of mouse T cells and that Foxa1 and Foxa2 have overlapping/compensatory roles. Conditional deletion of both Foxa1 and Foxa2 from DP thymocytes reduced positive selection and development of CD4SP, CD8SP and peripheral naïve CD4+ T cells. Foxa1 and Foxa2 regulated the expression of many genes encoding splicing factors and regulators, including Mbnl1, H1f0, Sf3b1, Hnrnpa1, Rnpc3, Prpf4b, Prpf40b and Snrpd3. Within the positively selecting CD69+DP cells, alternative RNA splicing was dysregulated in the double Foxa1/Foxa2 conditional knockout, leading to >850 differentially used exons. Many genes important for this stage of T-cell development (Ikzf1-3, Ptprc, Stat5a, Stat5b, Cd28, Tcf7) and splicing factors (Hnrnpab, Hnrnpa2b1, Hnrnpu, Hnrnpul1, Prpf8) showed multiple differentially used exons. Thus, Foxa1 and Foxa2 are required during positive selection to regulate alternative splicing of genes essential for T-cell development, and, by also regulating splicing of splicing factors, they exert widespread control of alternative splicing.


Subject(s)
Alternative Splicing/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , RNA Splicing/genetics , Thymocytes/physiology , Animals , Exons/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Splicing Factors/genetics , T-Lymphocytes/physiology , Thymus Gland/physiology
7.
Nat Immunol ; 13(8): 761-9, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772404

ABSTRACT

Thymopoiesis depends on the recruitment and expansion of bone marrow-derived progenitor populations; tight regulation of these processes is required for maintenance of the homeostasis of the T lineage. Lyl-1, a transcription factor that regulates hematopoietic progenitors, is expressed in thymocyte progenitors until T cell commitment. Here we demonstrate a requirement for Lyl-1 in lymphoid specification and the maintenance of early T lineage progenitors (ETPs). Lyl-1 deficiency resulted in profound defects in the generation of lymphoid-primed multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs) and ETPs. Lyl-1-deficient ETPs and thymocyte progenitors at the CD4(-)CD8(-) double-negative 2 (DN2) stage showed more apoptosis, blocked differentiation and impaired population expansion. We identified Gfi1 as a critical transcriptional target of Lyl-1-mediated lymphopoiesis of T cells. Thus, Lyl-1 is a pivotal component of a transcriptional program that controls the lymphoid specification and maintenance of ETPs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Lymphoid Progenitor Cells/physiology , Lymphopoiesis , Neoplasm Proteins/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/physiology , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Lineage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lymphoid Progenitor Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , T-Lymphocytes/physiology , Thymocytes/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Proc Natl Acad Sci U S A ; 117(51): 32574-32583, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288689

ABSTRACT

It is known that a subpopulation of T cells expresses two T cell receptor (TCR) clonotypes, though the extent and functional significance of this is not established. To definitively evaluate dual TCRα cells, we generated mice with green fluorescent protein and red fluorescent protein reporters linked to TCRα, revealing that ∼16% of T cells express dual TCRs, notably higher than prior estimates. Importantly, dual TCR expression has functional consequences, as dual TCR cells predominated response to lymphocytic choriomeningitis virus infection, comprising up to 60% of virus-specific CD4+ and CD8+ T cells during acute responses. Dual receptor expression selectively influenced immune memory, as postinfection memory CD4+ populations contained significantly increased frequencies of dual TCR cells. These data reveal a previously unappreciated contribution of dual TCR cells to the immune repertoire and highlight their potential effects on immune responses.


Subject(s)
Genes, T-Cell Receptor alpha/physiology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , CD5 Antigens/immunology , CD5 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chlorocebus aethiops , Female , Gene Expression , Green Fluorescent Proteins/genetics , Immunologic Memory/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymocytes/immunology , Thymocytes/physiology , Vero Cells
9.
J Immunol ; 205(7): 1830-1841, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32839237

ABSTRACT

The ß-catenin/Wnt signaling pathway plays an important role in all stages of T cell development. Nemo-like kinase (NLK) is an evolutionary conserved serine/threonine kinase and a negative regulator of the Wnt signaling pathway. NLK can directly phosphorylate histone deacetylase 1 (HDAC1), as well as T cell factor/lymphoid enhancer-binding factor (TCF/LEF), causing subsequent repression of target gene transcription. By engineering mice lacking NLK in early stages of T cell development, we set out to characterize the role NLK plays in T cell development and found that deletion of NLK does not affect mouse health or lymphoid tissue development. Instead, these mice harbored a reduced number of single-positive (SP) CD8+ thymocytes without any defects in the SP CD4+ thymocyte population. The decrease in SP CD8+ thymocytes was not caused by a block in differentiation from double-positive CD4+CD8+ cells. Neither TCR signaling nor activation was altered in the absence of NLK. Instead, we observed a significant increase in cell death and reduced phosphorylation of LEF1 as well as HDAC1 among NLK-deleted SP CD8+ cells. Thus, NLK seems to play an important role in the survival of CD8+ thymocytes. Our data provide evidence for a new function for NLK with regard to its involvement in T cell development and supporting survival of SP CD8+ thymocytes.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocyte Subsets/physiology , Thymocytes/physiology , Animals , Cell Differentiation , Cell Survival , Histone Deacetylase 1/metabolism , Lymphocyte Activation , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Wnt Proteins/metabolism
10.
J Immunol ; 205(1): 133-142, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32434937

ABSTRACT

It is becoming increasingly clear that unconventional T cell subsets, such as NKT, γδ T, mucosal-associated invariant T, and CD8αα T cells, each play distinct roles in the immune response. Subsets of these cell types can lack both CD4 and CD8 coreceptor expression. Beyond these known subsets, we identify CD4-CD8-TCRαß+, double-negative (DN) T cells, in mouse secondary lymphoid organs. DN T cells are a unique unconventional thymic-derived T cell subset. In contrast to CD5high DN thymocytes that preferentially yield TCRαß+ CD8αα intestinal lymphocytes, we find that mature CD5low DN thymocytes are precursors to peripheral DN T cells. Using reporter mouse strains, we show that DN T cells transit through the immature CD4+CD8+ (double-positive) thymocyte stage. Moreover, we provide evidence that DN T cells can differentiate in MHC-deficient mice. Our study demonstrates that MHC-independent thymic selection can yield DN T cells that are distinct from NKT, γδ T, mucosal-associated invariant T, and CD8αα T cells.


Subject(s)
Cell Differentiation/immunology , Major Histocompatibility Complex/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Female , Flow Cytometry , Male , Mice , Mice, Knockout , Models, Animal , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Thymocytes/physiology , Thymus Gland/cytology , Thymus Gland/physiology
11.
J Immunol ; 204(12): 3248-3261, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32358021

ABSTRACT

Thymocyte differentiation is a highly complex process that is accompanied by epigenetic changes. Ubiquitin-like containing PHD ring finger 1 (UHRF1) is a critical epigenetic modifier involved in various cellular processes. In this study, we demonstrated that it is highly expressed in T cell precursors of the thymus. Further, its deficiency results in significantly reduced thymocyte cellularity and thymus size in mice. Through systematic analysis based on single-cell RNA sequencing, we found that UHRF1 deficiency thwarts αß T cell lineage development, whereas biasing γδ T lineage differentiation dampens the progression of immature single-positive cells. UHRF1 deficiency promotes the IL-17 secreting and RORγt expression in γδ T cell, indicating a Tγδ17 phenotype. Further, the analysis of gene-regulatory networks demonstrated that UHRF1 controls the expression of early growth response 1 (EGR1). UHRF1 interacts with DNA methyltransferase 1 (DNMT1) at the CpG promoter region of Egr1 loci and affects the nearby chromatin modifications of H3K9me3 and H3K4me3. Taken together, our results demonstrate that UHRF1 is a key factor that mediates the epigenetic regulation of EGR1 and, consequently, thymocyte fate decisions.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Early Growth Response Protein 1/genetics , Epigenesis, Genetic/genetics , Thymocytes/physiology , Ubiquitin-Protein Ligases/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Gene Expression Regulation/genetics , Histones/genetics , Interleukin-17/genetics , Intraepithelial Lymphocytes/physiology , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Promoter Regions, Genetic/genetics , Thymus Gland/physiology
12.
Proc Natl Acad Sci U S A ; 116(37): 18537-18543, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451631

ABSTRACT

Deletion or Treg cell differentiation are alternative fates of autoreactive MHCII-restricted thymocytes. How these different modes of tolerance determine the size and composition of polyclonal cohorts of autoreactive T cells with shared specificity is poorly understood. We addressed how tolerance to a naturally expressed autoantigen of the central nervous system shapes the CD4 T cell repertoire. Specific cells in the tolerant peripheral repertoire either were Foxp3+ or displayed anergy hallmarks and, surprisingly, were at least as frequent as in the nontolerant repertoire. Despite this apparent lack of deletional tolerance, repertoire inventories uncovered that some T cell receptors (TCRs) were lost from the CD4 T cell pool, whereas others mediated Treg cell differentiation. The antigen responsiveness of these TCRs supported an affinity model of central tolerance. Importantly, the contribution of different diverter TCRs to the nascent thymic Treg cell population reflected their antigen reactivity rather than their frequency among precursors. This reveals a multilayered TCR hierarchy in CD4 T cell tolerance that separates deleted and diverted TCRs and assures that the Treg cell compartment is filled with cells of maximal permissive antigen reactivity.


Subject(s)
Autoantigens/immunology , Cell Differentiation/immunology , Clonal Deletion/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/genetics , Autoantigens/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Rearrangement, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/immunology , Myelin Proteolipid Protein/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/metabolism , Thymocytes/physiology
13.
Trends Immunol ; 39(2): 86-98, 2018 02.
Article in English | MEDLINE | ID: mdl-29162323

ABSTRACT

As they differentiate, thymocytes encounter spatially restricted cues critical for differentiation and selection of a functional, self-tolerant T cell repertoire. Sequential migration of developing T cells through distinct thymic microenvironments is enforced by the ordered expression of chemokine receptors. Herein, we provide an updated perspective on T cell differentiation through the lens of recent advances that illuminate the dynamics of chemokine-driven thymocyte migration, localization, and interactions with stromal cells. We consider these findings in the context of earlier groundwork exploring the contribution of chemokines to T cell development, recent advances regarding the specificity of chemokine signaling, and novel techniques for evaluating the T cell repertoire. We suggest future research should amalgamate visualization of localized cellular interactions with downstream molecular signals.


Subject(s)
Chemokines/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/physiology , Thymocytes/physiology , Thymus Gland/immunology , Animals , Cell Communication , Cell Differentiation , Cell Movement , Clonal Selection, Antigen-Mediated , Humans , Immune Tolerance
14.
Immunity ; 37(5): 840-53, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23159226

ABSTRACT

Proliferation and differentiation are tightly coordinated to produce an appropriate number of differentiated cells and often exhibit an antagonistic relationship. Developing T cells, which arise in the thymus from a minute number of bone-marrow-derived progenitors, undergo a major expansion upon pre-T cell receptor (TCR) expression. The burst of proliferation coincides with differentiation toward the αß T cell lineage-but the two processes were previously thought to be independent from one another, although both were driven by signaling from pre-TCR and Notch receptors. Here we report that proliferation at this step was not only absolutely required for differentiation but also that its ectopic activation was sufficient to substantially rescue differentiation in the absence of Notch signaling. Consistently, pharmacological inhibition of the cell cycle machinery also blocked differentiation in vivo. Thus the proliferation step is strictly required prior to differentiation of immature thymocytes.


Subject(s)
T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Division/immunology , Cell Division/physiology , Cell Growth Processes/physiology , Cell Lineage , Cells, Cultured , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Notch/immunology , Receptors, Notch/metabolism , Signal Transduction/immunology , Signal Transduction/physiology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/physiology , Transcription Factors/immunology , Transcription Factors/metabolism
15.
J Immunol ; 202(12): 3434-3446, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31068388

ABSTRACT

Development of lymphoid progenitors requires a coordinated regulation of gene expression, DNA replication, and gene rearrangement. Chromatin-remodeling activities directed by SWI/SNF2 superfamily complexes play important roles in these processes. In this study, we used a conditional knockout mouse model to investigate the role of Smarca5, a member of the ISWI subfamily of such complexes, in early lymphocyte development. Smarca5 deficiency results in a developmental block at the DN3 stage of αß thymocytes and pro-B stage of early B cells at which the rearrangement of Ag receptor loci occurs. It also disturbs the development of committed (CD73+) γδ thymocytes. The αß thymocyte block is accompanied by massive apoptotic depletion of ß-selected double-negative DN3 cells and premitotic arrest of CD4/CD8 double-positive cells. Although Smarca5-deficient αß T cell precursors that survived apoptosis were able to undergo a successful TCRß rearrangement, they exhibited a highly abnormal mRNA profile, including the persistent expression of CD44 and CD25 markers characteristic of immature cells. We also observed that the p53 pathway became activated in these cells and that a deficiency of p53 partially rescued the defect in thymus cellularity (in contrast to early B cells) of Smarca5-deficient mice. However, the activation of p53 was not primarily responsible for the thymocyte developmental defects observed in the Smarca5 mutants. Our results indicate that Smarca5 plays a key role in the development of thymocytes undergoing ß-selection, γδ thymocytes, and also B cell progenitors by regulating the transcription of early differentiation programs.


Subject(s)
Adenosine Triphosphatases/metabolism , B-Lymphocytes/physiology , Chromosomal Proteins, Non-Histone/metabolism , Lymphoid Progenitor Cells/physiology , T-Lymphocytes/physiology , Thymocytes/physiology , Adenosine Triphosphatases/genetics , Animals , Cell Differentiation , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Clonal Selection, Antigen-Mediated , Gene Rearrangement , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Ecotoxicol Environ Saf ; 207: 111272, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-32927162

ABSTRACT

Tobacco smoke is a common global environmental pollutant. Maternal tobacco smoke/nicotine exposure has long-term toxic effects on immune organs. We previously found that prenatal nicotine exposure (PNE)-induced programmed immune diseases caused by fetal thymic hypoplasia, but the mechanism still unknown. Autophagy has important functions in maintaining thymopoiesis, whether autophagy was involved in PNE-inhibited fetal thymocytes development is also obscure. Therefore, this study aimed to investigate how nicotine changed the development of fetal thymocytes from the perspective of autophagy in vivo and in vitro. PNE model was established by 3 mg/kg nicotine administration in Balb/c mice from gestational day 9 to 18. The results showed that PNE reduced the percentage and absolute number of CD69-CD4+SP cells, suggesting a block of fetal thymocytes mature. PNE promoted autophagosome formation, autophagy related proteins (Beclin1, LC3I/II) expression, and upregulated α7 nAChR as well as AMPK phosphorylation in fetal thymus. Moreover, PNE promoted Bcl10 degradation via autophagy-mediated proteolysis and inhibited p65 activation, blocking the transition of thymocytes between the DP to SP stage. Further, primary thymocytes were treated with nicotine in vitro and showed induced autophagy in a dose- and time-dependent manner. In addition, nicotine-inhibited CD69-CD4+SP cells and the Bcl10/p-p65 pathway have been reversed by an autophagy inhibitor. The α7 nAChR specific antagonist abrogated nicotine-induced AMPK phosphorylation and autophagy initiation. In conclusion, our findings showed that PNE repressed the Bcl10/p-p65 development pathway of CD4+SP cells by triggering autophagy, and illuminated the developmental origin mechanism of programmed immune diseases in PNE offspring.


Subject(s)
Hazardous Substances/toxicity , Nicotine/toxicity , Thymocytes/physiology , Animals , Autophagy/drug effects , B-Cell CLL-Lymphoma 10 Protein , Beclin-1 , Female , Fetus , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Pregnancy , Prenatal Exposure Delayed Effects , Thymocytes/drug effects , Thymocytes/immunology , Vitamins
17.
Immunol Rev ; 277(1): 9-20, 2017 05.
Article in English | MEDLINE | ID: mdl-28462532

ABSTRACT

The differentiation of hematopoietic precursors into the many functionally distinct T-cell types produced by the thymus is a complex process. It proceeds through a series of stages orchestrated by a variety of thymic microenvironments that shape the T-cell developmental processes. Numerous cytokine and cell surface receptors direct thymocyte differentiation but the primary determinant of cell fate is the engagement of the T-cell antigen receptor (TCR). The strength of the TCR signal and the maturation stage of the thymocyte receiving it can direct the various differentiation programs or, alternatively, end the process by inducing cell death. The regulation of thymocyte death is critical for the efficiency of thymic T-cell differentiation and the preservation of immune tolerance. A detailed knowledge of mechanisms that eliminate thymocytes from the T-cell repertoire is essential to understand the "logic" of T-cell selection in the thymus. This review focuses on the central role of the BCL-2 family of proteins in the apoptotic checkpoints that punctuate thymocyte differentiation and the consequences of defects in these processes.


Subject(s)
Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Thymus Gland/immunology , Animals , Cell Death , Cell Differentiation , Cellular Microenvironment , Central Tolerance , Hematopoiesis , Humans , Receptors, Antigen, T-Cell/metabolism
18.
J Membr Biol ; 253(4): 343-356, 2020 08.
Article in English | MEDLINE | ID: mdl-32725429

ABSTRACT

The effect of the natural saponin glycyrrhizic acid (GA) and polysaccharide arabinogalactan (AG) on the transmembrane potential of rat thymocytes was investigated using the potential-sensitive fluorescent probe 4-(p-dimethylaminostyryl)-1-methylpyridinium (DSM). Incubation of cells with GA in micellar form resulted in a decrease of the amplitude of observed fluorescence kinetics that points out to a decrease of the transmembrane potential. The proposed mechanism is an increase of membrane ion permeability (passive ion transport) of the plasma cell membrane due to GA incorporation. The incorporation of GA molecules into the cell membrane is extremely sensitive to the degree of GA dissociation. The neutral form of glycyrrhizic acid enters the lipid bilayer in contrast to the deprotonated anionic form. The incubation of rat thymocytes with anionic form of GA, namely with its disodium salt, has no effect on the fluorescence kinetics. The possible reasons of this phenomenon are discussed in the light of the nuclear magnetic resonance (NMR) and molecular dynamics (MD) data. The treatment of thymocytes with AG affects only the initial rate of the probe incorporation. The proposed mechanism is that AG covers the surface of the cell membrane and forms a barrier for the probe. Additionally, our experiments demonstrated that both polysaccharide AG and GA in the neutral form (but not Na2GA) effectively capture the cationic probe in an aqueous solution and then deliver it to the cell membrane.


Subject(s)
Galactans/pharmacology , Glycyrrhizic Acid/pharmacology , Membrane Potentials/drug effects , Thymocytes/drug effects , Thymocytes/physiology , Animals , Cell Membrane Permeability/drug effects , Fluorescent Dyes , Galactans/chemistry , Glycyrrhizic Acid/chemistry , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Male , Molecular Conformation , Molecular Dynamics Simulation , Rats
19.
Int Immunol ; 31(4): 211-223, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30561621

ABSTRACT

Sphingomyelin (SM) in combination with cholesterol forms specialized membrane lipid microdomains in which specific receptors and signaling molecules are localized or recruited to mediate intracellular signaling. SM-microdomain levels in mouse thymus were low in the early CD4+CD8+ double-positive (DP) stage prior to thymic selection and increased >10-fold during late selection. T-cell receptor (TCR) signal strength is a key factor determining whether DP thymocytes undergo positive or negative selection. We examined the role of SM-microdomains in thymocyte development and related TCR signaling, using SM synthase 1 (SMS1)-deficient (SMS1-/-) mice which display low SM expression in all thymocyte populations. SMS1 deficiency caused reduced cell numbers after late DP stages in TCR transgenic models. TCR-dependent apoptosis induced by anti-CD3 treatment was enhanced in SMS1-/- DP thymocytes both in vivo and in vitro. SMS1-/- DP thymocytes, relative to controls, showed increased phosphorylation of TCR-proximal kinase ZAP-70 and increased expression of Bim and Nur77 proteins involved in negative selection following TCR stimulation. Addition of SM to cultured normal DP thymocytes led to greatly increased surface expression of SM-microdomains, with associated reduction of TCR signaling and TCR-induced apoptosis. Our findings indicate that SM-microdomains are increased in late DP stages, function as negative regulators of TCR signaling and modulate the efficiency of TCR-proximal signaling to promote thymic selection events leading to subsequent developmental stages.


Subject(s)
Cell Membrane/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Apoptosis , Cell Differentiation , Cells, Cultured , Female , Immunomodulation , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transferases (Other Substituted Phosphate Groups)/genetics
20.
Trends Immunol ; 38(9): 622-632, 2017 09.
Article in English | MEDLINE | ID: mdl-28697966

ABSTRACT

THEMIS, a recently identified T-lineage-restricted protein, is the founding member of a large metazoan protein family. Gene inactivation studies have revealed a critical requirement for THEMIS during thymocyte positive selection, implicating THEMIS in signaling downstream of the T cell antigen receptor (TCR), but the mechanistic underpinnings of THEMIS function have remained elusive. A previous model posited that THEMIS prevents thymocytes from inappropriately crossing the positive/negative selection threshold by dampening TCR signaling. However, new data suggest an alternative model where THEMIS enhances TCR signaling enabling thymocytes to reach the threshold for positive selection, avoiding death by neglect. We review the data supporting each model and conclude that the preponderance of evidence favors an enhancing function for THEMIS in TCR signaling.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Models, Immunological , T-Lymphocytes/physiology , Thymocytes/physiology , Animals , Cell Differentiation , Cysteine/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation , Protein Domains/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL