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











Publication year range
1.
Elife ; 112022 02 21.
Article in English | MEDLINE | ID: mdl-35188458

ABSTRACT

Interactions of developing T cells with Aire+ medullary thymic epithelial cells expressing high levels of MHCII molecules (mTEChi) are critical for the induction of central tolerance in the thymus. In turn, thymocytes regulate the cellularity of Aire+ mTEChi. However, it remains unknown whether thymocytes control the precursors of Aire+ mTEChi that are contained in mTEClo cells or other mTEClo subsets that have recently been delineated by single-cell transcriptomic analyses. Here, using three distinct transgenic mouse models, in which antigen presentation between mTECs and CD4+ thymocytes is perturbed, we show by high-throughput RNA-seq that self-reactive CD4+ thymocytes induce key transcriptional regulators in mTEClo and control the composition of mTEClo subsets, including Aire+ mTEChi precursors, post-Aire and tuft-like mTECs. Furthermore, these interactions upregulate the expression of tissue-restricted self-antigens, cytokines, chemokines, and adhesion molecules important for T-cell development. This gene activation program induced in mTEClo is combined with a global increase of the active H3K4me3 histone mark. Finally, we demonstrate that these self-reactive interactions between CD4+ thymocytes and mTECs critically prevent multiorgan autoimmunity. Our genome-wide study thus reveals that self-reactive CD4+ thymocytes control multiple unsuspected facets from immature stages of mTECs, which determines their heterogeneity.


Subject(s)
Autoantigens/physiology , Epithelial Cells/physiology , Thymocytes/physiology , Thymus Gland , Animals , CD4-Positive T-Lymphocytes , DNA-Binding Proteins , Epithelium/physiology , Female , Gene Expression Profiling , Gene Expression Regulation , Histones , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins , Signal Transduction
2.
J Biosci ; 462021.
Article in English | MEDLINE | ID: mdl-34815373

ABSTRACT

The prolactin hormone (PRL), in addition to its known effects on breast development and lactation, exerts effects on the immune system, including pleiotropic effects on the thymus. The aim of this study was to evaluate the influence of PRL on the epithelial compartment of the thymus. Thymic epithelial cells (TECs) (2BH4 cells) and fresh thymocytes were used. Immunofluorescence assay revealed that PRL treatment (10 ng/ mL) increases the deposition of laminin and expression of the chemokine CXCL12 in 2BH4 cells. However, no change was observed in the deposition of fibronectin. Moreover, PRL altered F-actin polymerisation, allowing the formation of focal adhesion complexes in treated cells. When 2BH4 cells were pre-treated with PRL, thymocyte adhesion was not altered. However, in the cell migration assay, pre-treatment with PRL potentiated the chemotactic effect of CXCL12 on the migration of total, double-positive, CD4-positive, and CD8-positive thymocytes. Together, the results of this study demonstrate the effect of PRL on thymic epithelial cells, particularly on CXCL12-driven thymocyte migration, confirming that this hormone is a regulator of thymic physiology.


Subject(s)
Chemokine CXCL12/metabolism , Prolactin/pharmacology , Thymocytes/cytology , Thymus Gland/cytology , Actins/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Epithelial Cells , Extracellular Matrix/drug effects , Female , Fibronectins/metabolism , Male , Mice, Inbred C57BL , Thymocytes/drug effects , Thymocytes/physiology
3.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34726730

ABSTRACT

Analysis of the transcriptional profiles of developing thymocytes has shown that T lineage commitment is associated with loss of stem cell and early progenitor gene signatures and the acquisition of T cell gene signatures. Less well understood are the epigenetic alterations that accompany or enable these transcriptional changes. Here, we show that the histone demethylase Lsd1 (Kdm1a) performs a key role in extinguishing stem/progenitor transcriptional programs in addition to key repressive gene programs during thymocyte maturation. Deletion of Lsd1 caused a block in late T cell development and resulted in overexpression of interferon response genes as well as genes regulated by the Gfi1, Bcl6, and, most prominently, Bcl11b transcriptional repressors in CD4+CD8+ thymocytes. Transcriptional overexpression in Lsd1-deficient thymocytes was not always associated with increased H3K4 trimethylation at gene promoters, indicating that Lsd1 indirectly affects the expression of many genes. Together, these results identify a critical function for Lsd1 in the epigenetic regulation of multiple repressive gene signatures during T cell development.


Subject(s)
Epigenesis, Genetic , Histone Demethylases/genetics , T-Lymphocytes/physiology , Thymocytes/cytology , Animals , Cell Lineage/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Mice, Mutant Strains , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-6/genetics , Repressor Proteins/genetics , Thymocytes/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
4.
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
5.
Nat Commun ; 12(1): 3933, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168132

ABSTRACT

Thymic T cell development and T cell receptor repertoire selection are dependent on essential molecular cues provided by thymic epithelial cells (TEC). TEC development and function are regulated by their epigenetic landscape, in which the repressive H3K27me3 epigenetic marks are catalyzed by polycomb repressive complex 2 (PRC2). Here we show that a TEC-targeted deficiency of PRC2 function results in a hypoplastic thymus with reduced ability to express antigens and select a normal repertoire of T cells. The absence of PRC2 activity reveals a transcriptomically distinct medullary TEC lineage that incompletely off-sets the shortage of canonically-derived medullary TEC whereas cortical TEC numbers remain unchanged. This alternative TEC development is associated with the generation of reduced TCR diversity. Hence, normal PRC2 activity and placement of H3K27me3 marks are required for TEC lineage differentiation and function and, in their absence, the thymus is unable to compensate for the loss of a normal TEC scaffold.


Subject(s)
Epigenesis, Genetic , Epithelial Cells/cytology , Polycomb Repressive Complex 2/genetics , Thymus Gland/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Lineage , Epithelial Cells/physiology , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 2/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Thymocytes/cytology , Thymocytes/physiology , Thymus Gland/physiology
6.
Cell Death Dis ; 12(6): 526, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34023853

ABSTRACT

Thymic atrophy in sepsis is a critical disadvantage because it induces immunosuppression and increases the mortality rate as the disease progresses. However, the exact mechanism of thymic atrophy has not been fully elucidated. In this study, we discovered a novel role for VSIG4-positive peritoneal macrophages (V4(+) cells) as the principal cells that induce thymic atrophy and thymocyte apoptosis. In CLP-induced mice, V4(+) cells were activated after ingestion of invading microbes, and the majority of these cells migrated into the thymus. Furthermore, these cells underwent a phenotypic shift from V4(+) to V4(-) and from MHC II(low) to MHC II(+). In coculture with thymocytes, V4(+) cells mainly induced apoptosis in DP thymocytes via the secretion of TNF-α. However, there was little effect on CD4 or CD8 SP and DN thymocytes. V4(-) cells showed low levels of activity compared to V4(+) cells. Thymic atrophy in CLP-induced V4(KO) mice was much less severe than that in CLP-induced wild-type mice. In addition, V4(KO) peritoneal macrophages also showed similar activity to V4(-) cells. Taken together, the current study demonstrates that V4(+) cells play important roles in inducing immunosuppression via thymic atrophy in the context of severe infection. These data also suggest that controlling the function of V4(+) cells may play a crucial role in the development of new therapies to prevent thymocyte apoptosis in sepsis.


Subject(s)
Macrophages, Peritoneal/physiology , Receptors, Complement/metabolism , Sepsis/pathology , Thymocytes/physiology , Animals , Apoptosis/genetics , Cecum/pathology , Cecum/surgery , Disease Models, Animal , Female , Ligation , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Receptors, Complement/genetics , Sepsis/genetics , Sepsis/metabolism , Thymocytes/metabolism , Thymocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Sci Rep ; 11(1): 10439, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001954

ABSTRACT

The thymus facilitates mature T cell production by providing a suitable stromal microenvironment. This microenvironment is impaired by radiation and aging which lead to immune system disturbances known as thymic involution. Young adult thymus shows thymic recovery after such involution. Although various genes have been reported for thymocytes and thymic epithelial cells in such processes, the roles of stromal transcription factors in these remain incompletely understood. MafB (v-maf musculoaponeurotic fibrosarcoma oncogene homolog B) is a transcription factor expressed in thymic stroma and its expression was induced a day after radiation exposure. Hence, the roles of mesenchymal MafB in the process of thymic regeneration offers an intriguing research topic also for radiation biology. The current study investigated whether MafB plays roles in the adult thymus. MafB/green fluorescent protein knock-in mutant (MafB+/GFP) mice showed impaired thymic regeneration after the sublethal irradiation, judged by reduced thymus size, total thymocyte number and medullary complexity. Furthermore, IL4 was induced after irradiation and such induction was reduced in mutant mice. The mutants also displayed signs of accelerated age-related thymic involution. Altogether, these results suggest possible functions of MafB in the processes of thymic recovery after irradiation, and maintenance during aging.


Subject(s)
MafB Transcription Factor/metabolism , Regeneration/radiation effects , Thymocytes/physiology , Thymus Gland/physiology , Aging/genetics , Animals , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Gene Expression Regulation/radiation effects , Gene Knock-In Techniques , MafB Transcription Factor/genetics , Male , Mice , Mice, Transgenic , Mutation , Regeneration/genetics , Thymocytes/radiation effects , Thymus Gland/cytology , Thymus Gland/radiation effects , Whole-Body Irradiation
8.
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
9.
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
10.
Nat Commun ; 11(1): 6264, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293517

ABSTRACT

The molecular and cellular mechanisms mediating thymic central tolerance and prevention of autoimmunity are not fully understood. Here we show that B7-CD28 co-stimulation and B7 expression by specific antigen-presenting cell (APC) types are required for clonal deletion and for regulatory T (Treg) cell generation from endogenous tissue-restricted antigen (TRA)-specific thymocytes. While B7-CD28 interaction is required for both clonal deletion and Treg induction, these two processes differ in their CD28 signaling requirements and in their dependence on B7-expressing dendritic cells, B cells, and thymic epithelial cells. Meanwhile, defective thymic clonal deletion due to altered B7-CD28 signaling results in the accumulation of mature, peripheral TRA-specific T cells capable of mediating destructive autoimmunity. Our findings thus reveal a function of B7-CD28 co-stimulation in shaping the T cell repertoire and limiting autoimmunity through both thymic clonal deletion and Treg cell generation.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Central Tolerance , Clonal Deletion , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Antigen-Presenting Cells/metabolism , Autoimmunity/physiology , CD28 Antigens/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Gene Knock-In Techniques , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/physiology , Thymus Gland/cytology , Thymus Gland/metabolism
11.
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
12.
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
13.
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
14.
Bull Exp Biol Med ; 169(1): 60-62, 2020 May.
Article in English | MEDLINE | ID: mdl-32488775

ABSTRACT

We studied the formation of proliferative response of thymic lymphocytes to T-cell mitogen in rats exposed to endocrine disrupter DDT during prenatal and postnatal ontogeny. Developmental exposure to the endocrine disruptor was found to attenuate proliferative response during puberty and adulthood due to maintenance of higher proliferation rate of thymic lymphocytes in comparison with age-matched controls. Insufficient proliferative response to mitogens in rats developmentally exposed to the endocrine disrupter increases the risk of impairment of cell-mediated reactions of adaptive immunity.


Subject(s)
Cell Proliferation/drug effects , DDT/toxicity , Growth and Development/drug effects , T-Lymphocytes/drug effects , Thymocytes/drug effects , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/immunology , Embryo, Mammalian , Endocrine Disruptors/toxicity , Female , Growth and Development/immunology , Lymphopoiesis/drug effects , Male , Mitogens/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Rats , Rats, Wistar , T-Lymphocytes/physiology , Thymocytes/physiology , Toxicity Tests
15.
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
16.
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
17.
Pharmacol Rep ; 71(6): 1079-1087, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31629088

ABSTRACT

BACKGROUND: Cell volume regulation and volume-regulated anion channels are critical for cell survival in non-isosmotic conditions, and dysregulation of this system is detrimental. Although genes and proteins underlying this basic cellular machinery were recently identified, the pharmacology remains poorly explored. METHODS: We examined effects of 16 flavonoids on the regulatory volume decrease (RVD) of thymocytes under hypoosmotic stress assessed by light transmittance and on the activity of volume-sensitive chloride channel by patch-clamp technique. RESULTS: Comparison of effects of flavonoids on RVD revealed a group of four active substances with lehmannin being the strongest inhibitor (IC50 = 8.8 µM). Structure-functional comparison suggested that hydrophobicity brought about by methoxy, prenyl or lavandulyl groups as well as by the absence of glucosyl fragment together with localization of the phenyl ring B at the position C2 (which is at C3 in totally inactive isoflavones) are important structural determinants for the flavonoids activity as volume regulation inhibitors. All active flavonoids suppressed RVD under Gramicidin D-NMDG hypotonic stress conditions when cationic permeability was increased by an ionophore, gramicidin D, with all extracellular monovalent cations replaced with bulky NMDG+ suggesting that they target volume-sensitive anionic permeability. While effects of hispidulin and pulicarin were only partial, lehmannin and pinocembrin completely abolished RVD under Gramicidin D-NMDG conditions. In direct patch-clamp experiments, lehmannin and pinocembrin produced a strong inhibiting effect on the swelling-induced whole-cell chloride conductance in a voltage-independent manner. CONCLUSION: Lehmannin, pinocembrin, and possibly hispidulin and pulicarin may serve as leads for developing effective low-toxic immunomodulators.


Subject(s)
Chloride Channels/physiology , Flavonoids/pharmacology , Osmotic Pressure/drug effects , Thymocytes/physiology , Alkaloids/pharmacology , Animals , Cell Size/drug effects , Flavanones/pharmacology , Flavonoids/chemistry , Gramicidin , Patch-Clamp Techniques , Quinolizidines/pharmacology , Rats , Thymocytes/drug effects , Thymocytes/metabolism
18.
Front Immunol ; 10: 1911, 2019.
Article in English | MEDLINE | ID: mdl-31456807

ABSTRACT

Proper orchestration of T lymphocyte development is critical, as T cells underlie nearly all responses of the adaptive immune system. Developing thymocytes differentiate in response to environmental cues carried from cell surface receptors to the nucleus, shaping a distinct transcriptional program that defines their developmental outcome. Our recent work has identified a previously undescribed role for the vacuolar ATPase (V-ATPase) in facilitating the development of murine thymocytes progressing toward the CD4+ and CD8+ αß T cell lineages. Vav1Cre recombinase-mediated deletion of the a2 isoform of the V-ATPase (a2V) in mouse hematopoietic cells leads to a specific and profound loss of peripheral CD4+ and CD8+ αß T cells. Utilizing T cell-restricted LckCre and CD4Cre strains, we further traced this deficiency to the thymus and found that a2V plays a cell-intrinsic role throughout intrathymic development. Loss of a2V manifests as a partial obstruction in the double negative stage of T cell development, and later, a near complete failure of positive selection. These data deepen our understanding of the biological mechanisms that orchestrate T cell development and lend credence to the recent focus on V-ATPase as a potential chemotherapeutic target to combat proliferative potential in T cell lymphoblastic leukemias and autoimmune disease.


Subject(s)
Lymphopoiesis , T-Lymphocytes/physiology , Thymocytes/physiology , Thymus Gland/cytology , Thymus Gland/enzymology , Vacuolar Proton-Translocating ATPases/physiology , Animals , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Female , Gene Deletion , Leukopenia/genetics , Male , Mice , Mice, Inbred C57BL , Receptor, Notch1/metabolism , Signal Transduction , Thymus Gland/immunology , Vacuolar Proton-Translocating ATPases/deficiency , Vacuolar Proton-Translocating ATPases/genetics
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL