Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 557
Filter
1.
Nat Commun ; 15(1): 7784, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39237503

ABSTRACT

The structural components of the thymus are essential for guiding T cell development, but a thorough spatial view is still absent. Here we develop the TSO-his tool, designed to integrate multimodal data from single-cell and spatial transcriptomics to decipher the intricate structure of human thymus. Specifically, we characterize dynamic changes in cell types and critical markers, identifying ELOVL4 as a mediator of CD4+ T cell positive selection in the cortex. Utilizing the mapping function of TSO-his, we reconstruct thymic spatial architecture at single-cell resolution and recapitulates classical cell types and their essential co-localization for T cell development; additionally, previously unknown co-localization relationships such as that of CD8αα with memory B cells and monocytes are identified. Incorporating VDJ sequencing data, we also delineate distinct intermediate thymocyte states during αß T cell development. Overall, these insights enhance our understanding of thymic biology and may inform therapeutic interventions targeting T cell-mediated immune responses.


Subject(s)
Single-Cell Analysis , Thymocytes , Thymus Gland , Transcriptome , Humans , Thymocytes/metabolism , Thymocytes/cytology , Single-Cell Analysis/methods , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/immunology , Gene Expression Profiling/methods , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Membrane Proteins/metabolism , Membrane Proteins/genetics , Multiomics
2.
Front Immunol ; 15: 1376655, 2024.
Article in English | MEDLINE | ID: mdl-39328409

ABSTRACT

To further understand the impact of deficiency of the autoimmune regulator (Aire) gene during the adhesion of medullary thymic epithelial cells (mTECs) to thymocytes, we sequenced single-cell libraries (scRNA-seq) obtained from Aire wild-type (WT) (Airewt/wt ) or Aire-deficient (Airewt/mut ) mTECs cocultured with WT single-positive (SP) CD4+ thymocytes. Although the libraries differed in their mRNA and long noncoding RNA (lncRNA) profiles, indicating that mTECs were heterogeneous in terms of their transcriptome, UMAP clustering revealed that both mTEC lines expressed their specific markers, i.e., Epcam, Itgb4, Itga6, and Casp3 in resting mTECs and Ccna2, Pbk, and Birc5 in proliferative mTECs. Both cocultured SP CD4+ thymocytes remained in a homogeneous cluster expressing the Il7r and Ccr7 markers. Comparisons of the two types of cocultures revealed the differential expression of mRNAs that encode transcription factors (Zfpm2, Satb1, and Lef1), cell adhesion genes (Itgb1) in mTECs, and Themis in thymocytes, which is associated with the regulation of positive and negative selection. At the single-cell sequencing resolution, we observed that Aire acts on both Aire WT and Aire-deficient mTECs as an upstream controller of mRNAs, which encode transcription factors or adhesion proteins that, in turn, are posttranscriptionally controlled by lncRNAs, for example, Neat1, Malat1, Pvt1, and Dancr among others. Under Aire deficiency, mTECs dysregulate the expression of MHC-II, CD80, and CD326 (EPCAM) protein markers as well as metabolism and cell cycle-related mRNAs, which delay the cell cycle progression. Moreover, when adhered to mTECs, WT SP CD4+ or CD8+ thymocytes modulate the expression of cell activation proteins, including CD28 and CD152/CTLA4, and the expression of cellular metabolism mRNAs. These findings indicate a complex mechanism through which an imbalance in Aire expression can affect mTECs and thymocytes during adhesion.


Subject(s)
AIRE Protein , Cell Adhesion , Epithelial Cells , RNA, Long Noncoding , Thymocytes , Transcription Factors , Transcriptome , RNA, Long Noncoding/genetics , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Mice , Thymocytes/metabolism , Thymocytes/immunology , Thymocytes/cytology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Single-Cell Analysis , Gene Regulatory Networks , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Coculture Techniques , Gene Expression Profiling , Mice, Knockout
3.
Immunol Lett ; 269: 106904, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39117004

ABSTRACT

Thymic epithelial cells participate in the maturation and selection of T lymphocytes. This review explores recent insights from single-cell sequencing regarding classifying thymic epithelial cells in both normal and neoplastic thymus. Cortical thymic epithelial cells facilitate thymocyte differentiation and contribute to positive selection. Medullary epithelial cells are distinguished by their expression of AIRE. Cells progress from a pre-AIRE state, containing precursors with cortical and medullary characteristics, termed junctional cells. Mature medullary epithelial cells exhibit promiscuous gene expression and after that downregulate AIRE mRNA. Post-AIRE cells can adopt a Hassall corpuscle-like phenotype or exhibit distinctive differentiation characteristics including tuft cells, ionocytes, neuroendocrine cells, and myoid cells.


Subject(s)
Cell Differentiation , Epithelial Cells , Single-Cell Analysis , Thymus Gland , Transcription Factors , Humans , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/immunology , Epithelial Cells/metabolism , Single-Cell Analysis/methods , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , AIRE Protein , Thymocytes/metabolism , Thymocytes/cytology , Thymocytes/immunology
4.
Elife ; 132024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158947

ABSTRACT

The fate of developing T cells is determined by the strength of T cell receptor (TCR) signal they receive in the thymus. This process is finely regulated through the tuning of positive and negative regulators in thymocytes. The Family with sequence similarity 49 member B (Fam49b) protein is a newly discovered negative regulator of TCR signaling that has been shown to suppress Rac-1 activity in vitro in cultured T cell lines. However, the contribution of Fam49b to the thymic development of T cells is unknown. To investigate this important issue, we generated a novel mouse line deficient in Fam49b (Fam49b-KO). We observed that Fam49b-KO double positive (DP) thymocytes underwent excessive negative selection, whereas the positive selection stage was unaffected. Fam49b deficiency impaired the survival of single positive thymocytes and peripheral T cells. This altered development process resulted in significant reductions in CD4 and CD8 single-positive thymocytes as well as peripheral T cells. Interestingly, a large proportion of the TCRγδ+ and CD8αα+TCRαß+ gut intraepithelial T lymphocytes were absent in Fam49b-KO mice. Our results demonstrate that Fam49b dampens thymocytes TCR signaling in order to escape negative selection during development, uncovering the function of Fam49b as a critical regulator of the selection process to ensure normal thymocyte development and peripheral T cells survival.


Subject(s)
Receptors, Antigen, T-Cell , Signal Transduction , Thymocytes , Animals , Mice , Cell Survival , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Thymocytes/metabolism , Thymocytes/cytology
5.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38980292

ABSTRACT

In the vertebrate immune system, thymus stromal microenvironments support the generation of αßT cells from immature thymocytes. Thymic epithelial cells are of particular importance, and the generation of cortical and medullary epithelial lineages from progenitor stages controls the initiation and maintenance of thymus function. Here, we discuss the developmental pathways that regulate thymic epithelial cell diversity during both the embryonic and postnatal periods. We also examine how thymus microenvironments respond to injury, with particular focus on mechanisms that ensure regeneration of thymic epithelial cells for the restoration of thymus function.


Subject(s)
Epithelial Cells , Thymus Gland , Thymus Gland/cytology , Thymus Gland/immunology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Animals , Humans , Cell Differentiation , Regeneration/physiology , Thymocytes/cytology , Thymocytes/metabolism , Thymocytes/immunology
6.
Dev Cell ; 59(19): 2643-2658.e7, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38981469

ABSTRACT

Mitochondria and endoplasmic reticulum contacts (MERCs) control multiple cellular processes, including cell survival and differentiation. Based on the observations that MERCs were specifically enriched in the CD4-CD8- double-negative (DN) stage, we studied their role in early mouse thymocyte development. We found that T cell-specific knockout of Hspa9, which encodes GRP75, a protein that mediates MERC formation by assembling the IP3R-GRP75-VDAC complex, impaired DN3 thymocyte viability and resulted in thymocyte developmental arrest at the DN3-DN4 transition. Mechanistically, GRP75 deficiency induced mitochondrial stress, releasing mitochondrial DNA (mtDNA) into the cytosol and triggering the type I interferon (IFN-I) response. The IFN-I pathway contributed to both the impairment of cell survival and DN3-DN4 transition blockage, while increased lipid peroxidation (LPO) played a major role downstream of IFN-I. Thus, our study identifies the essential role of GRP75-dependent MERCs in early thymocyte development and the governing facts of cell survival and differentiation in the DN stage.


Subject(s)
Cell Differentiation , Cell Survival , Endoplasmic Reticulum , HSP70 Heat-Shock Proteins , Mitochondria , Thymocytes , Animals , Mice , Mitochondria/metabolism , Thymocytes/metabolism , Thymocytes/cytology , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Mice, Knockout , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Interferon Type I/metabolism
7.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38980291

ABSTRACT

During thymic development, thymocytes adjust their TCR response based on the strength of their reactivity to self-peptide MHC complexes. This tuning process allows thymocytes with a range of self-reactivities to survive positive selection and contribute to a diverse T cell pool. In this review, we will discuss recent advances in our understanding of how thymocytes tune their responsiveness during positive selection, and we present a "sequential selection" model to explain how MHC specificity influences lineage choice. We also discuss recent evidence for cell type diversity in the medulla and discuss how this heterogeneity may contribute to medullary niches for negative selection and regulatory T cell development.


Subject(s)
Cell Lineage , T-Lymphocytes, Regulatory , Thymus Gland , Animals , Thymus Gland/immunology , Thymus Gland/cytology , Humans , T-Lymphocytes, Regulatory/immunology , Cell Lineage/immunology , Cell Differentiation/immunology , CD8-Positive T-Lymphocytes/immunology , Thymocytes/immunology , Thymocytes/cytology , Thymocytes/metabolism , CD4-Positive T-Lymphocytes/immunology
8.
Oncogene ; 43(34): 2535-2547, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38907003

ABSTRACT

Malignant transformation of T-cell progenitors causes T-cell acute lymphoblastic leukemia (T-ALL), an aggressive childhood lymphoproliferative disorder. Activating mutations of Notch, Notch1 and Notch3, have been detected in T-ALL patients. In this study, we aimed to deeply characterize hyperactive Notch3-related pathways involved in T-cell dynamics within the thymus and bone marrow to propose these processes as an important step in facilitating the progression of T-ALL. We previously generated a transgenic T-ALL mouse model (N3-ICtg) demonstrating that aberrant Notch3 signaling affects early thymocyte maturation programs and leads to bone marrow infiltration by CD4+CD8+ (DP) T cells that are notably, Notch3highCXCR4high. Newly, our in vivo results suggest that an anomalous immature thymocyte subpopulation, such as CD4-CD8- (DN) over-expressing CD3ɛ, but with low CXCR4 expression, dominates N3-ICtg thymus-resident DN subset in T-ALL progression. MicroRNAs might be of significance in T-ALL pathobiology, however, whether required for leukemia maintenance is not fully understood. The selection of specific DN subsets demonstrates the inverse correlation between CXCR4 expression and a panel of Notch3-deregulated miRNAs. Interestingly, we found that within DN thymocyte subset hyperactive Notch3 inhibits CXCR4 expression through the cooperative effects of miR-139-5p and miR-150-5p, thus impinging on thymocyte differentiation with accumulation of DNCD3ɛ+CXCR4- cells. These data point out that deregulation of Notch3 in T-ALL, besides its role in sustaining dissemination of abnormal DP T cells, as we previously demonstrated, could play a role in selecting specific DN immature T cells within the thymus, thus impeding T cell development, to facilitate T-ALL progression inside the bone marrow.


Subject(s)
Disease Progression , MicroRNAs , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptor, Notch3 , Receptors, CXCR4 , Thymocytes , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Thymocytes/metabolism , Thymocytes/cytology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Mice , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Humans , Mice, Transgenic , Signal Transduction , Cell Differentiation/genetics
9.
Immunol Lett ; 267: 106861, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697225

ABSTRACT

Hematopoietic precursors (HPCs) entering into the thymus undergo a sequential process leading to the generation of a variety of T cell subsets. This developmental odyssey unfolds in distinct stages within the thymic cortex and medulla, shaping the landscape of T cell receptor (TCR) expression and guiding thymocytes through positive and negative selection. Initially, early thymic progenitors (ETPs) take residence in the thymic cortex, where thymocytes begin to express their TCR and undergo positive selection. Subsequently, thymocytes transition to the thymic medulla, where they undergo negative selection. Both murine and human thymocyte development can be broadly classified into distinct stages based on the expression of CD4 and CD8 coreceptors, resulting in categorizations as double negative (DN), double positive (DP) or single positive (SP) cells. Thymocyte migration to the appropriate thymic microenvironment at the right differentiation stage is pivotal for the development and the proper functioning of T cells, which is critical for adaptive immune responses. The journey of lymphoid progenitor cells into the T cell developmental pathway hinges on an ongoing dialogue between the differentiating cell and the signals emanating from the thymus niche. Herein, we review the contribution of the key factors mentioned above for the localization, migration and emigration of thymocytes.


Subject(s)
Cell Differentiation , Cell Movement , Thymocytes , Thymus Gland , Thymocytes/immunology , Thymocytes/cytology , Thymocytes/metabolism , Animals , Humans , Thymus Gland/cytology , Thymus Gland/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Receptors, Antigen, T-Cell/metabolism
11.
Turk J Pediatr ; 65(1): 73-80, 2023.
Article in English | MEDLINE | ID: mdl-36866987

ABSTRACT

BACKGROUND: Early diagnosis and effective treatment serve as life-saving procedures for primary immunodeficiencies (PIDs) which are very common and a major public health problem in Turkey. Severe combined immunodeficiency (SCID) is constitutively a T-cell defect in which naïve T-cell development is defective due to the mutations in genes responsible for the T cell differentiation and insufficient thymopoiesis. So, assessment of thymopoiesis is very important in the diagnosis of SCID and several combined immune deficiencies (CIDs). METHODS: The purpose of this study is to examine thymopoiesis in healthy children via measurement of recent thymic emigrants (RTE); T lymphocytes that express CD4, CD45RA and CD31 to establish the RTE reference values in Turkish children. RTE were measured in the peripheral blood (PB) of 120 healthy infants and children between 0-6 years including cord blood samples, by flow cytometry. RESULTS: The absolute count of RTE cells and their relative ratios were found to be higher during the first year of life, being highest at the 6th month and tending to decrease significantly by age following birth (p=0.001). In the cord blood group, both values were lower than those in the 6-month-old group. The absolute lymphocyte count (ALC) varying by age, was found to reduce to 1850/mm³ in 4-years and after. CONCLUSIONS: Here we evaluated normal thymopoiesis and established the normal reference levels of RTE cells in the peripheral blood of healthy children aged between 0-6 years. We believe that the collected data will contribute to early diagnosis and monitoring of immune reconstitution; serving as an additional fast and reliable marker for many PID patients especially for SCID including many other CIDs, especially in nations where newborn screening (NBS) via T cell receptor excision circles (TREC) has not yet become available.


Subject(s)
T-Lymphocytes , Thymocytes , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Fetal Blood , Leukocyte Common Antigens , Mutation , Turkey/epidemiology , Thymocytes/cytology , T-Lymphocytes/cytology , Reference Values
12.
Nature ; 613(7944): 565-574, 2023 01.
Article in English | MEDLINE | ID: mdl-36410718

ABSTRACT

Programming T cells to distinguish self from non-self is a vital, multi-step process that occurs in the thymus1-4. Signalling through the pre-T cell receptor (preTCR), a CD3-associated heterodimer comprising an invariant pTα chain and a clone-specific ß chain, is a critical early checkpoint in thymocyte development within the αß T cell lineage5,6. PreTCRs arrayed on CD4-CD8- double-negative thymocytes ligate peptides bound to major histocompatibility complex molecules (pMHC) on thymic stroma, similar to αß T cell receptors that appear on CD4+CD8+ double-positive thymocytes, but via a different molecular docking strategy7-10. Here we show the consequences of these distinct interactions for thymocyte progression using synchronized fetal thymic progenitor cultures that differ in the presence or absence of pMHC on support stroma, and single-cell transcriptomes at key thymocyte developmental transitions. Although major histocompatibility complex (MHC)-negative stroma fosters αß T cell differentiation, the absence of preTCR-pMHC interactions leads to deviant thymocyte transcriptional programming associated with dedifferentiation. Highly proliferative double-negative and double-positive thymocyte subsets emerge, with antecedent characteristics of T cell lymphoblastic and myeloid malignancies. Compensatory upregulation of diverse MHC class Ib proteins in B2m/H2-Ab1 MHC-knockout mice partially safeguards in vivo thymocyte progression, although disseminated double-positive thymic tumours may develop with ageing. Thus, as well as promoting ß chain repertoire broadening for subsequent αß T cell receptor utilization, preTCR-pMHC interactions limit cellular plasticity to facilitate normal thymocyte differentiation and proliferation that, if absent, introduce developmental vulnerabilities.


Subject(s)
Cell Dedifferentiation , Histocompatibility Antigens Class I , Receptors, Antigen, T-Cell , Thymocytes , Animals , Mice , Mice, Knockout , Molecular Docking Simulation , Peptides/immunology , Peptides/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism
13.
Eur J Immunol ; 52(8): 1228-1242, 2022 08.
Article in English | MEDLINE | ID: mdl-35491946

ABSTRACT

ICAP-1 regulates ß1-integrin activation and cell adhesion. Here, we used ICAP-1-null mice to study ICAP-1 potential involvement during immune cell development and function. Integrin α4ß1-dependent adhesion was comparable between ICAP-1-null and control thymocytes, but lack of ICAP-1 caused a defective single-positive (SP) CD8+ cell generation, thus, unveiling an ICAP-1 involvement in SP thymocyte development. ICAP-1 bears a nuclear localization signal and we found it displayed a strong nuclear distribution in thymocytes. Interestingly, there was a direct correlation between the lack of ICAP-1 and reduced levels in SP CD8+ thymocytes of Runx3, a transcription factor required for CD8+ thymocyte generation. In the spleen, ICAP-1 was found evenly distributed between cytoplasm and nuclear fractions, and ICAP-1-/- spleen T and B cells displayed upregulation of α4ß1-mediated adhesion, indicating that ICAP-1 negatively controls their attachment. Furthermore, CD3+ - and CD19+ -selected spleen cells from ICAP-1-null mice showed reduced proliferation in response to T- and B-cell stimuli, respectively. Finally, loss of ICAP-1 caused a remarkable decrease in marginal zone B- cell frequencies and a moderate increase in follicular B cells. Together, these data unravel an ICAP-1 involvement in the generation of SP CD8+ thymocytes and in the control of marginal zone B-cell numbers.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes , CD8-Positive T-Lymphocytes , Lymphocyte Activation , Thymocytes , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Integrin beta1/metabolism , Mice , Mice, Knockout , Spleen/cytology , Thymocytes/cytology , Thymus Gland/cytology
14.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055071

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulated T and B lymphocytes. Type I interferons (IFN-I) have been shown to play important pathogenic roles in both SLE patients and mouse models of lupus. Recent studies have shown that B cell intrinsic responses to IFN-I are enough to drive B cell differentiation into autoantibody-secreting memory B cells and plasma cells, although lower levels of residual auto-reactive cells remain present. We speculated that IFN-I stimulation of T cells would similarly drive specific T-cell associated lupus phenotypes including the upregulation of T follicular helper cells and Th17, thereby affecting autoantibody production and the development of glomerulonephritis. Using the B6.Nba2 mouse model of lupus, we evaluated disease parameters in T cell specific IFN-I receptor (IFNAR)-deficient mice (cKO). Surprisingly, all measured CD4+ T cell abnormalities and associated intra-splenic cytokine levels (IFNγ, IL-6, IL-10, IL-17, IL-21) were unchanged and thus independent of IFN-I. In contrast B6.Nba2 cKO mice displayed reduced levels of effector CD8+ T cells and increased levels of Foxp3+ CD8+ regulatory T cells, suggesting that IFN-I induced signaling specifically affecting CD8+ T cells. These data suggest a role for both pathogenic and immunosuppressive CD8+ T cells in Nba2-driven autoimmunity, providing a model to further evaluate the role of these cell subsets during lupus-like disease development in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Receptor, Interferon alpha-beta/metabolism , Animals , Antibodies, Antinuclear/immunology , Autoimmunity , Biomarkers , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cytokines , Disease Models, Animal , Germinal Center/immunology , Germinal Center/metabolism , Immunohistochemistry , Immunophenotyping , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Organ Specificity/immunology , Phenotype , Receptor, Interferon alpha-beta/genetics , Splenomegaly , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
15.
Nat Commun ; 13(1): 457, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075189

ABSTRACT

Maldevelopment of the pharyngeal endoderm, an embryonic tissue critical for patterning of the pharyngeal region and ensuing organogenesis, ultimately contributes to several classes of human developmental syndromes and disorders. Such syndromes are characterized by a spectrum of phenotypes that currently cannot be fully explained by known mutations or genetic variants due to gaps in characterization of critical drivers of normal and dysfunctional development. Despite the disease-relevance of pharyngeal endoderm, we still lack a comprehensive and integrative view of the molecular basis and gene regulatory networks driving pharyngeal endoderm development. To close this gap, we apply transcriptomic and chromatin accessibility single-cell sequencing technologies to generate a multi-omic developmental resource spanning pharyngeal endoderm patterning to the emergence of organ-specific epithelia in the developing mouse embryo. We identify cell-type specific gene regulation, distill GRN models that define developing organ domains, and characterize the role of an immunodeficiency-associated forkhead box transcription factor.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Developmental , Pharynx/embryology , Transcriptome , Animals , Chromatin/metabolism , Endoderm/embryology , Endoderm/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Organogenesis , Pharynx/metabolism , Single-Cell Analysis , Thymocytes/cytology , Thymocytes/metabolism
16.
J Immunol ; 208(2): 384-395, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34937744

ABSTRACT

CD4+CD25+FOXP3+ regulatory T (Treg) cells control immunological tolerance. Treg cells are generated in the thymus (tTreg) or in the periphery. Their superior lineage fidelity makes tTregs the preferred cell type for adoptive cell therapy (ACT). How human tTreg cells develop is incompletely understood. By combining single-cell transcriptomics and flow cytometry, we in this study delineated three major Treg developmental stages in the human thymus. At the first stage, which we propose to name pre-Treg I, cells still express lineage-inappropriate genes and exhibit signs of TCR signaling, presumably reflecting recognition of self-antigen. The subsequent pre-Treg II stage is marked by the sharp appearance of transcription factor FOXO1 and features induction of KLF2 and CCR7, in apparent preparation for thymic exit. The pre-Treg II stage can further be refined based on the sequential acquisition of surface markers CD31 and GPA33. The expression of CD45RA, finally, completes the phenotype also found on mature recent thymic emigrant Treg cells. Remarkably, the thymus contains a substantial fraction of recirculating mature effector Treg cells, distinguishable by expression of inflammatory chemokine receptors and absence of CCR7. The developmental origin of these cells is unclear and warrants caution when using thymic tissue as a source of stable cells for ACT. We show that cells in the major developmental stages can be distinguished using the surface markers CD1a, CD27, CCR7, and CD39, allowing for their viable isolation. These insights help identify fully mature tTreg cells for ACT and can serve as a basis for further mechanistic studies into tTreg development.


Subject(s)
Cell Differentiation/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymocytes/cytology , Thymus Gland/cytology , Cells, Cultured , Child, Preschool , Forkhead Box Protein O1/metabolism , Humans , Immune Tolerance/immunology , Kruppel-Like Transcription Factors/metabolism , Leukocyte Common Antigens/metabolism , Membrane Glycoproteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA-Seq/methods , Receptors, CCR7/metabolism , Single-Cell Analysis , Thymus Gland/immunology , Transcriptome/genetics , Exome Sequencing
17.
Sci Rep ; 11(1): 21550, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732767

ABSTRACT

Salt Inducible Kinases (SIKs), of which there are 3 isoforms, are established to play roles in innate immunity, metabolic control and neuronal function, but their role in adaptive immunity is unknown. To address this gap, we used a combination of SIK knockout and kinase-inactive knock-in mice. The combined loss of SIK1 and SIK2 activity did not block T cell development. Conditional knockout of SIK3 in haemopoietic cells, driven by a Vav-iCre transgene, resulted in a moderate reduction in the numbers of peripheral T cells, but normal B cell numbers. Constitutive knockout of SIK2 combined with conditional knockout of SIK3 in the haemopoietic cells resulted in a severe reduction in peripheral T cells without reducing B cell number. A similar effect was seen when SIK3 deletion was driven via CD4-Cre transgene to delete at the DP stage of T cell development. Analysis of the SIK2/3 Vav-iCre mice showed that thymocyte number was greatly reduced, but development was not blocked completely as indicated by the presence of low numbers CD4 and CD8 single positive cells. SIK2 and SIK3 were not required for rearrangement of the TCRß locus, or for low level cell surface expression of the TCR complex on the surface of CD4/CD8 double positive thymocytes. In the absence of both SIK2 and SIK3, progression to mature single positive cells was greatly reduced, suggesting a defect in negative and/or positive selection in the thymus. In agreement with an effect on negative selection, increased apoptosis was seen in thymic TCRbeta high/CD5 positive cells from SIK2/3 knockout mice. Together, these results show an important role for SIK2 and SIK3 in thymic T cell development.


Subject(s)
Protein Serine-Threonine Kinases/genetics , T-Lymphocytes/cytology , Alleles , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Cell Separation , Exons , Female , Flow Cytometry , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Isoforms , Signal Transduction/physiology , Spleen/metabolism , Thymocytes/cytology , Transgenes
18.
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
19.
Scand J Immunol ; 94(4): e13094, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34780092

ABSTRACT

The thymus produces self-limiting and self-tolerant T cells through the interaction between thymocytes and thymus epithelial cells (TECs), thereby generating central immune tolerance. The TECs are composed of cortical and medullary thymic epithelial cells, which regulate the positive and negative selection of T cells, respectively. During the process of negative selection, thymocytes with self-reactive ability are deleted or differentiated into regulatory T cells (Tregs). Tregs are a subset of suppressor T cells that are important for maintaining immune homeostasis. The differentiation and development of Tregs depend on the development of TECs and other underlying molecular mechanisms. Tregs regulated by thymic epithelial cells are closely related to human health and are significant in autoimmune diseases, thymoma and pregnancy. In this review, we summarize the current molecular and transcriptional regulatory mechanisms by which TECs affect the development and function of thymic Tregs. We also review the pathophysiological models of thymic epithelial cells regulating thymic Tregs in human diseases and specific physiological conditions.


Subject(s)
T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Epithelial Cells/classification , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Homeostasis , Humans , Male , Models, Immunological , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Pregnancy , Signal Transduction/immunology , T-Lymphocytes, Regulatory/classification , Thymocytes/classification , Thymocytes/cytology , Thymocytes/immunology , Thymoma/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Neoplasms/immunology
20.
Sci Rep ; 11(1): 22595, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799646

ABSTRACT

The nuclear receptors liver X receptor α (LXRα) and LXRß are lipid sensors that regulate lipid metabolism and immunity. Natural killer T (NKT) cells, a T cell subset expressing surface markers of both natural killer cells and T lymphocytes and involved in antitumor immunity, are another abundant immune cell type in the liver. The potential function of the metabolic regulators LXRα/ß in hepatic NKT cells remains unknown. In this study, we examined the role of LXRα and LXRß in NKT cells using mice deficient for LXRα and/or LXRß, and found that hepatic invariant NKT (iNKT) cells are drastically decreased in LXRα/ß-KO mice. Cytokine production stimulated by the iNKT cell activator α-galactosylceramide was impaired in LXRα/ß-KO hepatic mononuclear cells and in LXRα/ß-KO mice. iNKT cell-mediated antitumor effect was also disturbed in LXRα/ß-KO mice. LXRα/ß-KO mice transplanted with wild-type bone marrow showed decreased iNKT cells in the liver and spleen. The thymus of LXRα/ß-KO mice showed a decreased population of iNKT cells. In conclusion, LXRα and LXRß are essential for NKT cell-mediated immunity, such as cytokine production and hepatic antitumor activity, and are involved in NKT cell development in immune tissues, such as the thymus.


Subject(s)
Antineoplastic Agents/pharmacology , Killer Cells, Natural/metabolism , Liver X Receptors/metabolism , Liver/drug effects , Animals , Bone Marrow/metabolism , Bone Marrow Transplantation , Cytokines/metabolism , Galactosylceramides/metabolism , Leukocytes, Mononuclear/metabolism , Liver/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Spleen/cytology , Spleen/metabolism , Thymocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL