Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 13.604
1.
Immunol Lett ; 267: 106861, 2024 Jun.
Article En | MEDLINE | ID: mdl-38697225

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.


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
2.
Proc Natl Acad Sci U S A ; 121(20): e2320268121, 2024 May 14.
Article En | MEDLINE | ID: mdl-38709934

Insulin is a central autoantigen in the pathogenesis of T1D, and thymic epithelial cell expression of insulin under the control of the Autoimmune Regulator (Aire) is thought to be a key component of maintaining tolerance to insulin. In spite of this general working model, direct detection of this thymic selection on insulin-specific T cells has been somewhat elusive. Here, we used a combination of highly sensitive T cell receptor transgenic models for detecting thymic selection and sorting and sequencing of Insulin-specific CD4+ T cells from Aire-deficient mice as a strategy to further define their selection. This analysis revealed a number of unique t cell receptor (TCR) clones in Aire-deficient hosts with high affinity for insulin/major histocompatibility complex (MHC) ligands. We then modeled the thymic selection of one of these clones in Aire-deficient versus wild-type hosts and found that this model clone could escape thymic negative selection in the absence of thymic Aire. Together, these results suggest that thymic expression of insulin plays a key role in trimming and removing high-affinity insulin-specific T cells from the repertoire to help promote tolerance.


AIRE Protein , Insulin , Receptors, Antigen, T-Cell , Thymus Gland , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clone Cells , Immune Tolerance , Insulin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/cytology , Transcription Factors/metabolism , Transcription Factors/genetics
3.
J Med Invest ; 71(1.2): 29-39, 2024.
Article En | MEDLINE | ID: mdl-38735722

The establishment of an adaptive immune system is critical for protecting our bodies from neoplastic cancers and invading pathogens such as viruses and bacteria. As a primary lymphoid organ, the thymus generates lymphoid T cells that play a major role in the adaptive immune system. T cell generation in the thymus is controlled by interactions between thymocytes and other thymic cells, primarily thymic epithelial cells. Thus, the normal development and function of thymic epithelial cells are important for the generation of immunocompetent and self-tolerant T cells. On the other hand, the degeneration of the thymic epithelium due to thymic aging causes thymic involution, which is associated with the decline of adaptive immune function. Herein we summarize basic and current knowledge of the development and function of thymic epithelial cells and the mechanism of thymic involution. J. Med. Invest. 71 : 29-39, February, 2024.


Aging , Thymus Gland , Thymus Gland/immunology , Thymus Gland/growth & development , Humans , Aging/physiology , Aging/immunology , Animals , Epithelial Cells/physiology , Epithelium/immunology , T-Lymphocytes/immunology
4.
Nat Commun ; 15(1): 4248, 2024 May 18.
Article En | MEDLINE | ID: mdl-38762584

The naked mole-rat (Heterocephalus glaber) is a long-lived rodent species showing resistance to the development of cancer. Although naked mole-rats have been reported to lack natural killer (NK) cells, γδ T cell-based immunity has been suggested in this species, which could represent an important arm of the immune system for antitumor responses. Here, we investigate the biology of these unconventional T cells in peripheral tissues (blood, spleen) and thymus of the naked mole-rat at different ages by TCR repertoire profiling and single-cell gene expression analysis. Using our own TCR annotation in the naked mole-rat genome, we report that the γδ TCR repertoire is dominated by a public invariant Vγ4-2/Vδ1-4 TCR, containing the complementary-determining-region-3 (CDR3)γ CTYWDSNYAKKLF / CDR3δ CALWELRTGGITAQLVF that are likely generated by short-homology-repeat-driven DNA rearrangements. This invariant TCR is specifically found in γδ T cells expressing genes associated with NK cytotoxicity and is generated in both the thoracic and cervical thymus of the naked mole-rat until adult life. Our results indicate that invariant Vγ4-2/Vδ1-4 NK-like effector T cells in the naked mole-rat can contribute to tumor immunosurveillance by γδ TCR-mediated recognition of a common molecular signal.


Mole Rats , Receptors, Antigen, T-Cell, gamma-delta , Thymus Gland , Animals , Mole Rats/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Thymus Gland/immunology , Thymus Gland/cytology , Killer Cells, Natural/immunology , Spleen/immunology , Complementarity Determining Regions/genetics , Natural Killer T-Cells/immunology
5.
Toxicology ; 505: 153836, 2024 Jun.
Article En | MEDLINE | ID: mdl-38768702

Caramel color is a widely used food pigment, and 2-Acetyl-4-tetrahydroxybutylimidazole (THI) is a by-products of Class III caramel color. Some studies have shown that THI can reduce the number of peripheral blood lymphocytes. However, the comprehensive mechanism of THI immunotoxicity requires further study. In this study, the effects of THI on lymphocyte count, humoral immunity, cellular immunity and nonspecific immunity were determined and the effect of the nutritional status of VB6 on THI immunotoxicity was evaluated. Female BALB/c mice were divided into 3 groups and fed chow containing different doses of VB6: VB6-normal (6 mg/kg VB6), VB6-deprived (0.5 mg/kg VB6) or VB6-enhanced (12 mg/kg VB6) feed. Each group was further divided into 4 subgroups and treated with THI (0.5, 2.5 or 12.5 mg/kg bw) or the solvent control by gavage for 30 days. The thymic cortical thickness was measured with ViewPoint; the proportions of major immune cells and T cells in peripheral blood and tissues were detected via flow cytometry; the transformation and proliferation abilities of T and B cells were detected via T and B lymphocyte proliferation assays; NK cell activity was assessed via lactate dehydrogenase assays; humoral immune function was assessed via plaque-forming cell assays; and the immune function of T lymphocytes was assessed via delayed type hypersensitivity assays. The results showed that compared with those in the corresponding control group, the white blood cell count and lymphocyte count decreased significantly in all the VB6-deprived groups, in the 2.5 and 12.5 mg/kg VB6 groups, and in the 12.5 mg/kg VB6-enhanced group. With increasing THI dose, the thymic cortical layer became thinner. In the thymus, THI increased the proportions of CD3+ T cells and mature CD8+ T cells and decreased the proportions of immature double-positive, double-negative T cells and CD69-expressing lymphocytes. The proportions of naïve T cells and Tcm (central memory T) cells related to homing decreased. The proportion of mature T cells in the spleen decreased significantly. The proliferation of T cells stimulated by ConA decreased after THI exposure. VB6-deficient mice were more sensitive to THI immunotoxicity, and supplementation with VB6 had a certain protective effect on these mice. The results of the PFC and NK cell activity assays indicated that THI exposure might not affect humoral immune or innate immune function.


Imidazoles , Immunity, Humoral , Mice, Inbred BALB C , Vitamin B 6 , Animals , Female , Mice , Imidazoles/toxicity , Imidazoles/pharmacology , Immunity, Humoral/drug effects , Vitamin B 6/pharmacology , Vitamin B 6/administration & dosage , Lymphocyte Count , Nutritional Status/drug effects , Thymus Gland/drug effects , Thymus Gland/immunology , Immunity, Cellular/drug effects , Spleen/drug effects , Spleen/immunology , Food Coloring Agents/toxicity , Cell Proliferation/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
Front Immunol ; 15: 1364957, 2024.
Article En | MEDLINE | ID: mdl-38650932

Introduction: CARD11 is a lymphoid lineage-specific scaffold protein regulating the NF-κB activation downstream of the antigen receptor signal pathway. Defective CARD11 function results in abnormal development and differentiation of lymphocytes, especially thymic regulatory T cells (Treg). Method: In this study, we used patients' samples together with transgenic mouse models carrying pathogenic CARD11 mutations from patients to explore their effects on Treg development. Immunoblotting and a GFP receptor assay were used to evaluate the activation effect of CARD11 mutants on NF-κB signaling. Then the suppressive function of Tregs carrying distinct CARD11 mutations was measured by in vitro suppression assay. Finally, we applied the retroviral transduced bone marrow chimeras to rescue the Treg development in an NF-κB independent manner. Results and discuss: We found CARD11 mutations causing hyper-activated NF-κB signals also gave rise to compromised Treg development in the thymus, similar to the phenotype in Card11 deficient mice. This observation challenges the previous view that CARD11 regulates Treg lineage dependent on the NF-kB activation. Mechanistic investigations reveal that the noncanonical function CARD11, which negatively regulates the AKT/ FOXO1 signal pathway, is responsible for regulating Treg generation. Moreover, primary immunodeficiency patients carrying CARD11 mutation, which autonomously activates NF-κB, also represented the reduced Treg population in their peripheral blood. Our results propose a new regulatory function of CARD11 and illuminate an NF-κB independent pathway for thymic Treg lineage commitment.


CARD Signaling Adaptor Proteins , Guanylate Cyclase , Mutation , NF-kappa B , Signal Transduction , T-Lymphocytes, Regulatory , Thymus Gland , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , NF-kappa B/metabolism , Humans , Mice , Thymus Gland/immunology , Thymus Gland/cytology , Thymus Gland/metabolism , Mice, Transgenic , Cell Differentiation/immunology , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/genetics , Male
7.
Cell Rep ; 43(4): 114072, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38581680

Medullary thymic epithelial cells (mTECs) are essential for the establishment of self-tolerance in T cells. Promiscuous gene expression by a subpopulation of mTECs regulated by the nuclear protein Aire contributes to the display of self-genomic products to newly generated T cells. Recent reports have highlighted additional self-antigen-displaying mTEC subpopulations, namely Fezf2-expressing mTECs and a mosaic of self-mimetic mTECs including thymic tuft cells. In addition, a functionally different subset of mTECs produces chemokine CCL21, which attracts developing thymocytes to the medullary region. Here, we report that CCL21+ mTECs and Aire+ mTECs non-redundantly cooperate to direct self-tolerance to prevent autoimmune pathology by optimizing the deletion of self-reactive T cells and the generation of regulatory T cells. We also detect cooperation for self-tolerance between Aire and Fezf2, the latter of which unexpectedly regulates thymic tuft cells. Our results indicate an indispensable interplay among functionally diverse mTECs for the establishment of central self-tolerance.


AIRE Protein , Central Tolerance , Epithelial Cells , Nerve Tissue Proteins , Thymus Gland , Transcription Factors , Animals , Epithelial Cells/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/immunology , Transcription Factors/metabolism , Transcription Factors/genetics , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Self Tolerance
8.
Cell Mol Immunol ; 21(6): 546-560, 2024 Jun.
Article En | MEDLINE | ID: mdl-38641698

γδ T cells play a crucial role in immune surveillance and serve as a bridge between innate and adaptive immunity. However, the metabolic requirements and regulation of γδ T-cell development and function remain poorly understood. In this study, we investigated the role of liver kinase B1 (Lkb1), a serine/threonine kinase that links cellular metabolism with cell growth and proliferation, in γδ T-cell biology. Our findings demonstrate that Lkb1 is not only involved in regulating γδ T lineage commitment but also plays a critical role in γδ T-cell effector function. Specifically, T-cell-specific deletion of Lkb1 resulted in impaired thymocyte development and distinct alterations in γδ T-cell subsets in both the thymus and peripheral lymphoid tissues. Notably, loss of Lkb1 inhibited the commitment of Vγ1 and Vγ4 γδ T cells, promoted the maturation of IL-17-producing Vγ6 γδ T cells, and led to the occurrence of fatal autoimmune hepatitis (AIH). Notably, clearance of γδ T cells or blockade of IL-17 significantly attenuated AIH. Mechanistically, Lkb1 deficiency disrupted metabolic homeostasis and AMPK activity, accompanied by increased mTORC1 activation, thereby causing overactivation of γδ T cells and enhanced apoptosis. Interestingly, activation of AMPK or suppression of mTORC1 signaling effectively inhibited IL-17 levels and attenuated AIH in Lkb1-deficient mice. Our findings highlight the pivotal role of Lkb1 in maintaining the homeostasis of γδ T cells and preventing IL-17-mediated autoimmune diseases, providing new insights into the metabolic programs governing the subset determination and functional differentiation of thymic γδ T cells.


AMP-Activated Protein Kinases , Hepatitis, Autoimmune , Interleukin-17 , Mice, Inbred C57BL , Protein Serine-Threonine Kinases , Receptors, Antigen, T-Cell, gamma-delta , Animals , Interleukin-17/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Mice , AMP-Activated Protein Kinases/metabolism , Mice, Knockout , Cell Differentiation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/pathology , Signal Transduction , Mechanistic Target of Rapamycin Complex 1/metabolism
9.
Immunol Lett ; 267: 106857, 2024 Jun.
Article En | MEDLINE | ID: mdl-38604551

To control immune responses, regulatory CD4+CD25+Foxp3+ T cells (Treg) maintain their wide and diverse repertoire through continuous arrival of recent thymic emigrants (RTE). However, during puberty, the activity of RTE starts to decline as a natural process of thymic involution, introducing consequences, not completely described, to the repertoire. Type 1 diabetes (T1D) patients show quantitative and qualitative impairments on the Treg cells. Our aim was to evaluate peripheral Treg and RTE cell frequencies, in T1D patients from two distinct age groups (young and adults) and verify if HLA phenotypes are concomitant associated. To this, blood samples from Brazilian twenty established T1D patients (12 young and 8 adults) and twenty-one healthy controls (11 young and 10 adults) were analyzed, by flow cytometry, to verify the percentages of CD4, Treg (CD4+CD25+Foxp3+) and the subsets of CD45RA+ (naive) and CD31+(RTE) within then. Furthermore, the HLA typing was also set. We observed that the young established T1D patients feature decreased frequencies in total Treg cells and naive RTE within Treg cells. Significant prevalence of HLA alleles, associated with risk, in T1D patients, was also identified. Performing a multivariate analysis, we confirmed that the cellular changes described offers significant variables that distinct T1D patients from the controls. Our data collectively highlight relevant aspects about homeostasis imbalances in the Treg cells of T1D patients, especially in young, and disease prognosis; that might contribute for future therapeutic strategies involving Treg cells manipulation.


Diabetes Mellitus, Type 1 , Forkhead Transcription Factors , Interleukin-2 Receptor alpha Subunit , T-Lymphocytes, Regulatory , Thymus Gland , Humans , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adult , Brazil , Male , Female , Forkhead Transcription Factors/metabolism , Thymus Gland/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Young Adult , Adolescent , Immunophenotyping , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Child
11.
J Immunol ; 212(11): 1733-1743, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38656392

The thymus is the site of T lymphocyte development and T cell education to recognize foreign, but not self, Ags. B cells also reside and develop in the thymus, although their functions are less clear. During "thymic involution," a process of lymphoid atrophy and adipose replacement linked to sexual maturation, thymocytes decline. However, thymic B cells decrease far less than T cells, such that B cells comprise ∼1% of human neonatal thymocytes but up to ∼10% in adults. All jawed vertebrates possess a thymus, and we and others have shown zebrafish (Danio rerio) also have thymic B cells. In this article, we investigated the precise identities of zebrafish thymic T and B cells and how they change with involution. We assessed the timing and specific details of zebrafish thymic involution using multiple lymphocyte-specific, fluorophore-labeled transgenic lines, quantifying the changes in thymic T- and B-lymphocytes pre- versus postinvolution. Our results prove that, as in humans, zebrafish thymic B cells increase relative to T cells postinvolution. We also performed RNA sequencing on D. rerio thymic and marrow lymphocytes of four novel double-transgenic lines, identifying distinct populations of immature T and B cells. Collectively, this is, to our knowledge, the first comprehensive analysis of zebrafish thymic involution, demonstrating its similarity to human involution and establishing the highly genetically manipulatable zebrafish model as a template for involution studies.


B-Lymphocytes , Thymus Gland , Zebrafish , Animals , Zebrafish/immunology , Thymus Gland/immunology , Thymus Gland/cytology , B-Lymphocytes/immunology , Animals, Genetically Modified , T-Lymphocytes/immunology , Humans , Cell Differentiation/immunology , Models, Animal
12.
Oncologist ; 29(6): 473-483, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38520743

Thymic epithelial tumors (TETs) are rare tumors for which treatment options are limited. The ongoing need for improved systemic therapies reflects a limited understanding of tumor biology as well as the normal thymus. The essential role of the thymus in adaptive immunity is largely effected by its epithelial compartment, which directs thymocyte (T-cell) differentiation and immunologic self-tolerance. With aging, the thymus undergoes involution whereby epithelial tissue is replaced by adipose and other connective tissue, decreasing immature T-cell production. Against this natural drive toward involution, a fraction of thymuses will instead undergo oncologic transformation, leading to the formation of TETs, including thymoma and thymic carcinoma. The rarity of these tumors restricts investigation of the mechanisms of tumorigenesis and development of rational treatment options. To this end, the development of technologies which allow deep molecular profiling of individual tumor cells permits a new window through which to view normal thymic development and contrast the malignant changes that result in oncogenic transformation. In this review, we describe the findings of recent illuminating studies on the diversity of cell types within the epithelial compartment through thymic differentiation and aging. We contextualize these findings around important unanswered questions regarding the spectrum of known somatic tumor alterations, cell of origin, and tumor heterogeneity. The perspectives informed by single-cell molecular profiling offer new approaches to clinical and basic investigation of thymic epithelial tumors, with the potential to accelerate development of improved therapeutic strategies to address ongoing unmet needs in these rare tumors.


Neoplasms, Glandular and Epithelial , Thymus Gland , Thymus Neoplasms , Humans , Thymus Neoplasms/pathology , Thymus Gland/pathology , Thymus Gland/immunology , Neoplasms, Glandular and Epithelial/pathology , Single-Cell Analysis/methods , Cell Differentiation
13.
Neuroimmunomodulation ; 31(1): 78-88, 2024.
Article En | MEDLINE | ID: mdl-38527434

BACKGROUND: The brain and the immune systems represent the two primary adaptive systems within the body. Both are involved in a dynamic process of communication, vital for the preservation of mammalian homeostasis. This interplay involves two major pathways: the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system. SUMMARY: The establishment of infection can affect immunoneuroendocrine interactions, with functional consequences for immune organs, particularly the thymus. Interestingly, the physiology of this primary organ is not only under the control of the central nervous system (CNS) but also exhibits autocrine/paracrine regulatory circuitries mediated by hormones and neuropeptides that can be altered in situations of infectious stress or chronic inflammation. In particular, Chagas disease, caused by the protozoan parasite Trypanosoma cruzi (T. cruzi), impacts upon immunoneuroendocrine circuits disrupting thymus physiology. Here, we discuss the most relevant findings reported in relation to brain-thymic connections during T. cruzi infection, as well as their possible implications for the immunopathology of human Chagas disease. KEY MESSAGES: During T. cruzi infection, the CNS influences thymus physiology through an intricate network involving hormones, neuropeptides, and pro-inflammatory cytokines. Despite some uncertainties in the mechanisms and the fact that the link between these abnormalities and chronic Chagasic cardiomyopathy is still unknown, it is evident that the precise control exerted by the brain over the thymus is markedly disrupted throughout the course of T. cruzi infection.


Brain , Chagas Disease , Thymus Gland , Humans , Chagas Disease/immunology , Chagas Disease/physiopathology , Animals , Brain/immunology , Thymus Gland/immunology , Thymus Gland/physiology , Trypanosoma cruzi/physiology , Trypanosoma cruzi/immunology , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Neuroimmunomodulation/physiology , Neuroimmunomodulation/immunology , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/physiopathology , Pituitary-Adrenal System/metabolism
14.
Immunohorizons ; 8(3): 281-294, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38551395

Inhibitory proteins, such as programmed cell death protein 1 (PD-1), have been studied extensively in peripheral T cell responses to foreign Ags, self-Ags, and neoantigens. Notably, these proteins are first expressed during T cell development in the thymus. Reports suggest that PD-1 limits regulatory T cell (Treg) development, but the mechanism by which PD-1 exerts this function remains unknown. The present study expands the evaluation of murine PD-1 and its ligands in the thymus, demonstrating that some of the highest expressers of PD-1 and programmed death-ligand 1 are agonist selected cells. Surprisingly, we reveal a selective role for PD-1 in regulating the developmental niche only for Tregs because other agonist selected cell populations, such as NK T cells, remain unchanged. We also ruled out PD-1 as a regulator of proliferation or cell death of agonist selected Tregs and further demonstrated that PD-1-deficient Tregs have reduced TCR signaling. Unexpectedly, the data suggest that PD-1-deficient thymocytes produce elevated levels of IL-2, a Treg niche-limiting cytokine. Collectively, these data suggest a novel role for PD-1 in regulating IL-2 production and the concurrent agonist selection of murine thymic Tregs. This observation has implications for the use of checkpoint blockade in the context of cancer and infection.


Interleukin-2 , Programmed Cell Death 1 Receptor , T-Lymphocytes, Regulatory , Thymus Gland , Animals , Mice , Cytokines/metabolism , Interleukin-2/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology
15.
Eur J Immunol ; 54(5): e2350450, 2024 May.
Article En | MEDLINE | ID: mdl-38356202

The Wiskott-Aldrich syndrome protein (WASp) regulates actin cytoskeletal dynamics and function of hematopoietic cells. Mutations in the WAS gene lead to two different syndromes; Wiskott-Aldrich syndrome (WAS) caused by loss-of-function mutations, and X-linked neutropenia (XLN) caused by gain-of-function mutations. We previously showed that WASp-deficient mice have a decreased number of regulatory T (Treg) cells in the thymus and the periphery. We here evaluated the impact of WASp mutations on Treg cells in the thymus of WAS and XLN mouse models. Using in vitro Treg differentiation assays, WAS CD4 single-positive thymocytes have decreased differentiation to Treg cells, despite normal early signaling upon IL-2 and TGF-ß stimulation. They failed to proliferate and express CD25 at high levels, leading to poor survival and a lower number of Foxp3+ Treg cells. Conversely, XLN CD4 single-positive thymocytes efficiently differentiate into Foxp3+ Treg cells following a high proliferative response to IL-2 and TGF-ß, associated with high CD25 expression when compared with WT cells. Altogether, these results show that specific mutations of WASp affect Treg cell development differently, demonstrating a critical role of WASp activity in supporting Treg cell development and expansion.


Cell Differentiation , Cell Proliferation , T-Lymphocytes, Regulatory , Thymus Gland , Wiskott-Aldrich Syndrome Protein , Animals , T-Lymphocytes, Regulatory/immunology , Cell Differentiation/immunology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Mice , Thymus Gland/immunology , Thymus Gland/cytology , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Interleukin-2/metabolism , Interleukin-2/immunology , Mutation , Transforming Growth Factor beta/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Mice, Knockout , Mice, Inbred C57BL
16.
Nature ; 627(8003): 407-415, 2024 Mar.
Article En | MEDLINE | ID: mdl-38383779

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Aquaporin 4 , Autoantibodies , Autoantigens , B-Lymphocytes , Immune Tolerance , Neuromyelitis Optica , Animals , Humans , Mice , AIRE Protein , Aquaporin 4/deficiency , Aquaporin 4/genetics , Aquaporin 4/immunology , Aquaporin 4/metabolism , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/immunology , Germinal Center/cytology , Germinal Center/immunology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thyroid Epithelial Cells/immunology , Thyroid Epithelial Cells/metabolism , Transcriptome
17.
J Pharm Sci ; 113(6): 1572-1579, 2024 Jun.
Article En | MEDLINE | ID: mdl-38237668

Poor stability and difficult uptake of natural polysaccharides have been the main problems in their application. The purpose of this study was to optimize the preparation conditions of Polygonatum cyrtonema Hua polysaccharides liposomes (PCPL) and to investigate the immune enhancement activity of PCPL in vitro and in vivo, with a view to discovering new ways of natural polysaccharide application. The optimal preparation conditions of PCPL were as follows: the adding amount of Tween 80 of 0.5 %, the ultrasound time of 2 min and the ultrasound times of once. Under these conditions, the entrapment efficiency, drug loading rate and particle size of PCPL were 38.033 %±0.050, 2.172 %±0.003 and 146 nm, which indicated that PCPL with small particle size could be prepared by the reverse-phase evaporation method. Furthermore, PCPL promoted proliferation, phagocytosis, and secretion of nitric oxide and related cytokines in RAW264.7 cells. Moreover, PCPL improved spleen and thymus indices, increased the number or proportion of red blood cells, platelets, and lymphocytes in the blood, and ameliorated spleen and thymus atrophy in immunosuppressed mice. This study provides a new idea for applying Polygonatum cyrtonema Hua polysaccharides (PCP) and references for studying other polysaccharides.


Liposomes , Phagocytosis , Polygonatum , Polysaccharides , Animals , Mice , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polygonatum/chemistry , RAW 264.7 Cells , Phagocytosis/drug effects , Particle Size , Spleen/drug effects , Spleen/immunology , Nitric Oxide/metabolism , Thymus Gland/drug effects , Thymus Gland/immunology , Cell Proliferation/drug effects , Cytokines/metabolism , Male
18.
Science ; 382(6670): 534-541, 2023 11 03.
Article En | MEDLINE | ID: mdl-37917689

CD8 T cell tolerance is thought to result from clonal deletion of autoreactive thymocytes before they differentiate into mature CD8 T cells in the thymus. However, we report that, in mice, CD8 T cell tolerance instead results from premature thymic eviction of immature autoreactive CD8 thymocytes into the periphery, where they differentiate into self-tolerant mature CD8 T cells. Premature thymic eviction is triggered by T cell receptor (TCR)-driven down-regulation of the transcriptional repressor Gfi1, which induces expression of sphingosine-1-phosphate receptor-1 (S1P1) on negatively selected immature CD8 thymocytes. Thus, premature thymic eviction is the basis for CD8 T cell tolerance and is the mechanism responsible for the appearance in the periphery of mature CD8 T cells bearing autoreactive TCRs that are absent from the thymus.


CD8-Positive T-Lymphocytes , Clonal Deletion , Peripheral Tolerance , Thymus Gland , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/metabolism , Male , Female
19.
Nature ; 623(7988): 803-813, 2023 Nov.
Article En | MEDLINE | ID: mdl-37938781

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Autoantibodies , Genetic Predisposition to Disease , Interferon Type I , NF-kappa B , Humans , Autoantibodies/immunology , COVID-19/genetics , COVID-19/immunology , Gain of Function Mutation , Heterozygote , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Loss of Function Mutation , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p52 Subunit/deficiency , NF-kappa B p52 Subunit/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology , Thymus Gland/pathology , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , AIRE Protein , NF-kappaB-Inducing Kinase
20.
Nature ; 622(7981): 164-172, 2023 Oct.
Article En | MEDLINE | ID: mdl-37674082

Development of immunocompetent T cells in the thymus is required for effective defence against all types of pathogens, including viruses, bacteria and fungi. To this end, T cells undergo a very strict educational program in the thymus, during which both non-functional and self-reactive T cell clones are eliminated by means of positive and negative selection1.Thymic epithelial cells (TECs) have an indispensable role in these processes, and previous studies have shown the notable heterogeneity of these cells2-7. Here, using multiomic analysis, we provide further insights into the functional and developmental diversity of TECs in mice, and reveal a detailed atlas of the TEC compartment according to cell transcriptional states and chromatin landscapes. Our analysis highlights unconventional TEC subsets that are similar to functionally well-defined parenchymal populations, including endocrine cells, microfold cells and myocytes. By focusing on the endocrine and microfold TEC populations, we show that endocrine TECs require Insm1 for their development and are crucial to maintaining thymus cellularity in a ghrelin-dependent manner; by contrast, microfold TECs require Spib for their development and are essential for the generation of thymic IgA+ plasma cells. Collectively, our study reveals that medullary TECs have the potential to differentiate into various types of molecularly distinct and functionally defined cells, which not only contribute to the induction of central tolerance, but also regulate the homeostasis of other thymus-resident populations.


Self Tolerance , T-Lymphocytes , Thymus Gland , Animals , Mice , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Self Tolerance/immunology , Self Tolerance/physiology , T-Lymphocytes/classification , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Parenchymal Tissue , Muscle Cells , Endocrine Cells , Chromatin , Transcription, Genetic , Ghrelin
...