Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nat Commun ; 15(1): 6976, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143070

ABSTRACT

Regulatory T cells (Treg) are critical players of immune tolerance that develop in the thymus via two distinct developmental pathways involving CD25+Foxp3- and CD25-Foxp3lo precursors. However, the mechanisms regulating the recently identified Foxp3lo precursor pathway remain unclear. Here, we find that the membrane-bound lymphotoxin α1ß2 (LTα1ß2) heterocomplex is upregulated during Treg development upon TCR/CD28 and IL-2 stimulation. We show that Lta expression limits the maturational development of Treg from Foxp3lo precursors by regulating their proliferation, survival, and metabolic profile. Transgenic reporter mice and transcriptomic analyses further reveal that medullary thymic epithelial cells (mTEC) constitute an unexpected source of IL-4. We demonstrate that LTα1ß2-lymphotoxin ß receptor-mediated interactions with mTEC limit Treg development by down-regulating IL-4 expression in mTEC. Collectively, our findings identify the lymphotoxin axis as the first inhibitory checkpoint of thymic Treg development that fine-tunes the Foxp3lo Treg precursor pathway by limiting IL-4 availability.


Subject(s)
Forkhead Transcription Factors , Interleukin-4 , Lymphotoxin beta Receptor , Lymphotoxin-alpha , Signal Transduction , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Interleukin-4/metabolism , Mice , Lymphotoxin-alpha/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin beta Receptor/metabolism , Lymphotoxin beta Receptor/genetics , Thymus Gland/immunology , Thymus Gland/cytology , Thymus Gland/metabolism , Epithelial Cells/metabolism , Mice, Inbred C57BL , Cell Differentiation , Mice, Transgenic , Interleukin-2/metabolism , Cell Proliferation , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/genetics
2.
J Vis Exp ; (209)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39141542

ABSTRACT

Thymic epithelial cells (TECs) play an essential role in promoting the development and repertoire selection of T cells. Cortical TECs (cTECs) in the thymic cortex induce early T cell development and positive selection of cortical thymocytes. In contrast, medullary TECs (mTECs) in the thymic medulla attract positively selected thymocytes from the cortex and establish self-tolerance in T cells. A variety of molecules, including DLL4 and beta5t expressed in cTECs, as well as Aire and CCL21 expressed in mTECs, contribute to thymus function supporting T cell development and selection. Flow cytometric analysis of functionally relevant molecules in cTECs and mTECs is useful to improve our understanding of the biology of TECs, even though current methods for the preparation of single-cell suspensions of TECs can retrieve only a small fraction of TECs (approximately 1% for cTECs and approximately 10% for mTECs) from young adult mouse thymus. Because many of these functionally relevant molecules in TECs are localized within the cells, we describe our protocols for the preparation of single-cell suspension of mouse TECs and the staining of intracellular molecules for flow cytometric analysis.


Subject(s)
Epithelial Cells , Flow Cytometry , Thymus Gland , Animals , Mice , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/chemistry , Thymus Gland/cytology , Thymus Gland/metabolism , Flow Cytometry/methods
4.
J Immunol ; 213(5): 641-650, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38958395

ABSTRACT

Activation of ß-catenin in CD4+CD8+ double-positive (DP) thymocytes halts development before the thymic selection stage and predisposes to transformation. Leukemogenesis, but not the developmental block, depends on TCF-1, ß-catenin's DNA-binding partner. In this study, we show that ß-catenin activation directs the DNA-binding protein HEB to block DP thymocyte development. Conditional loss of HEB in DP thymocytes with stabilized ß-catenin restores the frequencies of postselection TCRßhi/CCR7+ and TCRßhi/CD69+ DPs and their cell-cycle profile. This recovery is associated with significant reversal of ß-catenin-induced expression changes, particularly those related to the CD69+ DP cell signature and to cell-cycle pathways. Stabilizing ß-catenin in DP thymocytes directs HEB binding to ≈11,000 novel DNA sites throughout the genome. Novel HEB sites mark most CD69+ DP cell signature genes that change expression upon activation of ß-catenin and then revert after loss of HEB. Moreover, many of the novel HEB sites occupy promoter regions of genes enriched in mitotic cell cycle pathways. HEB binding to those regions correlates with downregulation of the associated genes, and HEB inactivation restores expression to physiologic levels. These findings highlight a molecular interplay between HEB and ß-catenin that can impair thymic development.


Subject(s)
Thymocytes , Thymus Gland , beta Catenin , Animals , beta Catenin/metabolism , beta Catenin/genetics , Mice , Thymocytes/metabolism , Thymocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/immunology , Cell Differentiation/genetics , Protein Stability , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/genetics
5.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38958026

ABSTRACT

Thymic epithelial cells (TECs) are crucial to the ability of the thymus to generate T cells for the adaptive immune system in vertebrates. However, no in vitro system for studying TEC function exists. Overexpressing the transcription factor FOXN1 initiates transdifferentiation of fibroblasts into TEC-like cells (iTECs) that support T-cell differentiation in culture or after transplant. In this study, we have characterized iTEC programming at the cellular and molecular level in mouse to determine how it proceeds, and have identified mechanisms that can be targeted for improving this process. These data show that iTEC programming consists of discrete gene expression changes that differ early and late in the process, and that iTECs upregulate markers of both cortical and medullary TEC (cTEC and mTEC) lineages. We demonstrate that promoting proliferation enhances iTEC generation, and that Notch inhibition allows the induction of mTEC differentiation. Finally, we show that MHCII expression is the major difference between iTECs and fetal TECs. MHCII expression was improved by co-culturing iTECs with fetal double-positive T-cells. This study supports future efforts to improve iTEC generation for both research and translational uses.


Subject(s)
Cell Differentiation , Epithelial Cells , Fibroblasts , Forkhead Transcription Factors , Thymus Gland , Animals , Epithelial Cells/metabolism , Epithelial Cells/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/embryology , Fibroblasts/metabolism , Fibroblasts/cytology , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , Cell Proliferation , Cell Transdifferentiation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Coculture Techniques , Receptors, Notch/metabolism
6.
Immunohorizons ; 8(7): 492-499, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39008056

ABSTRACT

The transcription factor FOXN1 plays an established role in thymic epithelial development to mediate selection of maturing thymocytes. Patients with heterozygous loss-of-function FOXN1 variants are associated with T cell lymphopenia at birth and low TCR excision circles that can ultimately recover. Although CD4+ T cell reconstitution in these patients is not completely understood, a lower proportion of naive T cells in adults has suggested a role for homeostatic proliferation. In this study, we present an immunophenotyping study of fraternal twins with low TCR excision circles at birth. Targeted primary immunodeficiency testing revealed a heterozygous variant of uncertain significance in FOXN1 (c.1205del, p.Pro402Leufs*148). We present the immune phenotypes of these two patients, as well as their father who carries the same FOXN1 variant, to demonstrate an evolving immune environment over time. While FOXN1 haploinsufficiency may contribute to thymic defects and T cell lymphopenia, we characterized the transcriptional activity and DNA binding of the heterozygous FOXN1 variant in 293T cells and found the FOXN1 variant to have different effects across several target genes. These data suggest multiple mechanisms for similar FOXN1 variants pathogenicity that may be mutation specific. Increased understanding of how these variants drive transcriptional regulation to impact immune cell populations will guide the potential need for therapeutics, risk for infection or autoimmunity over time, and help inform clinical decisions for other variants that might arise.


Subject(s)
Forkhead Transcription Factors , Heterozygote , Immunophenotyping , Humans , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Male , Female , Lymphopenia/genetics , Lymphopenia/immunology , Mutation , Adult , Haploinsufficiency , T-Lymphocytes/immunology , HEK293 Cells , Infant, Newborn , Thymus Gland/immunology , Thymus Gland/metabolism
7.
Front Immunol ; 15: 1423488, 2024.
Article in English | MEDLINE | ID: mdl-39072332

ABSTRACT

T cell development in the thymus is dependent on the thymic microenvironment, in which thymic epithelial cells (TECs) are the major component. However, TECs undergo both a qualitative and quantitative loss during aging, which is believed to be the major factor responsible for age-dependent thymic atrophy. FOXN1 plays a critical role in TEC development and adult TECs maintenance. We have previously reported that intrathymic injection of a recombinant (r) protein containing murine FOXN1 and a protein transduction domain increases the number of TECs in mice, leading to enhanced thymopoiesis. However, intrathymic injection may not be an ideal choice for clinical applications. In this study, we produced a rFOXN1 fusion protein containing the N-terminal of CCR9, human FOXN1 and a protein transduction domain. When injected intravenously into 14-month-old mice, the rFOXN1 fusion protein enters the thymus and TECs, and enhances thymopoiesis, resulting in increased T cell generation in the thymus and increased number of T cells in peripheral lymphoid organ. Our results suggest that the rFOXN1 fusion protein has the potential to be used in preventing and treating T cell immunodeficiency in older adults.


Subject(s)
Forkhead Transcription Factors , Recombinant Fusion Proteins , T-Lymphocytes , Thymus Gland , Animals , Mice , Recombinant Fusion Proteins/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Thymus Gland/immunology , Thymus Gland/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Aging/immunology , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/immunology , Cell Differentiation
8.
Immunohorizons ; 8(7): 500-510, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39018546

ABSTRACT

The critical importance of the thymus for generating new naive T cells that protect against novel infections and are tolerant to self-antigens has led to a recent revival of interest in monitoring thymic function in species other than humans and mice. Nonhuman primates such as rhesus macaques (Macaca mulatta) provide particularly useful animal models for translational research in immunology. In this study, we tested the performance of a 15-marker multicolor Ab panel for flow cytometric phenotyping of lymphocyte subsets directly from rhesus whole blood, with validation by thymectomy and T cell depletion. Immunohistochemical and multiplex RNA expression analysis of thymus tissue biopsies and molecular assays on PBMCs were used to further validate thymus function. Results identify Ab panels that can accurately classify rhesus naive T cells (CD3+CD45RA+CD197+ or CD3+CD28+CD95-) and recent thymic emigrants (CD8+CD28+CD95-CD103+CD197+) using just 100 µl of whole blood and commercially available fluorescent Abs. An immunohistochemical panel reactive with pan-cytokeratin (CK), CK14, CD3, Ki-67, CCL21, and TdT provides histologic evidence of thymopoiesis from formalin-fixed, paraffin-embedded thymus tissues. Identification of mRNAs characteristic of both functioning thymic epithelial cells and developing thymocytes and/or molecular detection of products of TCR gene rearrangement provide additional complementary methods to evaluate thymopoiesis, without requiring specific Abs. Combinations of multiparameter flow cytometry, immunohistochemistry, multiplex gene expression, and TCR excision circle assays can comprehensively evaluate thymus function in rhesus macaques while requiring only minimal amounts of peripheral blood or biopsied thymus tissue.


Subject(s)
Flow Cytometry , Macaca mulatta , Thymus Gland , Animals , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/cytology , Immunohistochemistry , Immunophenotyping , Male , Female , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymectomy
9.
Microb Pathog ; 192: 106723, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823465

ABSTRACT

The Hedgehog (Hh) signaling pathway is involved in T cell differentiation and development and plays a major regulatory part in different stages of T cell development. A previous study by us suggested that prenatal exposure to staphylococcal enterotoxin B (SEB) changed the percentages of T cell subpopulation in the offspring thymus. However, it is unclear whether prenatal SEB exposure impacts the Hh signaling pathway in thymic T cells. In the present study, pregnant rats at gestational day 16 were intravenously injected once with 15 µg SEB, and the thymi of both neonatal and adult offspring rats were aseptically acquired to scrutinize the effects of SEB on the Hh signaling pathway. It firstly found that prenatal SEB exposure clearly caused the increased expression of Shh and Dhh ligands of the Hh signaling pathway in thymus tissue of both neonatal and adult offspring rats, but significantly decreased the expression levels of membrane receptors of Ptch1 and Smo, transcription factor Gli1, as well as target genes of CyclinD1, C-myc, and N-myc in Hh signaling pathway of thymic T cells. These data suggest that prenatal SEB exposure inhibits the Hh signaling pathway in thymic T lymphocytes of the neonatal offspring, and this effect can be maintained in adult offspring via the imprinting effect.


Subject(s)
Enterotoxins , Hedgehog Proteins , Signal Transduction , T-Lymphocytes , Thymus Gland , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Female , Pregnancy , Rats , Thymus Gland/metabolism , Thymus Gland/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Prenatal Exposure Delayed Effects/immunology , Cell Differentiation/drug effects , Rats, Sprague-Dawley , Male
10.
Environ Pollut ; 357: 124438, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38942270

ABSTRACT

Humans indirectly consume approximately 0.02 mg/kg/day of short-chained chlorinated paraffins (SCCPs) through the environment; however, the thymic senescence/damage induced by SCCPs has not been assessed. In this study, 16 female mice (4-week-old) per group were orally administered 0, 0.01, 0.1, and 1 mg/kg/day of SCCPs for 21 days, and the phenotypes and levels of superoxide dismutase (SOD), malondialdehyde (MDA), Tß4, αß TCR, SA-ß-Gal, GRP78, PERK/CHOP, P53/P21, and CASPASE-1 of the thymus were assessed as indicators. Another group comprising 16 mice was killed at 4-week-old and these indicators were assessed. Thereafter, the thymuses cultured in vitro were exposed to 0, 14, 140, and 1400 µg/L SCCPs, respectively, and the above indicators were measured after 7-day. Based on the results, the oral administration of ≥0.01 mg/kg/day SCCPs to mice and ≥14 µg/L of SCCPs in medium caused thymic aging features, such as a decrease in the ratio of cortex to medulla, gradual blurring of the boundary between the cortex and medulla, dose-dependent oxidative stress (decreased SOD and increased MDA), and decreased levels of Tß4 and αß TCRs in the thymus. The oral administration of ≥1 mg/kg/day of SCCPs also impeded the growth and development of female mice and their thymuses. Exposure to the low levels of SCCPs activated PERK-CHOP in the mouse thymus, which modulated increases in SA-ß-Gal, IL-1ß, P53, and CASPASE-1 in vivo and in vitro. Overall, environmental levels and human blood concentrations (14.8-1400 µg/L) of SCCPs may induce mouse thymus senescence by activating PERK-CHOP in vivo and in vitro, respectively.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Thymus Gland , Transcription Factor CHOP , Animals , Thymus Gland/drug effects , Thymus Gland/metabolism , Mice , Female , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism , Oxidative Stress/drug effects , Aging
11.
Microbiol Immunol ; 68(8): 281-293, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38886542

ABSTRACT

The thymus, a site to culture the naïve T lymphocytes, is susceptible to atrophy or involution due to aging, inflammation, and oxidation. Epigallocatechin-3-gallate (EGCG) has been proven to possess anti-inflammatory, antioxidant, and antitumor activity. Here, we investigate the effects of EGCG on thymic involution induced by lipopolysaccharide (LPS), an endotoxin derived from Gram-negative bacteria. The methodology included an in vivo experiment on female Kunming mice exposed to LPS and EGCG. Morphological assessment of thymic involution, immunohistochemical detection, and thymocyte subsets analysis by flow cytometry were further carried out to evaluate the potential role of EGCG on the thymus. As a result, we found that EGCG alleviated LPS-induced thymic atrophy, increased mitochondrial membrane potential and superoxide dismutase levels, and decreased malondialdehyde and reactive oxygen species levels. In addition, EGCG pre-supplement restored the ratio of thymocyte subsets, the expression of autoimmune regulator, sex-determining region Y-box 2, and Nanog homebox, and reduced the number of senescent cells and collagen fiber deposition. Western blotting results indicated that EGCG treatment elevated LPS-induced decrease in pAMPK, Sirt1 protein expression. Collectively, EGCG relieved thymus architecture and function damaged by LPS via regulation of AMPK/Sirt1 signaling pathway. Our findings may provide a new strategy on protection of thymus from involution caused by LPS by using EGCG. And EGCG might be considered as a potential agent for the prevention and treatment of thymic involution.


Subject(s)
AMP-Activated Protein Kinases , Catechin , Lipopolysaccharides , Signal Transduction , Sirtuin 1 , Thymus Gland , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Sirtuin 1/metabolism , Mice , Female , Thymus Gland/drug effects , Thymus Gland/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Membrane Potential, Mitochondrial/drug effects , Atrophy
12.
J Reprod Immunol ; 164: 104288, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38924811

ABSTRACT

Thymic atrophy affects T cell generation and migration to the periphery, thereby affecting T cell pool diversity. However, the mechanisms underlying thymic atrophy have not been fully elucidated. Here, gonadotropin-releasing hormone (GnRH) immunization and surgical castration did not affect thymocyte proliferation, but significantly reduced the apoptosis and increased the survival rate of CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+ thymocytes. Following testosterone supplementation in rats subjected to GnRH immunization and surgical castration, thymocyte proliferation remained unchange, but the apoptosis of CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+ thymocytes significantly increased. Transcriptome analyses of the thymus after GnRH immunization and surgical castration showed a significant reduction in the thymus's response to corticosterone. Cholesterol metabolism and the synthesis and secretion of corticosterone were significantly reduced. Analysis of the enzyme levels involved in the corticosterone synthesis pathway revealed that corticosterone synthesis in thymocytes was significantly reduced after GnRH immunization and surgical castration, whereas exogenous testosterone supplementation relieved this process. Testosterone promoted thymocyte apoptosis in a concentration-dependent manner, and induced corticosterone secretion in vitro. Blocking the intracellular androgen receptor (AR) signaling pathway did not significantly affect thymocyte apoptosis, but blocking the glucocorticoid receptor (GR) signaling pathway significantly reduced it. Our findings indicate that testosterone regulates thymus remodeling by affecting corticosterone synthesis in thymocytes, which activates GR signal transduction and promotes thymocyte apoptosis.


Subject(s)
Apoptosis , Receptors, Glucocorticoid , Signal Transduction , Testosterone , Thymocytes , Thymus Gland , Animals , Male , Testosterone/metabolism , Apoptosis/immunology , Rats , Signal Transduction/immunology , Signal Transduction/drug effects , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Receptors, Glucocorticoid/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Gonadotropin-Releasing Hormone/metabolism , Corticosterone/metabolism , Corticosterone/blood , Cells, Cultured , Rats, Sprague-Dawley , Receptors, Androgen/metabolism , Orchiectomy
13.
Sci Rep ; 14(1): 10045, 2024 05 02.
Article in English | MEDLINE | ID: mdl-38698013

ABSTRACT

Chronic stress has been implicated in mental illnesses and depressive behaviors. Somatostatin 4 receptor (SSTR4) has been shown to mediate anxiolytic and depression-like effects. Here, we aimed to explore the potential of SSTR4 as a diagnostic marker for chronic stress in mice. The mice were divided into single stress, chronic restraint stress, and control groups, and Sstr4 mRNA expression in the pituitary, lungs, and thymus, its protein expression in the thymus, were analyzed. Compared to controls, Sstr4 mRNA expression decreased significantly in the pituitary gland of the chronic and single-stress groups (P = 0.0181 and 0.0022, respectively) and lungs of the single-stress group (P = 0.0124), whereas it significantly increased in the thymus of the chronic-stress group (P = 0.0313). Thymic SSTR4 expression did not decrease significantly in stress groups compared to that in the control group (P = 0.0963). These results suggest that SSTR4 expression fluctuates in response to stress. Furthermore, Sstr4 mRNA expression dynamics in each organ differed based on single or chronic restraint stress-loading periods. In conclusion, this study suggests that investigating SSTR4 expression in each organ could allow for its use as a stress marker to estimate the stress-loading period and aid in diagnosing chronic stress.


Subject(s)
Biomarkers , Receptors, Somatostatin , Stress, Psychological , Thymus Gland , Animals , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/genetics , Mice , Stress, Psychological/metabolism , Male , Biomarkers/metabolism , Thymus Gland/metabolism , Pituitary Gland/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Lung/metabolism , Chronic Disease , Stress, Physiological , Restraint, Physical
14.
Proc Natl Acad Sci U S A ; 121(20): e2320268121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709934

ABSTRACT

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.


Subject(s)
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
15.
J Agric Food Chem ; 72(23): 13382-13392, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38814005

ABSTRACT

Cadmium (Cd) is a transition metal ion that is extremely harmful to human and animal biological systems. Cd is a toxic substance that can accumulate in the food chain and cause various health issues. Sulforaphane (SFN) is a natural bioactive compound with potent antioxidant properties. In our study, 80 1 day-old chicks were fed with Cd (140 mg/kg BW/day) and/or SFN (50 mg/kg BW/day) for 90 days. The blood-thymus barrier (BTB) is a selective barrier separating T-lymphocytes from blood and cortical capillaries in the thymus cortex. Our research revealed that Cd could destroy the BTB by downregulating Wnt/ß-catenin signaling and induce immunodeficiency, leading to irreversible injury to the immune system. The study emphasizes the health benefits of SFN in the thymus. SFN could ameliorate Cd-triggered BTB dysfunction and pyroptosis in the thymus tissues. SFN modulated the PI3K/AKT/FOXO1 axis, improving the level of claudin-5 (CLDN5) in the thymus to alleviate BTB breakdown. Our findings indicated the toxic impact of Cd on thymus, and BTB could be the specific target of Cd toxicity. The finding also provides evidence for the role of SFN in maintaining thymic homeostasis for Cd-related health issues.


Subject(s)
Cadmium , Chickens , Isothiocyanates , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sulfoxides , Thymus Gland , Animals , Isothiocyanates/pharmacology , Cadmium/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Thymus Gland/drug effects , Thymus Gland/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Signal Transduction/drug effects , Humans , Male
16.
Sci Rep ; 14(1): 10636, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724644

ABSTRACT

Gene-knockout animal models with organ-deficient phenotypes used for blastocyst complementation are generally not viable. Animals need to be maintained as heterozygous mutants, and homozygous mutant embryos yield only one-fourth of all embryos. In this study, we generated organ-deficient embryos using the CRISPR-Cas9-sgRNAms system that induces cell death with a single-guide RNA (sgRNAms) targeting multiple sites in the genome. The Cas9-sgRNAms system interrupted cell proliferation and induced cell ablation in vitro. The mouse model had Cas9 driven by the Foxn1 promoter with a ubiquitous expression cassette of sgRNAms at the Rosa26 locus (Foxn1Cas9; Rosa26_ms). It showed an athymic phenotype similar to that of nude mice but was not hairless. Eventually, a rat cell-derived thymus in an interspecies chimera was generated by blastocyst complementation of Foxn1Cas9; Rosa26_ms mouse embryos with rat embryonic stem cells. Theoretically, a half of the total embryos has the Cas9-sgRNAms system because Rosa26_ms could be maintained as homozygous.


Subject(s)
CRISPR-Cas Systems , Forkhead Transcription Factors , RNA, Guide, CRISPR-Cas Systems , Animals , Mice , Rats , RNA, Guide, CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Thymus Gland/metabolism , Models, Animal , Blastocyst/metabolism
17.
Cell Death Dis ; 15(5): 352, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773063

ABSTRACT

Within the thymus, thymic epithelial cells (TECs) create dedicated microenvironments for T cell development and selection. Considering that TECs are sensitive to distinct pathophysiological conditions, uncovering the molecular elements that coordinate their thymopoietic role has important fundamental and clinical implications. Particularly, medullary thymic epithelial cells (mTECs) play a crucial role in central tolerance. Our previous studies, along with others, suggest that mTECs depend on molecular factors linked to genome-protecting pathways, but the precise mechanisms underlying their function remain unknown. These observations led us to examine the role of Foxo3, as it is expressed in TECs and involved in DNA damage response. Our findings show that mice with TEC-specific deletion of Foxo3 (Foxo3cKO) displayed a disrupted mTEC compartment, with a more profound impact on the numbers of CCL21+ and thymic tuft mTEClo subsets. At the molecular level, Foxo3 controls distinct functional modules in the transcriptome of cTECs and mTECs under normal conditions, which includes the regulation of ribosomal biogenesis and DNA damage response, respectively. These changes in the TEC compartment resulted in a reduced total thymocyte cellularity and specific changes in regulatory T cell and iNKT cell development in the Foxo3cKO thymus. Lastly, the thymic defects observed in adulthood correlated with mild signs of altered peripheral immunotolerance in aged Foxo3cKO mice. Moreover, the deficiency in Foxo3 moderately aggravated the autoimmune predisposition observed in Aire-deficient mice. Our findings highlight the importance of Foxo3 in preserving the homeostasis of TECs and in supporting their role in T cell development and tolerance.


Subject(s)
Epithelial Cells , Forkhead Box Protein O3 , Homeostasis , Thymus Gland , Animals , Thymus Gland/metabolism , Thymus Gland/cytology , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Epithelial Cells/metabolism , Mice , Mice, Knockout , Cell Differentiation , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Mice, Inbred C57BL
18.
Elife ; 122024 Apr 18.
Article in English | MEDLINE | ID: mdl-38635416

ABSTRACT

Transposable elements (TEs) are repetitive sequences representing ~45% of the human and mouse genomes and are highly expressed by medullary thymic epithelial cells (mTECs). In this study, we investigated the role of TEs on T-cell development in the thymus. We performed multiomic analyses of TEs in human and mouse thymic cells to elucidate their role in T-cell development. We report that TE expression in the human thymus is high and shows extensive age- and cell lineage-related variations. TE expression correlates with multiple transcription factors in all cell types of the human thymus. Two cell types express particularly broad TE repertoires: mTECs and plasmacytoid dendritic cells (pDCs). In mTECs, transcriptomic data suggest that TEs interact with transcription factors essential for mTEC development and function (e.g., PAX1 and REL), and immunopeptidomic data showed that TEs generate MHC-I-associated peptides implicated in thymocyte education. Notably, AIRE, FEZF2, and CHD4 regulate small yet non-redundant sets of TEs in murine mTECs. Human thymic pDCs homogenously express large numbers of TEs that likely form dsRNA, which can activate innate immune receptors, potentially explaining why thymic pDCs constitutively secrete IFN ɑ/ß. This study highlights the diversity of interactions between TEs and the adaptive immune system. TEs are genetic parasites, and the two thymic cell types most affected by TEs (mTEcs and pDCs) are essential to establishing central T-cell tolerance. Therefore, we propose that orchestrating TE expression in thymic cells is critical to prevent autoimmunity in vertebrates.


Subject(s)
AIRE Protein , DNA Transposable Elements , Mice , Humans , Animals , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Thymocytes/metabolism , Epithelial Cells/metabolism , Cell Differentiation/genetics , Mice, Inbred C57BL
19.
Front Immunol ; 15: 1331846, 2024.
Article in English | MEDLINE | ID: mdl-38605970

ABSTRACT

Foxp3+ TREG cells have been at the focus of intense investigation for their recognized roles in preventing autoimmunity, facilitating tissue recuperation following injury, and orchestrating a tolerance to innocuous non-self-antigens. To perform these critical tasks, TREG cells undergo deep epigenetic, transcriptional, and post-transcriptional changes that allow them to adapt to conditions found in tissues both at steady-state and during inflammation. The path leading TREG cells to express these tissue-specialized phenotypes begins during thymic development, and is further driven by epigenetic and transcriptional modifications following TCR engagement and polarizing signals in the periphery. However, this process is highly regulated and requires TREG cells to adopt strategies to avoid losing their regulatory program altogether. Here, we review the origins of tissue-resident TREG cells, from their thymic and peripheral development to the transcriptional regulators involved in their tissue residency program. In addition, we discuss the distinct signalling pathways that engage the inflammatory adaptation of tissue-resident TREG cells, and how they relate to their ability to recognize tissue and pathogen-derived danger signals.


Subject(s)
Autoimmunity , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , Thymus Gland/metabolism
20.
Front Immunol ; 15: 1364957, 2024.
Article in English | MEDLINE | ID: mdl-38650932

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL