Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 68
Filtrer
1.
Immunol Res ; 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39042204

RÉSUMÉ

Age-related thymic involution is characterized by the loss of T cell development and the supporting epithelial network, which are replaced by adipose tissue. We previously showed that aging functionally impairs lymphohematopoietic progenitor cells, including thymic early T cell progenitors (ETPs), contributing to thymic involution. Considering that the thymic microenvironment is essential for thymocyte incubation, we aimed to investigate its role in age-related thymic involution and the mechanisms underlying these changes. The challenge in studying these processes led us to transplant T cell-depleted fetal thymus tissue into the kidney capsule of aged mice. This model allowed us to identify the mechanisms driving age-related changes in the thymic microenvironment and to assess whether these changes could be reversed. Flow cytometry was used to detect naïve T cells (CD62L+CD44-), including CD4 CD8 double-negative, double-positive, and single-positive T cells. Real-time PCR was used to detect and quantify signal-joint T cell receptor excision circles. We rearranged δRec-ΨJα in murine peripheral blood leukocytes to evaluate the thymic output of newly developed naïve T cells in the mice and gene expression in the thymus. Age-related thymic involution decreased naïve T cells and increased memory T cells, while fetal thymus transplantation improved thymic output and T cell production and reversed the impairment of thymopoiesis due to thymic involution in aged mice. Furthermore, the expression of key cytokines was restored and ETPs in the aged mice showed normal thymic T cell development. Our study suggests that degenerative changes in the thymic microenvironment are the primary cause of thymic dysfunction, leading to immunosenescence associated with age-related thymic involution.

2.
Development ; 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39036995

RÉSUMÉ

Although the advent of organoids opened unprecedented perspectives for basic and translational research, immune system-related organoids remain largely underdeveloped. Here we established organoids from the thymus, the lymphoid organ responsible for T cell development. We identified conditions enabling thymic epithelial progenitor cell proliferation and development into organoids with diverse cell populations and transcriptional profiles resembling in vivo thymic epithelial cells (TECs) more closely than traditional TEC cultures. Contrary to these two-dimensional cultures, thymic epithelial organoids maintained thymus functionality in vitro and mediated physiological T cell development upon reaggregation with T cell progenitors. The reaggregates showed in vivo-like epithelial diversity and ability to attract T cell progenitors. Thymic epithelial organoids are the first organoids originating from the stromal compartment of a lymphoid organ. They provide new opportunities to study TEC biology and T cell development in vitro, paving the way for future thymic regeneration strategies in ageing or acute injuries.

3.
Front Immunol ; 15: 1288045, 2024.
Article de Anglais | MEDLINE | ID: mdl-38629065

RÉSUMÉ

Thymic epithelial tumors (TETs) are rare mediastinal cancers originating from the thymus, classified in two main histotypes: thymoma and thymic carcinoma (TC). TETs affect a primary lymphoid organ playing a critical role in keeping T-cell homeostasis and ensuring an adequate immunological tolerance against "self". In particular, thymomas and not TC are frequently associated with autoimmune diseases (ADs), with Myasthenia Gravis being the most common AD present in 30% of patients with thymoma. This comorbidity, in addition to negatively affecting the quality and duration of patients' life, reduces the spectrum of the available therapeutic options. Indeed, the presence of autoimmunity represents an exclusion criteria for the administration of the newest immunotherapeutic treatments with checkpoint inhibitors. The pathophysiological correlation between TETs and autoimmunity remains a mystery. Several studies have demonstrated the presence of a residual and active thymopoiesis in adult patients affected by thymomas, especially in mixed and lymphocytic-rich thymomas, currently known as type AB and B thymomas. The aim of this review is to provide the state of art in regard to the histological features of the different TET histotype, to the role of the different immune cells infiltrating tumor microenvironments and their impact in the break of central immunologic thymic tolerance in thymomas. We discuss here both cellular and molecular immunologic mechanisms inducing the onset of autoimmunity in TETs, limiting the portfolio of therapeutic strategies against TETs and greatly impacting the prognosis of associated autoimmune diseases.


Sujet(s)
Myasthénie , Tumeurs épithéliales épidermoïdes et glandulaires , Thymome , Tumeurs du thymus , Adulte , Humains , Auto-immunité , Tumeurs du thymus/complications , Tumeurs épithéliales épidermoïdes et glandulaires/thérapie , Tumeurs épithéliales épidermoïdes et glandulaires/complications , Microenvironnement tumoral
4.
Hum Immunol ; 85(3): 110791, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38553383

RÉSUMÉ

BACKGROUND: A genetic polymorphism, rs2204985, has been reported to be associated with the diversity of T-cell antigen receptor repertoire and TREC levels, reflecting the function of the thymus. As the thymus function can be assumed to be an important factor regulating the outcome of stem cell transplantation (SCT), it was of great interest that rs2204985 showed a genetic association to disease-free and overall survival in a German SCT donor cohort. Tools to predict the outcome of SCT more accurately would help in risk assessment and patient safety. OBJECTIVE: To evaluate the general validity of the original genetic association found in the German cohort, we determined genetic associations between rs2204985 and the outcome of SCT in 1,473 SCT donors from four different populations. STUDY DESIGN: Genetic associations between rs2204985 genotype AA versus AG/GG and overall survival (OS) and disease-free survival (DFS) in 1,473 adult, allogeneic SCT from Finland, the United Kingdom, Spain, and Poland were performed using the Kaplan-Meier analysis and log-rank tests. We adjusted the survival models with covariates using Cox regression. RESULTS: In unrelated SCT donors (N = 425), the OS of genotype AA versus AG/GG had a trend for a similar association (p = 0.049, log-rank test) as previously reported in the German cohort. The trend did not remain significant in the Cox regression analysis with covariates. No other associations were found. CONCLUSION: Weak support for the genetic association between rs2204985, previously also associated with thymus function, and the outcome of SCT could be found in a cohort from four populations.


Sujet(s)
Thymus (glande) , Humains , Adulte , Mâle , Femelle , Adulte d'âge moyen , Études de cohortes , Polymorphisme de nucléotide simple , Génotype , Donneurs de tissus , Transplantation de cellules souches , Sujet âgé , Jeune adulte , Adolescent , Pologne , Résultat thérapeutique , Espagne , Transplantation de cellules souches hématopoïétiques , Royaume-Uni
5.
Front Immunol ; 15: 1321309, 2024.
Article de Anglais | MEDLINE | ID: mdl-38469297

RÉSUMÉ

Background: The thymus plays a central role in shaping human immune function. A mechanistic, quantitative description of immune cell dynamics and thymic output under homeostatic conditions and various patho-physiological scenarios are of particular interest in drug development applications, e.g., in the identification of potential therapeutic targets and selection of lead drug candidates against infectious diseases. Methods: We here developed an integrative mathematical model of thymocyte dynamics in human. It incorporates mechanistic features of thymocyte homeostasis as well as spatial constraints of the thymus and considerations of age-dependent involution. All model parameter estimates were obtained based on published physiological data of thymocyte dynamics and thymus properties in mouse and human. We performed model sensitivity analyses to reveal potential therapeutic targets through an identification of processes critically affecting thymic function; we further explored differences in thymic function across healthy subjects, multiple sclerosis patients, and patients on fingolimod treatment. Results: We found thymic function to be most impacted by the egress, proliferation, differentiation and death rates of those thymocytes which are most differentiated. Model predictions also showed that the clinically observed decrease in relapse risk with age, in multiple sclerosis patients who would have discontinued fingolimod therapy, can be explained mechanistically by decreased thymic output with age. Moreover, we quantified the effects of fingolimod treatment duration on thymic output. Conclusions: In summary, the proposed model accurately describes, in mechanistic terms, thymic output as a function of age. It may be further used to perform predictive simulations of clinically relevant scenarios which combine specific patho-physiological conditions and pharmacological interventions of interest.


Sujet(s)
Sclérose en plaques , Thymocytes , Humains , Souris , Animaux , Thymocytes/métabolisme , Chlorhydrate de fingolimod/pharmacologie , Chlorhydrate de fingolimod/usage thérapeutique , Chlorhydrate de fingolimod/métabolisme , Thymus (glande) , Différenciation cellulaire , Sclérose en plaques/métabolisme
6.
Immunol Rev ; 322(1): 178-211, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38228406

RÉSUMÉ

The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.


Sujet(s)
Déficits immunitaires , Lymphocytes T , Thymus (glande)/malformations , Nouveau-né , Humains , Différenciation cellulaire
7.
Eur J Immunol ; 53(12): e2350546, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37751619

RÉSUMÉ

Cryopreservation of mouse thymus depletes donor thymocytes but preserves thymus function when transplanted after thawing into athymic mice. No differences in immune reconstitution were observed between fresh and frozen/thawed transplants suggesting that donor thymocyte depletion does not affect outcome. Thus, cryopreservation of thymus may improve outcomes in thymus transplant patients.


Sujet(s)
Reconstitution immunitaire , Thymocytes , Humains , Animaux , Souris , Thymus (glande) , Cryoconservation
8.
Eur J Immunol ; 53(12): e2350725, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37724048

RÉSUMÉ

In mammals, T-cell development depends on the activity of the Foxn1 transcription factor in the thymic epithelium; mutations in the vertebrate-specific Foxn1 gene are associated with profound T-cell lymphopenia and fatal immunodeficiency. Here, we examined the extent of T-cell development in teleosts lacking a functional foxn1 gene. In zebrafish carrying a deleterious internal deletion of foxn1, reduced but robust lymphopoietic activity is maintained in the mutant thymus. Moreover, pseudogenization or loss of foxn1 in the genomes of deep-sea anglerfishes is independent of the presence or absence of the canonical signatures of the T-cell lineage. Thus, in contrast to the situation in mammals, the teleost thymus can support foxn1-independent lymphopoiesis, most likely through the activity of the Foxn4, an ancient metazoan paralog of Foxn1. Our results imply that during the early stages of vertebrate evolution, genetic control of thymopoiesis was functionally redundant and thus robust; in mammals, the genetic network was reorganized to become uniquely dependent on the FOXN1 transcription factor.


Sujet(s)
Réseaux de régulation génique , Danio zébré , Souris , Animaux , Souris transgéniques , Danio zébré/génétique , Lymphocytes T , Thymus (glande) , Facteurs de transcription/génétique , Facteurs de transcription Forkhead/génétique , Cellules épithéliales , Mammifères/génétique , Protéines de poisson-zèbre/génétique
9.
J Allergy Clin Immunol ; 152(5): 1273-1291.e15, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37419334

RÉSUMÉ

BACKGROUND: Thymus hypoplasia due to stromal cell problems has been linked to mutations in several transcription factors, including Forkhead box N1 (FOXN1). FOXN1 supports T-cell development by regulating the formation and expansion of thymic epithelial cells (TECs). While autosomal recessive FOXN1 mutations result in a nude and severe combined immunodeficiency phenotype, the impact of single-allelic or compound heterozygous FOXN1 mutations is less well-defined. OBJECTIVE: With more than 400 FOXN1 mutations reported, their impact on protein function and thymopoiesis remains unclear for most variants. We developed a systematic approach to delineate the functional impact of diverse FOXN1 variants. METHODS: Selected FOXN1 variants were tested with transcriptional reporter assays and imaging studies. Thymopoiesis was assessed in mouse lines genocopying several human FOXN1 variants. Reaggregate thymus organ cultures were used to compare the thymopoietic potential of the FOXN1 variants. RESULTS: FOXN1 variants were categorized into benign, loss- or gain-of-function, and/or dominant-negatives. Dominant negative activities mapped to frameshift variants impacting the transactivation domain. A nuclear localization signal was mapped within the DNA binding domain. Thymopoiesis analyses with mouse models and reaggregate thymus organ cultures revealed distinct consequences of particular Foxn1 variants on T-cell development. CONCLUSIONS: The potential effect of a FOXN1 variant on T-cell output from the thymus may relate to its effects on transcriptional activity, nuclear localization, and/or dominant negative functions. A combination of functional assays and thymopoiesis comparisons enabled a categorization of diverse FOXN1 variants and their potential impact on T-cell output from the thymus.


Sujet(s)
Lymphocytes T , Thymus (glande) , Animaux , Humains , Souris , Différenciation cellulaire , Cellules épithéliales/métabolisme , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Phénotype , Lymphocytes T/métabolisme
10.
Front Immunol ; 14: 1159341, 2023.
Article de Anglais | MEDLINE | ID: mdl-37251390

RÉSUMÉ

Robust human immune system (HIS) mice are created using human fetal thymus tissue and hematopoietic stem cells (HSCs). A HIS mouse model using neonatal human thymus tissue and umbilical cord blood (CB) HSCs (NeoHu) was recently described. We improved the model by removing the native murine thymus, which can also generate human T cells, and demonstrated definitively the capacity of human T cells to develop in a grafted neonatal human thymus. Human T cells derived from the neonatal thymus tissue appeared in peripheral blood early post-transplantation and CB-derived T cells appeared later. Naïve T cells were demonstrated in peripheral blood but effector memory and T peripheral helper phenotypes predominated later, in association with development of autoimmunity in some animals. Treatment of thymus grafts with 2-deoxyglucose (2-DG) increased the proportion of stem cells derived from injected HSCs, delayed onset of autoimmune disease, reduced early T cell reconstitution, and reduced effector/memory T cell conversion. Younger neonatal human thymus tissue was associated with improved T cell reconstitution. While the NeoHu model bypasses the need for fetal tissue, it has yet to demonstrate equivalent reconstitution to fetal tissue, though 2-DG can improve results by removing native thymocytes prior to transplantation.


Sujet(s)
Système immunitaire , Thymus (glande) , Humains , Animaux , Souris , Thymocytes , Cellules souches hématopoïétiques , Phénotype
11.
Turk J Pediatr ; 65(1): 73-80, 2023.
Article de Anglais | MEDLINE | ID: mdl-36866987

RÉSUMÉ

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.


Sujet(s)
Lymphocytes T , Thymocytes , Enfant , Enfant d'âge préscolaire , Humains , Nourrisson , Nouveau-né , Sang foetal , Antigènes CD45 , Mutation , Turquie/épidémiologie , Thymocytes/cytologie , Lymphocytes T/cytologie , Valeurs de référence
12.
Elife ; 122023 03 30.
Article de Anglais | MEDLINE | ID: mdl-36995951

RÉSUMÉ

T-cell receptors (TCRs) are formed by stochastic gene rearrangements, theoretically generating >1019 sequences. They are selected during thymopoiesis, which releases a repertoire of about 108 unique TCRs per individual. How evolution shaped a process that produces TCRs that can effectively handle a countless and evolving set of infectious agents is a central question of immunology. The paradigm is that a diverse enough repertoire of TCRs should always provide a proper, though rare, specificity for any given need. Expansion of such rare T cells would provide enough fighters for an effective immune response and enough antigen-experienced cells for memory. We show here that human thymopoiesis releases a large population of clustered CD8+ T cells harboring α/ß paired TCRs that (i) have high generation probabilities and (ii) a preferential usage of some V and J genes, (iii) which CDR3 are shared between individuals, and (iv) can each bind and be activated by multiple unrelated viral peptides, notably from EBV, CMV, and influenza. These polyspecific T cells may represent a first line of defense that is mobilized in response to infections before a more specific response subsequently ensures viral elimination. Our results support an evolutionary selection of polyspecific α/ß TCRs for broad antiviral responses and heterologous immunity.


Sujet(s)
Antigènes viraux , Lymphocytes T CD8+ , Humains , Antigènes viraux/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/génétique , Récepteurs aux antigènes des cellules T/génétique , Peptides
13.
Transpl Immunol ; 75: 101699, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35988896

RÉSUMÉ

Shikonin (SHK) has multifaceted physiological functions, including antitumor, anti-inflammatory, and antimicrobial effects. Recently, SHK has been shown to affect immune responses; however, its detailed immune modulatory function in vivo remains unclear. In this study, we demonstrated that SHK not only inhibited T cell proliferation in vitro, but also intensively inhibited thymopoiesis and eliminated CD4/CD8 double-positive thymic progenitor cells in vivo. Treatment of mice with SHK resulted in immune profile alterations, which promoted myelosis in the bone marrow and increased inhibitory immune cells in central immune organs. A decrease in T cells and B cells was observed in the spleen. Using a murine allogenic skin transplantation model, we revealed that short-term treatment of recipients with SHK significantly inhibited skin graft rejection, in which the levels of myeloid-derived suppressor cells (MDSC) were markedly increased. Taken together, our study suggests that SHK can efficiently eliminate proliferating T cells and inhibit thymopoiesis while promoting the generation of MDSC, indicating its potential role in alleviating immune responses in allogeneic organ/cell transplantation.


Sujet(s)
Cellules myéloïdes suppressives , Souris , Animaux , Souris de lignée C57BL , Activation des lymphocytes , Lymphocytes T , Prolifération cellulaire
14.
Cell Rep ; 40(1): 111050, 2022 07 05.
Article de Anglais | MEDLINE | ID: mdl-35793622

RÉSUMÉ

Many aspects of the porcine immune system remain poorly characterized, which poses a barrier to improving swine health and utilizing pigs as preclinical models. Here, we employ single-cell RNA sequencing (scRNA-seq) to create a cell atlas of the early-adolescent pig thymus. Our data show conserved features as well as species-specific differences in cell states and cell types compared with human thymocytes. We also describe several unconventional T cell types with gene expression profiles associated with innate effector functions. This includes a cell census of more than 11,000 differentiating invariant natural killer T (iNKT) cells, which reveals that the functional diversity of pig iNKT cells differs substantially from the iNKT0/1/2/17 subset differentiation paradigm established in mice. Our data characterize key differentiation events in porcine thymopoiesis and iNKT cell maturation and provide important insights into pig T cell development.


Sujet(s)
Cellules T tueuses naturelles , Animaux , Différenciation cellulaire/génétique , Souris , Analyse sur cellule unique , Suidae , Thymocytes
15.
Cell Immunol ; 377: 104554, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35636065

RÉSUMÉ

T-cell-mediated immune responses play indispensable roles in the defense against infectious pathogens including human immunodeficiency virus type 1 (HIV-1) which can establish a persistent infection that leads to many alterations in T-cell-mediated immunity. The latter include T-cell hyperactivation and depletion, both of which are essential for disease progression. Determining the factors and mechanisms pathways that lead to such abnormalities in T-cell mediated immunity during HIV-1 infection and ascertaining how the virus is able to evade immune responses elicited by T cells are critical for understanding the pathophysiology of HIV-1 infection, which in turn, could lead to new insights that may accelerate the development of novel and effective therapeutic strategies. To this end, we addressed the roles played by HIV-1 Tat protein, one of the first proteins to be expressed, in the pathogenesis of HIV-1 infection, focusing on the pathological effects of this protein in the cellular adaptive immune response in which T cells are intimately involved.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Lymphocytes T CD4+ , Humains , Produits du gène tat du virus de l'immunodéficience humaine
16.
Front Nutr ; 9: 878382, 2022.
Article de Anglais | MEDLINE | ID: mdl-35529463

RÉSUMÉ

Modern industrial practices have transformed the human diet over the last century, increasing the consumption of processed foods. Dietary imbalance of macro- and micro-nutrients and excessive caloric intake represent significant risk factors for various inflammatory disorders. Increased ingestion of food additives, residual contaminants from agricultural practices, food processing, and packaging can also contribute deleteriously to disease development. One common hallmark of inflammatory disorders, such as autoimmunity and allergies, is the defect in anti-inflammatory regulatory T cell (Treg) development and/or function. Treg represent a highly heterogeneous population of immunosuppressive immune cells contributing to peripheral tolerance. Tregs either develop in the thymus from autoreactive thymocytes, or in the periphery, from naïve CD4+ T cells, in response to environmental antigens and cues. Accumulating evidence demonstrates that various dietary factors can directly regulate Treg development. These dietary factors can also indirectly modulate Treg differentiation by altering the gut microbiota composition and thus the production of bacterial metabolites. This review provides an overview of Treg ontogeny, both thymic and peripherally differentiated, and highlights how diet and gut microbiota can regulate Treg development and function.

17.
Curr Protoc ; 2(4): e403, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-35384408

RÉSUMÉ

The generation of T cells is a complex, carefully orchestrated process that occurs in the thymus. The ability to mimic T cell differentiation in vitro has opened up avenues to better understand different stages of thymopoiesis but has also enabled the in vitro production of mature T cells suitable for immunotherapy. Among existing protocols, the artificial thymic organoid (ATO) system has been shown to be the most efficient at producing mature conventional T cells. In this serum-free model, human or murine hematopoietic stem and progenitor cells (HSPCs) are combined with a murine stromal cell line expressing a Notch ligand in a 3D cell aggregate. In ATOs, although only simple medium changes are required throughout the cultures, HSPCs differentiate into T cells with kinetics and phenotypes similar to those of endogenous thymopoiesis. This article describes protocols for the generation of ATOs from human and murine HSPCs. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Expansion and preparation of MS5-hDLL4 or MS5-mDLL4 cells Basic Protocol 2: Isolation of human hematopoietic stem and progenitor cells (HSPCs; CD34+ cells) Support Protocol 1: Transduction of human HSPCs (CD34+ cells) Basic Protocol 3: Production of thymic progenitors and mature T cells from human HSPCs in artificial thymic organoids (ATOs) Support Protocol 2: Phenotype analysis of human ATO cells by flow cytometry Basic Protocol 4: Isolation of murine HSPCs (Lin- Sca1+ cKit+; LSK) and hematopoietic stem cells (LSK CD150+ CD48-) Basic Protocol 5: Production of thymic progenitors and mature T cells from murine HSPCs in ATOs Support Protocol 3: Phenotype analysis of murine ATO cells by flow cytometry Alternate Protocol: Generation of ATOs from single HSPCs.


Sujet(s)
Cellules souches hématopoïétiques , Organoïdes , Animaux , Différenciation cellulaire , Hématopoïèse , Humains , Souris , Lymphocytes T
18.
J Leukoc Biol ; 112(4): 641-657, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-35258130

RÉSUMÉ

Thymocyte differentiation and lineage commitment is regulated by an extensive network of transcription factors and signaling molecules among which Erk plays a central role. However, Erk effectors as well as the molecular mechanisms underlying this network are not well understood. Erf is a ubiquitously expressed transcriptional repressor regulated by Erk-dependent phosphorylation. Here, we investigated the role of Erf in T cell maturation and lineage commitment, using a double-fluorescent Erf-floxed mouse to produce thymus-specific Erf knockouts. We observed significant accumulation of thymocytes in the CD4/CD8 DP stage, followed by a significant reduction in CD4SP cells, a trend for lower CD8SP cell frequency, and an elevated percentage of γδ expressing thymocytes in Erf-deficient mice. Also, an elevated number of CD69+ TCRß+ cells indicates that thymocytes undergoing positive selection accumulate at this stage. The expression of transcription factors Gata3, ThPOK, and Socs1 that promote CD4+ cell commitment was significantly decreased in Erf-deficient mice. These findings suggest that Erf is involved in T cell maturation, acting as a positive regulator during CD4 and eventually CD8 lineage commitment, while negatively regulates the production of γδ T cells. In addition, Erf-deficient mice displayed decreased percentages of CD4+ and CD8+ splenocytes and elevated levels of IL-4 indicating that Erf may have an additional role in the homeostasis, differentiation, and immunologic response of helper and cytotoxic T cells in the periphery. Overall, our results show, for the first time, Erf's involvement in T cell biology suggesting that Erf acts as a potential regulator during thymocyte maturation and thymocyte lineage commitment, in γδ T cell generation, as well as in Th cell differentiation.


Sujet(s)
Interleukine-4 , Thymocytes , Animaux , Lymphocytes T CD4+ , Lymphocytes T CD8+ , Différenciation cellulaire , Lignage cellulaire , Facteur de transcription GATA-3/métabolisme , Interleukine-4/métabolisme , Souris , Protéines de répression , Thymus (glande)
19.
Virology ; 567: 77-86, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35032866

RÉSUMÉ

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Sujet(s)
Atrophie/virologie , Interféron alpha/immunologie , Interféron bêta/immunologie , Chorioméningite lymphocytaire/virologie , Virus de la chorioméningite lymphocytaire/immunologie , Thymocytes/virologie , Thymus (glande)/virologie , Animaux , Atrophie/génétique , Atrophie/immunologie , Atrophie/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/virologie , Maladie chronique , Femelle , Régulation de l'expression des gènes , Humains , Mémoire immunologique , Interféron alpha/génétique , Interféron bêta/génétique , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Noeuds lymphatiques/virologie , Déplétion lymphocytaire , Chorioméningite lymphocytaire/génétique , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/anatomopathologie , Virus de la chorioméningite lymphocytaire/pathogénicité , Souris , Souris de lignée C57BL , Souris knockout , Récepteur à l'interféron alpha-bêta/déficit , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/immunologie , Transduction du signal/immunologie , Analyse sur cellule unique , Thymocytes/immunologie , Thymocytes/anatomopathologie , Thymus (glande)/immunologie , Thymus (glande)/anatomopathologie
20.
Front Immunol ; 12: 716661, 2021.
Article de Anglais | MEDLINE | ID: mdl-34394122

RÉSUMÉ

Although metabolic pathways have been shown to control differentiation and activation in peripheral T cells, metabolic studies on thymic T cell development are still lacking, especially in human tissue. In this study, we use transcriptomics and extracellular flux analyses to investigate the metabolic profiles of primary thymic and in vitro-derived mouse and human thymocytes. Core metabolic pathways, specifically glycolysis and oxidative phosphorylation, undergo dramatic changes between the double-negative (DN), double-positive (DP), and mature single-positive (SP) stages in murine and human thymus. Remarkably, despite the absence of the complex multicellular thymic microenvironment, in vitro murine and human T cell development recapitulated the coordinated decrease in glycolytic and oxidative phosphorylation activity between the DN and DP stages seen in primary thymus. Moreover, by inducing in vitro T cell differentiation from Rag1-/- mouse bone marrow, we show that reduced metabolic activity at the DP stage is independent of TCR rearrangement. Thus, our findings suggest that highly conserved metabolic transitions are critical for thymic T cell development.


Sujet(s)
Différenciation cellulaire , Métabolisme énergétique , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Thymocytes/cytologie , Thymocytes/métabolisme , Animaux , Évolution biologique , Marqueurs biologiques , Lignée cellulaire , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/métabolisme , Humains , Lymphopoïèse , Métabolome , Métabolomique/méthodes , Souris , Organoïdes , Thymocytes/immunologie , Techniques de culture de tissus
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE