4,544 results on '"Thymocytes"'
Search Results
2. Fam49b dampens TCR signal strength to regulate survival of positively selected thymocytes and peripheral T cells.
- Author
-
Park, Chan-Su, Guan, Jian, Rhee, Peter, Gonzalez, Federico, Lee, Hee-Sung, Park, Ji-Hyun, Coscoy, Laurent, Robey, Ellen, Shastri, Nilabh, and Sadegh-Nasseri, Scheherazade
- Subjects
Fam49b ,Rac ,T cell development ,cytoskeleton remodeling ,immunology ,inflammation ,intraepithelial T cells ,mouse ,negative selection ,Animals ,Mice ,Cell Survival ,Mice ,Knockout ,Receptors ,Antigen ,T-Cell ,Signal Transduction ,T-Lymphocytes ,Thymocytes - Abstract
The fate of developing T cells is determined by the strength of T cell receptor (TCR) signal they receive in the thymus. This process is finely regulated through the tuning of positive and negative regulators in thymocytes. The Family with sequence similarity 49 member B (Fam49b) protein is a newly discovered negative regulator of TCR signaling that has been shown to suppress Rac-1 activity in vitro in cultured T cell lines. However, the contribution of Fam49b to the thymic development of T cells is unknown. To investigate this important issue, we generated a novel mouse line deficient in Fam49b (Fam49b-KO). We observed that Fam49b-KO double positive (DP) thymocytes underwent excessive negative selection, whereas the positive selection stage was unaffected. Fam49b deficiency impaired the survival of single positive thymocytes and peripheral T cells. This altered development process resulted in significant reductions in CD4 and CD8 single-positive thymocytes as well as peripheral T cells. Interestingly, a large proportion of the TCRγδ+ and CD8αα+TCRαβ+ gut intraepithelial T lymphocytes were absent in Fam49b-KO mice. Our results demonstrate that Fam49b dampens thymocytes TCR signaling in order to escape negative selection during development, uncovering the function of Fam49b as a critical regulator of the selection process to ensure normal thymocyte development and peripheral T cells survival.
- Published
- 2024
3. Systemic immunostimulation induces glucocorticoid-mediated thymic involution succeeded by rebound hyperplasia which is impaired in aged recipients
- Author
-
Collins, Craig P, Khuat, Lam T, Sckisel, Gail D, Vick, Logan V, Minnar, Christine M, Dunai, Cordelia, Le, Catherine T, Curti, Brendan D, Crittenden, Marka, Merleev, Alexander, Sheng, Michael, Chao, Nelson J, Maverakis, Emanual, Rosario, Spencer R, Monjazeb, Arta M, Blazar, Bruce R, Longo, Dan L, Canter, Robert J, and Murphy, William J
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Infectious Diseases ,Aging ,Cancer ,1.1 Normal biological development and functioning ,Inflammatory and immune system ,Animals ,Thymus Gland ,Mice ,Humans ,Glucocorticoids ,Female ,Male ,Aged ,Middle Aged ,Interleukin-2 ,Adult ,Thymocytes ,Thymus Hyperplasia ,Mice ,Inbred C57BL ,Immunization ,Hyperplasia ,immune therapy ,thymic involution ,age ,viral infection ,stress ,glucocoricoids ,Medical Microbiology ,Biochemistry and cell biology ,Genetics - Abstract
The thymus is the central organ involved with T-cell development and the production of naïve T cells. During normal aging, the thymus undergoes marked involution, reducing naïve T-cell output and resulting in a predominance of long-lived memory T cells in the periphery. Outside of aging, systemic stress responses that induce corticosteroids (CS), or other insults such as radiation exposure, induce thymocyte apoptosis, resulting in a transient acute thymic involution with subsequent recovery occurring after cessation of the stimulus. Despite the increasing utilization of immunostimulatory regimens in cancer, effects on the thymus and naïve T cell output have not been well characterized. Using both mouse and human systems, the thymic effects of systemic immunostimulatory regimens, such as high dose IL-2 (HD IL-2) with or without agonistic anti-CD40 mAbs and acute primary viral infection, were investigated. These regimens produced a marked acute thymic involution in mice, which correlated with elevated serum glucocorticoid levels and a diminishment of naïve T cells in the periphery. This effect was transient and followed with a rapid thymic "rebound" effect, in which an even greater quantity of thymocytes was observed compared to controls. Similar results were observed in humans, as patients receiving HD IL-2 treatment for cancer demonstrated significantly increased cortisol levels, accompanied by decreased peripheral blood naïve T cells and reduced T-cell receptor excision circles (TRECs), a marker indicative of recent thymic emigrants. Mice adrenalectomized prior to receiving immunotherapy or viral infection demonstrated protection from this glucocorticoid-mediated thymic involution, despite experiencing a substantially higher inflammatory cytokine response and increased immunopathology. Investigation into the effects of immunostimulation on middle aged (7-12 months) and advance aged (22-24 months) mice, which had already undergone significant thymic involution and had a diminished naïve T cell population in the periphery at baseline, revealed that even further involution was incurred. Thymic rebound hyperplasia, however, only occurred in young and middle-aged recipients, while advance aged not only lacked this rebound hyperplasia, but were entirely absent of any indication of thymic restoration. This coincided with prolonged deficits in naïve T cell numbers in advanced aged recipients, further skewing the already memory dominant T cell pool. These results demonstrate that, in both mice and humans, systemic immunostimulatory cancer therapies, as well as immune challenges like subacute viral infections, have the potential to induce profound, but transient, glucocorticoid-mediated thymic involution and substantially reduced thymic output, resulting in the reduction of peripheral naive T cells. This can then be followed by a marked rebound effect with naïve T cell restoration, events that were shown not to occur in advanced-aged mice.
- Published
- 2024
4. The single-cell transcriptome of mTECs and CD4+ thymocytes under adhesion revealed heterogeneity of mTECs and a network controlled by Aire and lncRNAs.
- Author
-
Monteiro, Cíntia J., Duarte, Max J., Machado, Mayara Cristina V., Mascarenhas, Romário S., Bonini Palma, Patrícia V., García, Henry D. Mogollón, Nakaya, Helder I., Cunha, Thiago M., Donadi, Eduardo A., and Passos, Geraldo A.
- Subjects
LINCRNA ,GENE expression ,CELL metabolism ,CELL cycle ,THYMOCYTES - Abstract
To further understand the impact of deficiency of the autoimmune regulator (Aire) gene during the adhesion of medullary thymic epithelial cells (mTECs) to thymocytes, we sequenced single-cell libraries (scRNA-seq) obtained from Aire wild-type (WT) (Aire
wt/wt ) or Aire-deficient (Airewt/mut ) mTECs cocultured with WT single-positive (SP) CD4+ thymocytes. Although the libraries differed in their mRNA and long noncoding RNA (lncRNA) profiles, indicating that mTECs were heterogeneous in terms of their transcriptome, UMAP clustering revealed that both mTEC lines expressed their specific markers, i.e., Epcam, Itgb4, Itga6, and Casp3 in resting mTECs and Ccna2, Pbk, and Birc5 in proliferative mTECs. Both cocultured SP CD4+ thymocytes remained in a homogeneous cluster expressing the Il7r and Ccr7markers. Comparisons of the two types of cocultures revealed the differential expression of mRNAs that encode transcription factors (Zfpm2, Satb1, and Lef1), cell adhesion genes (Itgb1) inmTECs, and Themis in thymocytes, which is associated with the regulation of positive and negative selection. At the single-cell sequencing resolution, we observed that Aire acts on both Aire WT and Airedeficient mTECs as an upstream controller of mRNAs, which encode transcription factors or adhesion proteins that, in turn, are posttranscriptionally controlled by lncRNAs, for example, Neat1, Malat1, Pvt1, and Dancr among others. Under Aire deficiency, mTECs dysregulate the expression of MHC-II, CD80, and CD326 (EPCAM) protein markers as well as metabolism and cell cycle-related mRNAs, which delay the cell cycle progression. Moreover, when adhered to mTECs, WT SP CD4+ or CD8+ thymocytes modulate the expression of cell activation proteins, including CD28 and CD152/CTLA4, and the expression of cellular metabolism mRNAs. These findings indicate a complex mechanism through which an imbalance in Aire expression can affect mTECs and thymocytes during adhesion. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
5. Single-cell multiomic analysis of thymocyte development reveals drivers of CD4+ T cell and CD8+ T cell lineage commitment.
- Author
-
Steier, Zoë, Aylard, Dominik, McIntyre, Laura, Baldwin, Isabel, Kim, Esther, Lutes, Lydia, Ergen, Can, Huang, Tse-Shun, Yosef, Nir, Robey, Ellen, and Streets, Aaron
- Subjects
Mice ,Animals ,CD8-Positive T-Lymphocytes ,Cell Lineage ,CD4-Positive T-Lymphocytes ,Thymocytes ,Multiomics ,Mice ,Transgenic ,Cell Differentiation ,Receptors ,Antigen ,T-Cell ,Thymus Gland ,Histocompatibility Antigens Class I ,CD4 Antigens - Abstract
The development of CD4+ T cells and CD8+ T cells in the thymus is critical to adaptive immunity and is widely studied as a model of lineage commitment. Recognition of self-peptide major histocompatibility complex (MHC) class I or II by the T cell antigen receptor (TCR) determines the CD8+ or CD4+ T cell lineage choice, respectively, but how distinct TCR signals drive transcriptional programs of lineage commitment remains largely unknown. Here we applied CITE-seq to measure RNA and surface proteins in thymocytes from wild-type and T cell lineage-restricted mice to generate a comprehensive timeline of cell states for each T cell lineage. These analyses identified a sequential process whereby all thymocytes initiate CD4+ T cell lineage differentiation during a first wave of TCR signaling, followed by a second TCR signaling wave that coincides with CD8+ T cell lineage specification. CITE-seq and pharmaceutical inhibition experiments implicated a TCR-calcineurin-NFAT-GATA3 axis in driving the CD4+ T cell fate. Our data provide a resource for understanding cell fate decisions and implicate a sequential selection process in guiding lineage choice.
- Published
- 2023
6. Type B thymomas in patients with myasthenia gravis display a distinctive pattern of αβ TCR and IL-7 receptor α expression on CD4+CD8+ thymocytes.
- Author
-
Tianlai Wang, Boyu Wang, Xiaowu Fan, Yixin Cai, Lequn Li, and Shengling Fu
- Subjects
- *
MYASTHENIA gravis , *THYMOCYTES , *T cell receptors , *CELL populations , *LIGANDS (Biochemistry) , *T cells , *TRANSCRIPTION factors - Abstract
Thymoma is closely associated with myasthenia gravis (MG). However, due to the heterogeneity of thymoma and the intricate pathogenesis of MG, it remains unclear why some patients with thymoma develop MG and others do not. in this study, we conducted a comparative phenotype analysis of thymocytes in type B thymomas in patients with MG (MG (+) thymomas) and without MG (MG (-) thymomas) via fluorescence-activated cell sorting (FACS). Our results show that the developmental stages defined by the expression of CD3, CD4, and CD8 were largely maintained in both MG (+) and MG (-) thymomas, with CD4+CD8+ cells constituting the majority of thymocytes in type B thymoma, and no significant difference between this cell population was observed in MG (+) and MG (-) thymomas. We discovered that CD4+CD8+ thymocytes in MG (+) thymomas expressed low levels of αβ tcR and high levels of IL-7 receptor α (IL-7Rα), whereas in MG (-) thymomas, cD4+cD8+ thymocytes exhibited the opposite pattern of αβ TCR and il-7Rα expression. these results suggest that the positive and negative selection processes of CD4+CD8+ thymocytes might differ between MG (+) thymomas and MG (-) thymomas. the expression of the helios transcription factor is induced during negative selection and marks a group of t cells that have undergone negative selection and are likely to be deleted due to strong TCR binding with self-peptides/MHC ligands. We observed that the percentage of helios-positive CD4SP t cells was greater in MG (-) than in MG (+) thymomas. thus, the differentially regulated selection process of CD4+CD8+ thymocytes, which involves TCR and IL-7/IL-7Rα signaling, is associated with the presence of MG in type B thymomas. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Fam49b dampens TCR signal strength to regulate survival of positively selected thymocytes and peripheral T cells.
- Author
-
Chan-Su Park, Jian Guan, Rhee, Peter, Gonzalez, Federico, Hee-sung Lee, Ji-hyun Park, Coscoy, Laurent, Robey, Ellen A., Shastri, Nilabh, and Sadegh-Nasseri, Scheherazade
- Subjects
- *
T cells , *INTERLEUKIN-7 , *THYMOCYTES , *T cell receptors , *CELL survival - Abstract
The fate of developing T cells is determined by the strength of T cell receptor (TCR) signal they receive in the thymus. This process is finely regulated through the tuning of positive and negative regulators in thymocytes. The Family with sequence similarity 49 member B (Fam49b) protein is a newly discovered negative regulator of TCR signaling that has been shown to suppress Rac-1 activity in vitro in cultured T cell lines. However, the contribution of Fam49b to the thymic development of T cells is unknown. To investigate this important issue, we generated a novel mouse line deficient in Fam49b (Fam49b-KO). We observed that Fam49b-KO double positive (DP) thymocytes underwent excessive negative selection, whereas the positive selection stage was unaffected. Fam49b deficiency impaired the survival of single positive thymocytes and peripheral T cells. This altered development process resulted in significant reductions in CD4 and CD8 singlepositive thymocytes as well as peripheral T cells. Interestingly, a large proportion of the TCRγδ+ and CD8αα+TCRαβ+ gut intraepithelial T lymphocytes were absent in Fam49b-KO mice. Our results demonstrate that Fam49b dampens thymocytes TCR signaling in order to escape negative selection during development, uncovering the function of Fam49b as a critical regulator of the selection process to ensure normal thymocyte development and peripheral T cells survival. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. In Vivo Tracking and 3D Mapping of Cell Death in Regeneration and Cancer Using Trypan Blue.
- Author
-
Procel, Nicole, Camacho, Karen, Verboven, Elisabeth, Baroja, Isabel, Guerrero, Priscila A., Hillen, Hanne, Estrella-García, Carlos, Vizcaíno-Rodríguez, Nicole, Sansores-Garcia, Leticia, Santamaría-Naranjo, Ana, Romero-Carvajal, Andrés, Caicedo, Andrés, Halder, Georg, and Moya, Iván M.
- Subjects
- *
LIVER cells , *CELL death , *TRYPAN blue , *CANCER cells , *THYMOCYTES - Abstract
Tracking cell death in vivo can enable a better understanding of the biological mechanisms underlying tissue homeostasis and disease. Unfortunately, existing cell death labeling methods lack compatibility with in vivo applications or suffer from low sensitivity, poor tissue penetration, and limited temporal resolution. Here, we fluorescently labeled dead cells in vivo with Trypan Blue (TBlue) to detect single scattered dead cells or to generate whole-mount three-dimensional maps of large areas of necrotic tissue during organ regeneration. TBlue effectively marked different types of cell death, including necrosis induced by CCl4 intoxication in the liver, necrosis caused by ischemia-reperfusion in the skin, and apoptosis triggered by BAX overexpression in hepatocytes. Moreover, due to its short circulating lifespan in blood, TBlue labeling allowed in vivo "pulse and chase" tracking of two temporally spaced populations of dying hepatocytes in regenerating mouse livers. Additionally, upon treatment with cisplatin, TBlue labeled dead cancer cells in livers with cholangiocarcinoma and dead thymocytes due to chemotherapy-induced toxicity, showcasing its utility in assessing anticancer therapies in preclinical models. Thus, TBlue is a sensitive and selective cell death marker for in vivo applications, facilitating the understanding of the fundamental role of cell death in normal biological processes and its implications in disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Glucocorticoids impair T lymphopoiesis after myocardial infarction.
- Author
-
Shane, Danielle X., Konovalova, Daria M., Rajendran, Harishkumar, Yuan, Sarah Y., and Ma, Yonggang
- Subjects
- *
MYOCARDIAL infarction , *GLUCOCORTICOIDS , *THYMOCYTES , *ADRENAL glands , *T cells - Abstract
The thymus, where T lymphocytes develop and mature, is sensitive to insults such as tissue ischemia or injury. The insults can cause thymic atrophy and compromise T-cell development, potentially impairing adaptive immunity. The objective of this study was to investigate whether myocardial infarction (MI) induces thymic injury to impair T lymphopoiesis and to uncover the underlying mechanisms. When compared with sham controls, MI mice at day 7 post-MI exhibited smaller thymus, lower cellularity, as well as less thymocytes at different developmental stages, indicative of T-lymphopoiesis impairment following MI. Accordingly, the spleen of MI mice has less T cells and recent thymic emigrants (RTEs), implying that the thymus of MI mice releases fewer mature thymocytes than sham controls. Interestingly, the secretory function of splenic T cells was not affected by MI. Further experiments showed that the reduction of thymocytes in MI mice was due to increased thymocyte apoptosis. Removal of adrenal glands by adrenalectomy (ADX) prevented MI-induced thymic injury and dysfunction, whereas corticosterone supplementation in ADX + MI mice reinduced thymic injury and dysfunction, indicating that glucocorticoids mediate thymic damage triggered by MI. Eosinophils play essential roles in thymic regeneration postirradiation, and eosinophil-deficient mice exhibit impaired thymic recovery after sublethal irradiation. Interestingly, the thymus was fully regenerated in both wild-type and eosinophil-deficient mice at day 14 post-MI, suggesting that eosinophils are not critical for thymus regeneration post-MI. In conclusion, our study demonstrates that MI-induced glucocorticoids trigger thymocyte apoptosis and impair T lymphopoiesis, resulting in less mature thymocyte release to the spleen. NEW & NOTEWORTHY: The thymus is essential for maintaining whole body T-cell output. Thymic injury can adversely affect T lymphopoiesis and T-cell immune response. This study demonstrates that MI induces thymocyte apoptosis and compromises T lymphopoiesis, resulting in fewer releases of mature thymocytes to the spleen. This process is mediated by glucocorticoids secreted by adrenal glands. Therefore, targeting glucocorticoids represents a novel approach to attenuate post-MI thymic injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Epigallocatechin‐3‐gallate ameliorates lipopolysaccharide‐induced acute thymus involution in mice via AMPK/Sirt1 pathway.
- Author
-
Su, Qing, Yang, Shu‐Ping, Guo, Jun‐Ping, Rong, Yi‐Ren, Sun, Yun, and Chai, Yu‐Rong
- Subjects
SIRTUINS ,MEMBRANE potential ,REACTIVE oxygen species ,MITOCHONDRIAL membranes ,SUPEROXIDE dismutase - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. PIM1 is a potential therapeutic target for the leukemogenic effects mediated by JAK/STAT pathway mutations in T-ALL/LBL.
- Author
-
Lahera, Antonio, Vela-Martín, Laura, Fernández-Navarro, Pablo, Llamas, Pilar, López-Lorenzo, José L., Cornago, Javier, Santos, Javier, Fernández-Piqueras, José, and Villa-Morales, María
- Subjects
DRUG target ,BRAF genes ,GENETIC overexpression ,GENETIC mutation ,HEMATOLOGIC malignancies ,THYMOCYTES - Abstract
Precursor T-cell neoplasms (T-ALL/LBL) are aggressive hematological malignancies that arise from the malignant transformation of immature thymocytes. Despite the JAK/STAT pathway is recurrently altered in these neoplasms, there are not pharmacological inhibitors officially approved for the treatment of T-ALL/LBL patients that present oncogenic JAK/STAT pathway mutations. In the effort to identify potential therapeutic targets for those patients, we followed an alternative approach and focused on their transcriptional profile. We combined the analysis of molecular data from T-ALL/LBL patients with the generation of hematopoietic cellular models to reveal that JAK/STAT pathway mutations are associated with an aberrant transcriptional profile. Specifically, we demonstrate that JAK/STAT pathway mutations induce the overexpression of the PIM1 gene. Moreover, we show that the pan-PIM inhibitor, PIM447, significantly reduces the leukemogenesis, as well as the aberrant activation of c-MYC and mTOR pathways in cells expressing different JAK/STAT pathway mutations, becoming a potential therapeutic opportunity for a relevant subset of T-ALL/LBL patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. MHC heterozygosity limits T cell receptor variability in CD4 T cells.
- Author
-
Brown, Alexander J., White, Janice, Shaw, Laura, Gross, Jimmy, Slabodkin, Andrei, Kushner, Ella, Greiff, Victor, Matsuda, Jennifer, Gapin, Laurent, Scott-Browne, James, Kappler, John, and Marrack, Philippa
- Subjects
T cell receptors ,T cells ,CD4 antigen ,GENETIC code ,THYMOCYTES - Abstract
αβ T cell receptor (TCR) V(D)J genes code for billions of TCR combinations. However, only some appear on peripheral T cells in any individual because, to mature, thymocytes must react with low affinity but not high affinity with thymus expressed major histocompatibility (MHC)/peptides. MHC proteins are very polymorphic. Different alleles bind different peptides. Therefore, any individual might express many different MHC alleles to ensure that some peptides from an invader are bound to MHC and activate T cells. However, most individuals express limited numbers of MHC alleles. To explore this, we compared the TCR repertoires of naïve CD4 T cells in mice expressing one or two MHC alleles. Unexpectedly, the TCRs in heterozygotes were less diverse that those in the sum of their MHC homozygous relatives. Our results suggest that thymus negative selection cancels out the advantages of increased thymic positive selection in the MHC heterozygotes. Editor's summary: T cells undergo positive and negative selection in the thymus through T cell receptor (TCR) recognition of peptides presented on major histocompatibility (MHC) proteins. Although the genes encoding MHC proteins are among the most polymorphic, most vertebrates individually express relatively few MHC alleles. How the number of MHC alleles expressed affects thymic selection and the TCR repertoire on an individual level remains unclear. Using mice expressing different MHC loci on the same background, Brown et al. compared the TCR repertoires of naïve CD4 T cells from MHC homozygous and heterozygous mice. MHC heterozygotes expressed a less diverse TCR repertoire than expected compared with their MHC homozygous relatives, likely because of increased rates of negative selection. These findings suggest that the number of MHC alleles expressed in an individual animal has been evolutionarily optimized to minimize "holes" in the TCR repertoire. —Claire Olingy [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Transcriptomic profiling of thymic dysregulation and viral tropism after neonatal roseolovirus infection.
- Author
-
Belean, Andrei, Eden Xue, Cisneros, Benjamin, Roberson, Elisha D. O., Paley, Michael A., and Bigley, Tarin M.
- Subjects
NEONATAL infections ,VIRAL tropism ,TRANSCRIPTOMES ,VIRAL genes ,CELL populations ,RNA sequencing - Abstract
Introduction: Herpesviruses, including the roseoloviruses, have been linked to autoimmune disease. The ubiquitous and chronic nature of these infections have made it difficult to establish a causal relationship between acute infection and subsequent development of autoimmunity. We have shown that murine roseolovirus (MRV), which is highly related to human roseoloviruses, induces thymic atrophy and disruption of central tolerance after neonatal infection. Moreover, neonatal MRV infection results in development of autoimmunity in adult mice, long after resolution of acute infection. This suggests that MRV induces durable immune dysregulation. Methods: In the current studies, we utilized single-cell RNA sequencing (scRNAseq) to study the tropism of MRV in the thymus and determine cellular processes in the thymus that were disrupted by neonatal MRV infection. We then utilized tropism data to establish a cell culture system. Results: Herein, we describe how MRV alters the thymic transcriptome during acute neonatal infection. We found that MRV infection resulted in major shifts in inflammatory, differentiation and cell cycle pathways in the infected thymus. We also observed shifts in the relative number of specific cell populations. Moreover, utilizing expression of late viral transcripts as a proxy of viral replication, we identified the cellular tropism of MRV in the thymus. This approach demonstrated that double negative, double positive, and CD4 single positive thymocytes, as well as medullary thymic epithelial cells were infected by MRV in vivo. Finally, by applying pseudotime analysis to viral transcripts, which we refer to as “pseudokinetics,” we identified viral gene transcription patterns associated with specific cell types and infection status. We utilized this information to establish the first cell culture systems susceptible to MRV infection in vitro. Conclusion: Our research provides the first complete picture of roseolovirus tropism in the thymus after neonatal infection. Additionally, we identified major transcriptomic alterations in cell populations in the thymus during acute neonatal MRV infection. These studies offer important insight into the early events that occur after neonatal MRV infection that disrupt central tolerance and promote autoimmune disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. The promiscuous development of an unconventional Qa1b-restricted T cell population.
- Author
-
Manoharan Valerio, Michael, Arana, Kathya, Guan, Jian, Chan, Shiao, Yang, Xiaokun, Kurd, Nadia, Lee, Angus, Shastri, Nilabh, Coscoy, Laurent, and Robey, Ellen
- Subjects
IEL ,MHC-E ,T cell development ,non-classical MHC-1 ,unconventional T cells ,Animals ,Mice ,CD8-Positive T-Lymphocytes ,Peptides ,Receptors ,Antigen ,T-Cell ,alpha-beta ,Thymocytes ,Genes ,MHC Class II - Abstract
MHC-E restricted CD8 T cells show promise in vaccine settings, but their development and specificity remain poorly understood. Here we focus on a CD8 T cell population reactive to a self-peptide (FL9) bound to mouse MHC-E (Qa-1b) that is presented in response to loss of the MHC I processing enzyme ERAAP, termed QFL T cells. We find that mature QFL thymocytes are predominantly CD8αβ+CD4-, show signs of agonist selection, and give rise to both CD8αα and CD8αβ intraepithelial lymphocytes (IEL), as well as memory phenotype CD8αβ T cells. QFL T cells require the MHC I subunit β-2 microglobulin (β2m), but do not require Qa1b or classical MHC I for positive selection. However, QFL thymocytes do require Qa1b for agonist selection and full functionality. Our data highlight the relaxed requirements for positive selection of an MHC-E restricted T cell population and suggest a CD8αβ+CD4- pathway for development of CD8αα IELs.
- Published
- 2023
15. KMT2D regulates activation, localization, and integrin expression by T-cells.
- Author
-
Potter, Sarah J., Li Zhang, Kotliar, Michael, Yuehong Wu, Schafer, Caitlin, Stefan, Kurtis, Boukas, Leandros, Qu'd, Dima, Bodamer, Olaf, Simpson, Brittany N., Barski, Artem, Lindsley, Andrew W., and Bjornsson, Hans T.
- Subjects
INTEGRINS ,T cells ,CELLULAR signal transduction ,THYMOCYTES ,GENETIC variation ,CATECHOL-O-methyltransferase - Abstract
Individuals with Kabuki syndrome present with immunodeficiency; however, how pathogenic variants in the gene encoding the histone-modifying enzyme lysine methyltransferase 2D (KMT2D) lead to immune alterations remain poorly understood. Following up on our prior report of KMT2D-altered integrin expression in B-cells, we performed targeted analyses of KMT2D's influence on integrin expression in T-cells throughout development (thymocytes through peripheral T-cells) in murine cells with constitutive- and conditional-targeted Kmt2d deletion. Using high-throughput RNA-sequencing and flow cytometry, we reveal decreased expression (both at the transcriptional and translational levels) of a cluster of leukocyte-specific integrins, which perturb aspects of T-cell activation, maturation, adhesion/localization, and effector function. H3K4me3 ChIP-PCR suggests that these evolutionary similar integrins are under direct control of KMT2D. KMT2D loss also alters multiple downstream programming/signaling pathways, including integrin-based localization, which can influence Tcell populations. We further demonstrated that KMT2D deficiency is associated with the accumulation of murine CD8+ single-positive (SP) thymocytes and shifts in both human and murine peripheral T-cell populations, including the reduction of the CD4+ recent thymic emigrant (RTE) population. Together, these data show that the targeted loss of Kmt2d in the T-cell lineage recapitulates several distinct features of Kabuki syndrome-associated immune deficiency and implicates epigenetic mechanisms in the regulation of integrin signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. The CD4 Versus CD8 T Cell Fate Decision: A Multiomics-Informed Perspective.
- Author
-
Steier, Zoë, Kim, Esther Jeong Yoon, Aylard, Dominik A., and Robey, Ellen A.
- Abstract
The choice of developing thymocytes to become CD8+ cytotoxic or CD4+ helper T cells has been intensely studied, but many of the underlying mechanisms remain to be elucidated. Recent multiomics approaches have provided much higher resolution analysis of gene expression in developing thymocytes than was previously achievable, thereby offering a fresh perspective on this question. Focusing on our recent studies using CITE-seq (cellular indexing of transcriptomes and epitopes) analyses of mouse thymocytes, we present a detailed timeline of RNA and protein expression changes during CD8 versus CD4 T cell differentiation. We also revisit our current understanding of the links between T cell receptor signaling and expression of the lineage-defining transcription factors ThPOK and RUNX3. Finally, we propose a sequential selection model to explain the tight linkage between MHC-I versus MHC-II recognition and T cell lineage choice. This model incorporates key aspects of previously proposed kinetic signaling, instructive, and stochastic/selection models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis.
- Author
-
Shirafkan, Fatemeh, Hensel, Luca, and Rattay, Kristin
- Subjects
AUTOIMMUNE diseases ,IMMUNOLOGICAL tolerance ,REGULATORY T cells ,ANTIGEN presentation ,PREVENTIVE medicine ,HOMEOSTASIS - Abstract
The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. IgG from Dermatophagoides pteronyssinus (Der p)-atopic individuals modulates non-atopic thymic B cell phenotype (alfa-4/beta-7) and cytokine production (IFN-γ, IL-9, and IL-10) with direct membrane interaction.
- Author
-
de-Apoena Reche, Daniela Terra, Machado, Nicolle Rakanidis, Fagundes, Beatriz Oliveira, Bergamasco, Isabella Siuffi, de Sousa, Thamires Rodrigues, do Nascimento, Lais Alves, Cunha, Fernando Roberto Machado, de-Oliveira, Marilia Garcia, da-Ressureição Sgnotto, Fábio, França, Carolina Nunes, and Victor, Jefferson Russo
- Subjects
- *
DERMATOPHAGOIDES pteronyssinus , *B cells , *FC receptors , *INTERLEUKIN-10 , *THYMOCYTES , *IMMUNE system , *CYTOKINES - Abstract
Studies about thymic B cells are scarce in the literature, but it was suggested that they can exert modulatory and regulatory functions on the immune system. Thymic B cells can play some role in regulating the most frequent allergic background worldwide, the atopy induced by the mite Dermatophagoides pteronyssinus (Der p). Here, we aimed to evaluate if the polyclonal IgG repertoire produced by Der p-atopic individuals can influence the homing and cytokine profile of human thymic B derived from non-atopic children aged less than seven days. With this purpose, we produced polyclonal IgG formulations and cultivated human thymocytes in their presence. We also assessed IgG subclasses and the direct interaction of IgG with thymic B cell membranes. Our results could demonstrate that Der p-atopic IgG could not reduce the expression of α4β7 homing molecule as observed in response to the other IgG formulations and could reduce the frequency of IFN-γ- and IL-9-producing thymic B cells compared to the mock condition. Der p-atopic IgG could also induce thymic IL-10-producing B cells compared to control conditions. The IgG derived from Der p-atopic individuals failed to diminish the population of IL-13-producing thymic B cells, unlike the reduction observed with other IgG formulations when compared to the mock condition. All IgG formulations had similar levels of IgG subclasses and directly interacted with thymic B cell membranes. Finally, we performed experiments using peripheral non-atopic B cells where IgG effects were not observed. In conclusion, our observation demonstrates that IgG induced in allergic individuals can modulate non-atopic thymic B cells, potentially generating thymic B cells prone to allergy development, which seems to not occur in mature B cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. A Bcl11bN797K variant isolated from an immunodeficient patient inhibits early thymocyte development in mice.
- Author
-
Kazuaki Matsumoto, Kazuki Okuyama, Sidwell, Tom, Motoi Yamashita, Takaho Endo, Naoko Satoh-Takayama, Hiroshi Ohno, Tomohiro Morio, Rothenberg, Ellen V., and Ichiro Taniuchi
- Subjects
FETAL liver cells ,KILLER cells ,GENETIC variation ,MICE ,THYMOCYTES ,INTERLEUKIN-7 ,TRANSCRIPTION factors ,FETOFETAL transfusion - Abstract
BCL11B is a transcription factor with six C
2 H2 -type zinc-finger domains. Studies in mice have shown that Bcl11b plays essential roles in T cell development. Several germline heterozygous BCL11B variants have been identified in human patients with inborn errors of immunity (IEI) patients. Among these, two de novo mis-sense variants cause asparagine (N) to lysine (K) replacement in distinct zincfinger domains, BCL11BN441K and BCL11BN807K . To elucidate the pathogenesis of the BCL11BN807K variant, we generated a mouse model of BCL11BN807K by inserting the corresponding mutation, Bcl11bN797K , into the mouse genome. In Bcl11b+/N797K mice, the proportion of immature CD4- CD8+ single-positive thymocytes was increased, and the development of invariant natural killer cells was severely inhibited in a T-cell-intrinsic manner. Under competitive conditions, γδT cell development was outcompeted by control cells. Bcl11bN797K/N797K mice died within one day of birth. Recipient mice reconstituted with Bcl11bN797K/N797K fetal liver cells nearly lacked CD4+ CD8+ double-positive thymocytes, which was consistent with the lack of their emergence in culture from Bcl11bN797K/N797K fetal liver progenitors. Interestingly, Bcl11bN797K/N797K progenitors gave rise to aberrant c-Kit+ and CD44+ cells both in vivo and in vitro. The increase in the proportion of immature CD8 single-positive thymocytes in the Bcl11bN797K mutants is caused, in part, by the inefficient activation of the Cd4 gene due to the attenuated function of the two Cd4 enhancers via distinct mechanisms. Therefore, we conclude that immunodeficient patient-derived Bcl11bN797K mutant mice elucidated a novel role for Bcl11b in driving the appropriate transition of CD4- CD8- into CD4+ CD8+ thymocytes. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
20. Assembling the thymus medulla: Development and function of epithelial cell heterogeneity.
- Author
-
James, Kieran D., Cosway, Emilie J., Parnell, Sonia M., White, Andrea J., Jenkinson, William E., and Anderson, Graham
- Subjects
- *
EPITHELIAL cells , *CELL physiology , *THYMUS , *MYASTHENIA gravis , *HETEROGENEITY , *T cells , *T cell receptors - Abstract
The thymus is a unique primary lymphoid organ that supports the production of self‐tolerant T‐cells essential for adaptive immunity. Intrathymic microenvironments are microanatomically compartmentalised, forming defined cortical, and medullary regions each differentially supporting critical aspects of thymus‐dependent T‐cell maturation. Importantly, the specific functional properties of thymic cortical and medullary compartments are defined by highly specialised thymic epithelial cells (TEC). For example, in the medulla heterogenous medullary TEC (mTEC) contribute to the enforcement of central tolerance by supporting deletion of autoreactive T‐cell clones, thereby counterbalancing the potential for random T‐cell receptor generation to contribute to autoimmune disease. Recent advances have further shed light on the pathways and mechanisms that control heterogeneous mTEC development and how differential mTEC functionality contributes to control self‐tolerant T‐cell development. Here we discuss recent findings in relation to mTEC development and highlight examples of how mTEC diversity contribute to thymus medulla function. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. RORγt up-regulates RAG gene expression in DP thymocytes to expand the Tcra repertoire.
- Author
-
Naik, Abani Kanta, Dauphars, Danielle J., Corbett, Elizabeth, Simpson, Lunden, Schatz, David G., and Krangel, Michael S.
- Subjects
THYMOCYTES ,GENE expression ,PHYSIOLOGY ,ANTIGEN receptors ,TRANSCRIPTION factors ,INTERLEUKIN-7 - Abstract
Recombination activating gene (RAG) expression increases as thymocytes transition from the CD4
− CD8− double-negative (DN) to the CD4+ CD8+ double-positive (DP) stage, but the physiological importance and mechanism of transcriptional up-regulation are unknown. Here, we show that a DP-specific component of the recombination activating genes antisilencer (DPASE) provokes elevated RAG expression in DP thymocytes. Mouse DP thymocytes lacking the DPASE display RAG expression equivalent to that in DN thymocytes, but this supports only a partial Tcra repertoire due to inefficient secondary Vα-Jα rearrangement. These data indicate that RAG up-regulation is required for a replete Tcra repertoire and that RAG expression is fine-tuned during lymphocyte development to meet the requirements of distinct antigen receptor loci. We further show that transcription factor RORγt directs RAG up-regulation in DP thymocytes by binding to the DPASE and that RORγt influences the Tcra repertoire by binding to the Tcra enhancer. These data, together with prior work showing RORγt to control Tcra rearrangement by regulating DP thymocyte proliferation and survival, reveal RORγt to orchestrate multiple pathways that support formation of the Tcra repertoire. Editor's summary: Thymocytes show an increase in recombination activating gene (RAG) expression as they transition from CD4− CD8− double-negative (DN) to CD4+ CD8+ double-positive (DP) cells. Naik et al. examined the mechanism and effects of increased RAG expression in thymocytes. The presence of a DP-specific component of the RAG antisilencer (DPASE) in the RAG locus was linked to higher RAG expression in DP thymocytes and was needed for efficient Vα-Jα rearrangement and a sufficient Tcra repertoire. The RORγt transcription factor binds to the DPASE to up-regulate RAG expression in DP thymocytes and binds to the Tcra enhancer as well, which affects the Tcra repertoire. These data deepen our understanding of mechanisms involved in DP thymocyte development and how this affects the Tcra repertoire. —Christiana N. Fogg [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
22. Transcriptomic profiling of thymic dysregulation and viral tropism after neonatal roseolovirus infection
- Author
-
Andrei Belean, Eden Xue, Benjamin Cisneros, Elisha D. O. Roberson, Michael A. Paley, and Tarin M. Bigley
- Subjects
roseolovirus ,thymus ,central tolerance ,transcriptomics ,thymocytes ,medullary thymic epithelial cells (mTECs) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionHerpesviruses, including the roseoloviruses, have been linked to autoimmune disease. The ubiquitous and chronic nature of these infections have made it difficult to establish a causal relationship between acute infection and subsequent development of autoimmunity. We have shown that murine roseolovirus (MRV), which is highly related to human roseoloviruses, induces thymic atrophy and disruption of central tolerance after neonatal infection. Moreover, neonatal MRV infection results in development of autoimmunity in adult mice, long after resolution of acute infection. This suggests that MRV induces durable immune dysregulation.MethodsIn the current studies, we utilized single-cell RNA sequencing (scRNAseq) to study the tropism of MRV in the thymus and determine cellular processes in the thymus that were disrupted by neonatal MRV infection. We then utilized tropism data to establish a cell culture system.ResultsHerein, we describe how MRV alters the thymic transcriptome during acute neonatal infection. We found that MRV infection resulted in major shifts in inflammatory, differentiation and cell cycle pathways in the infected thymus. We also observed shifts in the relative number of specific cell populations. Moreover, utilizing expression of late viral transcripts as a proxy of viral replication, we identified the cellular tropism of MRV in the thymus. This approach demonstrated that double negative, double positive, and CD4 single positive thymocytes, as well as medullary thymic epithelial cells were infected by MRV in vivo. Finally, by applying pseudotime analysis to viral transcripts, which we refer to as “pseudokinetics,” we identified viral gene transcription patterns associated with specific cell types and infection status. We utilized this information to establish the first cell culture systems susceptible to MRV infection in vitro.ConclusionOur research provides the first complete picture of roseolovirus tropism in the thymus after neonatal infection. Additionally, we identified major transcriptomic alterations in cell populations in the thymus during acute neonatal MRV infection. These studies offer important insight into the early events that occur after neonatal MRV infection that disrupt central tolerance and promote autoimmune disease.
- Published
- 2024
- Full Text
- View/download PDF
23. Thymic macrophages consist of two populations with distinct localization and origin.
- Author
-
Zhou, Tyng-An, Hsu, Hsuan-Po, Tu, Yueh-Hua, Cheng, Hui-Kuei, Lin, Chih-Yu, Chen, Nien-Jung, Tsai, Jin-Wu, Robey, Ellen, Huang, Hsuan-Cheng, Hsu, Chia-Lin, and Dzhagalov, Ivan
- Subjects
cell proliferation ,developmental biology ,diversity ,immunology ,inflammation ,macrophages ,mouse ,ontology ,thymus ,transcriptional profile ,Mice ,Animals ,Thymus Gland ,Macrophages ,Thymocytes ,Hematopoietic Stem Cells ,Phenotype - Abstract
Tissue-resident macrophages are essential to protect from pathogen invasion and maintain organ homeostasis. The ability of thymic macrophages to engulf apoptotic thymocytes is well appreciated, but little is known about their ontogeny, maintenance, and diversity. Here, we characterized the surface phenotype and transcriptional profile of these cells and defined their expression signature. Thymic macrophages were most closely related to spleen red pulp macrophages and Kupffer cells and shared the expression of the transcription factor (TF) SpiC with these cells. Single-cell RNA sequencing (scRNA-Seq) showed that the macrophages in the adult thymus are composed of two populations distinguished by the expression of Timd4 and Cx3cr1. Remarkably, Timd4+ cells were located in the cortex, while Cx3cr1+ macrophages were restricted to the medulla and the cortico-medullary junction. Using shield chimeras, transplantation of embryonic thymuses, and genetic fate mapping, we found that the two populations have distinct origins. Timd4+ thymic macrophages are of embryonic origin, while Cx3cr1+ macrophages are derived from adult hematopoietic stem cells. Aging has a profound effect on the macrophages in the thymus. Timd4+ cells underwent gradual attrition, while Cx3cr1+ cells slowly accumulated with age and, in older mice, were the dominant macrophage population in the thymus. Altogether, our work defines the phenotype, origin, and diversity of thymic macrophages.
- Published
- 2022
24. Quercetin inhibits NF-kB and JAK/STAT signaling via modulating TLR in thymocytes and splenocytes during MSG-induced immunotoxicity: an in vitro approach.
- Author
-
Das, Debasmita, Banerjee, Arnab, Mukherjee, Sandip, and Maji, Bithin Kumar
- Abstract
Background: The most widely used food additive monosodium glutamate (MSG) has been linked to immunopathology. Conversely, quercetin (Q), a naturally occurring flavonoid has been demonstrated to have immunomodulatory functions. Therefore, the purpose of the study is to determine if quercetin can mitigate the deleterious effects of MSG on immune cells, and the possible involvement of TLR, if any. Methods and Results: This study was conducted on Q, to determine how it affects the inflammatory response triggered by MSG in primary cultured thymocytes and splenocytes from rats (n = 5). Q shielded cells by augmenting cell survival and decreasing lactate dehydrogenase leakage during MSG treatment. It decreased IL-1β, IL-6, IL-8, and TNF-α expression and release by hindering NF-kB activation and by inhibiting the JAK/STAT pathway. Moreover, Q prevented NLRP3 activation, lowered IL-1β production, and promoted an anti-inflammatory response by increasing IL-10 production. Q reduced MSG-induced cellular stress and inflammation by acting as an agonist for PPAR-γ and LXRα, preventing NF-kB activation, and lowering MMP-9 production via increasing TIMP-1. Additionally, Q neutralized free radicals, elevated intracellular antioxidants, and impeded RIPK3, which is involved in inflammation induced by oxidative stress, TNF-α, and TLR agonists in MSG-treated cells. Furthermore, it also modulated TYK2 and the JAK/STAT pathway, which exhibited an anti-inflammatory effect. Conclusions: MSG exposure is associated with immune cell dysfunction, inflammation, and oxidative stress, and Q modulates TLR to inhibit NF-kB and JAK/STAT pathways, providing therapeutic potential. Further research is warranted to understand Q's downstream effects and explore its potential clinical applications in inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Thymic-Epithelial-Cell-Dependent Microenvironment Influences Proliferation and Apoptosis of Leukemic Cells.
- Author
-
Patel, Sandesh Kumar, Zhdanovskaya, Nadezda, Sergio, Ilaria, Cardinale, Antonella, Rosichini, Marco, Varricchio, Claudia, Pace, Eleonora, Capalbo, Carlo, Locatelli, Franco, Macone, Alberto, Velardi, Enrico, Palermo, Rocco, and Felli, Maria Pia
- Subjects
- *
THYMOCYTES , *NOTCH genes , *CELL morphology , *CELL cycle regulation , *HEMATOLOGIC malignancies , *EPITHELIAL cells , *BONE marrow - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a hematological cancer characterized by the infiltration of immature T-cells in the bone marrow. Aberrant NOTCH signaling in T-ALL is mainly triggered by activating mutations of NOTCH1 and overexpression of NOTCH3, and rarely is it linked to NOTCH3-activating mutations. Besides the known critical role of NOTCH, the nature of intrathymic microenvironment-dependent mechanisms able to render immature thymocytes, presumably pre-leukemic cells, capable of escaping thymus retention and infiltrating the bone marrow is still unclear. An important challenge is understanding how leukemic cells shape their tumor microenvironment to increase their ability to infiltrate and survive within. Our previous data indicated that hyperactive NOTCH3 affects the CXCL12/CXCR4 system and may interfere with T-cell/stroma interactions within the thymus. This study aims to identify the biological effects of the reciprocal interactions between human leukemic cell lines and thymic epithelial cell (TEC)-derived soluble factors in modulating NOTCH signaling and survival programs of T-ALL cells and TECs. The overarching hypothesis is that this crosstalk can influence the progressive stages of T-cell development driving T-cell leukemia. Thus, we investigated the effect of extracellular space conditioned by T-ALL cell lines (Jurkat, TALL1, and Loucy) and TECs and studied their reciprocal regulation of cell cycle and survival. In support, we also detected metabolic changes as potential drivers of leukemic cell survival. Our studies could shed light on T-cell/stroma crosstalk to human leukemic cells and propose our culture system to test pharmacological treatment for T-ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. BCL6 is required for the thymic development of TCRαβ+CD8αα+ intraepithelial lymphocyte lineage.
- Author
-
Xing, Qi, Chang, Dehui, Xie, Shiyuan, Zhao, Xiaohong, Zhang, Hao, Wang, Xiaohu, Bai, Xue, and Dong, Chen
- Subjects
LYMPHOCYTES ,T cells ,INTESTINAL mucosa ,TRANSCRIPTION factors ,THYMOCYTES - Abstract
TCRαβ
+ CD8αα+ intraepithelial lymphocytes (CD8αα+ αβ IELs) are a specialized subset of T cells in the gut epithelium that develop from thymic agonist selected IEL precursors (IELps). The molecular mechanisms underlying the selection and differentiation of this T cell type in the thymus are largely unknown. Here, we found that Bcl6 deficiency in αβ T cells resulted in the near absence of CD8αα+ αβ IELs. BCL6 was expressed by approximately 50% of CD8αα+ αβ IELs and by the majority of thymic PD1+ IELps after agonist selection. Bcl6 deficiency blocked early IELp generation in the thymus, and its expression in IELps was induced by thymic TCR signaling in an ERK-dependent manner. As a result of Bcl6 deficiency, the precursors of IELps among CD4+ CD8+ double-positive thymocytes exhibited increased apoptosis during agonist selection and impaired IELp differentiation and maturation. Together, our results elucidate BCL6 as a crucial transcription factor during the thymic development of CD8αα+ αβ IELs. Editor's summary: Intraepithelial lymphocytes (IELs) reside within the intestinal epithelium, where they regulate immune tolerance. Unconventional CD8αα+ αβ IELs are selected from thymic IEL precursors (IELps) after strong TCR signaling; however, the transcription factors governing this process of agonist selection are not well defined. Xing et al. found that mice lacking the transcription factor BCL6 in αβ T cells almost did not develop CD8αα+ αβ IELs. Thymic IELps up-regulated BCL6 after TCR stimulation, and BCL6 expression was required for their survival, differentiation, and maturation. These findings identify that BCL6 is required for the development of unconventional CD8αα+ αβ IELs during agonist selection. —Hannah Isles [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
27. Modified live vaccine strains of porcine reproductive and respiratory syndrome virus cause immune system dysregulation similar to wild strains.
- Author
-
Stepanova, Katerina, Toman, Miroslav, Sinkorova, Jana, Sinkora, Simon, Pfeiferova, Sarka, Skalnikova, Helena Kupcova, Abuhajiar, Salim, Moutelikova, Romana, Salat, Jiri, Stepanova, Hana, Nechvatalova, Katerina, Leva, Lenka, Hermanova, Petra, Kratochvilova, Mirka, Dusankova, Blanka, Sinkora Jr, Marek, Horak, Vratislav, Hudcovic, Tomas, Butler, John E., and Sinkora, Marek
- Subjects
PORCINE reproductive & respiratory syndrome ,IMMUNE system ,WEIGHT gain ,ANTIBODY formation ,T cells ,THYMUS tumors - Abstract
Introduction: Porcine reproductive and respiratory syndrome virus (PRRSV) emerged about 30 years ago and continues to cause major economic losses in the pork industry. The lack of effective modified live vaccines (MLV) allows the pandemic to continue. Background and objective: We have previously shown that wild strains of PRRSV affect the nascent T cell repertoire in the thymus, deplete T cell clones recognizing viral epitopes essential for neutralization, while triggering a chronic, robust, but ineffective antibody response. Therefore, we hypothesized that the current MLV are inappropriate because they cause similar damage and fail to prevent viral-induced dysregulation of adaptive immunity. Methods: We tested three MLV strains to demonstrate that all have a comparable negative effect on thymocytes in vitro. Further in vivo studies compared the development of T cells in the thymus, peripheral lymphocytes, and antibody production in young piglets. These three MLV strains were used in a mixture to determine whether at least some of them behave similarly to the wild virus type 1 or type 2. Results: Both the wild and MLV strains cause the same immune dysregulations. These include depletion of T-cell precursors, alteration of the TCR repertoire, necrobiosis at corticomedullary junctions, low body weight gain, decreased thymic cellularity, lack of virus-neutralizing antibodies, and production of nonneutralizing anti-PRRSV antibodies of different isotypes. Discussion and conclusion: The results may explain why the use of current MLV in young animals may be ineffective and why their use may be potentially dangerous. Therefore, alternative vaccines, such as subunit or mRNA vaccines or improved MLV, are needed to control the PRRSV pandemic. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Thymosin β4 Regulates the Differentiation of Thymocytes by Controlling the Cytoskeletal Rearrangement and Mitochondrial Transfer of Thymus Epithelial Cells.
- Author
-
Ying, Yuyuan, Tao, Nana, Zhang, Fengjie, Wen, Xunuo, Zhou, Meiru, and Gao, Jianli
- Subjects
- *
THYMOSIN , *EPITHELIAL cells , *THYMOCYTES , *CYTOSKELETAL proteins , *MITOCHONDRIA , *THYMUS - Abstract
The thymus is one of the most crucial immunological organs, undergoing visible age-related shrinkage. Thymic epithelial cells (TECs) play a vital role in maintaining the normal function of the thymus, and their degeneration is the primary cause of age-induced thymic devolution. Thymosin β4 (Tβ4) serves as a significant important G-actin sequestering peptide. The objective of this study was to explore whether Tβ4 influences thymocyte differentiation by regulating the cytoskeletal rearrangement and mitochondrial transfer of TECs. A combination of H&E staining, immunofluorescence, transmission electron microscopy, RT-qPCR, flow cytometry, cytoskeletal immunolabeling, and mitochondrial immunolabeling were employed to observe the effects of Tβ4 on TECs' skeleton rearrangement, mitochondrial transfer, and thymocyte differentiation. The study revealed that the Tβ4 primarily regulates the formation of microfilaments and the mitochondrial transfer of TECs, along with the formation and maturation of double-negative cells (CD4−CD8−) and CD4 single-positive cells (CD3+TCRβ+CD4+CD8−) thymocytes. This study suggests that Tβ4 plays a crucial role in thymocyte differentiation by influencing the cytoskeletal rearrangement and mitochondrial transfer of TECs. These effects may be associated with Tβ4's impact on the aggregation of F-actin. This finding opens up new avenues for research in the field of immune aging. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Preclinical Study in Mouse Thymus and Thymocytes: Effects of Treatment with a Combination of Sodium Dichloroacetate and Sodium Valproate on Infectious Inflammation Pathways.
- Author
-
Stakišaitis, Donatas, Kapočius, Linas, Kilimaitė, Evelina, Gečys, Dovydas, Šlekienė, Lina, Balnytė, Ingrida, Palubinskienė, Jolita, and Lesauskaitė, Vaiva
- Subjects
- *
VALPROIC acid , *THYMOCYTES , *THYMUS , *GENE expression , *SODIUM - Abstract
The research presents data from a preclinical study on the anti-inflammatory effects of a sodium dichloroacetate and sodium valproate combination (DCA–VPA). The 2-week treatment with a DCA 100 mg/kg/day and VPA 150 mg/kg/day combination solution in drinking water's effects on the thymus weight, its cortex/medulla ratio, Hassall's corpuscles (HCs) number in the thymus medulla, and the expression of inflammatory and immune-response-related genes in thymocytes of male Balb/c mice were studied. Two groups of mice aged 6–7 weeks were investigated: a control (n = 12) and a DCA–VPA-treated group (n = 12). The treatment did not affect the body weight gain (p > 0.05), the thymus weight (p > 0.05), the cortical/medulla ratio (p > 0.05), or the number of HCs (p > 0.05). Treatment significantly increased the Slc5a8 gene expression by 2.1-fold (p < 0.05). Gene sequence analysis revealed a significant effect on the expression of inflammation-related genes in thymocytes by significantly altering the expression of several genes related to the cytokine activity pathway, the inflammatory response pathway, and the Il17 signaling pathway in thymocytes. Data suggest that DCA–VPA exerts an anti-inflammatory effect by inhibiting the inflammatory mechanisms in the mouse thymocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Interruption of Lymph Flow Worsens the Skin Inflammation Caused by Saprophytic Staphylococcus epidermidis.
- Author
-
Cąkała-Jakimowicz, Marta, Domaszewska-Szostek, Anna, and Puzianowska-Kuznicka, Monika
- Subjects
STAPHYLOCOCCUS epidermidis ,SKIN inflammation ,STEM cells ,T cells ,THYMOCYTES - Abstract
Lymphedema is often complicated by chronic inflammation, leading to fibrosis, fat deposition, and inhibition of lymphangiogenesis. This study aimed to verify whether lymphedema itself or together with commensal bacterial flora infection contributes to the severity of local inflammation. Edema was induced by interruption of the lymph flow in the rat's hind limb. Immune cell infiltrates were examined by flow cytometry and immunohistochemistry. Nine-day edema alone did not affect immune cell content in the skin but resulted in a decrease in CD4
+ T helper lymphocytes and monocytes in the draining popliteal lymph nodes. In turn, local saprophytic Staphylococcus epidermidis infection of the edematous limb resulted in dense infiltrates of CD68+ macrophages and monocytes, MHC class II antigen-presenting cells, CD90+ stem cells, thymocytes, and immature B cells in the skin, accompanied by a simultaneous reduction in density of CD4+ T helper lymphocytes and monocytes, OX62+ dendritic cells, CD68+ macrophages and monocytes, HiS48+ granulocytes, CD90+ stem cells, thymocytes, and immature B cells in the draining popliteal lymph nodes. These results indicate that the combination of edema and saprophytic bacteria infection induces severe inflammation in the peripheral tissues and results in a delay of antibacterial protection processes in neighboring lymphatic organs. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
31. Cryopreservation of mouse thymus depletes thymocytes but supports immune reconstitution on transplantation.
- Author
-
Chawda, Mira M., Ross, Susan, Lau, Ching‐In, Yánez, Diana C., Rowell, Jasmine, Kilbride, Peter, and Crompton, Tessa
- Subjects
THYMUS ,THYMOCYTES ,MICE ,TREATMENT delay (Medicine) ,CLINICAL immunology - Abstract
The article discusses the cryopreservation of mouse thymus and its impact on immune reconstitution after transplantation. The study found that cryopreservation depleted thymocytes but preserved thymus function, and there were no differences in immune reconstitution between fresh and frozen/thawed transplants. This suggests that donor thymocyte depletion does not affect the outcome of thymus transplantation. Cryopreservation of thymus may have advantages such as increasing availability, decreasing delays in treatment, and enabling MHC-matching, which could improve immune reconstitution and reduce autoimmunity. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
32. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis
- Author
-
Fatemeh Shirafkan, Luca Hensel, and Kristin Rattay
- Subjects
thymus ,autoimmune disease ,tolerance ,tissue homeostasis ,mTEC ,thymocytes ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions.
- Published
- 2024
- Full Text
- View/download PDF
33. Chromatin organizer SATB1 controls the cell identity of CD4+ CD8+ double-positive thymocytes by regulating the activity of super-enhancers
- Author
-
Feng, Delong, Chen, Yanhong, Dai, Ranran, Bian, Shasha, Xue, Wei, Zhu, Yongchang, Li, Zhaoqiang, Yang, Yiting, Zhang, Yan, Zhang, Jiarui, Bai, Jie, Qin, Litao, Kohwi, Yoshinori, Shi, Weili, Kohwi-Shigematsu, Terumi, Ma, Jing, Liao, Shixiu, and Hao, Bingtao
- Subjects
Human Genome ,Genetics ,HIV/AIDS ,CD4 Antigens ,CD4-Positive T-Lymphocytes ,CD8 Antigens ,CD8-Positive T-Lymphocytes ,Cell Differentiation ,Chromatin ,Matrix Attachment Region Binding Proteins ,Receptors ,Antigen ,T-Cell ,Receptors ,Antigen ,T-Cell ,alpha-beta ,Thymocytes ,Thymus Gland - Abstract
CD4+ and CD8+ double-positive (DP) thymocytes play a crucial role in T cell development in the thymus. DP cells rearrange the T cell receptor gene Tcra to generate T cell receptors with TCRβ. DP cells differentiate into CD4 or CD8 single-positive (SP) thymocytes, regulatory T cells, or invariant nature kill T cells (iNKT) in response to TCR signaling. Chromatin organizer SATB1 is highly expressed in DP cells and is essential in regulating Tcra rearrangement and differentiation of DP cells. Here we explored the mechanism of SATB1 orchestrating gene expression in DP cells. Single-cell RNA sequencing shows that Satb1 deletion changes the cell identity of DP thymocytes and down-regulates genes specifically and highly expressed in DP cells. Super-enhancers regulate the expressions of DP-specific genes, and our Hi-C data show that SATB1 deficiency in thymocytes reduces super-enhancer activity by specifically decreasing interactions among super-enhancers and between super-enhancers and promoters. Our results reveal that SATB1 plays a critical role in thymocyte development to promote the establishment of DP cell identity by globally regulating super-enhancers of DP cells at the chromatin architectural level.
- Published
- 2022
34. Deciphering the factors determining the differentiation of autoreactive thymocytes into regulatory T cells.
- Author
-
Benito‐Villalvilla, Cristina, Angelina, Alba, and Martín‐Cruz, Leticia
- Subjects
- *
REGULATORY T cells , *THYMOCYTES , *FORKHEAD transcription factors , *CD25 antigen - Abstract
This article discusses the factors that determine the differentiation of autoreactive thymocytes into regulatory T cells (Tregs). Tregs are a subset of CD4+ T cells that play a crucial role in maintaining peripheral tolerance and immune homeostasis. The majority of Tregs are generated in the thymus upon recognition of self-antigens, while others can be generated in specialized peripheral tissues. The differentiation of Tregs is influenced by factors such as T-cell receptor signaling, co-stimulation, and the presence of TGF-β and retinoic acid. The study also highlights the role of late agonist signaling and the disruption of TCR signaling in the generation of Tregs. The findings contribute to our understanding of thymocyte selection and central tolerance, and may have implications for future therapeutic interventions involving Tregs. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
35. Modified live vaccine strains of porcine reproductive and respiratory syndrome virus cause immune system dysregulation similar to wild strains
- Author
-
Katerina Stepanova, Miroslav Toman, Jana Sinkorova, Simon Sinkora, Sarka Pfeiferova, Helena Kupcova Skalnikova, Salim Abuhajiar, Romana Moutelikova, Jiri Salat, Hana Stepanova, Katerina Nechvatalova, Lenka Leva, Petra Hermanova, Mirka Kratochvilova, Blanka Dusankova, Marek Sinkora, Vratislav Horak, Tomas Hudcovic, and John E. Butler
- Subjects
Porcine respiratory and reproductive syndrome virus ,thymocytes ,T-cell precursors ,T lymphocytes ,B lymphocytes ,animals ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionPorcine reproductive and respiratory syndrome virus (PRRSV) emerged about 30 years ago and continues to cause major economic losses in the pork industry. The lack of effective modified live vaccines (MLV) allows the pandemic to continue.Background and objectiveWe have previously shown that wild strains of PRRSV affect the nascent T cell repertoire in the thymus, deplete T cell clones recognizing viral epitopes essential for neutralization, while triggering a chronic, robust, but ineffective antibody response. Therefore, we hypothesized that the current MLV are inappropriate because they cause similar damage and fail to prevent viral-induced dysregulation of adaptive immunity. MethodsWe tested three MLV strains to demonstrate that all have a comparable negative effect on thymocytes in vitro. Further in vivo studies compared the development of T cells in the thymus, peripheral lymphocytes, and antibody production in young piglets. These three MLV strains were used in a mixture to determine whether at least some of them behave similarly to the wild virus type 1 or type 2. ResultsBoth the wild and MLV strains cause the same immune dysregulations. These include depletion of T-cell precursors, alteration of the TCR repertoire, necrobiosis at corticomedullary junctions, low body weight gain, decreased thymic cellularity, lack of virus-neutralizing antibodies, and production of non-neutralizing anti-PRRSV antibodies of different isotypes. Discussion and conclusionThe results may explain why the use of current MLV in young animals may be ineffective and why their use may be potentially dangerous. Therefore, alternative vaccines, such as subunit or mRNA vaccines or improved MLV, are needed to control the PRRSV pandemic.
- Published
- 2024
- Full Text
- View/download PDF
36. The promiscuous development of an unconventional Qa1brestricted T cell population.
- Author
-
Valerio, Michael Manoharan, Arana, Kathya, Jian Guan, Shiao Wei Chan, Xiaokun Yang, Kurd, Nadia, Angus Lee, Shastri, Nilabh, Coscoy, Laurent, and Robey, Ellen A.
- Subjects
T cells ,CELL populations ,THYMOCYTES ,LYMPHOCYTES ,CD8 antigen - Abstract
MHC-E restricted CD8 T cells show promise in vaccine settings, but their development and specificity remain poorly understood. Here we focus on a CD8 T cell population reactive to a self-peptide (FL9) bound to mouse MHC-E (Qa-1
b ) that is presented in response to loss of the MHC I processing enzyme ERAAP, termed QFL T cells. We find that mature QFL thymocytes are predominantly CD8αβ+CD4-, show signs of agonist selection, and give rise to both CD8αα and CD8αβ intraepithelial lymphocytes (IEL), as well as memory phenotype CD8αβ T cells. QFL T cells require the MHC I subunit β-2 microglobulin (β2m), but do not require Qa1b or classical MHC I for positive selection. However, QFL thymocytes do require Qa1b for agonist selection and full functionality. Our data highlight the relaxed requirements for positive selection of an MHC-E restricted T cell population and suggest a CD8αβ+CD4- pathway for development of CD8αα IELs. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
37. CD8 T cell tolerance results from eviction of immature autoreactive cells from the thymus.
- Author
-
Badr, Mohamed Elsherif, Zhongmei Zhang, Xuguang Tai, and Singer, Alfred
- Subjects
- *
T cells , *CD8 antigen , *THYMUS , *EVICTION , *MYASTHENIA gravis , *THYMOCYTES , *T cell receptors - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. Human thymic putative CD8αα precursors exhibit a biased TCR repertoire in single cell AIRR-seq.
- Author
-
Heimli, Marte, Tennebø Flåm, Siri, Sagsveen Hjorthaug, Hanne, Bjørnstad, Pål Marius, Chernigovskaya, Maria, Le, Quy Khang, Tekpli, Xavier, Greiff, Victor, and Lie, Benedicte Alexandra
- Subjects
- *
T cell receptors , *REGULATORY T cells , *T cells , *CELL populations , *THYMOCYTES - Abstract
Thymic T cell development comprises T cell receptor (TCR) recombination and assessment of TCR avidity towards self-peptide-MHC complexes presented by antigen-presenting cells. Self-reactivity may lead to negative selection, or to agonist selection and differentiation into unconventional lineages such as regulatory T cells and CD8 α α T cells. To explore the effect of the adaptive immune receptor repertoire on thymocyte developmental decisions, we performed single cell adaptive immune receptor repertoire sequencing (scAIRR-seq) of thymocytes from human young paediatric thymi and blood. Thymic PDCD1+ cells, a putative CD8 α α T cell precursor population, exhibited several TCR features previously associated with thymic and peripheral ZNF683+ CD8 α α T cells, including enrichment of large and positively charged complementarity-determining region 3 (CDR3) amino acids. Thus, the TCR repertoire may partially explain the decision between conventional vs. agonist selected thymocyte differentiation, an aspect of importance for the development of therapies for patients with immune-mediated diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. CD1b glycoprotein, a crucial marker of thymocyte development during T cell maturation in cynomolgus monkeys.
- Author
-
Choi, Sung Min, Park, Hi Jung, Choi, Eun A, Jung, Kyeong Cheon, and Lee, Jae Il
- Subjects
- *
KRA , *T cells , *THYMOCYTES , *T cell receptors , *THYMUS , *CD8 antigen - Abstract
Phenotypic markers that denote different developmental stages of thymocytes are important for understanding T cell development in the thymus. Here, we show that CD1b is a critical discriminator of thymocyte maturation stage in cynomolgus monkeys. CD1b was expressed by immature thymocytes prior to β-selection, and its expression decreased as cells became fully mature in the thymus. MHC-I expression was lowest at the CD3loCD1b+ immature double-positive (DP) stage, while the ratio of CD1d:MHC-I expression was significantly higher at this stage than at other developmental stages. PLZF was expressed by < 0.2% of thymocytes; most PLZF+ thymocytes were CD3-/loCD1b+ immature DP thymocytes with the potential to produce IL-4. EOMES+ thymocytes, which accounted for > 2% of total thymocytes, were mostly CD3+CD1b- mature thymocytes and predominantly of the CD8 single-positive (SP) lineage. An unconventional CD8+ T cell subset expressing the NKG2AC+CXCR3+ innate-like T cell marker was identified within the EOMES+ CD8 SP lineage; these cells exhibited a memory phenotype. Taken together, these findings show that CD1b is a valuable discriminatory marker of thymocyte development. The data presented herein can be used to characterize the features of PLZF- and EOMES-associated unconventional T cells in the thymus. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. T cell self-reactivity during thymic development dictates the timing of positive selection.
- Author
-
Lutes, Lydia, Steier, Zoë, McIntyre, Laura, Pandey, Shraddha, Kaminski, James, Hoover, Ashley, Ariotti, Silvia, Streets, Aaron, Yosef, Nir, and Robey, Ellen
- Subjects
TCR ,developmental biology ,immunology ,inflammation ,ion channels ,mouse ,positive selection ,self-reactivity ,thymocyte ,Animals ,Cell Differentiation ,Cell Lineage ,Gene Expression Regulation ,Histocompatibility Antigens Class I ,Ion Channels ,Kinetics ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Phenotype ,Receptors ,Antigen ,T-Cell ,Self Tolerance ,Signal Transduction ,Thymocytes ,Thymus Gland ,Transcriptome - Abstract
Functional tuning of T cells based on their degree of self-reactivity is established during positive selection in the thymus, although how positive selection differs for thymocytes with relatively low versus high self-reactivity is unclear. In addition, preselection thymocytes are highly sensitive to low-affinity ligands, but the mechanism underlying their enhanced T cell receptor (TCR) sensitivity is not fully understood. Here we show that murine thymocytes with low self-reactivity experience briefer TCR signals and complete positive selection more slowly than those with high self-reactivity. Additionally, we provide evidence that cells with low self-reactivity retain a preselection gene expression signature as they mature, including genes previously implicated in modulating TCR sensitivity and a novel group of ion channel genes. Our results imply that thymocytes with low self-reactivity downregulate TCR sensitivity more slowly during positive selection, and associate membrane ion channel expression with thymocyte self-reactivity and progress through positive selection.
- Published
- 2021
41. Single-cell transcriptional profiling of human thymic stroma uncovers novel cellular heterogeneity in the thymic medulla.
- Author
-
Bautista, Jhoanne L, Cramer, Nathan T, Miller, Corey N, Chavez, Jessica, Berrios, David I, Byrnes, Lauren E, Germino, Joe, Ntranos, Vasilis, Sneddon, Julie B, Burt, Trevor D, Gardner, James M, Ye, Chun J, Anderson, Mark S, and Parent, Audrey V
- Subjects
Pericytes ,Thymus Gland ,T-Lymphocytes ,Endothelial Cells ,Epithelial Cells ,Mesoderm ,Animals ,Humans ,Mice ,Gene Expression Profiling ,Cell Differentiation ,Cell Lineage ,Genetic Heterogeneity ,Adult ,Single-Cell Analysis ,Thymocytes - Abstract
The thymus' key function in the immune system is to provide the necessary environment for the development of diverse and self-tolerant T lymphocytes. While recent evidence suggests that the thymic stroma is comprised of more functionally distinct subpopulations than previously appreciated, the extent of this cellular heterogeneity in the human thymus is not well understood. Here we use single-cell RNA sequencing to comprehensively profile the human thymic stroma across multiple stages of life. Mesenchyme, pericytes and endothelial cells are identified as potential key regulators of thymic epithelial cell differentiation and thymocyte migration. In-depth analyses of epithelial cells reveal the presence of ionocytes as a medullary population, while the expression of tissue-specific antigens is mapped to different subsets of epithelial cells. This work thus provides important insight on how the diversity of thymic cells is established, and how this heterogeneity contributes to the induction of immune tolerance in humans.
- Published
- 2021
42. The Metabolic Landscape of Thymic T Cell Development In Vivo and In Vitro
- Author
-
Sun, Victoria, Sharpley, Mark, Kaczor-Urbanowicz, Karolina E, Chang, Patrick, Montel-Hagen, Amélie, Lopez, Shawn, Zampieri, Alexandre, Zhu, Yuhua, de Barros, Stéphanie C, Parekh, Chintan, Casero, David, Banerjee, Utpal, and Crooks, Gay M
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Animals ,Biological Evolution ,Biomarkers ,Cell Differentiation ,Cell Line ,Computational Biology ,Energy Metabolism ,Gene Expression Profiling ,Hematopoietic Stem Cells ,Humans ,Lymphopoiesis ,Metabolome ,Metabolomics ,Mice ,Organoids ,T-Lymphocytes ,Thymocytes ,Tissue Culture Techniques ,thymus ,metabolism ,T cell ,organoid ,thymopoiesis ,human ,mouse ,in vitro ,Immunology ,Medical Microbiology ,Biochemistry and cell biology ,Genetics - Abstract
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.
- Published
- 2021
43. TCRα reporter mice reveal contribution of dual TCRα expression to T cell repertoire and function
- Author
-
Yang, Letitia, Jama, Burhan, Wang, Huawei, Labarta-Bajo, Lara, Zúñiga, Elina I, and Morris, Gerald P
- Subjects
Prevention ,Infectious Diseases ,Emerging Infectious Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Animals ,CD4-Positive T-Lymphocytes ,CD5 Antigens ,CD8-Positive T-Lymphocytes ,Chlorocebus aethiops ,Female ,Gene Expression ,Genes ,T-Cell Receptor alpha ,Green Fluorescent Proteins ,Immunologic Memory ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis virus ,Male ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Receptors ,Antigen ,T-Cell ,alpha-beta ,T-Lymphocytes ,Thymocytes ,Vero Cells ,T cell receptor ,TCR ,T cell ,LCMV - Abstract
It is known that a subpopulation of T cells expresses two T cell receptor (TCR) clonotypes, though the extent and functional significance of this is not established. To definitively evaluate dual TCRα cells, we generated mice with green fluorescent protein and red fluorescent protein reporters linked to TCRα, revealing that ∼16% of T cells express dual TCRs, notably higher than prior estimates. Importantly, dual TCR expression has functional consequences, as dual TCR cells predominated response to lymphocytic choriomeningitis virus infection, comprising up to 60% of virus-specific CD4+ and CD8+ T cells during acute responses. Dual receptor expression selectively influenced immune memory, as postinfection memory CD4+ populations contained significantly increased frequencies of dual TCR cells. These data reveal a previously unappreciated contribution of dual TCR cells to the immune repertoire and highlight their potential effects on immune responses.
- Published
- 2020
44. New insights into developmental fate decisions by autoreactive CD4 thymocytes.
- Author
-
Tai, Xuguang and Singer, Alfred
- Subjects
- *
THYMOCYTES , *T cell receptors , *CD4 antigen , *REGULATORY T cells , *TRANSFORMING growth factors-beta - Abstract
This article discusses the process of developmental fate decisions by autoreactive CD4 thymocytes in the thymus. The thymus plays a crucial role in selecting T cells for immune responses and establishing self-tolerance. The study found that clonal deletion and autoreactive T-cell differentiation are agonist signalled at different times during thymic selection, with early agonist signals inducing clonal deletion and late agonist signals inducing the differentiation of surviving autoreactive thymocytes into either regulatory T cells (Tregs) or effector T cells (Teffs). The fate of these cells depends on the duration of late agonist signalling. The study also explored the role of transforming growth factor beta (TGFβ) in promoting Treg differentiation by disrupting agonist signalling. Overall, the findings provide new insights into the mechanisms of autoreactive CD4 thymocyte development and Treg differentiation. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
45. Impact of in vitro HIV infection on human thymic regulatory T cell differentiation.
- Author
-
Swaminathan, Sharada, Scorza, Tatiana, Yero, Alexis, Farnos, Omar, Schinke, Stephanie C. Burke, Ange, Jonathan B., and Jenabian, Mohammad-Ali
- Subjects
REGULATORY T cells ,HIV infections ,T cell differentiation ,T cells ,SUPPRESSOR cells ,LYMPHOID tissue ,THYMOCYTES - Abstract
Background: The differentiation and function of immunosuppressive regulatory T cells (Tregs) is dictated by the master transcription factor FoxP3. During HIV infection, there is an increase in Treg frequencies in the peripheral blood and lymphoid tissues. This accentuates immune dysfunction and disease progression. Expression of FoxP3 by thymic Tregs (tTregs) is partially controlled by TGF-β. This cytokine also contributes to Treg development in the peripheral blood and lymphoid tissues. Although TGF-β mediates lymphoid tissue fibrosis and peripheral Treg differentiation in HIV-infected individuals, its role in the induction and maintenance of Tregs within the thymus during HIV infection remains unclear. Methods: Thymocytes were isolated from fresh human thymic tissues obtained from pediatric patients undergoing cardiac surgery. Infection by both R5- and X4- tropic HIV-1 strains and TGF-β treatment of human thymocytes was performed in an in vitro co-culture model with OP9-DL1 cells expressing Notch ligand deltalike 1 without T cell receptor (TCR) activation. Results: Despite high expression of CCR5 and CXCR4 by tTregs, FoxP3 + CD3
high CD8- thymocytes were much less prone to in vitro infection with R5- and X4-tropic HIV strains compared to FoxP3-CD3high CD8- thymocytes. As expected, CD3high CD4+ thymocytes, when treated with TGF-β1, upregulated CD127 and this treatment resulted in increased FoxP3 expression and Treg differentiation, but did not affect the rate of HIV infection. FoxP3 expression and Treg frequencies remained unchanged following in vitro HIV infection alone or in combination with TGF-β1. Conclusion: FoxP3 expression and tTreg differentiation is not affected by in vitro HIV infection alone or the combination of in vitro HIV infection and TGF-β treatment. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
46. E protein binding at the Tcra enhancer promotes Tcra repertoire diversity.
- Author
-
Mihai, Ariana, Roy, Sumedha, Krangel, Michael S., and Yuan Zhuang
- Subjects
PROTEIN binding ,BINDING sites ,ANTIGEN receptors ,T cells ,THYMOCYTES - Abstract
V(D)J recombination of antigen receptor loci is a highly developmentally regulated process. During T lymphocyte development, recombination of the Tcra gene occurs in CD4
+ CD8+ double positive (DP) thymocytes and requires the Tcra enhancer (Eα). E proteins are known regulators of DP thymocyte development and have three identified binding sites in Eα. To understand the contribution of E proteins to Eα function, mutants lacking one or two of the respective binding sites were generated. The double-binding site mutant displayed a partial block at the positive selection stage of αβ T cell development. Further investigation revealed loss of germline transcription within the Tcra locus at the Ja array, along with dysregulated primary and impaired secondary Va-Ja rearrangement. Eα E protein binding increases Tcra locus accessibility and regulates TCRa recombination, thus directly promoting Tcra repertoire diversity. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
47. Combined multidimensional single-cell protein and RNA profiling dissects the cellular and functional heterogeneity of thymic epithelial cells.
- Author
-
Klein, Fabian, Veiga-Villauriz, Clara, Börsch, Anastasiya, Maio, Stefano, Palmer, Sam, Dhalla, Fatima, Handel, Adam E., Zuklys, Saulius, Calvo-Asensio, Irene, Musette, Lucas, Deadman, Mary E., White, Andrea J., Lucas, Beth, Anderson, Graham, and Holländer, Georg A.
- Subjects
EPITHELIAL cells ,THYMOCYTES ,RNA ,HETEROGENEITY ,T cells ,MACHINE learning - Abstract
The network of thymic stromal cells provides essential niches with unique molecular cues controlling T cell development and selection. Recent single-cell RNA sequencing studies have uncovered previously unappreciated transcriptional heterogeneity among thymic epithelial cells (TEC). However, there are only very few cell markers that allow a comparable phenotypic identification of TEC. Here, using massively parallel flow cytometry and machine learning, we deconvoluted known TEC phenotypes into novel subpopulations. Using CITEseq, these phenotypes were related to corresponding TEC subtypes defined by the cells' RNA profiles. This approach allowed the phenotypic identification of perinatal cTEC and their physical localisation within the cortical stromal scaffold. In addition, we demonstrate the dynamic change in the frequency of perinatal cTEC in response to developing thymocytes and reveal their exceptional efficiency in positive selection. Collectively, our study identifies markers that allow for an unprecedented dissection of the thymus stromal complexity, as well as physical isolation of TEC populations and assignment of specific functions to individual TEC subtypes. T cell development requires functionally diverse thymic epithelial cell (TEC) populations performing specific functions. Here, using massively parallel flow cytometry and machine learning, the authors examine in mice the TEC compartment from the perinatal period to adulthood, identify novel phenotypic markers and characterize the function of perinatal cortical TEC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
48. Paxbp1 is indispensable for the survival of CD4 and CD8 double-positive thymocytes.
- Author
-
Wenting Li, Yang Yang, Shenglin Liu, Dongsheng Zhang, Xuanyao Ren, Mindan Tang, Wei Zhang, Xiaofan Chen, Cong Huang, and Bo Yu
- Subjects
THYMOCYTES ,CD8 antigen ,T cells ,CD4 antigen ,CELL populations - Abstract
The lifespan of double-positive (DP) thymocytes is critical for intrathymic development and shaping the peripheral T cell repertoire. However, the molecular mechanisms that control DP thymocyte survival remain poorly understood. Paxbp1 is a conserved nuclear protein that has been reported to play important roles in cell growth and development. Its high expression in T cells suggests a possible role in T cell development. Here, we observed that deletion of Paxbp1 resulted in thymic atrophy in mice lacking Paxbp1 in the early stages of T cell development. Conditional loss of Paxbp1 resulted in fewer CD4
+ CD8+ DP T cells, CD4 and CD8 single positive (SP) T cells in the thymus, and fewer T cells in the periphery. Meanwhile, Paxbp1 deficiency had limited effects on the CD4- CD8- double negative (DN) or immature single-positive (ISP) cell populations. Instead, we observed a significant increase in the susceptibility of Paxbp1-deficient DP thymocytes to apoptosis. Consistent with this, RNA-Seq analysis revealed a significant enrichment of the apoptotic pathway within differentially expressed genes in Paxbp1-deficient DP cells compared to control DP cells. Together, our results suggest a new function for Paxbp1, which is an important mediator of DP thymocyte survival and critical for proper thymic development. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
49. Pleiotropic Roles of VEGF in the Microenvironment of the Developing Thymus
- Author
-
de Barros, Stephanie C, Suterwala, Batul T, He, Chongbin, Ge, Shundi, Chick, Brent, Blumberg, Garrett K, Kim, Kenneth, Klein, Sam, Zhu, Yuhua, Wang, Xiaoyan, Casero, David, and Crooks, Gay M
- Subjects
Pediatric ,Aetiology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Animals ,Cell Adhesion ,Cell Movement ,Endothelium ,Mesoderm ,Mice ,Mice ,Inbred C57BL ,Neovascularization ,Pathologic ,Pericytes ,Thymocytes ,Thymus Gland ,Vascular Endothelial Growth Factor A ,Vascular Endothelial Growth Factor Receptor-2 ,Immunology - Abstract
Neonatal life marks the apogee of murine thymic growth. Over the first few days after birth, growth slows and the murine thymus switches from fetal to adult morphology and function; little is known about the cues driving this dramatic transition. In this study, we show for the first time (to our knowledge) the critical role of vascular endothelial growth factor (VEGF) on thymic morphogenesis beyond its well-known role in angiogenesis. During a brief window a few days after birth, VEGF inhibition induced rapid and profound remodeling of the endothelial, mesenchymal and epithelial thymic stromal compartments, mimicking changes seen during early adult maturation. Rapid transcriptional changes were seen in each compartment after VEGF inhibition, including genes involved in migration, chemotaxis, and cell adhesion as well as induction of a proinflammatory and proadipogenic signature in endothelium, pericytes, and mesenchyme. Thymocyte numbers fell subsequent to the stromal changes. Expression patterns and functional blockade of the receptors VEGFR2 and NRP1 demonstrated that VEGF mediates its pleiotropic effects through distinct receptors on each microenvironmental compartment of the developing mouse thymus.
- Published
- 2020
50. N-Glycan Branching Is Required for Development of Mature B Cells.
- Author
-
Mortales, Christie-Lynn, Lee, Sung-Uk, and Demetriou, Michael
- Subjects
Animals ,Antigens ,CD19 ,Carbohydrate Conformation ,Cell Differentiation ,Mice ,Mice ,Transgenic ,N-Acetylglucosaminyltransferases ,Polysaccharides ,Precursor Cells ,B-Lymphoid ,Receptors ,Antigen ,B-Cell ,Signal Transduction ,Thymocytes ,Immunology - Abstract
Galectins have been implicated in inhibiting BCR signaling in mature B cells but promoting pre-BCR signaling during early development. Galectins bind to branched N-glycans attached to cell surface glycoproteins to control the distribution, clustering, endocytosis, and signaling of surface glycoproteins. During T cell development, N-glycan branching is required for positive selection of thymocytes, inhibiting both death by neglect and negative selection via enhanced surface retention of the CD4/CD8 coreceptors and limiting TCR clustering/signaling, respectively. The role of N-glycan branching in B cell development is unknown. In this study, we report that N-glycan branching is absolutely required for development of mature B cells in mice. Elimination of branched N-glycans in developing B cells via targeted deletion of N-acetylglucosaminyl transferase I (Mgat1) markedly reduced cellularity in the bone marrow and/or spleen and inhibited maturation of pre-, immature, and transitional stage 2 B cells. Branching deficiency markedly reduced surface expression of the pre-BCR/BCR coreceptor CD19 and promoted spontaneous death of pre-B cells and immature B cells in vitro. Death was rescued by low-dose pre-BCR/BCR stimulation but exacerbated by high-dose pre-BCR/BCR stimulation as well as antiapoptotic BclxL overexpression in pre-B cells. Branching deficiency also enhanced Nur77 induction, a marker of negative selection. Together, these data suggest that, as in T cells, N-glycan branching promotes positive selection of B cells by augmenting pre-BCR/BCR signaling via CD19 surface retention, whereas limiting negative selection from excessive BCR engagement.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.