scholarly journals Developing T cells form an immunological synapse for passage through the β−selection checkpoint

2019 ◽  
Author(s):  
Amr H. Allam ◽  
Mirren Charnley ◽  
Kim Pham ◽  
Sarah M. Russell

AbstractThe β-selection checkpoint of T cell development tests whether the cell has recombined its genomic DNA to produce a functional T Cell Receptor β (TCRβ) receptor. Passage through the β-selection checkpoint requires the nascent TCRβ protein to mediate signaling through a pre-TCR complex. In this study, we show that developing T cells at the β-selection checkpoint establish an immunological synapse in in vitro & in situ, resembling that of the mature T cell. The immunological synapse is dependent on two key signaling pathways known to be critical for the transition beyond the β-selection checkpoint, Notch and CXCR4 signaling. In vitro and in situ analyses indicate that the immunological synapse promotes passage through the β-selection checkpoint. Collectively, these data indicate that developing T cells regulate pre-TCR signaling through the formation of an immunological synapse. This signaling platform integrates cues from Notch, CXCR4, and MHC on the thymic stromal cell, to allow transition beyond the β-selection checkpoint.SummaryT cell development requires testing whether genomic rearrangement has produced a T cell receptor capable of transmitting signals. Most T cells fail this test. Here, we show that passage through the β-selection checkpoint requires assembly of a platform to support TCR signaling.

2021 ◽  
Vol 220 (3) ◽  
Author(s):  
Amr H. Allam ◽  
Mirren Charnley ◽  
Kim Pham ◽  
Sarah M. Russell

The β-selection checkpoint of T cell development tests whether the cell has recombined its genomic DNA to produce a functional T cell receptor β (TCRβ). Passage through the β-selection checkpoint requires the nascent TCRβ protein to mediate signaling through a pre-TCR complex. In this study, we show that developing T cells at the β-selection checkpoint establish an immunological synapse in in vitro and in situ, resembling that of the mature T cell. The immunological synapse is dependent on two key signaling pathways known to be critical for the transition beyond the β-selection checkpoint, Notch and CXCR4 signaling. In vitro and in situ analyses indicate that the immunological synapse promotes passage through the β-selection checkpoint. Collectively, these data indicate that developing T cells regulate pre-TCR signaling through the formation of an immunological synapse. This signaling platform integrates cues from Notch, CXCR4, and MHC on the thymic stromal cell to allow transition beyond the β-selection checkpoint.


1999 ◽  
Vol 189 (10) ◽  
pp. 1679-1684 ◽  
Author(s):  
Sylvie Degermann ◽  
Giuseppina Sollami ◽  
Klaus Karjalainen

The striking and unique structural feature of the T cell receptor (TCR) β chain is the bulky solvent-exposed FG loop on the Cβ domain, the size of almost half an immunoglobulin domain. The location and size of this loop suggested immediately that it could be a crucial structural link between the invariant CD3 subunits and antigen-recognizing α/β chains during TCR signaling. However, functional analysis does not support the above notion, since transgene coding for TCR β chain lacking the complete FG loop supports normal α/β T cell development and function.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-30-SCI-30
Author(s):  
Jennifer Puck

Abstract Early diagnosis of rare immune disorders is important for clinical care and of great interest for the study of immune pathways. Severe combined immunodeficiency (SCID) is a collective term for the most profound inherited defects of T cell development combined with B cell defects or dysfunction. While fatal without treatment, SCID is treatable by allogeneic hematopoietic cell transplantation, or in certain genotypes by enzyme or gene therapy. Avoidance of life-threatening infections to provide optimal treatment and outcomes for affected infants has led to population-based SCID newborn screening (NBS). Infants with SCID fail to generate a diverse repertoire of functional T cells, and consequently have very low numbers of T cells and T cell receptor excision circles (TRECs), DNA byproducts of T cell receptor gene rearrangement. TRECs are readily measured in DNA isolated from newborn dried blood spots (DBS) collected for population based screening. Thus newborn screening for insufficient TRECs identifies SCID before infections occur. As SCID NBS has become widespread, new disease definitions are required for healthy-appearing affected infants without failure to thrive or opportunistic infections. Typical SCID cases have <300 autologous T cells/uL, <10% of the lower range of normal proliferation to the mitogen phytohemmaglutinin A, and/or detectable maternal T cell engraftment, most often with deleterious mutations in recognized SCID genes. One fourth of all SCID cases are "leaky" due to hypomorphic SCID gene mutations; these cases are also detected by TREC testing; they may have >300 T cells/uL, but have impaired T cell function and lack naïve CD4 T cells expressing CD45RA. A subset of infants with leaky SCID have Omenn syndrome, with expansion of oligoclonal, dysregulated T cells leading to adenopathy, erythroderma, hepatosplenomegaly, eosinophilia, and elevated IgE. In addition to these primary target disorders of population screening for SCID, the TREC test identifies infants with additional conditions due to either impaired production or increased loss of T cells. Non-SCID congenital syndromes with variable degrees of T cell lymphopenia (TCL) include DiGeorge syndrome/22q11.2 deletion, CHARGE syndrome, trisomy 21, and ataxia telangiectasia, among others. TREC NBS also finds infants with secondary TCL, in which T cell generating capacity is intrinsically normal, but circulating T cells are diminished as a consequence of other factors, including hydrops, congenital heart disease, chylothorax, neonatal leukemia, maternal immunosuppressive medications taken during pregnancy, or extreme preterm birth. The T cells of these infants normalize once their primary problems resolve. A particularly challenging group of infants are those with abnormal TREC screen results and non-SCID TCL, but no immediate diagnosis. About half of these have syndromes, such as DiGeorge/22q deletion, but with mild or initially unapparent features; others experience resolution of TCL over time, while still others prove to have previously unknown immune disorders that may be diagnosed after deep sequencing and research functional studies. It is important to remember that many serious disorders of T cells are not identified by TREC screening if the block in T cell development or function occurs at a later stage than T cell receptor rearrangement; combined immunodeficiencies (CIDs) with TRECs that are often normal include Zap-70 deficiency and MHC class I and II non-expression. Thus, while virtually completely sensitive and highly specific for the intended target, SCID, population based TREC screening leaves us with both diagnostic dilemmas presented by non-symptomatic infants with low T cells and inability to capture individuals with the >300 non-SCID primary immunodeficiency disorders that would also benefit from early intervention. Deep sequencing is not yet clinically useful, not only due to cost, turnaround time and technical limitations, but primarily due to problems of interpretation, given the extraordinary number of genomic variants of uncertain significance. Disclosures Puck: InVitae, a clinical DNA sequencing company: Other: Spouse employment and stock options; UpToDate: Patents & Royalties: Recieve royalties to write and edit entries on primary immune disorders.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 917-917
Author(s):  
Richard XuFeng ◽  
Qiong Yang ◽  
Youzhong Yuan ◽  
Binfeng Lu ◽  
Tao Cheng ◽  
...  

Abstract Abstract 917 Post-transcriptional regulation such as RNA editing in hematopoiesis and lymphopoiesis is poorly understood. ADAR1 (adenosine deaminase acting on RNA-1) is a RNA editing enzyme essential for embryonic development. Disruption of the ADAR1 gene was shown to cause defective embryonic hematopoiesis (Wang Q et al, Science 2000). Moreover, we have recently obtained direct evidence for the preferential effect of ADAR1 deletion on adult hematopoietic progenitor cells as opposed to the more primitive cells via a RNA-editing dependent mechanism by different conditional gene deletion strategies (Xufeng R et al PNAS 2009, in press). To further determine the role of ADAR1 in T cell development, we generated a mouse model in which ADAR1 was deleted specifically in T lymphocytes by interbreeding ADAR1lox/lox mice with Lck-Cre transgenic mice. In our current study, we report that ADAR1 is essential for T cell differentiation at the late progenitor stage in the thymus, coincident with T cell receptor-α/β expression. In ADAR1lox/loxLck-Cre mice, mature T cells decreased dramatically in peripheral blood, spleen and lymph nodes in comparison to littermate controls. In the thymus, the production of CD4+/CD8+ double positive cells was severely impaired and massive cell death was observed in pre-T cell populations. Within the pro-T cells, ADAR1 deletion resulted in a significant decrease of late progenitor cells but not early progenitor subsets. In both pro-T and pre-T cell stages, defective T cell development preferentially occurred in the beta chain positive cells, but was not apparent in gamma/delta T cells. Our data demonstrated an indispensable role of ADAR1 in early T cell differentiation that correlated with T cell receptor beta chain expression, thereby indicating that RNA editing by ADAR1 is an essential event in T cell development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1467-1467
Author(s):  
Tianjiao Wang ◽  
Ye Lu ◽  
Avery Polk ◽  
Carlos Murga Zamalloa ◽  
Ryan A Wilcox

Abstract Background: Most (≈ 95%) T-cell lymphomas (TCL) express an intact T-cell receptor (TCR), suggesting that malignant T cells, like their B-cell counterparts, may benefit from antigen-receptor signaling. TCR engagement culminates in the activation of pathways required for T-cell proliferation and survival. The Tec family kinase and BTK homologue ITK is required for optimal TCR-dependent signaling. Therefore, we sought to understand whether TCR activation promotes chemotherapy resistance in TCL, and whether this may be overcome upon inhibition of ITK. Methods: TCL cell lines, mouse models and primary patient specimens were utilized. TCR signaling was engaged by anti-CD3/CD28 beads. ITK and GATA3 were inhibited by lentiviral-mediated shRNA knockdown and by ibrutinib, an ITK inhibitor. The chemoresistance of TCL cells was investigated in vitro and in vivo. Results: We have previously shown by gene expression profiling, proliferation, cytokine release, and signaling pathway analysis that TCR signaling remains intact in TCL [Blood, 2014, 124(21), 2959]. To further investigate the effect of TCR on chemoresistance, T8ML1 (a PTCL, NOS cell line) and primary cells from TCL patients (n=4) were treated with either vincristine or romidepsin in vitro. For T8ML1, the viability of cells treated with either vincristine or romidepsin increased by 2.9+/-0.14 fold or 1.4 +/- 0.035 fold respectively (p<0.01) upon TCR engagement. The enhanced viability of T8ML1 by TCR engagement under either vincristine or romidepsin treatment was abolished by shRNA-mediated ITK knockdown and significantly inhibited by ibrutinib (p<0.01). Similarly for primary TCL patient cells, the viability of cells treated with romidepsin increased by 2.4 +/- 0.35 fold upon TCR engagement, which was also significantly inhibited by ibrutinib treatment (p<0.01). To study the mechanism of TCR signaling in TCL chemoresistance, downstream targets of TCR signaling (NFκB, GATA3) were examined. Upon TCR engagement, NFκB activity increased by 1.7 +/- 0.22 fold in T8ML1 and 1.8 +/- 0.36 fold in primary cells from TCL patients (p<0.01) as shown by DNA-binding and nuclear localization. The activation of NFκB in T8ML1 and primary TCL patient cells was significantly inhibited by ibrutinib (p<0.01). We and others have previously shown that GATA3 identifies a distinct subset of PTCL, NOS that is characterized by inferior progression-free survival following anthracycline-based chemotherapy. [Blood, 2014, 123 (19), 3007-3015; Blood, 2014, 123(19), 2915-2923]. Furthermore, GATA-3 regulates the homeostatic survival of normal T cells following TCR engagement. In addition to NFκB, GATA3 protein increased by 3.0 +/- 0.45 fold in T8ML1 (p<0.01) and by 3.6 +/- 3.0 fold in primary TCL patient cells (p<0.03) upon TCR engagement, which was also inhibited by ibrutinib treatment. Furthermore, in T8ML1 cells, GATA3 upregulation by TCR engagement was abolished by shRNA-mediated ITK knockdown. To study the effect of GATA3 on chemoresistance, GATA3 was knocked down by lentiviral-mediated shRNAs in TCL cell lines (T8ML1, H9 and MyLa). The viability of TCL lines following GATA3 knockdown decreases by 2-4 fold (p<0.01) following treatment with either vincristine, romidepsin or 4-hydroxycyclophosphamide. In comparison to tumor xenografts generated fromTCL lines transduced with a non-targeting shRNA, GATA3 deficient tumor xenografts were significantly more sensitive to vincristine alone or combined vincristine/cyclophosphamide (p<0.01). Conclusions: TCR engagement promotes resistance to chemotherapy in T-cell lymphomas in an ITK- and GATA-3-dependent manner. Furthermore, chemotherapy resistance following TCR engagement is significantly impaired by ibrutinib. Therefore, ibrutinib may warrant further investigation in the T-cell lymphomas. Disclosures Off Label Use: We used ibrutinib to inhibit T-cell receptor signaling and discussed its clinical implication in T-cell lymphomas..


2007 ◽  
Vol 204 (11) ◽  
pp. 2537-2544 ◽  
Author(s):  
Gabrielle M. Siegers ◽  
Mahima Swamy ◽  
Edgar Fernández-Malavé ◽  
Susana Minguet ◽  
Sylvia Rathmann ◽  
...  

The γδ T cell receptor for antigen (TCR) comprises the clonotypic TCRγδ, the CD3 (CD3γε and/or CD3δε), and the ζζ dimers. γδ T cells do not develop in CD3γ-deficient mice, whereas human patients lacking CD3γ have abundant peripheral blood γδ T cells expressing high γδ TCR levels. In an attempt to identify the molecular basis for these discordant phenotypes, we determined the stoichiometries of mouse and human γδ TCRs using blue native polyacrylamide gel electrophoresis and anti-TCR–specific antibodies. The γδ TCR isolated in digitonin from primary and cultured human γδ T cells includes CD3δ, with a TCRγδCD3ε2δγζ2 stoichiometry. In CD3γ-deficient patients, this may allow substitution of CD3γ by the CD3δ chain and thereby support γδ T cell development. In contrast, the mouse γδ TCR does not incorporate CD3δ and has a TCRγδCD3ε2γ2ζ2 stoichiometry. CD3γ-deficient mice exhibit a block in γδ T cell development. A human, but not a mouse, CD3δ transgene rescues γδ T cell development in mice lacking both mouse CD3δ and CD3γ chains. This suggests important structural and/or functional differences between human and mouse CD3δ chains during γδ T cell development. Collectively, our results indicate that the different γδ T cell phenotypes between CD3γ-deficient humans and mice can be explained by differences in their γδ TCR composition.


1999 ◽  
Vol 190 (7) ◽  
pp. 973-982 ◽  
Author(s):  
Joonsoo Kang ◽  
Mark Coles ◽  
David H. Raulet

Mice lacking the interleukin 7 receptor (IL-7R) generate α/β T cells at a detectable but greatly reduced rate, but γ/δ T cells are completely absent. The special role of IL-7R signaling in γ/δ T cell development has remained unclear. IL-7Rα−/− mice exhibit a paucity of γ gene rearrangements. This striking observation can be explained by a defect in T cell receptor (TCR)-γ gene rearrangement, a defect in TCR-γ gene transcription leading to death of γ/δ lineage cells, and/or a requirement for IL-7R in commitment of cells to the γ/δ lineage. To determine the role of IL-7R signaling in γ/δ T cell development, we examined transcription of a prerearranged TCR-γ transgene in IL-7Rα−/− mice, as well as the effects of IL-7 on transcription of endogenous, rearranged TCR-γ genes in α/β lineage cells. The results demonstrate that IL-7R–mediated signals are necessary for the normal expression of rearranged TCR-γ genes. Equally significant, the results show that the poor expression of TCR-γ genes in IL-7Rα−/− mice is responsible for the selective deficit in γ/δ cells in these mice, since a high copy TCR-γ transgene exhibited sufficient residual expression in IL-7Rα−/− mice to drive γ/δ cell development. The results indicate that the absence of γ/δ T cells in IL-7Rα−/− mice is due to insufficient TCR-γ gene expression.


2001 ◽  
Vol 194 (7) ◽  
pp. 903-914 ◽  
Author(s):  
Isabelle Corre ◽  
Manuel Gomez ◽  
Susina Vielkind ◽  
Doreen A. Cantrell

Loss of function of the guanine nucleotide binding protein RhoA blocks pre-T cell differentiation and survival indicating that this GTPase is a critical signaling molecule during early thymocyte development. Previous work has shown that the Rho family GTPase Rac-1 can initiate changes in actin dynamics necessary and sufficient for pre-T cell development. The present data now show that Rac-1 actions in pre-T cells require Rho function but that RhoA cannot substitute for Rac-1 and induce the actin cytoskeletal changes necessary for pre-T cell development. Activation of Rho is thus not sufficient to induce pre-T cell differentiation or survival in the absence of the pre-T cell receptor (TCR). The failure of RhoA activation to impact on pre-TCR–mediated signaling was in marked contrast to its actions on T cell responses mediated by the mature TCR α/β complex. Cells expressing active RhoA were thus hyperresponsive in the context of TCR-induced proliferation in vitro and in vivo showed augmented positive selection of thymocytes expressing defined TCR complexes. This reveals that RhoA function is not only important for pre-T cells but also plays a role in determining the fate of mature T cells.


Blood ◽  
2005 ◽  
Vol 106 (5) ◽  
pp. 1726-1733 ◽  
Author(s):  
Delphine Goux ◽  
Jérôme D. Coudert ◽  
Diane Maurice ◽  
Leonardo Scarpellino ◽  
Grégoire Jeannet ◽  
...  

Abstract Intrathymic T-cell maturation critically depends on the selective expansion of thymocytes expressing a functionally rearranged T-cell receptor (TCR) β chain. In addition, TCR-independent signals also contribute to normal T-cell development. It is unclear whether and how signals from the 2 types of pathways are integrated. Here, we show that T-cell factor-1 (TCF-1), a nuclear effector of the canonical wingless/int (wnt)/catenin signaling pathway, ensures the survival of proliferating, pre-TCR+ thymocytes. The survival of pre-TCR+ thymocytes requires the presence of the N-terminal catenin-binding domain in TCF-1. This domain can bind the transcriptional coactivator β-catenin and may also bind γ-catenin (plakoglobin). However, in the absence of γ-catenin, T-cell development is normal, supporting a role for β-catenin. Signaling competent β-catenin is present prior to and thus arises independently from pre-TCR signaling and does not substantially increase on pre-TCR signaling. In contrast, pre-TCR signaling significantly induces TCF-1 expression. This coincides with the activation of a wnt/catenin/TCF reporter transgene in vivo. Collectively, these data suggest that efficient TCF-dependent transcription requires that pre-TCR signaling induces TCF-1 expression, whereas wnt signals may provide the coactivator such as β-catenin. The 2 pathways thus have to cooperate to ensure thymocyte survival at the pre-TCR stage. (Blood. 2005;106:1726-1733)


2018 ◽  
Vol 115 (34) ◽  
pp. E8027-E8036 ◽  
Author(s):  
Wei-Chan Hsu ◽  
Ming-Yu Chen ◽  
Shu-Ching Hsu ◽  
Li-Rung Huang ◽  
Cheng-Yuan Kao ◽  
...  

Activated T cells undergo metabolic reprogramming and effector-cell differentiation but the factors involved are unclear. Utilizing mice lacking DUSP6 (DUSP6−/−), we show that this phosphatase regulates T cell receptor (TCR) signaling to influence follicular helper T (TFH) cell differentiation and T cell metabolism. In vitro, DUSP6−/− CD4+ TFH cells produced elevated IL-21. In vivo, TFH cells were increased in DUSP6−/− mice and in transgenic OTII-DUSP6−/− mice at steady state. After immunization, DUSP6−/− and OTII-DUSP6−/− mice generated more TFH cells and produced more antigen-specific IgG2 than controls. Activated DUSP6−/− T cells showed enhanced JNK and p38 phosphorylation but impaired glycolysis. JNK or p38 inhibitors significantly reduced IL-21 production but did not restore glycolysis. TCR-stimulated DUSP6−/− T cells could not induce phosphofructokinase activity and relied on glucose-independent fueling of mitochondrial respiration. Upon CD28 costimulation, activated DUSP6−/− T cells did not undergo the metabolic commitment to glycolysis pathway to maintain viability. Unexpectedly, inhibition of fatty acid oxidation drastically lowered IL-21 production in DUSP6−/− TFH cells. Our findings suggest that DUSP6 connects TCR signaling to activation-induced metabolic commitment toward glycolysis and restrains TFH cell differentiation via inhibiting IL-21 production.


Sign in / Sign up

Export Citation Format

Share Document