scholarly journals Essential Role of Survivin, an Inhibitor of Apoptosis Protein, in T Cell Development, Maturation, and Homeostasis

2003 ◽  
Vol 199 (1) ◽  
pp. 69-80 ◽  
Author(s):  
Zheng Xing ◽  
Edward M. Conway ◽  
Chulho Kang ◽  
Astar Winoto

Survivin is an inhibitor of apoptosis protein that also functions during mitosis. It is expressed in all common tumors and tissues with proliferating cells, including thymus. To examine its role in apoptosis and proliferation, we generated two T cell–specific survivin-deficient mouse lines with deletion occurring at different developmental stages. Analysis of early deleting survivin mice showed arrest at the pre–T cell receptor proliferating checkpoint. Loss of survivin at a later stage resulted in normal thymic development, but peripheral T cells were immature and significantly reduced in number. In contrast to in vitro studies, loss of survivin does not lead to increased apoptosis. However, newborn thymocyte homeostatic and mitogen-induced proliferation of survivin-deficient T cells were greatly impaired. These data suggest that survivin is not essential for T cell apoptosis but is crucial for T cell maturation and proliferation, and survivin-mediated homeostatic expansion is an important physiological process of T cell development.

2007 ◽  
Vol 3 (1) ◽  
pp. 57-75 ◽  
Author(s):  
Ross La Motte-Mohs ◽  
Geneve Awong ◽  
Juan Carlos Zuniga-Pflucker

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2980-2980
Author(s):  
Christian Reimann ◽  
Liliane Liliane Dal-Cortivo ◽  
Emmanuelle M. Six ◽  
Andrea Schiavo ◽  
Marina Cavazzana-Calvo ◽  
...  

Abstract Abstract 2980 Notchligand-based culture systems such as OP9-DL1 cells induce HSC to engage towards the T-cell developmental program and allow generation of T-lymphoid progenitors in vitro. In vitro generated murine T-lymphoid progenitors accelerated T-cell reconstitution in vivo. In consistency, human T-lymphoid progenitors generated in co-culture with OP9-DL1 cells enhanced thymic repopulation when injected into NOD/SCID/gc−/− mice (NSG). However, positive effects of human T-lymphoid progenitors on peripheral T-cell reconstitution have not been reported yet. Besides, Notchligand-based culture systems, consisting of genetically modified murine cells might raise safety concern for clinical use. It has been described that exposure of CD34+ cells to immobilized DL4 induces the T-cell developmental program even in absence of stromal cell support. Recently, we have made use of this system to generate T-lymphoid progenitors in vitro. In the present study we have further characterized their T-lymphoid potential in vitro and in vivo. Exposure of human CB-derived CD34+ cells to immobilized DL4 allowed generation of CD34+CD7+ and CD34−CD7++CD5+ progenitors displaying a similar phenotype as early thymic progenitors (ETP) and the prethymocytes (pre-T). Within the DL-4 derived ETP- and preT-like progenitors we observed subsequent up regulation of genes involved in T-cell development and silencing of genes implied in B-cell and myeloid differentiation. T-cell commitment of DL-4 progenitors could be further confirmed by early and intermediate rearrangement events within the TCR d/g/b genes. The pattern of gene expression profile and TCR-rearrangement events displayed a pattern similar to what we observed in corresponding intrathymic developmental stages. DL4-progenitors obtained after 7 days of culture displayed a 30-fold increased in vitro T-lymphoid potential as compared to untreated CD34+ CB progenitors. DL4 ETP-like and preT-like progenitors further completed T-cell differentiation in vitro (in OP9DL1 co-culture) faster than native CD34+ CB progenitors. When transferred into NSG, DL4 progenitors obtained after 7 days of culture were able to repopulate the recipients' thymus and to give rise to mature, polyclonal intrathymic and peripheral T-cells. Two months after transfer recipients of DL4 progenitors displayed advanced intrathymic T-cell development as compared to recipients of CD34+ CB cells. Furthermore, peripheral T-cells could be observed in a number of DL-4 progenitor recipients but not in control mice. Our experiments provide further evidence that DL4 allows in vitro induction of T-cell development and generation of early T-lymphoid progenitors in a system devoid of stromal cell support. These progenitors feature phenotypical and molecular characteristics of immature thymic developmental stages. Moreover, they are able to accelerate T-cell development in vitro and when transferred into NSG. This work provides further evidence of the ability of in vitro -generated human T-cell progenitors to accelerate T-cell reconstitution and simultaneously introduces a culture technique that could be rapidly transferred into a clinical setting. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
John M. Edgar ◽  
Peter W. Zandstra

ABSTRACTT-cell development from hematopoietic stem and progenitor cells (HSPCs) is tightly regulated through Notch pathway activation by the Notch ligands Delta-like (DL) 1 and 4 and Jagged-2. Other molecules, such as stem cell factor (SCF), FMS-like tyrosine kinase 3 ligand (Flt3L) and interleukin (IL)-7, play a supportive role in regulating the survival, differentiation, and proliferation of developing progenitor (pro)T-cells. Numerous other signaling molecules are known to instruct T-lineage development in vivo, but little work has been done to optimize their use for T-cell production in vitro. Using a defined T-lineage differentiation assay consisting of plates coated with the Notch ligand DL4 and adhesion molecule VCAM-1, we performed a cytokine screen that identified IL-3 and tumor necrosis factor α (TNFα) as enhancers of proT-cell differentiation and expansion. Mechanistically, we found that TNFα induced T-lineage differentiation through the positive regulation of T-lineage genes GATA3, TCF7, and BCL11b. TNFα also synergized with IL-3 to induce proliferation by upregulating the expression of the IL-3 receptor on CD34+ HSPCs, yielding 753.2 (532.4-1026.9; 5-95 percentile)-fold expansion of total cells after 14 days compared to 8.9 (4.3-21.5)-fold expansion in conditions without IL-3 and TNFα. We then optimized cytokine concentrations for T-cell maturation. Focusing on T-cell maturation, we used quantitative models to optimize dynamically changing cytokine requirements and used these to construct a three-stage assay for generating CD3+CD4+CD8+ and CD3+CD4−CD8+ T-cells. Our work provides new insight into T-cell development and a robust in vitro assay for generating T-cells to enable clinical therapies for treating cancer and immune disorders.


2006 ◽  
Vol 26 (9) ◽  
pp. 3639-3648 ◽  
Author(s):  
Uwe Kölsch ◽  
Börge Arndt ◽  
Dirk Reinhold ◽  
Jonathan A. Lindquist ◽  
Nicole Jüling ◽  
...  

ABSTRACT The transmembrane adaptor molecule TRIM is strongly expressed within thymus and in peripheral CD4+ T cells. Previous studies suggested that TRIM is an integral component of the T-cell receptor (TCR)/CD3 complex and might be involved in regulating TCR cycling. To elucidate the in vivo function of TRIM, we generated TRIM-deficient mice by homologous recombination. TRIM−/− mice develop normally and are healthy and fertile. However, the animals show a mild reduction in body weight that appears to be due to a decrease in the size and/or cellularity of many organs. The morphology and anatomy of nonlymphoid as well as primary and secondary lymphoid organs is normal. The frequency of thymocyte and peripheral T-cell subsets does not differ from control littermates. In addition, a detailed analysis of lymphocyte development revealed that TRIM is not required for either positive or negative selection. Although TRIM−/− CD4+ T cells showed an augmented phosphorylation of the serine/threonine kinase Akt, the in vitro characterization of peripheral T cells indicated that proliferation, survival, activation-induced cell death, migration, adhesion, TCR internalization and recycling, TCR-mediated calcium fluxes, tyrosine phosphorylation, and mitogen-activated protein family kinase activation are not affected in the absence of TRIM. Similarly, the in vivo immune response to T-dependent and T-independent antigens as well as the clinical course of experimental autoimmune encephalomyelitis, a complex Th1-mediated autoimmune model, is comparable to that of wild-type animals. Collectively, these results demonstrate that TRIM is dispensable for T-cell development and peripheral immune functions. The lack of an evident phenotype could indicate that TRIM shares redundant functions with other transmembrane adaptors involved in regulating the immune response.


2007 ◽  
Vol 204 (13) ◽  
pp. 3077-3084 ◽  
Author(s):  
Huiling Liang ◽  
Andrew H. Coles ◽  
Zhiqing Zhu ◽  
Jennifer Zayas ◽  
Roland Jurecic ◽  
...  

The Wnt–β-catenin signaling pathway has been shown to govern T cell development by regulating the growth and survival of progenitor T cells and immature thymocytes. We explore the role of noncanonical, Wnt–Ca2+ signaling in fetal T cell development by analyzing mice deficient for Wnt5a. Our findings reveal that Wnt5a produced in the thymic stromal epithelium does not alter the development of progenitor thymocytes, but regulates the survival of αβ lineage thymocytes. Loss of Wnt5a down-regulates Bax expression, promotes Bcl-2 expression, and inhibits apoptosis of CD4+CD8+ thymocytes, whereas exogenous Wnt5a increases apoptosis of fetal thymocytes in culture. Furthermore, Wnt5a overexpression increases apoptosis in T cells in vitro and increases protein kinase C (PKC) and calmodulin-dependent kinase II (CamKII) activity while inhibiting β-catenin expression and activity. Conversely, Wnt5a deficiency results in the inhibition of PKC activation, decreased CamKII activity, and elevation of β-catenin amounts in thymocytes. These results indicate that Wnt5a induction of the noncanonical Wnt–Ca2+ pathway alters canonical Wnt signaling and is critical for normal T cell development.


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 1074
Author(s):  
Veronica Della Chiara ◽  
Lucia Daxinger ◽  
Frank J. T. Staal

Hematopoietic multipotent progenitors seed the thymus and then follow consecutive developmental stages until the formation of mature T cells. During this process, phenotypic changes of T cells entail stage-specific transcriptional programs that underlie the dynamic progression towards mature lymphocytes. Lineage-specific transcription factors are key drivers of T cell specification and act in conjunction with epigenetic regulators that have also been elucidated as crucial players in the establishment of regulatory networks necessary for proper T cell development. In this review, we summarize the activity of transcription factors and epigenetic regulators that together orchestrate the intricacies of early T cell development with a focus on regulation of T cell lineage commitment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3304-3304
Author(s):  
Fukun Guo ◽  
David Hildeman ◽  
David A. Williams ◽  
Yi Zheng

Abstract The Rac subfamily GTPases of the Rho family have been implicated in the control of actin dynamics, cell proliferation, apoptosis, adhesion and migration of many blood cell types including hematopoietic stem/progenitors, neutrophils and macrophages, but their role in T cell development remains poorly understood. T cells from the Rac2 deficient mice appear to mostly undergo normal development, whereas previous constitutively active mutant Rac2 or Rac1 overexpression studies suggest Rac GTPases are required for CD4+ and CD8+ T cell maturation. Using conditional gene targeting, we have achieved specific deletion of Rac1 or Rac1 together with Rac2 in the T cell lineage by cross-breeding the Lck-Cre transgenic mice with the Rac1flox/flox mice that contain a pair of loxP sites sandwiching the exon 1 sequences of Rac1 or the Rac1flox/flox;Rac2−/− mice. We show that similar to Rac2 deficiency, inactivation of Rac1 alone had little effect on various developmental stages of T cells in the animal. However, deletion of both Rac1 and Rac2 significantly affected both the immature CD4−CD8− (2.3 fold increase) and CD4+CD8+ (13% decrease) populations in the mouse thymus and the mature CD4+ and CD8+ populations in the thymus and spleen (Table). These developmental defects are associated with proliferation defects of thymocytes and splenocytes in response to ConA or PMA/ionomycin stimulation and deficient survival of various T cell populations at different developmental stages (Table). Together, these data show that Rac1 and Rac2 play overlapping and obligatory roles in T-cell development and serve as important cell survival regulators at various stages. Table. Frequency and apoptosis rate of different T-cell subsets in thymocytes and splenocytes T cell subsets WT (n=10) Rac1−/− (n=6) Rac1−/−Rac2−/−(n=6) Total thymocyte number (×106) 101.3±30.0 98.0±25.0 44.7±25.5 CD4−CD8− thymocyte frequency (%) 5.5±1.9 4.5±0.7 12.7±4.3 apoptosis rate (%) 20.1±2.2 15.0±1.3 CD4+CD8+ thymocyte frequency (%) 76.2±3.2 77.3±4.1 66.2±5.4 apoptosis rate (%) 18.8±4.3 27.9±2.8 CD4+ thymocyte frequency (%) 14.5±3.4 14.4±2.4 7.9±2.3 apoptosis rate (%) 13.3±2.3 21.5±4.5 CD8+ thymocyte frequency (%) 3.9±1.2 3.8±1.0 13.2±2.2 apoptosis rate (%) 12.5±2.2 8.8±1.1 Total splenocyte number (×106) 60.4±21.8 62.0±13.0 51.1±28.9 CD4+TCRβ+ splenocyte frequency (%) 9.7±2.2 8.0±2.3 3.2±1.1 apoptosis rate (%) 15.1±3.1 27.5±6.9 CD8+TCRβ+ splenocyte frequency (%) 3.2±0.8 2.4±0.9 0.6±0.4 apoptosis rate (%) 13.1±3.0 24.5±6.4


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3712-3712
Author(s):  
Christian Reimann ◽  
Andrea Schiavo ◽  
Julien Rouiller ◽  
Elodie Vidal ◽  
Kheira Beldjord ◽  
...  

Abstract Abstract 3712 Injection of donor derived T-cellular precursors has been proposed as a novel strategy to shorten delayed reconstitution of the T-lymphoid compartment following HSCT. In the past years, several research groups have successfully generated murine and human T-cellular precursors in vitro using Notchligand-based coculture systems such as OP9-DL1 or Tst-DL4. Murine T-cellular precursors generated in vitro, promoted reconstitution of the T-cellular compartment when applied in murine HSCT-models. In consistency, transfer of human T-cellular precursors, generated in vitro in coculture with OP9-DL1 or Tst-DL4 resulted in enhanced thymic repopulation in NOD/SCID/gc−/− mice. Yet, positive effects on peripheral T-cell reconstitution have not been reported. Moreover, clinical application of OP9-DL1 or Tst-DL4 coculture systems is limited, since they consist of murine stromal cells transduced with either DL1 or DL4. It has been described that exposure of CD34+ cells to immobilized DL4 induces T-cell differentiation in vitro and allows expansion human T-cellular precursors even in absence of stromal cell support. However, the hypothesis that DL4 alone can drive hematopoietic progenitors towards a T-cell fate in vitro, requires more evidence. Here, we further characterized the in vitro and in vivo potential of T-cellular precursors generated by single exposure to DL4. We exposed human CD34+ progenitors to immobilized DL4 in the presence of different cytokine combinations implicated in human haematopoiesis. Within 7 days, CD34+CD7+ and CD34−CD7++ T-cellular precursors emerged in the presence of DL4, but not under control conditions. After 7 days the CD34+CD7+ population subsequently declined while the CD34−CD7++ population further expanded. Two distinct progenitor subsets, CD5+ and CD5-, emerged within the CD34−CD7++ population. The CD34−CD7++CD5+ subset partially acquired CD1a, corresponding to a developmental stage between the early thymic progenitor (ETP) and the prethymocyte (pre-T) stage. Conversely to what observed in the OP9-DL1 system, T-cell development did not progress beyond the pre-T-stage. Indeed, we neither observed more advanced stages of T-cell development, such as immature single positive CD4+ cells, nor complete TCR-rearrangements. 7-day exposure to immobilized DL4 induced a 90-fold increase of T-precursor frequency in CD34+ progenitors (1/8800 before culture vs. 1/90 after culture) as confirmed by limiting dilution assays on OP9-DL1. All T-cellular precursor activity was restricted to cells expressing CD34, CD7 or both (frequency: 1/9). In particular, elevated T-cellular precursor levels were found in the subsets expressing CD7 (CD34+/CD7+ and CD34−/CD7+), while the T-cellular precursor frequency in the CD34+/CD7− subset was equal to that seen in non-cultured CD34+ progenitors. In consistency the CD34−CD7− population did not contain any detectable T-cellular precursors. After 7 day exposure to DL4, cells phenotypically corresponding to T-cellular precursors were transferred into NOD/SCID/gc−/− mice. Within 2 months following HSCT, cells exposed to DL4 were able to reconstitute the recipients' thymus and partially gave rise to peripheral T-cells. When injecting non-cultured CD34+ progenitors, thymic reconstitution was likewise seen 2 months after HSCT. However, intrathymic T-cell development was less advanced and peripheral T-cells were absent. In contrast, cells cultured in presence of a control peptide did not retain any potential to repopulate the recipients' thymus. Our experiments provide further evidence that exposure DL4 induces early human T-cell development and allows generation of large numbers of T-cellular precursors in vitro. These precursors feature phenotypical and molecular properties corresponding to early precursors found in the human thymus. Furthermore, they have an increased potential to further differentiate into mature T-cells in vitro and when transferred into immunodeficient mice. Our preliminary data suggest, that injection of T-cellular precursors accelerates T-cell reconstitution after HSCT and provides further evidence for the feasibility of this novel strategy of immunotherapy. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Wook-Jin Chae ◽  
Alfred L. M. Bothwell

The control of inflammatory diseases requires functional regulatory T cells (Tregs) with significant Gata-3 expression. Here we address the inhibitory role of Tregs on intestinal tumorigenesis in theApc/Min+mouse model that resembles human familial adenomatous polyposis (FAP).Apc/Min+mice had a markedly increased frequency of Foxp3+ Tregs and yet decreased Gata-3 expression in the lamina propria. To address the role of heterozygousApcgene mutation in Tregs, we generatedFoxp3-Cre,Apcflox/+mice. Tregs from these mice effectively inhibited tumorigenesis comparable to wild type Tregs after adoptive transfer intoApc/Min+mice, demonstrating that the heterozygousApcgene mutation in Tregs does not induce the loss of control over tumor microenvironment. Adoptive transfer of in vitro generatedApc/Min+iTregs (inducible Tregs) failed to inhibit intestinal tumorigenesis, suggesting that naïve CD4 T cells generated fromApc/Min+mice thymus were impaired. We also showed that adoptively transferred IL-17A-deficientApc/Min+Tregs inhibited tumor growth, suggesting that IL-17A was critical to impair the tumor regression function ofApc/Min+Tregs. Taken together, our results suggest that both T cell development in a functional thymus and IL-17A control the ability of Treg to inhibit intestinal tumorigenesis inApc/Min+mice.


1995 ◽  
Vol 182 (6) ◽  
pp. 1973-1983 ◽  
Author(s):  
D A Grillot ◽  
R Merino ◽  
G Núñez

The survival of T lymphocytes is tightly controlled during development. Here, we show that Bcl-xL, a protein homologue of Bcl-2, is highly regulated in the thymus in a pattern different than that of Bcl-2. The maximum expression was in CD4+CD8+ thymocytes, a developmental stage where Bcl-2 is downregulated. To assess the role of Bcl-xL in thymocyte apoptosis, we generated mice overexpressing an E mu-bcl-x transgene within the T cell compartment. Constitutive expression of Bcl-xL resulted in accumulation of thymocytes and mature T cells in lymphoid organs. Thymocytes overexpressing Bcl-xL exhibited increased viability in vitro and were resistant to apoptosis induced by different signals, including glucocorticoid, gamma irradiation, calcium ionophore, and CD3 cross-linking. However, Bcl-xL was unable to block clonal deletion of thymocytes reactive with self-superantigens or H-Y antigen. These studies demonstrate that Bcl-2 and Bcl-xL, two functionally related proteins, are regulated independently during T cell development. In contrast to Bcl-2, which has been implicated in the maintenance of mature T cells, Bcl-xL appears to provide a survival signal for the maintenance of more immature CD4+CD8+ thymocytes before positive selection.


Sign in / Sign up

Export Citation Format

Share Document