scholarly journals Loss of CD44dim Expression from Early Progenitor Cells Marks T-Cell Lineage Commitment in the Human Thymus

2017 ◽  
Vol 8 ◽  
Author(s):  
Kirsten Canté-Barrett ◽  
Rui D. Mendes ◽  
Yunlei Li ◽  
Eric Vroegindeweij ◽  
Karin Pike-Overzet ◽  
...  
Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 548-548
Author(s):  
Fabian Zohren ◽  
George Souroullas ◽  
Min Luo ◽  
Ulrike Gerdemann ◽  
Nicola K Wilson ◽  
...  

Abstract Abstract 548 Long-term thymopoiesis crucially depends on the recruitment and expansion of bone marrow derived progenitor cells. Therefore, a tight regulation of the thymus-settling progenitor cells is required to maintain T cell lineage homeostasis. Lyl1, a transcription factor involved in homeostatic control of immature hematopoietic cells, remains expressed in early T-lineage progenitors (ETPs) until T cell lineage commitment. Here we demonstrate a critical requirement for Lyl1 in lymphoid priming of bone marrow (BM) progenitors and in the maintenance of ETPs. Lyl1 deficient hematopoiesis was unable to generate sufficient numbers of lymphoid-primed progenitor populations such as LMPPs, CLPs and in particular ETPs. We found a significant (p>0.001) 4-fold reduction of LMPPs in the BM and a 20-fold reduction (p<0.001) of ETPs in the thymus of Lyl1 KO mice compared to wildtype controls. Transplantation assays revealed a selective defect of Lyl1−/− LMPPs for thymic engraftment and T lineage development. Intra-thymic injections of Lyl1−/− LMPPs demonstrated, in absence of homing requirements, a cell autonomous defect of Lyl1−/− progenitors in their ability to undergo the ETP to DN2 transition and to expand in response to Notch. Injection of Lyl1+/+ LMPPs resulted in a 5-fold higher recovery of total donor cells compared to injections of Lyl1−/− LMPPs (p<0.001). 40% of the donor cells derived after intra-thymic injection of Lyl1+/+ LMPPs were committed to the T cell lineage (DN3, DP, SP), whereas T lineage commitment after injection of Lyl1−/− cells was only seen in 20% of the recovered cells (p<0.01). By absolute numbers, wildtype LMPPs had generated more DN3 (8-fold), more DP (21-fold) and more SP (9-fold) T cell lineage committed thymocytes, and fewer Lyl1+/+ cells remained in the c-kit positive “ETP/LMPP-like” stage (-0.5-fold). In contrast, reintroduction of Lyl1 cDNA into Lyl1−/− BM progenitors using retroviral vectors restored the thymic progenitor pool and enhanced T cell lineage output. At 12 weeks after transplantation of MIG-Lyl1 transduced cells we observed a significant expansion of transfected (GFPpos) cells in the peripheral blood solely attributable to expansion of normal, mature and poly-clonal T cells (p<0.001). The thymuses of MIG-Lyl1 transplanted recipients showed significantly greater overall cellularity (p<0.01) attributable to a significantly higher proportion of GFPpos thymocytes (p<0.01) compared to the MIG-GFP transplanted control group. To gain a more detailed understanding of the underlying molecular mechanisms of Lyl1-mediated T-lymphoid specification, we performed global gene expression profiling of wildtype and Lyl1−/− LMPPs as well as whole genome ChIP-seqencing in HPC-7 cells after pull-down with anti-Lyl1 antibodies. Here, we identified the lymphoid-promoting factor Gfi1 as a critical transcriptional target of Lyl1-mediated T lymphopoiesis. We found that Gfi1 expression was decreased in Lyl1−/− LMPPs and ETPs by 2- and 7.5- fold respectively. ChIP assays using ckitpos BM cells from Lyl1+/+ and Lyl1−/− mice revealed a strong enrichment of Lyl1 at a known enhancer region of the Gfi1 locus located 35kb upstream of Gfi1 (p<0.001). Binding of Lyl1 activated the Gfi1 35kb enhancer element in transactivation assays (p<0.001). Finally, intra-thymic injection of Lyl1−/− progenitors after retroviral transduction with Gfi1 cDNA allowed lymphoid development and enhanced the T cell lineage output compared to GFP-transduced controls (p=0.08). Collectively, our data provide evidence that pro-T cell expansion in the thymus is regulated through intrinsic control of thymus progenitor cells that employ a transcriptional program already established in hematopoietic stem and progenitor cells. We identify Lyl1 as a critical component of this regulatory network, which is vital for the maintenance of T cell lineage homeostasis. Finally, identification of important downstream mediators of Lyl1 function not only illuminates the molecular mechanisms underlying early T-cell development, but also suggests previously unrecognized pathways likely to play a role in Lyl1-mediated development of leukemia and lymphoma. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1995 ◽  
Vol 86 (5) ◽  
pp. 1850-1860 ◽  
Author(s):  
TA Moore ◽  
A Zlotnik

The earliest steps of intrathymic differentiation recently have been elucidated. It has been reported that both CD4lo (CD44+ CD25- c-kit+ CD3- CD4lo CD8-) and pro-T cells (CD44+ CD25+ c-kit+ CD3- CD4- CD8-, representing the next step in maturation) exhibit germline T-cell receptor beta and gamma loci, suggesting that neither population is exclusively committed to the T-cell lineage. Several groups have shown that CD4lo cells retain the capacity to generate multiple lymphoid lineages in vivo; however, the lineage commitment status of pro-T cells is unknown. To determine when T-cell lineage commitment occurs, we examined the ability of sorted CD4lo and pro-T cells to generate lymphoid lineage cells in vivo or in fetal thymic organ cultures (FTOCs). When intravenously injected into scid mice, CD4lo cells generated both T and B cells, whereas the progeny of pro-T cells contained T cells exclusively. Fetal thymic organ cultures repopulated with CD4lo cells contained both T and natural killer (NK) cells, whereas cultures repopulated with pro-T cells contained T cells almost exclusively. These observations strongly suggest that T-cell lineage commitment occurs during the transition of CD4lo to pro-T cells. Because it is likely that the thymic microenvironment plays a critical role in T-cell commitment, we compared the responses of CD4lo and pro-T cells to various cytokine combinations in vitro, as well as the ability of the cultured cells to repopulate organ cultures. Cytokine combinations that maintained T-cell repopulation potential for both CD4lo and pro-T cells were found. CD4lo cells proliferated best in response to the combination containing interleukin-1 (IL-1), IL-3, IL- 6, IL-7, and stem cell factor (SCF). Unlike CD4lo cells, pro-T cells were much more dependent upon IL-7 for proliferation and FTOC repopulation. However, combinations of cytokines lacking IL-7 were found that maintained the T-cell repopulating potential of pro-T cells, suggesting that, whereas this cytokine is clearly very important for normal pro-T cell function, it is not an absolute necessity during early T-cell expansion and differentiation.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 30-30
Author(s):  
Sjoukje van der Stegen ◽  
Pieter Lindenbergh ◽  
Roseanna Petrovic ◽  
Benjamin Whitlock ◽  
Raedun Clarke ◽  
...  

Chimeric Antigen Receptor (CAR) T cells are a new treatment paradigm for relapsed/refractory hematopoietic malignancies. However, their autologous nature imposes manufacturing constraints that can delay CAR T cell availability and increase their cost. We previously established proof of principle that αβ T cell-derived induced pluripotent stem cells (TiPSCs) can provide a self-renewing source for in vitro CAR T cell production (Themeli, Nat Biotechnol, 2013). The use of cloned TiPSC further enhances the feasibility of verifying genome integrity of the genetically engineered stem cells and should in principle yield highly homogenous cell products. Using αβ T cell-derived TiPSCs transduced with a well-defined CD19-specific CAR (1928z; Park, NEJM, 2018), we previously demonstrated that TiPSCs can be differentiated into CAR T cells. These T cells retained their endogenous T cell receptor (TCR) and also displayed characteristics of innate lymphoid cells. We have now examined how the timing of CAR expression as well as the CAR signaling strength influence T cell lineage commitment, enabling better control towards αβ T cell lineage commitment. αβ T cell lineage development depends in part on a precisely orchestrated interactions between NOTCH and (pre)TCR signaling, the timing and strength of which are crucial for αβ lineage commitment. Because TiPSCs harbor rearranged TCRα and TCRβ genes, mature TCR expression occurs earlier than if it required VDJ recombination, skewing differentiation towards acquiring innate features including CD4-CD8- double-negative or CD8αα single-positive phenotypes. We show that providing strong NOTCH stimulation counteracts the effects of early antigen receptor expression, facilitating CD4+CD8αβ+ double positive (DP) formation. We hypothesized that CAR signaling in the absence of ligand binding (tonic signaling) may mimic a TCR signal, the strength and timing of which could re-direct lineage commitment. We therefore investigated CARs providing different levels of signaling strength and the impact of delaying the onset of CAR expression. Tonic CAR signaling was measured in peripheral blood T cells expressing 1928z or 1928z-1XX, a construct in which the second and third ITAM in the CD3ζ domain have been mutated to be non-functional (Feucht, Nat Med, 2019), following either retroviral transduction (SFG vector) orTRAC-targeted cDNA integration, placing CAR expression under the transcriptional control of the TCRα promoter (Eyquem, Nature, 2017). CAR signaling in the absence of antigen exposure, measured by phosphorylation of ITAM3, ERK1/2 and ZAP70, was reduced by bothTRAC-targeting and reduction of functional ITAMs, with additive effects when combined inTRAC-1928z-1XX. Three of these engineering strategies (virally expressed 1928z,TRAC-1928z andTRAC-1928z-1XX) were evaluated in the context of TiPSC-derived T cell differentiation. Virally expressed 1928z (resulting in constitutive CAR expression throughout differentiation) resulted in the predominant generation of innate-like CD8αα T cells, associated with the absence of early T cell lineage markers such as CD5, CD2 and CD1a. Delayed expression of 1928z throughTRACtargeting resulted in increased CD5, CD2 and CD1a, but did not yield any more CD4+CD8αβ+ DP cells. In TiPSC expressingTRAClocus-encoded 1928z-1XX, a greater DP population emerged, from which CD8αβ single-positive T cells could be induced. Phenotypic analyses of clonal TRAC-1928z-1XX TiPSC lines further establish the interplay between CAR and NOTCH1 in determining αβ lineage commitment. Together these data show that early TCR and CAR expression skew T cell lineage commitment towards an innate-like T cell fate, which can be overcome by controlling the strength and timing of NOTCH, TCR and CAR signaling. These studies pave the way for the predetermined generation of a variety of CAR T cell types endowed with different functional attributes. Disclosures Whitlock: Fate Therapeutics Inc.:Current Employment, Current equity holder in publicly-traded company.Clarke:Fate Therapeutics Inc.:Current Employment, Current equity holder in publicly-traded company.Valamehr:Fate Therapeutics, Inc:Current Employment, Current equity holder in publicly-traded company.Riviere:Juno Therapeutics:Other: Ownership interest, Research Funding;Takeda:Research Funding;Fate Therapeutics Inc.:Consultancy, Other: Ownership interest , Research Funding;FloDesign Sonics:Consultancy, Other: Ownership interest;Atara:Research Funding.Sadelain:Atara:Patents & Royalties, Research Funding;Fate Therapeutics:Patents & Royalties, Research Funding;Mnemo:Patents & Royalties;Takeda:Patents & Royalties, Research Funding;Minerva:Other: Biotechnologies , Patents & Royalties.


1999 ◽  
Vol 11 (4) ◽  
pp. 239-249 ◽  
Author(s):  
Adrian C. Hayday ◽  
Domingo F. Barber ◽  
Nataki Douglas ◽  
Eric S. Hoffman

Immunity ◽  
2000 ◽  
Vol 12 (3) ◽  
pp. 313-322 ◽  
Author(s):  
Gabriela Hernández-Hoyos ◽  
Sue J Sohn ◽  
Ellen V Rothenberg ◽  
José Alberola-Ila

Sign in / Sign up

Export Citation Format

Share Document