scholarly journals Lunatic Fringe Controls T Cell Differentiation through Modulating Notch Signaling

2006 ◽  
Vol 177 (12) ◽  
pp. 8365-8371 ◽  
Author(s):  
Shin-ichi Tsukumo ◽  
Kayo Hirose ◽  
Yoichi Maekawa ◽  
Kenji Kishihara ◽  
Koji Yasutomo
PLoS ONE ◽  
2019 ◽  
Vol 14 (4) ◽  
pp. e0215012 ◽  
Author(s):  
Dave Maurice De Sousa ◽  
Frédéric Duval ◽  
Jean-François Daudelin ◽  
Salix Boulet ◽  
Nathalie Labrecque

2015 ◽  
Vol 194 (12) ◽  
pp. 5654-5662 ◽  
Author(s):  
Mélissa Mathieu ◽  
Frédéric Duval ◽  
Jean-François Daudelin ◽  
Nathalie Labrecque

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1699-1699
Author(s):  
Keizo Kato ◽  
Barbara Varnum-Finney ◽  
Irwin D. Bernstein

Abstract Notch Signaling in Hematopoietic Precursor Cells Maintains the Expression of Genes Required for Stem Cell Self-renewal and Promotes the Expression of Genes Associated with T cell Differentiation. Keizo Kato, Barbara Varnum-Finney, Irwin D. Bernstein Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA We have previously shown that Notch signaling promotes the self-renewal of hematopoietic precursors, including short-term repopulating cells, and induces early T-cell differentiation. Here we evaluate gene expression in murine Lin-Sca-1+c-kit+ Hoechst side population (SKSP) bone marrow cells during culture with the immobilized Notch ligand, Delta1ext-IgG, consisting of the extracellular domain of Delta1 fused to the Fc domain of human-IgG1 for 28 days with SCF, IL-6, IL-11 and Flt-3-ligand. We analyzed hematopoietic stem cell (HSC) associated genes, including polycomb genes, Bmi-1 and Rae28, required for HSC self-renewal, and Rex-1 required for embryonic stem cell self-renewal, together with early T-lymphoid differentiation associated genes such as GATA-3, pre-Ta and CD3e, by semi-quantitative or real-time RT-PCR. After culture for 7 or 14 days with Delta1ext-IgG, the expression of Bmi-1, Rae28 and Rex-1 was greater in cultures containing Delta1ext-IgG compared to those without. Bmi-1, Rae28 and Rex-1 were likely not direct targets of Notch signaling since the expression in SKSP cells was equivalent after 3 hours culture in the presence or absence of Notch ligand, whereas rapid up-regulation of the direct Notch target Hes1 was observed 3 hours after incubation with Delta1ext-IgG. Expression of GATA-3, pre-Ta and CD3e was induced by Notch signaling since their expression was seen by 7-14 days with but not without Delta1ext-IgG. Furthermore, the expression of these genes was dependent on Notch signaling since removal of cells from Notch ligand after culture for 28 days led to a rapid reduction of Hes1 expression within 3 hours, and a slower reduction in genes associated with self-renewal observed after 2 days. Our results suggest that Notch signaling regulates the self-renewal of hematopoietic precursors by maintaining the expression of genes known to be required for stem cell self-renewal, while also promoting the expression of T-cell differentiation-associated genes.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3156-3156
Author(s):  
Hongfang Wang ◽  
L. Jeanne Pierce ◽  
Gerald J. Spangrude

Abstract Notch signaling plays a critical role in T lineage commitment during lymphoid differentiation. However, Notch signaling alone is not sufficient to support T cell development through the CD4/CD8 double positive (DP) stage in vitro. We here report distinct effects of several cytokines on T cell differentiation in the OP9-DL1 cell culture model. Our studies show that Flt3 ligand enhances the proliferation of progenitors but has no obvious effect on differentiation. In contrast, stem cell factor (SCF) favors the proliferation of CD4/CD8 double negative (DN) lymphoid progenitors and inhibits differentiation to the DP stage in a dose-dependent manner. Differentiation of the NK lineage is promoted under these conditions. Conversely, blocking the function of SCF that is expressed endogenously by OP9-DL1 cells inhibits proliferation of lymphoid progenitors and accelerates T lineage differentiation. IL-7 is necessary for differentiation from the DP to the CD8 single positive (SP) stage, and is also required for γδ T lineage development. We also find a dosage effect of IL-7 during T cell development. OP9 and OP9-DL1 stromal cells produce endogenous levels of IL-7 that are sufficient to support B and DP T cell differentiation. However, the amount of endogenous IL-7 is not sufficient to support T cell differentiation from the DP to the SP stage. Addition of exogenous IL-7 (1–10 ng/ml) to the cultures promotes SP differentiation, while blocking endogenous IL-7 with anti-IL-7 antibody inhibits both B and T cell development. We conclude that activation through the Notch pathway is sufficient to suppress B lineage differentiation and thereby promote T lineage commitment, but is not sufficient to promote the subsequent stages of T cell development. SCF promotes expansion and directs NK lineage differentiation at the expense of T cell development, while IL-7 provides both proliferation as well as T lineage differentiation signals. T cell development from the DN to the DP stage requires a low amount of IL-7, while differentiation from the DP to the SP stage requires a higher level of IL-7. The balance between the effects mediated by these cytokines, along with Notch signaling, plays a critical role in regulating development of the T and NK lineages.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2470-2470
Author(s):  
James A Kennedy ◽  
Renata Teixeira ◽  
Sara Berthiaume ◽  
Frederic Barabe

Abstract Abstract 2470 LMO2 is overexpressed in a significant percentage of human T cell acute lymphoblastic leukemia (T-ALL) and its locus has been the target of insertional mutagenesis in gene therapy trials. In the past years, 4 X-linked severe combined immunodeficiency (X-linked SCID) and one Wiskott-Aldrich syndrome (WAS) patients who were treated by retrovirus-mediated gene therapy developed T-ALL as a result of retroviral integration in the LMO2 locus. In these patients, leukemia developed 2 to 3 years after gene therapy without prior significant haematological abnormalities. However, both the latency of disease and the finding of additional somatic mutations and/or translocations in these leukemias suggest that the overexpression of LMO2 alone is insufficient to generate leukemia, a notion that has been supported by studies in mouse. Though LMO2 is typically recognized as a T-cell oncogene, reports have shown that it is also aberrantly expressed in acute myeloid leukemias (AML), chronic myeloid leukemia (CML), B-ALL and some non-hodgkin B cell lymphomas. In order to study the impact of LMO2 overexpression on human hematopoietic stem/progenitor cells, a lentiviral vector was used to express this oncogene together with EGFP in lineage-depleted umbilical cord blood. In myeloid-promoting cultures, LMO2 had no effect on either differentiation or proliferation. Moreover, the expression of LMO2 did not modify the frequency or lineage distribution of colony forming progenitors compared to controls. However, significant differences were noted when transduced cells were assayed on OP9-Delta-Like 1 (DL1) stroma, an in vitro system that promotes T cell proliferation and differentiation. Cells overexpressing LMO2 were blocked at the double negative stage (CD4-CD8-) of differentiation and proliferated 50 to 100 times more than control cells. However, these cells were not immortalized as they proliferated for a median of 75 days, versus 50 days for controls. Immunodeficient mice transplanted with primitive human hematopoietic cells expressing LMO2 (hereafter referred as LMO2 mice) had bone marrow engraftment levels comparable to controls at 20–24 weeks post-transplant. Neither B-lymphoid nor myeloid development were affected by LMO2 overexpression. Strikingly, in the thymus, the percentage of EGFP+ cells was significantly increased in LMO2 mice compared to controls (mean of 47.7% versus 8.8%, p=0.0001), clearly indicating that expression of this oncogene enhances thymic T-cell engraftment. We next analyzed the phenotype of LMO2-expressing T cells in the thymus and peripheral blood of these mice. Surprisingly, unlike our in vitro studies, there was no evidence of a block at the DN-stage of differentiation. Instead, there were significantly less EGFP+ DN cells in the thymi of LMO2 mice compared to controls (mean of 7.5% vs 14.5%, p=0.035). These results clearly demonstrate that unlike what was observed in OP9-DL1 co-cultures, LMO2 overexpression does not induce a block in T-cell differentiation in our in vivo system. One possible explanation for this difference is the constitutive NOTCH signaling provided via DL1 on stroma compared to the in vivo setting where LMO2-expressing cells would encounter different levels and forms of NOTCH signaling throughout development. To test this hypothesis, LMO2 cells were cultured on OP9-DL1 stroma for 50 days then switched onto OP9 stroma lacking NOTCH ligand. Upon transfer, the DN cells promptly stopped proliferating and differentiated into DP (CD4+CD8+) cells expressing CD3 and TCRαβ. Thus, our results suggest that in the in vivo setting, as cells migrate through the thymus and face a decrease in NOTCH signaling, LMO2 overexpression alone can promote proliferation, but is not sufficient to maintain a differentiation block. However, constitutive NOTCH signaling can cooperate with LMO2 overexpression to block T cell differentiation at a proliferative DN stage. Thus, one can postulate that LMO2 exerts a proliferative effect on developing T-cells in thymic regions with high levels of NOTCH signaling, potentially providing a setting for the development of secondary leukemogenic events. NOTCH mutations are common in human T-ALL and can therefore allow for LMO2 overexpressing cells to become independent of the stromal niche. Taken together, our results suggest cooperation between LMO2 overexpression and NOTCH signaling in human T-cell leukemogenesis. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 191 (5) ◽  
pp. 2344-2350 ◽  
Author(s):  
Floriane Auderset ◽  
Steffen Schuster ◽  
Nicolas Fasnacht ◽  
Manuel Coutaz ◽  
Mélanie Charmoy ◽  
...  

2005 ◽  
Vol 95 (3) ◽  
pp. 543-558 ◽  
Author(s):  
Mirentxu Santos ◽  
Paula Río ◽  
Sergio Ruiz ◽  
Jesús Martínez-Palacio ◽  
Carmen Segrelles ◽  
...  

Author(s):  
H. Alasam

The possibility that intrathymic T-cell differentiation involves stem cell-lymphoid interactions in embryos led us to study the ultrastructure of epithelial cell in normal embryonic thymus. Studies in adult thymus showed that it produces several peptides that induce T-cell differentiation. Several of them have been chemically characterized, such as thymosin α 1, thymopoietin, thymic humoral factor or the serum thymic factor. It was suggested that most of these factors are secreted by populations of A and B-epithelial cells.Embryonic materials were obtained from inbred matings of Swiss Albino mice. Thymuses were disected from embryos 17 days old and prepared for transmission electron microscopy. Our studies showed that embryonic thymus at this stage contains undifferentiated and differentiated epithelial cells, large lymphoblasts, medium and few small lymphocytes (Fig. 5). No differences were found between cortical and medullary epithelial cells, in contrast to the findings of Van Vliet et al,. Epithelial cells were mostly of the A-type with low electron density in both cytoplasm and nucleus. However few B-type with high electron density were also found (Fig. 7).


2001 ◽  
Vol 120 (5) ◽  
pp. A517-A517
Author(s):  
A MIZOGUCHI ◽  
E MIZOGUCHI ◽  
Y DEJONG ◽  
H TAKEDATSU ◽  
F PREFFER ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document