scholarly journals Genome-Wide Analysis Identifies Rag1 and Rag2 as Novel Notch1 Transcriptional Targets in Thymocytes

Author(s):  
Yang Dong ◽  
Hao Guo ◽  
Donghai Wang ◽  
Rongfu Tu ◽  
Guoliang Qing ◽  
...  

Recombination activating genes 1 (Rag1) and Rag2 are expressed in immature lymphocytes and essential for generating the vast repertoire of antigen receptors. Yet, the mechanisms governing the transcription of Rag1 and Rag2 remain to be fully determined, particularly in thymocytes. Combining cDNA microarray and ChIP-seq analysis, we identify Rag1 and Rag2 as novel Notch1 transcriptional targets in acute T-cell lymphoblastic leukemia (T-ALL) cells. We further demonstrate that Notch1 transcriptional complexes directly bind the Rag1 and Rag2 locus in not only T-ALL but also primary double negative (DN) T-cell progenitors. Specifically, dimeric Notch1 transcriptional complexes activate Rag1 and Rag2 through a novel cis-element bearing a sequence-paired site (SPS). In T-ALL and DN cells, dimerization-defective Notch1 causes compromised Rag1 and Rag2 expression; conversely, dimerization-competent Notch1 achieves optimal upregulation of both. Collectively, these results reveal Notch1 dimerization-mediated transcription as one of the mechanisms for activating Rag1 and Rag2 expression in both primary and transformed thymocytes. Our data suggest a new role of Notch1 dimerization in compelling efficient TCRβ rearrangements in DN progenitors during T-cell development.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2389-2389
Author(s):  
Lars Fransecky ◽  
Martin Neumann ◽  
Sandra Heesch ◽  
Marco Seehawer ◽  
Cornelia Schlee ◽  
...  

Abstract Abstract 2389 Introduction The transcription factor GATA3 plays an important role in normal T cell development. Its role in mature T cells is well understood, but its function in earlier stages of T cell development remains unclear. Whereas GATA3 levels are precisely regulated for the T cell differentiation program, aberrant expression of GATA3 has been linked to tumorigenesis. Based on these observations, we investigated the role of GATA3 in Early Thymic Progenitor Acute Lymphoblastic Leukemia (ETP-ALL), a newly defined high-risk subgroup of T-ALL, characterized by a specific gene expression profile and distinct immunophenotype. Patients and Methods Eighty-six bone marrow samples from adult patients with newly diagnosed T-ALL, including ETP-ALL (n=17) enrolled into the German Multicenter Acute Lymphoblastic Leukemia (GMALL) trials, were studied for GATA3 expression by oligonucleotide expression arrays (HG-U133 plus 2.0) within the Microarray Innovations in LEukemia study. We identified additional 71 ETP-ALL adult patients and 94 T-ALL patients enrolled on the GMALL protocol, in which GATA3 mRNA expression was measured by quantitative polymerase chain reaction (RT-PCR). Combining ETP-ALL and T-ALL cases (n=165), we defined two GATA3 expression groups GATA3null and GATA3high based on a biological gap (GATA3 expression of 0.2). DNA methylation was analyzed in both T-ALL (n=11) and ETP-ALL (n=69) samples by pyrosequencing with primers designed to include seven CpG sites of Exon 2/Intron 3 of GATA3. Samples were grouped into GATA3 high vs. low methylation according to their mean methylation being below or above 40%. Results Based on gene expression arrays we observed a high proportion of ETP-ALL (11/17) that lacked GATA3 expression, whereas only a small fraction of the remaining T-ALL cases (3/69) had no GATA3 expression. These results were validated by RT-PCR in a larger cohort: 26% of ETP-ALL (19/71) were GATA3null, but only 2% of T-ALL (2/94) were in the GATA3null expression group. To explore the regulation of this specific expression pattern, epigenetic regulation of GATA3 was analyzed by pyrosequencing. While unselected T-ALL samples were hypomethylated (< 6% methylated CpG), ETP-ALL samples had a higher GATA3 methylation status (28% methylated CpG, p<0.001). ETP-ALL cases were further categorized into high methylated (18/69) and low methylated samples (51/69) and correlated to mRNA expression. GATA3null samples showed a higher degree of GATA3 methylation (41% methylated CpGs) compared to GATA3high samples (8% methylated CpGs, p < 0.001). In an in-vitro assay of T-cell leukemia cell lines demethylating agents increased GATA3 mRNA expression by up to 5-fold. In murine hematopoetic stem cells it was shown that loss of DNMT3A induced GATA3 expression via hypomethylation. In ETP-ALL, we identified 11 DNMT3A mutations in 69 samples (16%) and correlated the DNMT3A mutation status to GATA3 methylation. Ten of 11 (91%) DNMT3A mutated samples showed low level GATA3 methylation, whereas 17 (29%) of the 58 DNMT3A wildtype cases had high methylation. Conclusion ETP-ALL is a subgroup of adult T-ALL with a distinct molecular profile. Here we show that within ETP-ALL a separate molecular entity can be defined by GATA3 silencing due to DNA methylation. In-vitro studies showed that GATA3 expression can be restored by the use of demethylating agents. As loss of function mutations in DNMT3A correlate with low GATA3 methylation in ETP-ALL, a potential role of DNMT3A in the epigenetic silencing of GATA3 is suspected. So far, the number of targeted drugs available for T-ALL is limited. Therefore, incorporating demethylating agents may resolve the T-cell differentiation block in T-ALL by increasing GATA3 expression. Future work will explore downstream effects of GATA3 in acute leukemia. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 9 ◽  
Author(s):  
Ana Rosa Pérez ◽  
Alexandre Morrot ◽  
Vinicius Frias Carvalho ◽  
Juliana de Meis ◽  
Wilson Savino

2003 ◽  
Vol 191 (1) ◽  
pp. 119-138 ◽  
Author(s):  
Julie A. Lucas ◽  
Andrew T. Miller ◽  
Luana O. Atherly ◽  
Leslie J. Berg

2018 ◽  
Vol 11 (12) ◽  
pp. 945-956 ◽  
Author(s):  
Mattia Algeri ◽  
Francesca Del Bufalo ◽  
Federica Galaverna ◽  
Franco Locatelli

PLoS ONE ◽  
2015 ◽  
Vol 10 (7) ◽  
pp. e0134100 ◽  
Author(s):  
Heather L. Evans-Marin ◽  
Anthony T. Cao ◽  
Suxia Yao ◽  
Feidi Chen ◽  
Chong He ◽  
...  

2013 ◽  
Vol 2013 ◽  
pp. 1-8 ◽  
Author(s):  
Mark Engel ◽  
Tom Sidwell ◽  
Ajithkumar Vasanthakumar ◽  
George Grigoriadis ◽  
Ashish Banerjee

Regulatory T cells (Tregs) are a subset of CD4 T cells that are key mediators of immune tolerance. Most Tregs develop in the thymus. In this review we summarise recent findings on the role of diverse signalling pathways and downstream transcription factors in thymic Treg development.


2008 ◽  
Vol 22 (S1) ◽  
Author(s):  
Guo Fu ◽  
Nathalie Niederberger ◽  
Nicholas RJ Gascoigne

Blood ◽  
1989 ◽  
Vol 73 (8) ◽  
pp. 2133-2138
Author(s):  
A Biondi ◽  
E Champagne ◽  
V Rossi ◽  
G Giudici ◽  
A Cantu-Rajnoldi ◽  
...  

During the development of functional T lymphocytes, a variety of genes involved in antigen recognition undergo somatic rearrangement. These include the alpha, beta, and gamma chain genes. Recently a fourth rearranging gene, the delta chain gene, embedded in the alpha chain locus, has been described. We have determined the structure of the beta, gamma, and delta chain genes in 15 cases of T-cell acute lymphoblastic leukemia (T-ALL) representing stage I (CD7+, CD1-, CD3-) and stage II (CD7+, CD1+, CD3-) of intrathymic T-cell development. The alpha-delta locus was rearranged in 14 of the 15 cases. In three cases the delta constant region was deleted on both chromosomes, suggesting biallelic V-J alpha rearrangement. A limited pattern of rearrangement of the delta locus was observed in the remaining 11 cases. When the alpha-delta region was rearranged, there was rearrangement of the beta and gamma TcR in all cases except two; in these cases the beta chain was in the germline configuration. These findings support the hypothesis that delta chain gene rearrangement is an early event in T- cell development, possibly contemporary to gamma gene rearrangement, and that the delta locus has a limited repertoire.


Cells ◽  
2020 ◽  
Vol 9 (7) ◽  
pp. 1726
Author(s):  
Valentina Saccomani ◽  
Angela Grassi ◽  
Erich Piovan ◽  
Deborah Bongiovanni ◽  
Ludovica Di Martino ◽  
...  

T-cell acute lymphoblastic leukemia (T-ALL) is a rare, aggressive disease arising from T-cell precursors. NOTCH1 plays an important role both in T-cell development and leukemia progression, and more than 60% of human T-ALLs harbor mutations in components of the NOTCH1 signaling pathway, leading to deregulated cell growth and contributing to cell transformation. Besides multiple NOTCH1 target genes, microRNAs have also been shown to regulate T-ALL initiation and progression. Using an established mouse model of T-ALL induced by NOTCH1 activation, we identified several microRNAs downstream of NOTCH1 activation. In particular, we found that NOTCH1 inhibition can induce miR-22-3p in NOTCH1-dependent tumors and that this regulation is also conserved in human samples. Importantly, miR-22-3p overexpression in T-ALL cells can inhibit colony formation in vitro and leukemia progression in vivo. In addition, miR-22-3p was found to be downregulated in T-ALL specimens, both T-ALL cell lines and primary samples, relative to immature T-cells. Our results suggest that miR-22-3p is a functionally relevant microRNA in T-ALL whose modulation can be exploited for therapeutic purposes to inhibit T-ALL progression.


Sign in / Sign up

Export Citation Format

Share Document