Transcription factors in hematopoietic differentiation and leukemia

Author(s):  
Michael Chapman ◽  
Anthony Green
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2997-2997
Author(s):  
Irene Riz ◽  
Sergey S. Akimov ◽  
Shannon S. Eaker ◽  
Leonardo Marino-Ramirez ◽  
David Landsman ◽  
...  

Abstract The diverged TLX1/HOX11 homeobox gene is frequently transcriptionally activated in T-cell acute lymphoblastic leukemia as a result of two recurrent chromosomal translocations or by as yet unknown mechanisms. Sporadic expression of TLX1 has also been detected in some other human tumors, raising the possibility that TLX1 played a role in the etiology of these malignancies. In this context, it is noteworthy that TLX1 is aberrantly expressed in the human erythroleukemia cell line K562. We previously reported that enforced TLX1 expression immortalizes myeloid progenitors in murine bone marrow and erythroid progenitors originating from murine embryonic stem cell-derived embryoid bodies. Based on these findings, we speculated that dysregulated TLX1 expression contributes to neoplastic transformation by interfering with hematopoietic differentiation programs. Here we carried out genome-wide expression profiling on these model systems to elucidate the mechanism of TLX1-mediated differentiation arrest. Surprisingly, these investigations uncovered a latent erythroid phenotype of the TLX1+ bone marrow progenitor cell lines. Transcriptome comparison with murine GATA-1-null G1E-ER4 erythroblast cells (GEO Accession No. GDS568) showed nonrandom overlap (P = 2.3 × 10−5) with a set of genes that immediately respond to GATA-1 activation. Included among the genes for which a positive correlation was observed were the erythropoietin (Epo) receptor and several functionally-associated downstream signaling components such as Lyn and Dok-1, as well as the SCL and FOG-1 transcription factor genes. We confirmed the biological relevance of these findings by demonstrating that TLX1-immortalized bone marrow progenitors proliferated in response to erythropoietin, synthesizing beta-globin mRNA. We next extended this approach to iEBHX1S-4 cells, an embryoid body-derived cell line generated by conditional (doxycycline-controlled) TLX1 expression (manuscript in preparation). iEBHX1S-4 cells require IL-3 plus stem cell factor for survival and proliferation. However, when doxycycline is removed from the culture medium, the cells undergo Epo-dependent erythroid differentiation characterized by up-regulation of the TER119 surface antigen and hemoglobin synthesis. Interestingly, whereas there was statistically significant overlap of gene expression profiles between constitutive and conditional TLX1+ cells (P = 2.5 × 10−5), there were no significant similarities between the iEBHX1S-4 transcriptome and the G1E-ER4 dataset until 6–24 hours after release of the TLX1 differentiation block. Bioinformatics analysis of the upstream regulatory regions of the genes identified implicated transcriptional networks involving GATA-1 as well as the p53, NF-kappaB and Egr-1 transcription factors. All of these transcription factors are substrates of the acetyltransferase CREB-binding protein (CBP), suggesting that inappropriate TLX1 expression might interfere with CBP activity. In agreement with this notion, acetylation of GATA-1, a key substrate of CBP-modulated erythroid differentiation, was increased upon down-regulation of TLX1 expression in iEBHX1S-4 cells. Experiments to directly confirm TLX1 inhibition of CBP as a central facet of TLX1 transforming function are ongoing, the results of which will be presented.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 550-550
Author(s):  
Marc A Kerenyi ◽  
Jessica Hsu ◽  
Zhen Shao ◽  
Stuart H Orkin

Abstract Abstract 550 Lysine specific demethylase 1 (LSD1) is a demethylase that acts on mono- and dimethylated H3K4 (H3K4me1/2). Consistent with H3K4me2 (an active marker of transcription) as a substrate, LSD1 is part of a core complex with the co-repressor, CoREST and HDAC1/2. Previously our lab demonstrated that regulation of hematopoietic differentiation depends in part on the interaction of the growth factor independent transcription factors (= Gfi1 and Gfi1b) with the LSD1/CoREST/HDAC complex. We generated a conditional knock out mouse for LSD1 (LSD1fl/fl) to study its roles in hematopoiesis. Inducible deletion of LSD1fl/fl mice in all hematopoietic lineages with Mx-Cre resulted in severe neutropenia. Flow cytometric analysis showed that LSD1fl/fl Mx-Cre mice lacked Gr-1high Mac-1high double positive mature neutrophilic granulocytes in the bone marrow and the peripheral blood; however, the frequency of Gr-1dim Mac-1high (mainly consisting of promyelocytes and myeloblasts but not mature neutrophils) increased in frequency. To reveal the mechanism responsible for the observed neutropenia, we performed global mRNA expression profiling and chromatin immunoprecipitation sequencing (ChIPSeq) for H3K4 methylation states in Gr-1dim Mac-1high cells from LSD1fl/fl Mx-Cre and LSD1fl/fl mice. Five hundred ninety-eight genes (412 up / 186 down; p≤0.01, 2-fold cutoff) were differentially expressed in the absence of LSD1. Although we did not detect changes in expression of established myeloid transcription factors, including Pu.1, C/EBPα, C/EBPε or Gfi1, gene set enrichment analysis (GSEA) of Gr-1dim Mac-1high cells from LSD1fl/fl Mx-Cre using gene signatures for mature myeloid cells clearly showed that LSD1 deficient Gr-1dim Mac-1high cells failed to display a gene signature of differentiated myeloid cells (NES: 1.88; p≤0.003). Among the most highly upregulated genes, we observed genes highly expressed in hematopoietic stem and progenitor cells (HSPCs; i.e.: CD34 36.2-fold; HoxA9 26.3-fold; Sca-1 10.8-fold; Meis 1 2.6-fold). Therefore we performed GSEA using signatures from HSPCs (encompassing over 200 genes); the stem/progenitor gene set was highly significantly enriched (NES: −1.9; p<10−4) in LSD1 deficient Gr-1dim Mac-1high cells. Chromatin immunoprecipitation sequencing did not reveal any global changes in the amount of H3K4me2/3 histone methylation, however many genes critical for HSPCs, including Meis1 and the entire HoxA gene locus, where more strongly H3K4me2/3 marked than in control cells, which is in concord with the gene expression data. To determine if LSD1 represses stem/progenitor genes in additional lineages, we analyzed the effects of LSD1 loss in erythroid cell development through breeding with EpoR-Cre. Wild type, as well as control embryos, were recovered at Mendalian ratios up to E12.5, but no live LSD1fl/fl EpoR-Cre embryos were observed after E15.5. At E13.5, LSD1-deficient embryos were smaller and paler as compared to control embryos. Flow cytometry revealed a severe differentiation defect at the transition from pro-erythroblasts to basophilic erythroblasts, resulting in a paucity of more mature erythroid cells. To unravel molecular mechanisms responsible for this deficit, we performed gene expression profiling of wild type and knock out CD71+ c-kit+ Ter119lo pro-erythroblasts. Again, we did not detect changes in the expression levels of established erythroid transcription factors, including Gata-1, Klf1, SCL/Tal1, NF-E1, Ldb1, Lmo2 or Myb. By GSEA analysis we observed that LSD1 deficient CD71+ c-kit+ Ter119lo pro-erythroblasts displayed higher expression of the hematopoietic stem and progenitor cell gene signatures (NES: −2.4; p<10−4), a finding strikingly similar to the data in myeloid cells. Therefore, LSD1 is required in multiple hematopoietic lineages to repress stem/progenitor gene expression programs in maturing cells. We propose that repression of these early programs is essential for subsequent hematopoietic differentiation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (20) ◽  
pp. 4219-4228 ◽  
Author(s):  
Lorrie L. Delehanty ◽  
Grant C. Bullock ◽  
Adam N. Goldfarb

Abstract In red cell development, the differentiation program directed by the transcriptional regulator GATA1 requires signaling by the cytokine erythropoietin, but the mechanistic basis for this signaling requirement has remained unknown. Here we show that erythropoietin regulates GATA1 through protein kinase D activation, promoting histone deacetylase 5 (HDAC5) dissociation from GATA1, and subsequent GATA1 acetylation. Mice deficient for HDAC5 show resistance to anemic challenge and altered marrow responsiveness to erythropoietin injections. In ex vivo studies, HDAC5−/− progenitors display enhanced entry into and passage through the erythroid lineage, as well as evidence of erythropoietin–independent differentiation. These results reveal a molecular pathway that contributes to cytokine regulation of hematopoietic differentiation and offer a potential mechanism for fine tuning of lineage-restricted transcription factors by lineage-specific cytokines.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 110-110
Author(s):  
Eun-Young Ahn ◽  
Tsunehito Higashi ◽  
Ming Yan ◽  
Shinobu Matsuura ◽  
Miao-Chia Lo ◽  
...  

Abstract Abstract 110 While characterizing AML1-ETO domains important for leukemia development and identifying proteins interacting with these domains, we discovered SON as a novel AML1-ETO binding protein (PNAS, 2008, 105:17103). SON is a large, poorly characterized serine/arginine rich SR protein localized to nuclear speckles. SON has DNA, single stranded RNA, and double stranded RNA binding domains and long repeats of amino acids. Overexpression of a partial fragment of SON in a transformed cell line decreased the tumorigenic potential of the cell in nude mice and protected yeast from apoptosis. However, partially due to its large size, the SON protein has not been well characterized. Recently, we reported that SON plays an important role in RNA splicing of a specific set of cell cycle related genes: ones that possess weak splice sites (Molecular Cell, 2011, 42:185). While SON is expressed ubiquitously, its expression level is noticeably higher in hematopoietic organs/tissues and blood cells compared to other tissues, suggesting important roles of SON in the hematopoietic system. To examine whether SON expression is regulated during hematopoietic differentiation, we measured relative mRNA level of mouse Son in different stages of hematopoiesis. Son mRNA level is higher in lineage marker negative (Lin-) bone marrow cells when compared to total bone marrow cells. Macrophages showed less expression of Son, suggesting that Son is down-regulated along the hematopoietic differentiation. We further sorted Lin- cells and measured the Son level in LSK (Lin-, Sca1+, cKit+), CMP (commom myeloid progenitors), GMP (granulocyte/monocyte progenitors) and MEP (megakaryocyte/erythroid progenitors) populations. LSK cells, which precede other progenitors, showed the highest level of Son. In addition, we confirmed that SON is down-regulated during TPA-induced monocytic differentiation of U937 myeloid cells. Taken together, SON is more abundantly expressed in immature hematopoietic cells and down-regulated during differentiation. Since SON is differentially expressed during hematopoietic differentiation, we examined whether SON is involved in regulation of hematopoietic transcription factors that are key dictators of hematopoietic differentiation. Among the several transcription factors analyzed, we found that Gata-2 mRNA was consistently reduced by two different Son shRNAs in Lin- bone marrow cells. Down regulation of the GATA-2 mRNA level was further confirmed in human K562 leukemic cell line. More interestingly, while having 20∼40% reduction of mRNA level, the GATA-2 protein level is more remarkably down-regulated upon SON knockdown, resulting in 75∼90% reduction. These results indicate that upon SON knockdown, GATA-2 protein level is mainly regulated at the post-transcriptional steps. Sequence analysis of the 3' UTR of the human GATA-2 gene predicted several candidates of targeting microRNAs. Among them, we confirmed that the mature form of miR-27a and miR-24 are up-regulated upon SON knockdown. Next, we tested the effect of over-expression of miR-27a and miR-24 on GATA-2 expression using a GATA-2 3' UTR-containing luciferase reporter construct and demonstrated that miR-27a indeed inhibits GATA-2 mRNA level. miR-27a is a member of the miR-23a∼27a∼24-2 cluster. RTqPCR showed that primary miRNA of the miR-23a∼27a∼24 cluster is upregulated upon SON knockdown. To test whether the increase of pri-miR of this cluster is due to promoter activation, we used a reporter construct containing the promoter sequence of the miR-23a∼27a∼24-2 cluster fused to the luciferase reporter. The expression of luciferase driven by this promoter is significantly elevated upon SON knockdown, suggesting that that SON functions to repress transcription of the miR-23a-27a-24-2 cluster, thereby relieving GATA-2 mRNA from targeting by miR-27a, and contributes to maintaining the GATA-2 protein level. Taken together, our results reveal a previously unidentified function of SON in microRNA transcription and controlling the GATA-2 protein level in hematopoietic cells. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 20 (6) ◽  
pp. 438-452 ◽  
Author(s):  
Khaled Alsayegh ◽  
Lorena V. Cortés-Medina ◽  
Gerardo Ramos-Mandujano ◽  
Heba Badraiq ◽  
Mo Li

Numerous human disorders of the blood system would directly or indirectly benefit from therapeutic approaches that reconstitute the hematopoietic system. Hematopoietic stem cells (HSCs), either from matched donors or ex vivo manipulated autologous tissues, are the most used cellular source of cell therapy for a wide range of disorders. Due to the scarcity of matched donors and the difficulty of ex vivo expansion of HSCs, there is a growing interest in harnessing the potential of pluripotent stem cells (PSCs) as a de novo source of HSCs. PSCs make an ideal source of cells for regenerative medicine in general and for treating blood disorders in particular because they could expand indefinitely in culture and differentiate to any cell type in the body. However, advancement in deriving functional HSCs from PSCs has been slow. This is partly due to an incomplete understanding of the molecular mechanisms underlying normal hematopoiesis. In this review, we discuss the latest efforts to generate human PSC (hPSC)-derived HSCs capable of long-term engraftment. We review the regulation of the key transcription factors (TFs) in hematopoiesis and hematopoietic differentiation, the Homeobox (HOX) and GATA genes, and the interplay between them and microRNAs. We also propose that precise control of these master regulators during the course of hematopoietic differentiation is key to achieving functional hPSC-derived HSCs.


2018 ◽  
Author(s):  
Manu Setty ◽  
Vaidotas Kiseliovas ◽  
Jacob Levine ◽  
Adam Gayoso ◽  
Linas Mazutis ◽  
...  

AbstractRecent studies using single cell RNA-seq (scRNA-seq) data derived from differentiating systems have raised fundamental questions regarding the discrete vs continuous nature of both differentiation and cell fate. Here we present Palantir, an algorithm that models trajectories of differentiating cells, which treats cell-fate as a probabilistic process, and leverages entropy to measure the changing nature of cell plasticity along the differentiation trajectory. Palantir generates a high resolution pseudotime ordering of cells, and assigns each cell state with its probability to differentiate into each terminal state. We apply Palantir to human bone marrow scRNA-seq data and detect key landmarks of hematopoietic differentiation. Palantir’s resolution enables identification of key transcription factors driving lineage fate choices, as these TFs closely track when cells lose plasticity. We demonstrate that Palantir is generalizable to diverse tissue types and well-suited to resolve less studied differentiating systems.


2021 ◽  
Vol 219 (1) ◽  
Author(s):  
Nouraiz Ahmed ◽  
Martin Etzrodt ◽  
Philip Dettinger ◽  
Tobias Kull ◽  
Dirk Loeffler ◽  
...  

Transcription factors (TFs) regulate cell fates, and their expression must be tightly regulated. Autoregulation is assumed to regulate many TFs’ own expression to control cell fates. Here, we manipulate and quantify the (auto)regulation of PU.1, a TF controlling hematopoietic stem and progenitor cells (HSPCs), and correlate it to their future fates. We generate transgenic mice allowing both inducible activation of PU.1 and noninvasive quantification of endogenous PU.1 protein expression. The quantified HSPC PU.1 dynamics show that PU.1 up-regulation occurs as a consequence of hematopoietic differentiation independently of direct fast autoregulation. In contrast, inflammatory signaling induces fast PU.1 up-regulation, which does not require PU.1 expression or its binding to its own autoregulatory enhancer. However, the increased PU.1 levels induced by inflammatory signaling cannot be sustained via autoregulation after removal of the signaling stimulus. We conclude that PU.1 overexpression induces HSC differentiation before PU.1 up-regulation, only later generating cell types with intrinsically higher PU.1.


2020 ◽  
Vol 117 (38) ◽  
pp. 23626-23635
Author(s):  
Jingmei Hsu ◽  
Hsuan-Ting Huang ◽  
Chung-Tsai Lee ◽  
Avik Choudhuri ◽  
Nicola K. Wilson ◽  
...  

Hematopoietic stem and progenitor cell (HSPC) formation and lineage differentiation involve gene expression programs orchestrated by transcription factors and epigenetic regulators. Genetic disruption of the chromatin remodeler chromodomain-helicase-DNA-binding protein 7 (CHD7) expanded phenotypic HSPCs, erythroid, and myeloid lineages in zebrafish and mouse embryos. CHD7 acts to suppress hematopoietic differentiation. Binding motifs for RUNX and other hematopoietic transcription factors are enriched at sites occupied by CHD7, and decreased RUNX1 occupancy correlated with loss of CHD7 localization. CHD7 physically interacts with RUNX1 and suppresses RUNX1-induced expansion of HSPCs during development through modulation of RUNX1 activity. Consequently, the RUNX1:CHD7 axis provides proper timing and function of HSPCs as they emerge during hematopoietic development or mature in adults, representing a distinct and evolutionarily conserved control mechanism to ensure accurate hematopoietic lineage differentiation.


Sign in / Sign up

Export Citation Format

Share Document