Developmental stage–specific epigenetic control of human β-globin gene expression is potentiated in hematopoietic progenitor cells prior to their transcriptional activation

Blood ◽  
2003 ◽  
Vol 102 (12) ◽  
pp. 3989-3997 ◽  
Author(s):  
Stefania Bottardi ◽  
Angélique Aumont ◽  
Frank Grosveld ◽  
Eric Milot

Abstract To study epigenetic regulation of the human β-globin locus during hematopoiesis, we investigated patterns of histone modification and chromatin accessibility along this locus in hematopoietic progenitor cells (HPCs) derived from both humans and transgenic mice. We demonstrate that the developmentally related activation of human β-like globin genes in humans and transgenic mice HPCs is preceded by a wave of gene-specific histone H3 hyperacetylation and K4 dimethylation. In erythroid cells, expression of β-like globin genes is associated with histone hyperacetylation along these genes and, surprisingly, with local deacetylation at active promoters. We also show that endogenous mouse β major and human β-like genes are subject to different epigenetic control mechanisms in HPCs. This difference is likely due to intrinsic properties of the human β-globin locus since, in transgenic mice, this locus is epigenetically regulated in the same manner as in human HPCs. Our results suggest that a defined pattern of histone H3 acetylation/dimethylation is important for specific activation of human globin promoters during development in human and transgenic HPCs. We propose that this transient acetylation/dimethylation is involved in gene-specific potentiation in HPCs (ie, before extensive chromatin remodeling and transcription take place in erythroid cells).

1998 ◽  
Vol 18 (11) ◽  
pp. 6634-6640 ◽  
Author(s):  
Denise E. Sabatino ◽  
Amanda P. Cline ◽  
Patrick G. Gallagher ◽  
Lisa J. Garrett ◽  
George Stamatoyannopoulos ◽  
...  

ABSTRACT During development, changes occur in both the sites of erythropoiesis and the globin genes expressed at each developmental stage. Previous work has shown that high-level expression of human β-like globin genes in transgenic mice requires the presence of the locus control region (LCR). Models of hemoglobin switching propose that the LCR and/or stage-specific elements interact with globin gene sequences to activate specific genes in erythroid cells. To test these models, we generated transgenic mice which contain the human Aγ-globin gene linked to a 576-bp fragment containing the human β-spectrin promoter. In these mice, the β-spectrin Aγ-globin (βsp/Aγ) transgene was expressed at high levels in erythroid cells throughout development. Transgenic mice containing a 40-kb cosmid construct with the micro-LCR, βsp/Aγ-, ψβ-, δ-, and β-globin genes showed no developmental switching and expressed both human γ- and β-globin mRNAs in erythroid cells throughout development. Mice containing control cosmids with the Aγ-globin gene promoter showed developmental switching and expressed Aγ-globin mRNA in yolk sac and fetal liver erythroid cells and β-globin mRNA in fetal liver and adult erythroid cells. Our results suggest that replacement of the γ-globin promoter with the β-spectrin promoter allows the expression of the β-globin gene. We conclude that the γ-globin promoter is necessary and sufficient to suppress the expression of the β-globin gene in yolk sac erythroid cells.


1987 ◽  
Vol 7 (11) ◽  
pp. 4024-4029
Author(s):  
M Trudel ◽  
J Magram ◽  
L Bruckner ◽  
F Costantini

The human G gamma-globin and beta-globin genes are expressed in erythroid cells at different stages of human development, and previous studies have shown that the two cloned genes are also expressed in a differential stage-specific manner in transgenic mice. The G gamma-globin gene is expressed only in murine embryonic erythroid cells, while the beta-globin gene is active only at the fetal and adult stages. In this study, we analyzed transgenic mice carrying a series of hybrid genes in which different upstream, intragenic, or downstream sequences were contributed by the beta-globin or G gamma-globin gene. We found that hybrid 5'G gamma/3'beta globin genes containing G gamma-globin sequences upstream from the initiation codon were expressed in embryonic erythroid cells at levels similar to those of an intact G gamma-globin transgene. In contrast, beta-globin upstream sequences were insufficient for expression of 5'beta/3'G gamma hybrid globin genes or a beta-globin-metallothionein fusion gene in adult erythroid cells. However, beta-globin downstream sequences, including 212 base pairs of exon III and 1,900 base pairs of 3'-flanking DNA, were able to activate a 5'G gamma/3'beta hybrid globin gene in fetal and adult erythroid cells. These experiments suggest that positive regulatory elements upstream from the G gamma-globin and downstream from the beta-globin gene are involved in the differential expression of the two genes during development.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 129-129 ◽  
Author(s):  
Virryan Banzon ◽  
Vinzon Ibanez ◽  
Kestis Vaitkus ◽  
Tatiana Kousnetzova ◽  
Joseph Desimone ◽  
...  

Abstract The development of new therapies to increase fetal hemoglobin (HbF) levels in patients with sickle cell disease and β-thalassemia depends on an increased understanding of the mechanism responsible for the developmental regulation of globin gene expression. A role for epigenetic modifications in the mechanism of of globin gene regulation is suggested by the presence of high levels of DNA methylation near the 5’ regions of developmentally silenced ε- and γ-globin genes and the ability of pharmacological inhibitors of DNA methyltransferase (DNMTase) to reactivate ε- and γ-globin expression in adults. Whether additional epigenetic modifications associated with gene silencing and DNA methylation, such as histone H3 (lys9) dimethylation, are also involved is unknown. To investigate the hypothesis that histone H3 (lys9) dimethylation may function in the mechanism of developmental globin gene silencing, chromatin immunopreciptation assays were performed to determine the distribution of histone H3 (lys9) dimethyl and histone H3 (lys9) acetyl throughout the β-globin gene complex in purified primary baboon bone marrow (BM) erythroid cells from phlebotomized baboons expressing low levels (5–10%) of HbF and purified erythroid cells from erythroid progenitor cell cultures expressing high levels of HbF (30–50%). In BM erythroid cells, the level of histone H3 (lys9) acetyl associated with the β-globin gene was 10–20 fold higher than with the ε- and γ-globin genes, while the level of histone H3 (lys9) dimethyl associated with the ε- and γ-globin genes was 2–4 fold higher than with the β-globin gene. In erythroid cells from day 12 erythroid progenitor cell cultures, the level of histone H3 (lys9) acetyl associated with the highly expressed γ- and β-globin genes was 10–20 fold higher than with the silent ε-globin gene, while the level of histone H3 (lys9) dimethyl associated with the ε-globin gene was 2–4 fold higher than with the γ- and β-globin genes. Therefore a reciprocal relationship was observed between levels of histone H3 (lys9) acetylation and dimethylation associated with active and inactive globin genes. Experiments were performed to further investigate the role of histone H3 (lys9) dimethyl in ε-globin gene silencing by determining the effect of the G9A histone methyltransferase inhibitor BIX-01294 on ε-globin expression. Erythroid progenitor cell cultures derived from CD34+ BM cells of three individual baboons were treated with the varying doses of the DNMTase inhibitor decitabine (0.125–1.0μM), and BIX-01294 (1.25–5μM), alone and in combination. Changes in ε- globin were assessed by real time PCR using the ΔΔCT method with α-globin as the standard. Decitabine (0.5μM) increased ε-globin 25.8±7.7 fold while BIX-01294 (2.5μM) increased ε-globin 3.09±1.16 fold. Decitabine (1μM) and BIX-01294 (2.5μM) in combination increased ε-globin 55.7±24.9 fold. BIX-01294 enhanced ε-globin expression approximately twofold at all decitabine doses ranging from 0.125–1.0μM (mean increase=103± 44.7%). BIX-01294 also blocked terminal erythroid differentiation and allowed expansion of more primitive cells as evidenced by the presence of a large population of basophilic erythroblasts at late stages of culture (day 14). These results demonstrate that BIX-01294 reactivates expression of the silenced ε-globin gene and that synergistic reactivation can be achieved using combinations of BIX-01294 and decitabine. While these results are consistent with the hypothesis that epigenetic modifications are important in the mechanism of developmental globin gene silencing, the observation that BIX-01294 blocks erythroid differentiation suggests the possible involvement of a reprogramming mechanism.


2020 ◽  
Author(s):  
Xianjiang Lan ◽  
Ren Ren ◽  
Ruopeng Feng ◽  
Lana C. Ly ◽  
Yemin Lan ◽  
...  

SummaryMetazoan transcription factors typically regulate large numbers of genes. Here we identify via a CRISPR-Cas9 genetic screen ZNF410, a pentadactyl DNA binding protein that in human erythroid cells directly and measurably activates only one gene, the NuRD component CHD4. Specificity is conveyed by two highly evolutionarily conserved clusters of ZNF410 binding sites near the CHD4 gene with no counterparts elsewhere in the genome. Loss of ZNF410 in adult-type human erythroid cell culture systems and xenotransplant settings diminishes CHD4 levels and derepresses the fetal hemoglobin genes. While previously known to be silenced by CHD4, the fetal globin genes are exposed here as among the most sensitive to reduced CHD4 levels. In vitro DNA binding assays and crystallographic studies reveal the ZNF410-DNA binding mode. ZNF410 is a remarkably selective transcriptional activator in erythroid cells whose perturbation might offer new therapeutic opportunities in the treatment of hemoglobinopathies.HighlightsA CRISPR screen implicates ZNF410 in fetal globin gene repressionThe CHD4 gene is the singular direct ZNF410 target in erythroid cellsThe fetal globin genes are exquisitely sensitive to CHD4 levelsFive C2H2 zinc fingers of ZNF410 recognize the major groove of a 14 base pair sequence


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1768-1768
Author(s):  
Donald Lavelle ◽  
Janet Chin ◽  
Mahipal Singh ◽  
Kestis Vaitkus ◽  
Virryan Banzon ◽  
...  

Abstract Elevated levels of fetal hemoglobin (HbF) reduce the symptoms of sickle cell disease and increase the life span of patients. Future pharmacologic therapies to increase HbF will depend on increased knowledge of the mechanism(s) regulating γ-globin gene expression. To investigate the relationship between DNA methylation, chromatin structure, and γ-globin gene regulation, DNA methylation of 5 CpG sites within γ-globin gene promoter, levels of acetyl-histone H3 and H4 and histone H3 lys4 trimethyl throughout the β-globin gene complex, and the pattern of γ-globin polypeptide chain synthesis were analyzed in primary baboon fetal liver (FL) erythroid cells, primary baboon bone marrow (ABM) erythroid cells from phlebotomized adults expressing low levels of HbF, ABM erythroid cells from adults expressing high levels of HbF following treatment in vivo with the DNA demethylating drug decitabine, and erythroid cells expressing high levels of HbF generated from CD34+ baboon BM erythroid progenitors in a liquid culture system. High levels of histone acetylation were associated with the ε- and γ-globin genes and low levels with the β-globin gene in FL erythroid cells while in ABM erythroid cells expressing low levels of HbF (6.69±1.93%) high levels were associated with the β-globin gene and low levels with the γ-globin gene. Histone H3 lys 4 trimethyl was enriched near the γ-globin gene in FL and the β-globin gene in ABM. The γ-globin gene was not methylated in FL. The level of methylation was similar in bled (75.1±8.26%) and normal (82.1±7.51%) ABM erythroid cells. The ratio of expression of 5′ Iγ- and 3′ Vγ-globin genes in the fetus (1.85) differed from bled adults (0.65). Expression of γ-globin was reactivated to similar levels in decitabine-treated baboons (51.5±4.50%) and in erythroid progenitor cell cultures (45.3±12.1%; d14). The Iγ- and Vγ-globin chains were expressed at the characteristic fetal ratio in erythroid progenitor cell cultures (1.76±0.21), but not in decitabine-treated baboons (0.76±0.32). Distribution of histone acetylation and histone H3 lys4 trimethyl was nearly identical in erythroid cells expressing high levels of HbF from decitabine-treated animals and from erythroid progenitor cultures and was characterized by high levels of histone H3 lys4 trimethyl associated with both the active γ- and β-globin genes and enrichment of histone H3 and H4 acetylation near the ε-, γ-, and β-globin genes. HbF reactivation in decitabine-treated baboons was associated with a reduction in the level of γ-globin gene methylation (33.5±9.43% dmC). Surprisingly, the level of γ-globin gene methylation in erythroid cells purified from erythroid progenitor cell cultures (79.5±9.80% dmC; d11) synthesizing high levels of γ-globin was not significantly different than in ABM erythroid cells expressing <2% HbF. These results demonstrate that reactivation of γ-globin expression in erythroid progenitor cell cultures is uniquely characterized by the Iγ/Vγ-globin chain ratio of fetal development. In contrast to the absence of γ-globin gene methylation in FL and reduced levels associated with reactivation of HbF following decitabine treatment, the level of γ-globin gene methylation in erythroid progenitor cell cultures was the same as in ABM. Our data strongly suggests that reactivation of γ-globin gene expression in erythroid progenitor cell cultures is achieved through a novel mechanism not dependent on loss of DNA methylation.


1997 ◽  
Vol 185 (4) ◽  
pp. 755-766 ◽  
Author(s):  
Malte Peters ◽  
Peter Schirmacher ◽  
Jutta Goldschmitt ◽  
Margarete Odenthal ◽  
Christian Peschel ◽  
...  

Soluble cytokine receptors modulate the activity of their cognate ligands. Interleukin (IL)-6 in association with the soluble IL-6 receptor (sIL-6R) can activate cells expressing the gp130 signal transducer lacking the specific IL-6R. To investigate the function of the IL-6–sIL-6R complex in vivo and to discriminate the function of the IL-6–sIL-6R complex from the function of IL-6 alone, we have established a transgenic mouse model. Double-transgenic mice coexpressing IL-6 and sIL-6R were generated and compared with IL-6 and sIL-6R single-transgenic mice. The main phenotype found in IL-6–sIL-6R mice was a dramatic increase of extramedullary hematopoietic progenitor cells in liver and spleen but not in the bone marrow. In IL-6 single-transgenic mice and sIL-6R single-transgenic mice no such effects were observed. The high numbers of hematopoietic progenitor cells were reflected by a strong increase of peripheral blood cell numbers. Therefore, activators of the gp130 signal transducer like the IL-6–IL-6R complex may represent most powerful stimulators for extramedullary hematopoietic progenitor cells. gp130 activators may become important for the expansion of hematopoietic progenitor cells in vivo and in vitro.


Nature ◽  
1985 ◽  
Vol 314 (6009) ◽  
pp. 377-380 ◽  
Author(s):  
Kiran Chada ◽  
Jeanne Magram ◽  
Kathryn Raphael ◽  
Glenn Radice ◽  
Elizabeth Lacy ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1307-1307
Author(s):  
Alqadi Yarob Wael ◽  
Tohru Fujiwara ◽  
Yoko Okitsu ◽  
Yasushi Onishi ◽  
Kenichi Ishizawa ◽  
...  

Abstract Abstract 1307 Background: Developmental control mechanisms often utilize multimeric complexes containing transcription factors, coregulators, and additional non-DNA binding components. It is challenging to ascertain how such components contribute to complex function at endogenous loci. We recently analyzed the function of components of a complex containing master regulators of hematopoiesis (GATA-1 and Scl/TAL1) and the non-DNA binding components ETO2, the LIM domain protein LMO2, and the chromatin looping factor LDB1. We revealed that ETO2 and LMO2 regulate distinct target gene ensembles in erythroid cells. Furthermore, it was found that ETO2 commonly represses GATA-1 function via suppressing histone H3 acetylation, and also regulates methylation of histone H3 at lysine 27 (H3-trimeK27) at select loci, which suggested that ETO2 might be an important determinant of the erythroblast epigenome (Fujiwara et al. PNAS. 2010). Here, we investigated the role of ETO2 in the epigenetic regulation of erythroid genes. Methods: CBFA2T3 mRNA (which encodes ETO2 protein) was cloned into pcDNA3.1 (Clontech) and Flexi HaloTag vector (Promega), and ETO2 was transiently overexpressed in K562 cells using Amaxa nucleofection technology™ (Amaxa Inc.). For ETO2 knockdown, pGIPZ lentiviral shRNAmir (Open Biosystems) was used. Quantitative ChIP analysis was performed using antibodies for acetylated H3K9 (abcam), trimethyl H3K27 (Millipore), ETO2, c-Myc (Santa Cruz), and HaloCHIP™ system (Promega). To obtain human primary erythroblasts, CD34-positive cells isolated from cord blood were induced in liquid suspension culture. For transcription profiling, human whole expression array was used (Agilent), and the data was analyzed with GeneSpring GX software. Results: First, we conducted microarray analysis to characterize ETO2 target gene ensemble using erythroid cell line (K562 cells). The analysis demonstrated that 598 genes were downregulated in the ETO2-overexpressed cells (> 2 fold). To test what percentages of ETO2-repressed genes could be direct target genes of GATA-1 or GATA-2 in K562 cells, we merged the microarray results with ChIP-seq profile (n= 5,749 and n=21,167 for GATA-1 and GATA-2 ChIP-seq, respectively) (Fujiwara et al. Mol Cell. 2009), and demonstrated that 23.1% and 40.5% of ETO2-repressed genes contained significant GATA-1 and GATA-2 peaks in their loci, respectively. Gene Ontology analysis among ETO2-repressed genes revealed significant enrichment of genes related to “oxygen transporter” and “hemoglobin complex” (p=0.00128), corresponding to HBG, HBB, HBE, HBA, HBQ, HBM and HBZ. We also confirmed that shRNA-mediated knockdown of ETO2 de-repressed globin genes in K562 cells. Quantitative ChIP analysis confirmed endogeneous and exogeneous ETO2 protein occupancy at beta-globin locus control region (LCR) and alpha-globin HS-40 in K562 cells. Furthermore, the overexpression significantly increased H3-trimeK27 and reduced acetylated H3K9 at these loci. Co-immunoprecipitation analysis revealed the interaction of ETO2 with EZH2/SUZ12, known as components of histone H3K27 methyltransferase complex, polycomb repressor complex 2 (PRC2), implying that the complex might be involved in ETO2-mediated transcriptional repression. To test if ETO2-mediated repression of globin genes is also observed in primary erythroblasts, we conducted shRNA-mediated knockdown of ETO2 in cord blood cell-derived primary erythroblasts, and demonstrated that ETO2 knockdown significantly de-repressed HBB and HBA expression. We are currently analyzing the mechanism of ETO2-dependent transcriptional repression and how ETO2-dependent histone marks are established in erythroid cells. Conclusion: In conjunction with the evidence that ETO2 binds histone deacetylases and associates with GATA-Scl/TAL1 complex that binds epigenetic modifiers, our results suggest that ETO2 appears to have important roles in establishing the erythroblast epigenome. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document