A 200 kb Survey of Chromatin in the ANK-1 Locus Demonstrates an Erythroid-Specific Chromatin Hub That Activates the Erythrocyte Ankyrin (ANK-1E) Promoter.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 536-536
Author(s):  
Ashley N. Owen ◽  
Tyra Wolfsberg ◽  
Karina Laflamme ◽  
Clara Wong ◽  
Yelena Maksimova ◽  
...  

Abstract Mammals express a variety of erythroid and nonerythroid ankyrin-1 isoforms generated by alternate mRNA splicing and by expression from promoters upstream of 3 known alternate first exons. ANK-1 Exon 1B is located 138 kb 5′ of exon 2 and is expressed only in neuronal and muscle cells. Exon 1E is located 39 kb 5′ of exon 2 and is expressed only in erythroid cells. Exon 1A is located 27 kb 5′ of exon 2 and is expressed in many cell types. We have previously shown that the ANK-1E promoter is flanked by DNase I Hypersensitive Sites (HS), one immediately upstream of the RNA initiation sites (5′HS) and a pair of closely spaced HS 5kb downstream (3′HS1 and 3′HS2). To determine the location of additional HS in the ANK-1 locus, we designed PCR primers spaced ~250 bp apart that span a 200 kb region from exon 2 to 60 kb upstream of Exon 1B for use in a high throughput DNase I HS assay. We identified the HS surrounding Exon 1E, as well as HS that flank Exons 1A and 1B. In both the Exon 1A and Exon 1B promoters, the 5′HS is located immediately upstream of the mRNA initiation sites and the 3′HS is located 4–7 kb downstream. An additional pair of HS were identified 70 kb upstream of exon 2 between the ANK-1B and ANK-1E promoters. This region contains the 5′ ends of at least 5 human ESTs. We used 5′ RACE to show that the homologous region in the mouse is transcribed and splices to exon 2. This putative promoter is designated ANK-1C. All 4 ANK-1 promoters lack consensus promoter sequences involved in the binding of the transcription initiation complex, TFIID, including TATA, InR, DPE or DCE elements. We have recently identified a novel consensus sequence that binds TFIID: (T/G)(G/C)(G/C)GGTGAG. This sequence is present multiple times in all 4 ANK-1 promoters as well as in 22% of >4000 mammalian promoters lacking TFIID-binding consensus sequences, strong evidence of functional significance. To understand the relationship of the flanking HS to the ANK-1 promoters we used the activation of ANK-1E promoter in erythroid cells as a model and have undertaken a molecular dissection of the elements in the ANK-1E region. Using transgenic mice and K562-based assays we have shown that both ANK-1E 5′HS and 3′HS2 are barrier elements that prevent gene silencing. In K562 cells, ANK-1E 3′HS1 increases expression only when located adjacent to the ANK-1E or thymidine kinase promoters (p=0.0009), but not in SY5Y neuronal cells (p=0.35). DNase I footprinting, gel shift, and reporter gene assays demonstrated that 3′HS1 binds the erythroid-specific transcription factor NF-E2. Mutation of the NF-E2 binding site abolished the ability of 3′HS1 to increase gene expression (p=0.08) in K562 cells. Chromatin Conformation Capture (3C) analysis demonstrated the formation of a 5 kb erythroid-specific chromatin loop that brings 5′HS into close proximity with 3′HS1/2. In agreement with the 3C results, Chromatin Immune Precipitation analysis demonstrated a hub in which Brg-1 and CTCF (associated with barrier elements), NF-E2, GATA-1 and RNA Pol II occupy both 5′HS and 3′HS1/2, despite the lack of consensus sites for NF-E2 in 5′HS, or GATA-1 or RNA initiation sites in 3′HS1/2. Our current model is that the formation of the erythroid-specific ANK-1E chromatin loop is mediated by the binding of the erythroid-specific transcription factors GATA-1 to 5′HS and NF-E2 to 3′HS1.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 803-803
Author(s):  
Ashley N. Owen ◽  
Robert I. Liem ◽  
Andre M. Pilon ◽  
Patrick G. Gallagher ◽  
David M. Bodine

Abstract Ankyrin forms the bridge between the spectrin/actin network of the erythrocyte membrane skeleton and the red cell membrane by binding to both β-spectrin and band 3. The erythrocyte ankyrin promoter (Ank-1E) is active only in erythroid cells, while two other Ank-1 promoters located 20 kb downstream and 40 kb upstream of Ank-1E are active in the cerebellum and muscle cells respectively. We have been studying the mechanism by which the Ank-1E promoter becomes active in erythroid cells by studying the cis acting regulatory elements and the chromatin structure of the Ank-1 promoter region. We have previously shown that the sequences between −296 and −15 of the Ank-1E promoter are fully sufficient for erythroid specific, copy number dependent uniform expression of reporter genes in transgenic mice. We have also mapped a DNase I Hypersensitive site (5′HS) between −300 and −100 of the human and mouse Ank-1E promoters in human K562 and mouse fetal liver cells. Both the mouse and human 5′HS are capable of preventing the silencing of a β-globin/GFP reporter gene in K562 cells, establishing that they function as barrier elements. Consistent with this observation, the human and mouse 5′HS are hyperacetylated in erythroid cells. The chromatin 10 kb 5′ to the 5′HS is DNase I resistant (associated with inactive chromatin) in human and mouse erythroid and non-erythroid cells. Approximately 6 kb 3′ to 5′HS are two adjacent HS (3′HS1, 3′HS2). Beyond 3′HS2 the chromatin is also DNase I resistant in both human and mouse erythroid and non-erythroid cells. Between 5′HS and 3′HS1 the 6kb region is DNase I sensitive (active) in erythroid cells but not in other cell types. We hypothesized that this 6 kb region contains regulatory elements that activate the Ank-1E promoter. To screen for regulatory elements we isolated overlapping segments of a 10 kb region extending from 2 kb upstream of 5′HS to 2 kb downstream of 3′HS2. We inserted these fragments into a plasmid vector containing the Ank-1E promoter linked to a luciferase reporter gene and transfected these constructs into K562 cells. A single region up regulated Ank-1E/luciferase expression. This region mapped to a 211bp segment that included 3′HS1, but did not include 3′HS2. A fragment containing only 3′HS2 did not up regulate an Ank-1E/luciferase reporter gene, but 3′HS2 was capable of preventing the silencing of a β-globin/Green Fluorescent Protein reporter gene in K562 cells, demonstrating barrier activity. The region around 3′HS1 and 2 was also a site of histone hyperacetylation. The sequence of the 211 bp fragment containing 3′HS1 does not contain consensus sequences for any known erythroid-specific transcription factors, but does contain potential binding sites fro Sp1, AP-1 and E-box binding proteins. Using the Chromatin Conformation Capture assay we demonstrated that 5′HS and 3′HS1 and 2 are in close proximity in K562 chromatin, but are not closely associated in chromatin from other cell types. We propose that an erythroid-specific chromatin loop brings 3′HS1 and 2 into proximity with 5′HS, adjacent to the Ank-1E promoter. This interaction translocates the positive regulatory element in 3′HS1 to the Ank-1E promoter allowing the Ank-1E promoter to become active in erythroid cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2432-2432
Author(s):  
Ashley N. Owen ◽  
Karina Laflamme ◽  
Andre M. Pilon ◽  
Lisa J. Garrett ◽  
Patrick G. Gallagher ◽  
...  

Abstract Fewer than 20,000 protein-coding genes in the human genome generate more than 100,000 proteins. This diversity results from the selective use of alternative promoters and alternative mRNA splicing. Ankyrins are multifunctional linker/adapter proteins with isoforms expressed in cell-, tissue-, and developmental stage-specific patterns. The ANK-1 gene, which encodes a series of proteins that connect the red blood cell (RBC) membrane to the RBC skeleton, is an excellent system to study how specific promoters are selected for expression and others suppressed. The human ANK-1 locus has two tissue-specific promoters/first exons (erythroid, 1E; brain/muscle, 1B) and one ubiquitous promoter/first exon (1A). We have previously shown that the ANK-1E promoter sequences are contained in the 300 base pairs (bp) immediately upstream of exon 1E (including a critical GATA-1 binding site) are necessary for erythroid-specific expression in transgenic mice. We have recently reported a novel 9 base consensus sequence ([G/T][G/C][G/C]GGTGAG) located between +7 and +15 that serves as a binding site for the transcription initiation complex. This consensus is present in the other ANK-1 promoters, 30% of all mammalian promoters, and is highly enriched in those that lack known consensus elements (i.e, TATA box; Laflamme et al. submitted). We hypothesized that variation within this consensus sequence controls the level of mRNA transcription. We evaluated altered consensus sequences in the ANK-1E promoter linked to luciferase or gamma-globin reporter genes in transient transfection assays in erythroid K562 cells or transgenic mice, respectively. In both assays, the GCGGGTGAG sequence generated 7-fold higher levels of expression than the wild type sequence (TGCGGTGAG; p<0.01), while other variations gave similar or lower levels of expression. We concluded that while erythroid specificity of the minimal ANK-1E promoter is conferred by GATA-1 binding, the level of expression is controlled by the ([G/T][G/C][G/C]GGTGAG) box. In transient transfection assays in vitro, where the constraints of chromatin are released, the sequences adjacent to ANK-1E and ANK-1A promoters directed equivalent levels of expression in both erythroid and non-erythroid cells. We hypothesized that the activity of the ANK-1E promoter in vivo is controlled by both the core promoter sequence and the local chromatin architecture. Transcriptionally active regions of chromatin show increased sensitivity to DNase I digestion, which we have analyzed across a 200 kb region encompassing all three ANK-1 promoters. A region between the ANK-1E and ANK-1A promoters was sensitive to DNase I digestion only in erythroid cells, while the upstream (1B) and downstream (1A) regions were DNase I resistant. The 1E to 1A region is flanked by DNase hypersensitive sites (HS): one immediately 5′ to 1E (5′HS), and two adjacent HS (3′HS1, 3′HS2) located ~6 kb downstream. Histone acetylation is also associated with active chromatin. Chromatin Immunoprecipitation (ChIP) of the ANK-1E region showed erythroid-specific histone acetylation of the 6kb region between 5′HS and 3′HS1&2, with hyperacetylation at all three HS in all cell types. Barrier elements are found at the boundary between open and condensed chromatin. 5′HS provides a barrier against transgene silencing in cell lines and transgenic mice (p<0.01). 3′HS2 contains barrier activity in transfected cells (p<0.01), while the combination of 3′HS1 and 3′HS2 prevents silencing in transgenic mice (p<0.02). ChIP, EMSA (Mobility Shift Assay) and in vitro DNase I footprinting demonstrated that 3′HS1 binds the erythroid transcription factor NF-E2. In transient assays in erythroid cells, 3′HS1 increased reporter gene activity 5-fold when adjacent to the ANK-1E promoter. We hypothesized that NF-E2 could be translocated to the ANK-1E promoter by the formation of an internal chromatin loop. Chromatin Conformation Capture (3C) demonstrated the formation of a loop structure in which 5′HS and 3′HS1&2 are brought into physical proximity in erythroid, but not non-erythroid cells. In agreement with the 3C results, ChIP demonstrated that both ends of the ANK-1E chromatin loop bind GATA-1, NF-E2 and RNA Pol II. Our current model predicts that the 5′ HS barrier allows the ANK-1E promoter to function in transgenic mice, but in the native locus, ANK-1E promoter activity requires the formation of a chromatin loop mediated by GATA-1 and NF-E2.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3443-3443
Author(s):  
AI Inoue ◽  
Tohru Fujiwara ◽  
Yoko Okitsu ◽  
Noriko Fukuhara ◽  
Yasushi Onishi ◽  
...  

Abstract Abstract 3443 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. LMO2 (LIM-only protein 2) is a non-DNA binding transcriptional coregulator, and is an important regulator of hematopoietic stem cell development and erythropoiesis, as mice lacking this gene show defects in blood formation as well as fetal erythropoiesis (Warren et al. Cell. 1994). In the context of erythropoiesis, LMO2 has been demonstrated to be a part of multimetric complex, including master regulators of hematopoiesis (GATA-1 and SCL/TAL1), chromatin looping factor LDB1 and hematopoietic corepressor ETO2 (referred as GATA-SCL/TAL1 complex). As LMO2 controls hematopoiesis, its dysregulation is leukemogenic, and its influence on GATA factor function is still not evident, we investigated here the transcriptional regulatory mechanism via LMO2 in erythroid cells. Methods: For LMO2 knockdown, anti-LMO2 siRNA (Thermo Scientific Dharmacon) and pGIPZ lentiviral shRNAmir system (Open Biosystems) were used. Western blotting and Quantitative ChIP analysis were performed using antibodies for GATA-1, LMO2 (abcam), GATA-2, TAL1 and LDB1 (Santa Cruz). 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. To induce erythroid differentiation of K562 cells, hemin was treated at a concentration of 30 uM for 24h. Results: siRNA-mediated LMO2 knockdown in hemin-treated K562 cells results in significantly decreased ratio of benzidine-staining positive cells, suggesting that LMO2 has an important role in the erythroid differentiation of K562 cells. Next, we conducted microarray analysis to characterize LMO2 target gene ensemble in K562 cells. In contrast to the predominantly repressive role of LMO2 in murine G1E-ER-GATA-1 cells (Fujiwara et al. PNAS. 2010), the analyses (n = 2) demonstrated that 177 and 78 genes were upregulated and downregulated (>1.5-fold), respectively, in the LMO2-knockdowned K562 cells. Downregulated gene ensemble contained prototypical erythroid genes such as HBB and SLC4A1 (encodes erythrocyte membrane protein band 3). To test what percentages of LMO2-regulated genes could be direct target genes of GATA-1 in K562 cells, we merged the microarray results with ChIP-seq profile (n= 5,749, Fujiwara et al. Mol Cell. 2009), and demonstrated that 26.4% and 23.1% of upregulated and downregulated genes, respectively, contained significant GATA-1 peaks in their loci. Furthermore, whereas LMO2 knockdown in K562 cells did not affect the expression of GATA-1, GATA-2 and SCL/TAL1 based on quantitative RT-PCR as well as Western blotting, the knockdown resulted in the significantly decreased chromatin occupancy of GATA-1, GATA-2, SCL/TAL1 and LDB1 at beta-globin locus control region and SLC4A1 locus. We subsequently analyzed the consequences of LMO2 knockdown in primary erythroblasts. Endogeneous LMO2 expression was upregulated along with the differentiation of cord blood cell-derived primary erythroblasts. shRNA-mediated knockdown of LMO2 in primary erythroblasts resulted in significant downregulation of HBB, HBA and SLC4A1. Conclusion: Our results suggest that LMO2 contributes to the expression of GATA-1 target genes in a context-dependent manner, through modulating the assembly of the components of GATA-SCL/TAL1 complex at endogeneous loci. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1988 ◽  
Vol 72 (2) ◽  
pp. 606-612 ◽  
Author(s):  
JM Gimble ◽  
EE Max ◽  
TJ Ley

Abstract We performed high-resolution mapping studies of the DNAse I- hypersensitive sites located just 5′ to the human G gamma- and A gamma- globin genes of K562 erythroleukemia cells, in which these genes are constitutively expressed at low levels. This analysis revealed that the hypersensitive site extends from approximately -210 +/- 5 to -25 +/- 5 base pairs (bp) upstream from the transcription initiation site. Within this region, a GC-rich region located between the proximal CCAAT box and the TATA box is particularly accessible to nuclease digestion; however, the 5′ end of the hypersensitive site is less accessible to nucleases. The pattern of DNAse I cleavage does not change on either strand with hemin induction of K562 cells, which increases the rate of gamma-globin gene transcription about threefold. The region within the hypersensitive site includes all the consensus promoter elements of the gamma-globin genes as well as an octamer sequence located between -182 and -175, and a region associated with a variety of mutations that may cause hereditary persistence of fetal hemoglobin (HPFH).


1993 ◽  
Vol 13 (6) ◽  
pp. 3272-3281 ◽  
Author(s):  
S M Jane ◽  
D L Gumucio ◽  
P A Ney ◽  
J M Cunningham ◽  
A W Nienhuis

The human gamma-globin gene promoter contains a stage selector element (SSE) responsible for preferential interaction of the promoter with a powerful erythroid-specific enhancer in the fetal developmental stage (S.M. Jane, P.A. Ney, E.F. Vanin, D.L. Gumucio, and A.W. Nienhuis. EMBO J. 11:2691-2699, 1992). The element binds two proteins, the ubiquitous activator Sp1 and a protein previously known as -50 gamma and now named the stage selector protein (SSP). Binding of the second protein correlates with SSE activity in transient-transfection assays. We now report that a de novo binding site for the SSP is created by the -202(C-->G) mutation that causes hereditary persistence of fetal hemoglobin (HPFH). This site functions in an analogous manner to the SSE in hybrid beta-promoter/reporter gene constructs transfected into K562 cells. In contrast, the wild-type -202 sequence, which fails to bind the SSP, is incapable of activating the beta-gene promoter. Both the -50 and -202 HPFH sites for SSP binding overlap a consensus sequence for the transcriptional regulator Sp1. In addition, both sites contain CpG dinucleotides that are contact bases for SSP. Since the gamma promoter is known to be hypomethylated in fetal cells but fully methylated at CpG residues in adult erythroid cells, we examined the effects of this DNA modification on protein binding to the two regions. Gel mobility shift assays with nuclear extract from K562 cells (which contain both Sp1 and SSP) demonstrate preferential binding of SSP to the SSE and HPFH sites under conditions in which probe was limiting. Methylation of the CpG residues reverses this preference only in the SSE site, with a marked increase in the binding of Sp1 at the expense of the SSP. Purified Sp1 binds with 10-fold higher affinity to the methylated than to the nonmethylated -50 probe but with the same affinity to the -202 HPFH probe. The methylation-induced preferential binding of Sp1 to the SSE at the expense of SSP may be part of the mechanism by which the gamma genes are repressed in normal adult erythroid cells. In cells containing the -202 HPFH mutation, the inability of Sp1 to displace SSP in the methylated state may explain the persistence of gamma-promoter activity and gamma-gene expression observed in adults with this mutation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 215-215 ◽  
Author(s):  
Tohru Fujiwara ◽  
Ryoyu Niikuni ◽  
Koji Okamoto ◽  
Yoko Okitsu ◽  
Noriko Fukuhara ◽  
...  

Abstract (Backgroun d) Congenital sideroblastic anemia (CSA) is an inherited disease; it is a microcytic type of anemia characterized by bone marrow sideroblasts with excess iron deposition in the mitochondria. The most common form of CSA is XLSA (X-linked sideroblastic anemia), which is attributed to mutations in the X-linked gene ALAS2 (erythroid-specific 5-aminolevulinate synthase). ALAS2 encodes the first and rate-limiting enzyme involved in heme biosynthesis in erythroid cells, which utilizes glycine and acetyl-coenzyme A to form 5-aminolevulinic acid (ALA) and also requires pyridoxal 5'-phosphate (PLP, vitamin B6) as a cofactor. Based on the evidence that half of the XLSA cases were unresponsive to PLP (Ohba et al. Ann Hematol 2013), ALA supplementation could emerge as an alternative therapeutic strategy to restore heme synthesis in CSA caused by ALAS2 defects. As a preclinical study, we focused our study on the effect of ALA on human erythroid cells. Furthermore, we investigated the molecular mechanism by which ALA is transported into erythroid cells. (Method ) Human K562 erythroid cells as well as human induced pluripotent stem-derived erythroid progenitor (HiDEP) cells (Kurita et al. PLoS ONE 2013) were used for the analysis. We investigated the effects of ALA (0.01, 0.1, and 0.5 mM for 72 h) on heme content, hemoglobinization, and erythroid-related gene expression. Heme content was determined fluorometrically at 400 nm (excitation) and 662 nm (emission). Small interfering RNA (siRNA)-mediated knockdown of ALAS2 was conducted using Amaxa Nucleofector™ (Amaxa Biosystems, Koln, Germany). For transcription profiling, Human Oligo chip 25K (Toray, Tokyo, Japan) was used for control and ALAS2 siRNA-treated HiDEP cells. Gamma-aminobutyric acid (GABA) (Sigma, St. Louis, MO, USA) was used at concentrations of 10 and 20 mM. (Results) First, we demonstrated that ALA treatment resulted in significant dose-dependent accumulation of heme in K562 cells. Concomitantly, the treatment substantially induces erythroid differentiation as assessed using hemoglobin (benzidine) staining. Quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis confirmed significant upregulation of heme-regulated genes such as the globin genes (HBA and HBG) and the heme oxygenase 1 (HMOX1) gene in K562 cells. To investigate the mechanism by which ALA was transported into erythroid cells, we conducted quantitative RT-PCR analysis for previously reported ALA transporters, including SLC15A1, SLC15A2, SLC36A1, and SLC6A13 (Frølund et al. Br J Pharmacol 2010; Ahlin et al. Drug Metab Dispos 2009; Moretti et al. Br J Cancer 2002). The analysis revealed that SLC36A1 was abundantly expressed in K562 and HiDEP cells. Thus, GABA was added to K562 cells to competitively inhibit SLC36A1-mediated transport (Frølund et al. Br J Pharmacol 2010). GABA treatment significantly impeded the ALA-mediated increase in the number of hemoglobinized cells. Next, siRNA-mediated knockdown of ALAS2 in HiDEP cells resulted in a significant decrease in the expression of globin genes as well as HMOX1; however, ringed sideroblasts were not observed. Microarray analysis revealed >2-fold up- and down-regulation of 38 and 68 genes caused by ALAS2 knockdown, respectively. The downregulated gene ensemble included globins (HBZ, HBG, HBE, HBD, and HBM) as well as genes involved in iron metabolism (ferritin heavy chain 1: FTH1, transferrin receptor: TFRC and glutaredoxin-1: GLRX5). Gene ontology analysis revealed significant enrichment of cellular iron ion homeostasis (p = 0.000076), cell division (p = 0.00062), DNA repair (p = 0.0006) and translation (p = 0.018), implying that heme was involved in various biological processes in erythroid cells. Interestingly, ALA treatment significantly improved the consequences of ALAS2 knockdown-mediated downregulation of HBA, HBG, and HMOX1. (Conclusion) ALA appears to enter into erythroid cells mainly by SLC36A1 and utilized to generate heme precursor. Thus,ALA may represent a novel therapeutic option for CSA, particularly for cases harboring ALAS2 mutations. Disclosures Fujiwara: Chugai Pharmaceutical, CO., LTD.: Research Funding.


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.


Blood ◽  
1988 ◽  
Vol 72 (2) ◽  
pp. 606-612
Author(s):  
JM Gimble ◽  
EE Max ◽  
TJ Ley

We performed high-resolution mapping studies of the DNAse I- hypersensitive sites located just 5′ to the human G gamma- and A gamma- globin genes of K562 erythroleukemia cells, in which these genes are constitutively expressed at low levels. This analysis revealed that the hypersensitive site extends from approximately -210 +/- 5 to -25 +/- 5 base pairs (bp) upstream from the transcription initiation site. Within this region, a GC-rich region located between the proximal CCAAT box and the TATA box is particularly accessible to nuclease digestion; however, the 5′ end of the hypersensitive site is less accessible to nucleases. The pattern of DNAse I cleavage does not change on either strand with hemin induction of K562 cells, which increases the rate of gamma-globin gene transcription about threefold. The region within the hypersensitive site includes all the consensus promoter elements of the gamma-globin genes as well as an octamer sequence located between -182 and -175, and a region associated with a variety of mutations that may cause hereditary persistence of fetal hemoglobin (HPFH).


1993 ◽  
Vol 13 (6) ◽  
pp. 3272-3281
Author(s):  
S M Jane ◽  
D L Gumucio ◽  
P A Ney ◽  
J M Cunningham ◽  
A W Nienhuis

The human gamma-globin gene promoter contains a stage selector element (SSE) responsible for preferential interaction of the promoter with a powerful erythroid-specific enhancer in the fetal developmental stage (S.M. Jane, P.A. Ney, E.F. Vanin, D.L. Gumucio, and A.W. Nienhuis. EMBO J. 11:2691-2699, 1992). The element binds two proteins, the ubiquitous activator Sp1 and a protein previously known as -50 gamma and now named the stage selector protein (SSP). Binding of the second protein correlates with SSE activity in transient-transfection assays. We now report that a de novo binding site for the SSP is created by the -202(C-->G) mutation that causes hereditary persistence of fetal hemoglobin (HPFH). This site functions in an analogous manner to the SSE in hybrid beta-promoter/reporter gene constructs transfected into K562 cells. In contrast, the wild-type -202 sequence, which fails to bind the SSP, is incapable of activating the beta-gene promoter. Both the -50 and -202 HPFH sites for SSP binding overlap a consensus sequence for the transcriptional regulator Sp1. In addition, both sites contain CpG dinucleotides that are contact bases for SSP. Since the gamma promoter is known to be hypomethylated in fetal cells but fully methylated at CpG residues in adult erythroid cells, we examined the effects of this DNA modification on protein binding to the two regions. Gel mobility shift assays with nuclear extract from K562 cells (which contain both Sp1 and SSP) demonstrate preferential binding of SSP to the SSE and HPFH sites under conditions in which probe was limiting. Methylation of the CpG residues reverses this preference only in the SSE site, with a marked increase in the binding of Sp1 at the expense of the SSP. Purified Sp1 binds with 10-fold higher affinity to the methylated than to the nonmethylated -50 probe but with the same affinity to the -202 HPFH probe. The methylation-induced preferential binding of Sp1 to the SSE at the expense of SSP may be part of the mechanism by which the gamma genes are repressed in normal adult erythroid cells. In cells containing the -202 HPFH mutation, the inability of Sp1 to displace SSP in the methylated state may explain the persistence of gamma-promoter activity and gamma-gene expression observed in adults with this mutation.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2039-2039
Author(s):  
Caryn S Gonsalves ◽  
Clinton H. Joiner

Abstract Abstract 2039 The K-Cl co-transporters are key components of volume regulation in human reticulocytes. K-Cl transport activity is increased in sickle red blood cells (SS RBC) and is thought to contribute to SS RBC dehydration, a process that potentiates sickling. Four mammalian KCC isoforms have been cloned and characterized, of which three are expressed in erythroid cells (Crable et al. Exp Hematol. 2005; 33:624). Studies in sickle and normal reticulocytes indicate substantial variation in the relative expression of these genes among individuals, suggesting a potential source of phenotypic variability in sickle cell disease (SCD). Relatively little is known about the regulation of KCC expression. Previous studies have shown an increase in mRNA levels of KCC3a in HUVEC cells treated with the angiogenic factor vascular endothelial growth factor (VEGF) (Hiki, et.al. (1999) J. Biol. Chem. 274, 10661–10667). Because VEGF and the related placenta growth factor (PlGF) levels are elevated in SCD, we evaluated the potential effects of the vascular growth factors on KCC expression in erythroid cells. RT-PCR revealed that erythroid K562 cells expressed the VEGF receptor-1 (VEGF-R1, or Flt-1) but not VEGF receptor-2, (VEGF-R2 or Flk-1). Treatment of K562 cells with VEGF (50 ng/ml) showed an increase in KCC4 mRNA levels at 8 hours, with no significant change in KCC1, KCC3a and KCC3b expression. An inhibitor specific for the VEGF receptor-1 (VEGF-R1), SU5416, ablated the effect of VEGF, suggesting a role for this receptor in the signaling pathway. Studies with pharmacological inhibitors for specific kinases indicated that VEGF-stimulated KCC4 expression involves PI3 kinase, p38 MAP kinase, mTOR, JNK kinase and the transcription factor hypoxia inducible factor-1α (HIF-1α), which have been implicated in other VEGF effects. Analysis of the KCC4 promoter revealed binding sites for transcription factor SP-1 at positions -35 to -44 and -56 to -64, relative to the transcriptional start site. In addition, seven putative binding sites for transcription factor HIF-1α were found in the KCC4 promoter, suggesting a role for HIF-1α in VEGF-stimulated KCC4 expression, which occurs under non-hypoxic conditions. We examined KCC4 promoter activity using KCC4 luciferase promoter constructs expressed in K562 cells. Luciferase activity was stimulated by VEGF treatment, with maximum activity from the promoter constructs spanning 1200 bp and 875 bp from the start site. Minimal promoter activity was seen in the -65bp construct. Additionally, a mutation in the HIF-1α binding site at -73 to -76bp significantly inhibited promoter activity. Site-directed mutagenesis of the SP-1 sites at position -35 to -44 and position -56 to -64 also attenuated promoter activities upon VEGF treatment. These results suggest that activation of VEGF-R1 by VEGF, and presumably its other ligand, PlGF, leads to non-hypoxic activation of HIF-1α and SP-1-mediated up-regulation of KCC4 expression in erythroid K562 cells via its canonical signaling pathways. Variation in KCC gene expression and its modulation by cytokines and growth factors may be a source of inter-individual variation in SS RBC volume regulation and thus of phenotypic variability of SCD. Identifying the factors that modulate transcriptional control of KCC4 expression is important to understanding volume regulation in reticulocytes and its dysregulation in SS RBC. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document