Multiple Defects in Erythroid Gene Expression in Erythroid Krupple-Like Factor (EKLF) Target Genes in EKLF-Deficient Mice.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1602-1602
Author(s):  
Patrick G. Gallagher ◽  
Andre M. Pilon ◽  
Murat O. Arcasoy ◽  
David M. Bodine

Abstract EKLF is the founding member of the KLF family of transcription factors. EKLF-deficient (−/−) mice die at d14-15 of gestation of severe anemia, attributed to decreased expression of the β-globin gene. Morphology of fetal-liver derived circulating erythroid cells in EKLF −/− mice does not mimic that seen in thalassemia, and mice expressing high levels of γ-globin bred onto an EKLF −/− background do not correct the anemia or rescue the −/− mice. These data suggest additional red cell defects are present in EKLF −/− mice contributing to the anemia. To address this hypothesis, we performed subtractive hybridization and microarray analyses with RNA isolated from d13.5 fetal livers of wild type (WT,+/+) and EKLF-deficient mice. In subtractive hybridization, WT (1X) was the tester population and EKLF −/− (30X) was the driver. >200 differentially expressed clones were sequenced. 122 clones were β-globin; 21 alpha hemoglobin stabilizing protein (AHSP); and the remainder were RBC membrane proteins including band 3 and β-spectrin. Microarray analyses were performed with Affymetrix GeneChip Mouse Genome 430 2.0 arrays; 3 independent EKLF +/+ and −/− RNA samples were analyzed. AHSP had the most significantly decreased expression in −/− samples, reduced to 5% of WT (p<0.0001). Other significantly down regulated genes, in addition to those identified by subtractive hybridization, included pyruvate kinase and ankyrin. Reduced expression in EKLF −/− RNA was confirmed using ribonuclease protection assay and/or real-time RT-PCR. AHSP mRNA was decreased by 75–90%; band 3 and β-spectrin were decreased by ~40%; ankyrin by 45%, and pyruvate kinase 15%. Flow cytometry of +/+ and −/ − fetal liver cells using TER119 revealed a TER119Hi population of cells absent in −/ − fetal liver cells, suggesting a block in differentiation to more mature erythroid progenitors. To ensure that the potential EKLF target genes were expressed in the TER119Lo population and were not identified because they were present only in the TER119Hi, the expression of each selected gene was analyzed in TER119Lo and TER119Hi cells by real time PCR. Target gene/α-globin ratios indicated the selected genes were expressed at levels >2 fold higher in TER119Lo than TER119Hi cells. Studies of the AHSP locus identified a strong DNaseI hypersensitive site (HS) in WT fetal liver nuclei between 5′ of the AHSP promoter, that was absent in −/ − chromatin. Chromatin immunoprecipitation analysis of WT fetal liver chromatin spanning 3.5kb of the AHSP locus using 13 primer pairs (~300bp intervals from >1kb 5′ and 3′) demonstrated 2 regions of hyperacetylation of histones H3 and H4. The 5′ region corresponded to the DNaseI HS, and the other region maps 3′ of the AHSP polyA signal. Histones H3 and H4 were also acetylated in the interval between these hyperacetylated regions, while the chromatin upstream and downstream of these regions were hypoacetylated. In chromatin from −/ − fetal liver cells, all sites were hypoacetylated, correlating with the severe reduction in AHSP gene expression. These results support the hypothesis that the anemia in EKLF −/ − mice is due to both decreased expression of the β-globin gene and other erythroid genes including those involved in membrane integrity, stabilization of α-globin protein, and glycolysis leading to defects in erythrocyte structure, function, and metabolism. They also suggest that EKLF may act as a transcription factor and a chromatin modulator for genes other than β-globin.

Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3459-3466 ◽  
Author(s):  
Florence Martin ◽  
Jan M. van Deursen ◽  
Ramesh A. Shivdasani ◽  
Carl W. Jackson ◽  
Amber G. Troutman ◽  
...  

NF-E2 binding sites, located in distant regulatory sequences, may be important for high level α- and β-globin gene expression. Surprisingly, targeted disruption of each subunit of NF-E2 has either little or no effect on erythroid maturation in mice. For p18 NF-E2, this lack of effect is due, at least in part, to the presence of redundant proteins. For p45 NF-E2, one possibility is that NF-E2–related factors, Nrf-1 or Nrf-2, activate globin gene expression in the absence of NF-E2. To test this hypothesis for Nrf-2, we disrupted the Nrf-2 gene by homologous recombination. Nrf-2–deficient mice had no detectable hematopoietic defect. In addition, no evidence was found for reciprocal upregulation of NF-E2 or Nrf-2 protein in fetal liver cells deficient for either factor. Fetal liver cells deficient for both NF-E2 and Nrf-2 expressed normal levels of α- and β-globin. Mature mice with combined deficiency of NF-E2 and Nrf-2 did not exhibit a defect in erythroid maturation beyond that seen with loss of NF-E2 alone. Thus, the presence of a mild erythroid defect in NF-E2–deficient mice is not the result of compensation by Nrf-2.


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3459-3466 ◽  
Author(s):  
Florence Martin ◽  
Jan M. van Deursen ◽  
Ramesh A. Shivdasani ◽  
Carl W. Jackson ◽  
Amber G. Troutman ◽  
...  

Abstract NF-E2 binding sites, located in distant regulatory sequences, may be important for high level α- and β-globin gene expression. Surprisingly, targeted disruption of each subunit of NF-E2 has either little or no effect on erythroid maturation in mice. For p18 NF-E2, this lack of effect is due, at least in part, to the presence of redundant proteins. For p45 NF-E2, one possibility is that NF-E2–related factors, Nrf-1 or Nrf-2, activate globin gene expression in the absence of NF-E2. To test this hypothesis for Nrf-2, we disrupted the Nrf-2 gene by homologous recombination. Nrf-2–deficient mice had no detectable hematopoietic defect. In addition, no evidence was found for reciprocal upregulation of NF-E2 or Nrf-2 protein in fetal liver cells deficient for either factor. Fetal liver cells deficient for both NF-E2 and Nrf-2 expressed normal levels of α- and β-globin. Mature mice with combined deficiency of NF-E2 and Nrf-2 did not exhibit a defect in erythroid maturation beyond that seen with loss of NF-E2 alone. Thus, the presence of a mild erythroid defect in NF-E2–deficient mice is not the result of compensation by Nrf-2.


1989 ◽  
Vol 83 (3) ◽  
pp. 1032-1038 ◽  
Author(s):  
T J Ley ◽  
K A Maloney ◽  
J I Gordon ◽  
A L Schwartz

2007 ◽  
Vol 30 (11) ◽  
pp. 2091-2097 ◽  
Author(s):  
Masataka Maruyama ◽  
Tamihide Matsunaga ◽  
Eri Harada ◽  
Shigeru Ohmori

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 526-526
Author(s):  
Patrick G. Gallagher ◽  
Murat O. Arcasoy ◽  
Serena E. Vayda ◽  
Holly K. Dressman ◽  
James J. Bieker ◽  
...  

Abstract Mice deficient in the erythroid specific zinc-finger transcription factor EKLF die ~d14-15 of gestation of severe anemia, attributed to decreased expression of β-globin. The morphology of fetal-liver derived erythroid cells in EKLF-deficient mice does not mimic that seen in thalassemia, but instead shows hemolysis with uniform, nucleated erythroid progenitor cells. This has led to the hypothesis that a block in erythroid differentiation contributes to the anemia in EKLF-deficient mice. To address this, we performed microarray analyses with Affymetrix GeneChip Mouse Genome 430 2.0 arrays and RNA from d13.5 fetal livers of wild type (WT) and EKLF-deficient mice. Three independent EKLF +/+ and −/− RNA samples were analyzed. Numerous genes were down regulated including AHSP, pyruvate kinase, ankyrin, β spectrin and band 3. Verification of reduced expression of selected genes demonstrated that expression levels of many genes identified as down regulated via microarray analyses were minimally reduced in EKLF −/− RNA (&lt;20%) compared to normal (Rh 30, protein 4.2, protein 4.9, p55, AQP1, and ALAS-E). Flow cytometry of WT d14.5 fetal liver cells using TER 119 and CD71 was performed. In WT fetal livers, this identifies 5 populations, designated R1-R5, with R1/R2 composed of primitive progenitors and proerythroblasts and R3, R4, and R5 composed of more mature erythroblasts (Blood102:3938, 2003). In EKLF −/− fetal livers, R3, R4, and R5, populations involved in terminal erythroid differentiation, were completely absent, suggesting many of the genes identified by microarray analyses were differentially expressed because of a bias introduced by a differentiation block to more mature erythroid cells. Confirming this hypothesis, we demonstrated that genes with &lt;20% difference in expression between WT and EKLF-deficient fetal liver mRNA had 4-fold or higher levels in wild type R3+R4+R5 RNA compared to R1+R2 RNA. To better understand how differentially expressed genes were integrated into specific regulatory and signaling pathway networks, we used Ingenuity Pathway Analysis. A subset of focus genes incorporated into a biological network with highly a significant scores (&gt;40) was generated containing 35 focus genes. The biological function of this network involved cell cycle and DNA replication. At the central nodes of this network were E2F1 and E2F2, transcription factors involved in cell cycle control. Cell cycle analysis demonstrated that EKLF-deficient R1 cells exhibited a significant delay exiting G0+G1 and entering S phase and both R1 and R2 cells exhibited a defect in exiting S and entering G2+M. Colony assays with R1 and R2 cells revealed that EKLF-deficient fetal liver cells had decreased frequency of CFU-E, but similar absolute numbers of CFU-E as WT. As predicted by the cell cycle defect, EKLF−/− FL cells were severely (~10 fold) deficient in their ability to generate BFU-E. Flow cytometry with annexin V revealed no difference between WT and EKLF-deficient cells indicated that apoptosis was not contributing to the differentiation block. These results support the hypothesis that the failure of definitive erythropoiesis in EKLF deficient mice is due to decreased expression of many erythroid genes involved in erythroid differentiation, stabilization of α-globin protein, membrane stability, and glycolysis, not simply decreased transcription of the β-globin gene.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2404-2404
Author(s):  
Elena Bibikova ◽  
Shuo Lin ◽  
Kathleen M Sakamoto

Abstract Abstract 2404 Diamond-Blackfan Anemia (DBA) is characterized by defective erythropoiesis, congenital abnormalities, and predisposition to cancer. Approximately 25% of DBA patients have a defect in the RPS19 gene, which encodes a ribosomal protein on the 40S ribosomal subunit. Previous studies in zebrafish and mice have shown that knockdown of RPS19 is correlated with the upregulation of p53, a tumor suppressor gene that regulates cell cycle and apoptosis. However, the molecular pathways that link ribosome dysfunction and defective erythropoiesis remain unknown. Among the targets of p53 is microRNA34a (miR34a). Overexpression of miR34a in human myeloid leukemia K562 cells inhibits cell proliferation, leads to cell cycle arrest at the G1 stage, and promotes differentiation toward the megakaryocytic lineage in a p53-independent manner. MiR34a downregulates numerous proteins, including c-Myb, which is essential for normal erythropoiesis and has been found to be expressed at lower levels in bone marrow cells from DBA patients. We hypothesize that miR34a induction downstream of p53 activation in RPS19 insufficient normal hematopoietic progenitor cells contributes to defective erythropoiesis observed in DBA patients. To study the effects of RPS19 insufficiency and downstream signaling pathways, we transduced primary CD34+ fetal liver cells with two different shRNA constructs against RPS19. Knockdown was confirmed by Western blot analysis and by qRT-PCR (73%, p< 0.0001 for shRNA1, and 87%, p< 0.0001 for shRNA2, respectively), compared with a scrambled control shRNA. Cells were sorted for a GFP marker 72 hours post-transduction and either collected for downstream analysis or propagated further in methylcellulose. Our results demonstrated that knockdown of RPS19 in primary human CD34+ fetal liver cells leads to an increase in p53 protein by Western blot analysis, and upregulation of miR34a mRNA (3-fold, p<0.01). This upregulation correlates with downregulation of miR34a target genes including c-Myb (p<0.01) and c-Myc (p<0.01). Induction of miR34a and downregulation of c-Myb and c-Myc, occurs through a p53-dependent pathway since the expression of those genes was not altered in p53-null K562 cells transduced with RPS19 shRNA lentivirus. To correlate molecular pathways with the differentiation potential of RPS19 insufficient CD34+ fetal liver cells, methylcellulose colony assays were performed. Total colony formation was reduced by 87% (p<0.01) in cells treated with RPS19 shRNA compared to scrambled control shRNA. Erythroid colony formation in methylcellulose containing IL-3, SCF, GM-CSF, and EPO, decreased by 90% (p<0.01) in shRNA transduced cells compared to control shRNA. FACS analysis for erythroid markers CD71 and Glycophorin A showed a similar pattern, with the CD71+GlyA+ population representing 2.6% of RPS19 shRNA transduced cells, compared to 9.9% in cells transduced with the scrambled control. Preliminary data with lymphoblastoid cell lines (LCLs) from DBA patients with RPS19 insufficiency demonstrate 2-fold upregulation of miR34a. Therefore, we conclude that RPS19 insufficiency in human CD34+ fetal liver cells leads to p53-dependent upregulation of miR34a, down-regulation of miR34a target genes c-Myc and c-Myb, and defects in erythroid differentiation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2001 ◽  
Vol 97 (6) ◽  
pp. 1861-1868 ◽  
Author(s):  
Elise Coghill ◽  
Sarah Eccleston ◽  
Vanessa Fox ◽  
Loretta Cerruti ◽  
Clark Brown ◽  
...  

Erythroid Kruppel-like factor (EKLF) is a transcription factor of the C2H2 zinc-finger class that is essential for definitive erythropoiesis. We generated immortal erythroid cell lines from EKLF−/− fetal liver progenitor cells that harbor a single copy of the entire human β-globin locus and then reintroduced EKLF as a tamoxifen-inducible, EKLF–mutant estrogen receptor (EKLF-ER™) fusion protein. Addition of tamoxifen resulted in enhanced differentiation and hemoglobinization, coupled with reduced proliferation. Human β-globin gene expression increased significantly, whereas γ-globin transcripts remained elevated at levels close to endogenous mouse α-globin transcript levels. We conclude that EKLF plays a role in regulation of the cell cycle and hemoglobinization in addition to its role in β-globin gene expression. The cell lines we used will facilitate structural and functional analyses of EKLF in these processes and provide useful tools for the elucidation of nonglobin EKLF target genes.


2012 ◽  
Vol 27 (4) ◽  
pp. 398-404 ◽  
Author(s):  
Eiji Suzuki ◽  
Tamihide Matsunaga ◽  
Akiko Aonuma ◽  
Takamitsu Sasaki ◽  
Kiyoshi Nagata ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document