scholarly journals Molecular cloning and characterization of ZFF29: a protein containing a unique Cys2His2 zinc-finger motif

2004 ◽  
Vol 384 (3) ◽  
pp. 647-653 ◽  
Author(s):  
Haruhiko ASANO ◽  
Takashi MURATE ◽  
Tomoki NAOE ◽  
Hidehiko SAITO ◽  
George STAMATOYANNOPOULOS

We have cloned a gene, ZFF29 (zinc-finger protein of human fetal liver erythroid cells 29), from human fetal liver erythroid cells. Two types of mature mRNA were identified and designated ZFF29a and ZFF29b. In human genome the ZFF29 gene is on chromosome 9q, and the two forms are splice variants. There is a unique transcription start site, which predicts major mRNAs composed of 2485 bases for ZFF29a and 1801 bases for ZFF29b. The anticipated mRNAs were demonstrated in K562 cells, but not in any adult human tissues examined by Northern blotting. In the mouse, reverse transcription–PCR revealed that the ZFF29 mRNA is present in adult bone marrow and ovary at a higher level than in any other tissues examined. These findings suggest that ZFF29 proteins are expressed in embryonic/fetal erythroid tissues. The deduced polypeptide chains of ZFF29a and ZFF29b are composed of 306 and 350 amino acids respectively. A unique zinc-finger motif composed of two contiguous Cys2His2-type fingers is common to both forms of ZFF29. They are nuclear proteins and ZFF29b, but not ZFF29a, is an activator of erythroid gene promoters.

1999 ◽  
Vol 19 (5) ◽  
pp. 3571-3579 ◽  
Author(s):  
Haruhiko Asano ◽  
Xi Susan Li ◽  
George Stamatoyannopoulos

ABSTRACT A cDNA encoding a novel Krüppel-type zinc finger protein, FKLF, was cloned from fetal globin-expressing human fetal erythroid cells. The deduced polypeptide sequence composed of 512 amino acids revealed that, like Sp1 and EKLF, FKLF has three contiguous zinc fingers at the near carboxyl-terminal end. A long amino-terminal domain is characterized by the presence of two acidic and two proline-rich regions. Reverse transcription (RT)-PCR assays using various cell lines demonstrated that the FKLF mRNA is expressed predominantly in erythroid cells. FKLF message is detectable by RT-PCR in fetal liver but not in adult bone marrow cells. As predicted from its structural features, FKLF is a transcriptional activator. In luciferase assays FKLF activated the γ- and ɛ-globin gene promoters, and, to a much lower degree, the β-globin promoter. Studies of HS2-γ gene reporter constructs carrying CACCC box deletions revealed that the CACCC box sequence of the γ gene promoter mediates the activation of the γ gene by FKLF. Other erythroid promoters (GATA-1, glycophorin B, ferrochelatase, porphobilinogen deaminase, and 5-aminolevulinate synthase) containing CACCC elements or GC-rich potential Sp1-binding sites were activated minimally, if at all, by FKLF, indicating that FKLF is not a general activator of genes carrying the CACCC motifs. Transfection of K562 cells with FKLF cDNA enhanced the expression of the endogenous ɛ- and γ-globin genes, suggesting an in vivo role of FKLF in fetal and embryonic globin gene expression. Our results indicate that the protein potentially encoded by the FKLF cDNA acts as a transcriptional activator of embryonic and fetal β-like globin genes.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 433-433
Author(s):  
Jeremy W Rupon ◽  
Wulan Deng ◽  
Hongxin Wang ◽  
Philip D Gregory ◽  
Andreas Reik ◽  
...  

Abstract The β-genes undergo developmental activation and silencing in part by competing for their upstream enhancer, the locus control region (LCR). In adult erythroid cells, the LCR contacts the β-globin gene promoter by forming a loop that precludes interaction with the embryonic and fetal β-type globin genes. Reversing this developmental gene expression switch in favor of embryonic/fetal genes has therapeutic implications for patients with hemoglobinopathies. Here we employed a forced chromatin looping approach to activate the silenced murine embryonic βh1-globin gene and the human fetal γ-globin gene in adult erythroid cells. We have previously shown that forced recruitment via artificial zinc finger proteins of Ldb1, a protein necessary for long-range chromatin interactions at the β-globin locus, can trigger chromatin loop formation and transcription initiation. Here, we designed a zinc finger protein targeting the βh1 promoter, fused it to the self-association domain of Ldb1 (βh1-Ldb1), and introduced it into an adult murine erythroid cell line that normally produces nearly 100% adult β-globin. βh1-Ldb1 expression activated βh1-globin transcription up to 3000-fold accounting for ∼20% of total β-globin expression. βh1-Ldb1 similarly increased expression of βh1-globin in fetal liver derived primary erythroid cells. These results are striking given the degree to which murine embryonic globin genes are normally repressed. To test whether the activity βh1-Ldb1 was due to a looped interaction of the βh1 promoter with the LCR, we introduced βh1-Ldb1 into fetal liver derived erythroblasts from mice in which the LCR had been deleted. βh1-Ldb1 was virtually inactive in the absence of the LCR, demonstrating the dependence on the LCR and, by inference, long range looping of βh1-Ldb1 function. We next extended this approach to the human β-globin locus in an effort to activate expression of the fetal γ-globin gene in adult erythroid cells. Ldb1 was fused to a previously described γ-globin promoter binding zinc finger protein, GG1, to generate GG1-Ldb1. Introduction of GG1-Ldb1 into adult primary human erythroid cells strongly activated γ-globin expression with a concomitant reduction in β-globin transcription. Strikingly, γ-globin accounted for nearly 90% of total β-type globin transcription. Furthermore, fetal hemoglobin expression was nearly pan-cellular as determined by flow cytometry. These results demonstrate the power of forced chromatin looping to reprogram developmental regulation of gene expression, and provide a novel proof of concept for activating the γ-globin gene for the benefit of patients with hemoglobinopathies. Disclosures: Gregory: Sangamo BioSciences: Employment. Reik:Sangamo BioSciences: Employment.


Genomics ◽  
2001 ◽  
Vol 73 (2) ◽  
pp. 194-202 ◽  
Author(s):  
Kirsten Dahm ◽  
Peter J. Nielsen ◽  
Albrecht M. Müller

1977 ◽  
Vol 55 (5) ◽  
pp. 571-575 ◽  
Author(s):  
L. F. Congote ◽  
F. Bruno ◽  
S. Solomon

α-Fetoprotein and the synthesis of heme associated with hemoglobin were measured simultaneously in short-term cultures of human fetal liver cells to correlate the relationship of α-fetoprotein to erythroid cell function. Both synthetic processes decreased exponentially during the first 5 days of culture. The use of media supplemented with different batches of fetal calf serum and porcine portal vein serum indicated that the optimal conditions for the production of α-fetoprotein were different from those required for the synthesis of heme associated with hemoglobin. Moreover, the α-fetoprotein-producing cells could be separated from erythroid cells after velocity sedimentation in Ficoll gradients. Although it is well known that erythropoiesis and α-fetoprotein production occur simultaneously during ontogenesis, α-fetoprotein itself (0.01–100 μg/ml) did not stimulate heme synthesis in liver erythroid cells. Erythropoietin did not stimulate α-fetoprotein production. It is concluded that there is no cause–effect relationship between α-fetoprotein production and erythroid cell function in human fetal liver cells and that the two processes occur independently in different cell types.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1462-1462
Author(s):  
Michael Tallack ◽  
Thomas Whitington ◽  
Brooke Gardiner ◽  
Eleanor Wainwright ◽  
Janelle Keys ◽  
...  

Abstract Abstract 1462 Poster Board I-485 Klf1/Eklf regulates a diverse suite of genes to direct erythroid cell differentiation from bi-potent progenitors. To determine the local cis-regulatory contexts and transcription factor networks in which Klf1 works, we performed Klf1 ChIP-seq using the SOLiD deep sequencing platform. We mapped more than 10 million unique 35mer tags and found ∼1500 sites in the genome of primary fetal liver erythroid cells are occupied by endogenous Klf1. Many reside within well characterised erythroid gene promoters (e.g. b-globin) or enhancers (e.g. E2f2 intron 1), but some are >100kb from any known gene. We tested a number of Klf1 bound promoter and intragenic sites for activity in erythroid cell lines and zebrafish. Our data suggests Klf1 directly regulates most aspects of terminal erythroid differentiation including synthesis of the hemoglobin tetramer, construction of a deformable red cell membrane and cytoskeleton, bimodal regulation of proliferation, and co-ordination of anti-apoptosis and enucleation pathways. Additionally, we suggest new mechanisms for Klf1 co-operation with other transcription factors such as those of the gata, ets and myb families based on over-representation and spatial constraints of their binding motifs in the vicinity of Klf1-bound promoters and enhancers. Finally, we have identified a group of ∼100 Klf1-occupied sites in fetal liver which overlap with Klf4-occupied sites in ES cells defined by Klf4 ChIP-seq. These sites are associated with genes controlling the cell cycle and proliferation and are Klf4-dependent in skin, gut and ES cells, suggesting a global paradigm for Klfs as regulators of differentiation in many, if not all, cell types. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 6 (1) ◽  
pp. 61 ◽  
Author(s):  
Sergio Juárez-Méndez ◽  
Alejandro Zentella-Dehesa ◽  
Vanessa Villegas-Ruíz ◽  
Oscar Pérez-González ◽  
Mauricio Salcedo ◽  
...  

2010 ◽  
Vol 37 (4) ◽  
pp. 381-388
Author(s):  
Chao YANG ◽  
Lei JI ◽  
Shuang-Shuang SHI ◽  
Wen YUE ◽  
Li-Juan HE ◽  
...  

2000 ◽  
Vol 20 (9) ◽  
pp. 3178-3186 ◽  
Author(s):  
Takuro Nakamura ◽  
Yukari Yamazaki ◽  
Yuriko Saiki ◽  
Masatsugu Moriyama ◽  
David A. Largaespada ◽  
...  

ABSTRACT Evi9 is a common site of retroviral integration in BXH2 murine myeloid leukemias. Here we show that Evi9 encodes a novel zinc finger protein with three tissue-specific isoforms: Evi9a (773 amino acids [aa]) contains two C2H2-type zinc finger motifs, a proline-rich region, and an acidic domain; Evi9b (486 aa) lacks the first zinc finger motif and part of the proline-rich region; Evi9c (239 aa) lacks all but the first zinc finger motif. Proviral integration sites are located in the first intron of the gene and lead to increased gene expression. Evi9a and Evi9c, but not Evi9b, show transforming activity for NIH 3T3 cells, suggesting thatEvi9 is a dominantly acting proto-oncogene. Immunolocalization studies show that Evi9c is restricted to the cytoplasm whereas Evi9a and Evi9b are located in the nucleus, where they form a speckled localization pattern identical to that observed for BCL6, a human B-cell proto-oncogene product. Coimmunoprecipitation and glutathione S-transferase pull-down experiments show that Evi9a and Evi9b, but not Evi9c, physically interact with BCL6, while deletion mutagenesis localized the interaction domains in or near the second zinc finger and POZ domains of Evi9 and BCL6, respectively. These results suggest that Evi9 is a leukemia disease gene that functions, in part, through its interaction with BCL6.


Haematologica ◽  
2020 ◽  
pp. 0-0
Author(s):  
Daniel C. J. Ferguson ◽  
Juraidah Haji Mokim ◽  
Marjolein Meinders ◽  
Edmund R. R. Moody ◽  
Tom A. Williams ◽  
...  

Human ZNF648 is a novel poly C-terminal C2H2 zinc finger protein identified amongst the most dysregulated proteins in erythroid cells differentiated from iPSC. Its nuclear localisation and structure indicate it is likely a DNA-binding protein. Using a combination of ZNF648 overexpression in an iPSC line and primary adult erythroid cells, ZNF648 knockdown in primary adult erythroid cells and megakaryocytes, comparative proteomics and transcriptomics we show that ZNF648 is required for both erythroid and megakaryocyte differentiation. Orthologues of ZNF648 were detected across Mammals, Reptilia, Actinopterygii, in some Aves, Amphibia and Coelacanthiformes suggesting the gene originated in the common ancestor of Osteichthyes (Euteleostomi or bony fish). Conservation of the C-terminal zinc finger domain is higher, with some variation in zinc finger number but a core of at least six zinc fingers conserved across all groups, with the N-terminus recognisably similar within but not between major lineages. This suggests the N-terminus of ZNF648 evolves faster than the C-terminus, however this is not due to exon-shuffling as the entire coding region of ZNF648 is within a single exon. As for other such transcription factors, the N-terminus likely carries out regulatory functions, but showed no sequence similarity to any known domains. The greater functional constraint on the zinc finger domain suggests ZNF648 binds at least some similar regions of DNA in the different organisms. However, divergence of the N-terminal region may enable differential expression, allowing adaptation of function in the different organisms.


Sign in / Sign up

Export Citation Format

Share Document