Submit Paper

Article Processing Fee

Pay Online

           

Crossref logo

  DOI Prefix   10.20431


 

ARC Journal of Hematology
Volume-2 Issue-2, 2017, Page No: 23-37

Importance and Presentation of Transcription Factor Fli-1 in Hematopoiesis and in Hematological and Other Malignancies

Ota Fuchs

Institute of Hematology and Blood Transfusion, Prague, Czech Republic.

Citation : Ota Fuchs, "Importance and Presentation of Transcription Factor Fli-1 in Hematopoiesis and in Hematological and Other Malignancies" ARC Journal of Hematology. 2017 ; 2(2) : 23-37.

Copyright : © 2017 . This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Abstract:

Transcription factor Friend leukemia virus integration 1 (Fli-1) comes under 28 members in human Ets family of transcription factors. These members are involved in development of different tissues, proliferation, differentiation and apoptosis as well as in cancer progression. Avian erythroblastosis transforming retrovirus E26 (v-Ets), which produced leukemias in chicken, was initially detected in 1982. Its characteristic wing-turn-helix Ets DNA binding domain (DBD) was described in 1985. Fli-1 was first identified in 1990 as a proto-oncogene near a common site for retroviral integration in Friend virus-induced erythroleukemia. Fli-1 functions as both a transcriptional activator and repressor. The nuclear transcription factor Fli-1 plays an important role in hematopoiesis by coordinating the balance between proliferation and differentiation. Fli-1 is engaged in both erythroid and megakaryocytic development. Abnormal expressions of Fli-1 were detected in hematological malignancies such as acute myeloid leukemia and diffuse large B-cell lymphoma and also in other malignancies such as astrocytoma, melanoma, colon cancer, endometrial cancer, epithelial ovarian cancer, nasopharyngeal carcinoma, head and neck squamous cell cancer and is connected with progression of these diseases. Fli-1 negatively regulates the tumor suppressor p53 by directly binding to the promoter of MDM2 gene and stimulating of its transcription and up-regulating MDM2. Fli-1 acts as oncogene by promoting proliferation and inhibiting p53 function. Therefore, Fli-1 can be a potential prognostic biomarker and molecular target in hematological and other malignancies. Mithramycin, a DNA binding aureolic acid natural product targets oncogenic Fli-1. Cardenolides, calcium ionophores, topoisomerase I inhibitors and several other small compounds are also capable to inhibit Fli-1.


Keywords: ETS transcription factors, Fli-1, differentiation, proliferation, BET inhibitors, mithramycin,Hematology


1. Introduction


Friend murine leukemia virus (F-MuLV) – induced murine erythroleukemia is a very good model to study multi-step leukemogenesis, which means the multi-stage nature of cancer [1-3]. This model was used for identification of several oncogenes and tumor suppressors such as Fli-1, Spi-1/PU.1, TP53, Fli-2/p45 NFE2, and Fli-3/miR-17-92 [4-16]. F-MuLV – induced erythroleukemia is characterized by proerythroblast expansion leading to anemia, splenomegaly, and eventually death within eight weeks of viral infection in susceptible strains of newborn mice. Integration of the provirus at the Fli-1 locus drives enhancer-mediated Fli-1 over expression [4,5,13-15]. Fli-1 viral integration induces expression of anti-apoptotic genes (Bcl- 2 /B-cell lymphoma 2/ and Bcl-xL) [17] and tumor suppressor genes (TP53 and p45 NFE2) [6-8,19]. However, this Fli-1 viral integration decreases Rb (retinobloastoma gene) expression [20] and changes Epo (erythropoietin gene) locus arrangement resulting in the constitutive activation of the Epo receptor signal transduction pathway and in growth and survival advantage in vitro and in vivo [21,22].

Fli-1 inhibits the apoptotic cell death program normally activated in erythroblasts with Epo deficiency. Fli-1 inhibits also the terminal differentiation program induced in these cells in response to Epo and induces their proliferation. An abnormally activated form of the receptor for Epo (EpoR) is not needed for effects of Fli-1 on erythroblast [12]. Erythroblasts expressing Fli-1 have enhanced survival which correlates with the increase of Bcl2 expression. The rapid down regulation of cyclin D2 and D3 expression typical for Epo-induced differentiation is prevented by Fli-1. Transcription factor Fli-1 blocks also the down regulation of several other genes involved in cell cycle or cell proliferation control. These results show that over expression of Fli-1 causes the deregulation of the normal balance between differentiation and proliferation in primary erythroblasts [12]. Thus, the activation of Fli-1 expression in murine Friend erythroleukemia cells may provide a proliferative advantage to these cells and inhibit terminal differentiation or cell death. The envelope glycoprotein gp55 is present in spleen focus-forming virus (SFFV) and binds and activates EpoR [23-26]. However, gp55 is not present in F-MuLV. Erythroid cells expressing gp55 proliferate in the absence of their normal regulator Epo. While Fli-1 is activated in about 90% of F- MuLV-induced erythroleukemias, small percentage of infected mice had retroviral insertion within Fli-3 locus, leading to activation of a micro RNA (miRNA, miR) cluster labeled miR17-92 [27]. Both Fli-1 and miR17-92 are over expressed in various cancer cell types and activate similar signaling pathways associated with erythroid transformation [28-45].

Transcription factor Fli-1 is very important in megakaryocytic differentiation [46-58]. Fli-1 binds to and directly regulates the genes encoding several proteins that are essential for terminal megakaryocytic differentiation. Homozygous loss of functional Fli-1 alleles in mice leads to embryonic lethality because of severe defects in fetal megakaryopoiesis and a high incidence of embryonic hemorrhaging. This is the result of impaired megakaryopoiesis as well as inefficient blood vessel formation. Thus, Fli-1 functions as a master regulator of blood and endothelial development [59-61].

Moreover, transcription factor Fli-1 is a key component together with GATA-2 and SCL in the control of hematopoietic stem cell (HSC) specification [62]. Gata2, Fli1, and Scl (Stem cell leukemia)/Tal1 (T-cell acute lymphocytic leukemia protein 1) are expressed in embryonic day-11.5 dorsal aorta where HSCs originate and in fetal liver where they multiply. The three HSC enhancers in these tissues and in ES cell-derived hemangioblast equivalents are bound by each of these transcription factors. Pimkin et al.[57] mapped the genome-wide chromatin occupancy of four master hematopoietic transcription factors (GATA1, GATA2, TAL1, and FLI1) and three histone methylation marks (H3K4me1, H3K4me3, and H3K27me3) in lineage-committed erythroid and megakaryocytic cells. In addition, they defined global gene expression changes that accompany the development of these mature lineages from hematopoietic stem and progenitor cells (HSPCs). By correlating gene expression with transcription factor occupancy, they produced a global functional annotation of chromatin dynamics during erythro-megakaryopoiesis.

Their findings provided new insights into GATA protein functions and reveal a robust, genome-wide mechanism of megakaryocytic lineage priming in multi potential hematopoietic progenitors. Specification and differentiation of erythroid cells and megakaryocytes from the common megakaryocyte-erythroid progenitor (MEP) are governed by coordinated regulation of a precise balance of members of several transcription factors, adaptors and micro RNAs including GATA-binding transcription factors (GATA-1 and GATA-2), Ets factors (Fli-1 and GABPα /GA-binding protein α/), Krűppel-containing factors (KLF1/EKLF and Leukemia/lymphoma Related Factor /LRF/), basic helix-loop-helix factors (SCL), multiple adaptors (Friend of GATA-1 /FOG-1/, LIM domain-binding protein 1 /LDB1/ and ) and micro RNAs (miR-150 and miR-451) [63-77]. Recent studies have also provided support for a model in which murine HSCs rapidly commit to unipotent megakaryocytic progenitors not derived from bipotent megakaryocytic and erythroid-committed progenitors [78].

A novel and simple technology to obtain platelets using trans differentiation of human bone marrow erythroblasts to mega karyocytes with over expression of the Fli-1 and Erg genes has been recently described [79]. To generate Fli-1 and Erg over expressing erythroblasts, the isolated erythroblasts were transduced with lentiviral vectors containing the full coding sequences of human Fli-1 and Erg genes, respectively.

1.1. Friend Leukemia Integration-1 (Fli-1)
Fli-1 is located on mouse chromosome 9 and human chromosome 11q24, a region of frequent abnormalities in human disease [5,80]. Both the murine and human homologs of the Fli-1 gene are approximately 120 kb, consist of nine exons, end encode two protein isoforms, two protein isoforms, p51 (452 amino acid residues /aa/) and p48 (419 aa) [5,81,82]. The Fli-1 gene is on the same chromosome as Ets-1 gene located in the distance about 240 kb from Ets-1 gene locus. Both genes arose probably by gene duplication from a common ancestral gene [5]. Two initiation codons of transation are localized to nucleotide 245 within exon 1 and 344 within exon 2 of the mouse Fli-1 mRNA sequence (NCBI Reference Sequence: NM_008026.5 [82]. These translation initiation codons are responsible for the synthesis of two Fli-1 protein isoforms. Similar situation is in human Fli-1 mRNA where two ATG translation initiation sites have been localized to nucleotides 344 (exon 1) and 443 (exon 2) of the human Fli-1 mRNA sequence (NCBI Reference Sequence: NM_002017.4). These translation initiation codons are responsible for the synthesis of two Fli-1 protein isoforms p51 and p48[82]. The second isoform, p48, has a shorter N-terminus and contains a distinct 5´ untranslated region (UTR) in comparison with isoform 1, p51. Alternative exons 1 (1a and 1b) of murine and human Fli-1 gene as the result of alternative splicing and translation of alternative Fli-1 mRNAs were also detected [83,84]. Exon 1b was not conserved in the mouse but was detected in every analyzed human cell, whereas exon 1a was present only in a part of analyzed human and murine cells. These results suggested that both mouse and human Fli-1 gene expression might be under the control of several independent promoter regions. Fli-1 gene expression was detected in lymphoid, myeloid and endothelial cells, spleen, thymus and at lower levels in lung, heart and ovaries [85]. Fli-1 gene expression is potently down regulated during Epo-induced erythroid differentiation [20].

Several cis-regulatory elements within a 234 bp region (sequence from −271 to −37) that contibute to Fli-1 gene promoter activity was found in a murine B cell line and T cell line [86,87]. This region contains a GATA consensus site as well as two Ets binding sites. Indeed, introducing mutations within these Ets binding sites abolished the promoter activity. Transcription factor Spi-1/PU.1 binds to this region and positively regulates Fli-1 gene expression on the transcription level [9].

Ets factors Elf1, Tel and Fli-1 bind in vitro to this region and increase endogenous Fli-1 expression in a T cell line. These Ets transcription factors also stimulate Fli-1 gene expression in vivo . This mechanism and polymorphic microsatellite of GA dinucleotide repeats located at the 5´ end of exon 1 have the important influence on Fli-1 expression. The presence of a shorter microsatellite resulted in higher Fli1 promoter activity . A significant association of this polymorphism was observed with systemic lupus erythematosus (SLE) of patients without nephritis [88].

The functional domains located within the Fli-1 protein include the 5´ Ets domain, and Fli-1- specific region (FLS), both together referred to as the amino-terminal transcriptional activation domain (ATA), and a 3´ Ets domain and carboxy-terminal transcriptional activation domain (CTA). The 5´ Ets domain is located within amino acids 121-196 and the FLS, which is absent in the Ets-related protein Erg with 81% homology to the Fli-1 protein, is localized within amino acids 205-292. The 3´ Ets domain was determined witin amino acids 277-360 and is important for sequence specific DNA-binding activity of Fli-1. The CTA domain, found within amino acids 402-452, is also involved in transcriptional activation and protein-protein interactions with other transcription factors. Both, the 5´ and 3´ Ets domains contain sequences of helix-loop-helix (H-L-H) secondary structures, while the FLS and CTA domains contain sequences of turn-loop-turn (T-L-T) secondary structures. Transcriptional activity of Fli-1 is regulated by ATA and CTA domains. CTA domain may function also as transcriptional repressor [89]. Moussa et al. [90] generated mutant Fli-1 mice that expressed a truncated Fli-1 protein lacking CTA region. Heterozygous mice with this defect in Fli-1 were viable, while homozygous mice with this truncated Fli-1 had reduced viability and reduced platelet number. Platelet aggregation and activation were also impaired. Homozygous mutants had the thrombocytopenia phenotype with extensive bleeding.

Both isoforms of Fli-1, p51 and p48, are phosphorylated primarily on serine residues in human T cells [91]. Phosphorylated Fli-1 has a short half-life (p51 isoform-105min and p48 isoform-80 min). In contrast, the phosphorylated Erg protein is much more stable with a half-life of 21 h. Fli-1 phosphorylation is dependent on the concentration of intracellular calcium. Protein phosphatase 2A is involved in Fli-1 dephosphorylation. Transforming growth factor beta (TGF-β) stimulated phosphorylation and acetylation cascade of Fli-1 in dermal fibroblasts [92]. In this case, Fli-1 phosphorylation was localized to threonine 312 located in the DNA binding domain of Fli-1. Protein kinase C delta (PKCȍ) caused this phosphorylation and was controlled by the c-Abl tyrosine kinase, which is required for nuclear localization of PKCȍ [93].

1.2. Fli-1 Target Genes
Fli-1 functions as both a transcriptional activator and repressor [17,20,48,49,59,64,70,87,90,94-117]. A great part of results in this area was gained in F-MuLV – induced murine erythroleukemia model system as I described in Introduction. Fli-1 induced transcriptional up regulation of Bcl-2 and Mdm2 (the RING-E3 ubiquitin ligase) and blocks apoptosis. Mdm2 mediates deregulation of p53 by stabilizing Mdm2 via ubiquitination of p53 and its degradation in proteasomes. Therefore, direct upregulation of Mdm2 by Fli-1 [94] destabilizes the anti-apoptotic protein p53, which plays an important role in the initial transformation of erythroblasts by Friend virus [18] and accelerates tumor progression by inducing genomic instability. Fli-1 down regulates EKLF/KLF1 and GATA-1 genes expression . Moreover, Fli-1 negatively regulates phosphatidyl-inositol polyphosphate 5-phosphatase (Ship1) gene expression. Down-regulation of Ship1 resulted in higher phosphorylation of AKT/PKB by PI3K and erythroid proliferation [96]. Fli-1 stimulated ribosomal gene expression and ribosome biogenesis in erythroleukemogenesis [97].

Fli-1 has emerged as a critical regulator of inflammatory mediators, including monocyte chemoattractant protein-1(MCP-1) [107,108], macrophage inflammatory protein-2 (CXCL2) [111], chemokine C-C motif ligand 5 (CCL5) / RANTES (Regulated on Activation, Normal T cell Expressed and Secreted [112], G-protein-coupled receptor 9 in the CXC chemokine receptor (CXCR3) [113], interleukin 6 (IL-6) [115], and endothelial protein C receptor (EPCR) [116]. Infiltration of T cells into the kidney is a typical feature of human and experimental lupus nephritis that contributes to renal injury. All these inflammatory mediators are promising therapeutic targets in lupus nephritis, where Fli-1 expression is elevated. The reducing the levels of Fli-1 in SLE and similar autoimmune kidney diseases may be protective against development of nephritis through down regulation of desribed inflammatory mediators.

The cytokine, granulocyte colony stimulating factor (G-CSF) regulates neutrophil precursor maturation and survival, and activates mature neutrophils. Fli-1 drives transcription from the G-CSF promoter and mutation of the Fli-1 DNA binding domain resulted in abolut 90% loss of transcriptional activity . Acetylation of Fli-1 by histone acetyltransferases p300/CBP and p300/CBP associated factor (PCAF) significantly decreased Fli-1 specific activation of the G-CSF promoter [114]. On the other hand, G-CSF induces stabilization of Fli-1 during myeloid development [118].

Fli1 recruits HDAC1/p300 to the COL1A2 promoter and suppresses the expression of the COL1A2 gene by chromatin remodeling through histone deacetylation . TGF-β-dependent phosphorylation of Fli1 at threonine 312 is a critical step regulating the remodeling of the Fli1 transcription repressor complex, leading to transcriptional activation of the COL1A2 gene .

Fli-1 DBD dimerization plays a role in transcriptional activation and repression by Fli-1 and its fusion proteins at promoters containing Fli-1 binding sites [119]. The homodimerization interface is helix-swapped and dominated by hydrophobic interactions, including those between two interlocking Phe 362 residues.

1.3. The Role Of Fli-1 In Malignant Transformation
Fli-1 is over expressed in various cancer cell types [28-39]. A number of virally induced leukemias, including Friend virus-induced erythroleukemia, are associated with Fli-1 over expression [12-15]. Fli-1 activates the Rho GTPase pathway and is associated with metastasis in breast cancer [120]. The high expression of Fli-1 associated with adverse prognosis of endometrial cancer correlated with a high differentiation grade and mutated P53 expression [38]. The high expression of Fli-1 connected with the progression of epithelial ovarian cancer correlated with advanced tumor stage, positive lymph nodal involvement, and poor overall survival (OS) and disease-free survival (DFS) [36]. Deregulation of ETS1 and Fli-1 c ontributes to the pathogenesis of diffuse large B-cell lymphoma [34]. DLBCL is the most common type of non-Hodgkin lymphoma. ETS1 and Fli-1 sustained cell viability of a DLBCL cell line. Down regulation of ETS1 and Fli-1 through correspondent lentiviral shRNAs induced lower proliferation rate and decreased mRNA levels of target genes of ETS a Fli-1. High expression of FLI1 protein is an adverse prognostic factor in acute myeloid leukemia (AML) [33]. AML patients with high Fli-1 had lower complete remission CR) rate and shorter CR duration, resulting in a significantly shorter overall survival. However, high Fli-1 expression is good prognostic marker in the human breast cell lines and in a small sample of human primary breast tumors [121]. Loss of Fli-1 has similar effects as increased migration and invasion in the breast cancer cells and may promote tumor progression. Contrary to these results, another report showed that the expression of Fli-1 contributes to the progression of human breast cancer by up-regulation of the bcl-2 gene and inhibition of apoptosis [30].

Gene fusion is another contribution of Fli-1 to malignant transformation. Fli-1 is the most frequently implicated in chromosomal translocation t(11;22)(q24;q12) [122-128]. Ewing sarcoma is defined by this chromosomal translocations that fuse EWS (EWSR1), located at 22q12, and a gene of the Ets family of transcription factors. In 85-90% of cases, the fusion gene is EWS-Fli-1, which encodes the N-terminal portion of EWS and the C-terminal portion of Fli-1, including the ETS DNA-binding domain. The resultant EWS-Fli-1 chimeric protein includes transactivation domain of EWS and DNA-binding domain of Fli-1. EWS-Fli-1 regulates the expression of a number of genes important for cancer progression and is the causative oncogene. EWS-Fli-1 affects the transcriptome, epigenome and proteome to reprogram cells in disease development. EWS-Fli-1 up regulates target genes such as c-myc [123], DNA-binding protein inhibitor 2 (ID2), cyclin D1 (CCND1), glioma-associated oncogene homolog 1 (Gli1) [127], Forkhead box family gene (FOXM1) , vascular endothelial growth factor A (VEGFA), matrix metallopeptidase 3 (MMP-3), nuclear receptor subfamily 0, group B, member 1 (NR0B1, alias DAX-1) and histone methyl-transferase Enhancer of Zeste (EZH2), implicated in cell cycle, invasion and proliferation pathways. EWS-Fli-1 functions also as transcriptional repressor and inhibits the expression of some tumor suppressor genes such as p21Cip1, Waf1, p57Kip1, transforming growth factor beta receptor II (TGF-βRII), insulin-like growth factor binding protein 3 (IGFBP3) and lysine-6-oxidase (LOX) [127].

Direct pharmacological inhibition of EWS-Fli-1 to eradicate this cancer is unsuccessful but some new approaches downregulate important target genes. The bromodomain and extra-terminal domain (BET) inhibitors repressed these target genes but EWS-Fli-1 expression was not significantly affected [129-131]. BET proteins (bromodomain /BRD/ containing proteins BRD2, BRD3, BRD4, and the testis-specific BRDT) were found as an important class of epigenetic readers of histone acetylation involved in chromatin remodeling and transcriptional regulation. The human proteome contains 46 bromodomain-containing proteins with a total of 61 bromodomains with conserved structural features [132]. BET inhibitors led to inhibition of Ewing sarcoma cells proliferation and survival. Specific BET inhibitors such as dimethylisoxazole-based compound I-BET151 or triazolodiazepines-based compounds JQ1 and I-BET762 resulted in displacement of BRDs from chromatin and inhibition of transcription at key genes such as BCL2, MYC, and CDK6 [129].

Preclinical data suggest that the histone lysine specific demethylase 1 (LSD1) inhibition [133] also disrupts the function of EWS-Fli-1. Fli-1 is a novel Ets transcription factor involved in gene fusions in prostate cancer. SLC45A3-Fli-1 fusion was described in prostate cancer [134]. SLC45A3 exon 1 is fused with Fli-1 exon 3. As the currently available prostate cancer cell lines do not harbor Fli-1 overexpression, the development of a specific biological model of the SLC45A3-Fli-1 fusion is needed to further clarify the oncogenic role Fli-1 in prostate cancer.

1.4. Fli-1 Inhibitors
Transcription factors have been considered undrugable, but this concept has been recently changed. Several Fli-1 inhibitors are presently used to treat cancers. To identify small molecular weight compounds capable of inhibiting the transactivation ability of Fli-1, luciferase reporter assays were used [135]. The luciferase reporter assay is commonly used as a tool to study gene expression at the transcriptional level. It is widely used because it is convenient, relatively inexpensive, and gives quantitative measurements. A vector with the Fli-1 Ets DNA binding site cloned in front of a minimal promoter, immediately upstream of the luciferase reporter gene was utilized. The construct was co-transfected with the EWS/Fli-1 vector and control empty vector binto human embryonic kidney epithelial cell line 293T cells. Chemical library of about 4500 compounds was screened in this way . Thirty compounds that down regulated EWS/Fli-1 luciferase activity by at least 50% in three independent experiments were found. The six functional drug groups were established from these 30 effective compounds. Two compounds from the group of chemotherapeutic drugs, etoposide and dactinomycin (actinomycin D), are topoisomerase II inhibitors used to treat diverged cancer types including Ewing sarcoma [136-138]. These drugs kill tumor cells by inhibition of EWS/Fli-1 and by inducing DNA damage. Topoisomerases I and II are enzymes that control the changes in DNA structure by catalyzing the breaking and rejoining of the phosphodiester backbone of DNA strands during the cell cycle. Topoisomerase inhibitors block the ligation step of the cell cycle that leads to apoptosis and cell death. Dactinomycin intercalates between adjacent guanine-cytosine base pairs, blocking the transcription of DNA by RNA polymerase. It also causes single-strand DNA breaks, possibly via a free-radical intermediate or an interaction with topoisomerase II.

Most cardiac glycosides (e.g., digitoxin, digoxin, ouabain, and oleandrin) have been isolated from plants, including Digitalis purpurea, Digitalis lanata, Strophanthus gratus, and Nerium oleander. Some cardiac glycosides have also been found in amphibians and mammals, including digoxin, ouabain, bufalin, marinobufagenin, and telecinobufagin [139]. Ten compounds from this group inhibit EWS/Fli-1. Calcimycin (A23187) from the group of calcium ionophores is a rare divalent cation specific ionophore antibiotic that has many biochemical and pharmaceutical applications. Calcimycin inhibits binding of Fli-1 to DNA by regulation of PKCδ phosphorylation of Fli-1.

An aureolic acid natural product mithramycin (MTM, Mithracin®) was validated as a potent antagonists of the oncogenic transcription factor EWS-Fli-1 [140-142]. MTM (also known as plicamycin, trade name Mithracin) is member of tricyclic polyketides family and is produced by Sreptomyces plicatus. MTM binds in the minor groove of GC-rich DNA, thereby disrupting transcription of protooncogenes. MTM antagonizes EWS-Fli-1 without affecting its expression levels. MTM likely directly interferes with the EWS-Fli-1 function on DNA. Sub-micromolar concentrations MTMs stabilize Fli-1-DNAcomplex on GGAA repeats, which are critical for the oncogenic function of hybrid protein EWS-Fli-1 [141]. Formation of a ternary complex Fli-1-DNA-MTM on a single GGAA Fli-1/MTM binding site was also demonstrated by nuclear magnetic resonance. Phase I/II trial NCT 01610570 of mithramycin in children and adults with refractory extracranial solid tumors or Ewing sarcoma has been recently done.

1.5. Increased Level Of Fli-1 Messenger Rna In Bone Marrow And Blood Mononuclear Cells Of Mds Patients With 5q- Syndrome
Patients with the 5q- syndrome, a subtype of myelodysplastic syndrome (MDS) have macrocytic anemia and thrombocythemia with effective through dysplastic megakaryopoiesis. We found an increased level of Fli-1 mRNA in the mononuclear blood and bone marrow cells [143]. We therefore proposed that in this MDS subtype, high Fli-1 preferentially induces differentiation of a common megakaryocytic and erythroid progenitor (MEP) into the megakaryocytic lineage and probably allows effective megakaryopoiesis leading to normal or even increased platelet numbers. Boultwood et al. [144] localized 40 genes to the commonly deleted region (CDR) of the long arm of chromosome 5. Some of the genes on the distal part of the CDR encode micro RNAs (miR-145 and miR-146a) [145]. Kumar et al. [146] identified Fli-1 as a miR-145 target gene, which was confirmed in colon carcinoma [147] and Ewing sarcoma [148]. Our finding of a high Fli-1 mRNA level in 5q- syndrome was consistent with haploinsufficiency of this miR-145 in 5q-syndrome and stabilization of Fli-1 mRNA level and increased Fli-1 protein levels. Haploinsufficiency of miR-145 and miR-146a leads to increased expression of their target genes, TIRAP (Toll/interleukin-1 receptor /TIR/ homology domain-containing adaptor protein) and TRAF6 (Tumor necrosis factor /TNF/ receptor associated factor 6), and consequently to an upregulation of IL-6 expression [149]. IL-6 stimulates megakaryocyte differentiation and platelet production [150].


2. Conclusions And Perspectives


Transcription factor Fli-1 is a member of the Ets (E-twenty-six) family of winged helix-turn-helix transcription factors that bind a purine-rich consensus sequence GGA (A/T). Transcriptional activation of the Fli-1 gene by either chromosomal translocation or proviral insertion leads to Ewing sarcoma in humans and erythroleukemia in mice, respectively. In Ewing sarcoma, the DNA binding (Ets domain) of Fli- 1 is translocated from chromosome 11 to the EWS locus on chromosome 22, forming a fusion protein with altered expression and transactivation properties. In mice infected with Friend murine leukemia virus, about 75% of the resulting erythroleukemias have insertional activation of the Fli-1 gene. Thus, dysregulation of the Fli-1 gene is a common factor in the progression of at least two distinct tumor types.

Transcription factor Fli-1 has important role in normal development and malignant transformation. Fli-1 is necessary for stem cell maintenance and differentiation, hematopoiesis, vasculogenesis and angiogenesis. Fli-1 is required for murine vascular and megakaryocytic development and is frequently hemizygously deleted in patients with thrombocytopenia (Jacobsen and Paris-Trousseau syndrome). The level of Fli-1 during megakaryopoiesis affects thrombopoiesis and platelet biology [151]. It is unknown how deregulated Fli-1 expression alters the balance between erythroid differentiation and proliferation. Fli-1 induces de-differentiation by reverting erythroid colony-forming (CFU-E) cells to erythroid burst-forming (BFU-E) cells in mouse erythroleukemia cells [152]. The nuclear transcription factor Fli-1 has been shown to increase cellular proliferation and tumorigenesis in many types of cancer [29-39,85,120,153-156].

Transcription factors have long been counted as undruggable targets for therapeutics. Enhanced recognition of protein biochemistry as well as the need to have more targeted approaches to treat cancer has taken transcription factors approachable for therapy.It is possible to block protein-DNA or protein-protein interactions. Specific enantiomers, DNA-binding small molecular weight compounds (for exammple YK-4-279 and its individual enantiomers, etoposide, actinomycin D, calcium ionophores, mithramycin) are used or studied in pre-clinical trials. The main problem is that many these drugs are not specific for targeting Fli-1 or EWS-Fli-1 fusion proteins. The bromodomain and extra-terminal domain (BET) inhibitors repressed these target genes but EWS-Fli-1 expression was not significantly affected. BET proteins bind to the N-acetylated lysine residues on histone tails and induce an opened-chromatin structure to enhance transcription of several oncogenes (for example myc). Further progress in this area is required.


Acknowledgements


This work was supported by the research project for conceptual development of research organization (00023736; Institute of Hematology and Blood Transfusion, Prague) from the Ministry of Health of the Czech Republic.


References


  1. Tambourin, P.; Wendling, F.; Moreau-Gachelin, F. Friend leukemia as a multiple-step disease. Blood Cells 1981, 7, 133-44
  2. Ben-David, Y.; Bernstein, A. Friend virus-induced erythroleukemia: A multistage malignancy. Ann NY Acad Sci U S A 1989, 567, 165-70.
  3. Ben-David, Y.; Bernstein, A. Friend virus-induced erythroleukemia and the multistage nature of cancer. Cell 1991, 66, 831-4.
  4. Ben-David, Y.; Giddens, E.B.; Bernstein, A. Identification and mapping of a common proviral integration site Fli-1 in erythroleukemia cells induced by Friend murine erythroleukemia virus. Proc Natl Acad Sci U S A 1990, 87, 1332-6.
  5. Ben-David, Y.; Giddens, E.B.; Letwin, K.; Bernstein, A. Erythroleukemia induction by Friend murine erythroleukemia virus: Insertional activation of a new member of the ets gene family, Fli-1, closely linked to c-ets-1. Genes Dev 1991, 5, 908-18.
  6. Ben-David, Y.; Lavigueur, A.; Cheong, G.Y.; Bernstein, A. Insertional inactivation of the p53 gene during Friend leukemia: A new stratégy for identifying tumor suppressor genes. New Biologist 1990, 2, 1015-23.
  7. Johnson, P; Benchimol, S. Friend virus induced murine erythroleukemia: the p53 locus. Cancer Surv 1992, 12, 137-51.
  8. Prasher, J.M.; Elenitoba-Johnson, K.S.; Kelley, L.L. Loss of p53 tumor suppressor function is required for in vivo progression of Friend erythroleukemia. Oncogene 2001, 20, 2946-55.
  9. Starck, J.; Doubeikovski, A.; Sarrazin, S.; Gonnet, C.; Rao, C.; Skoultchi, A.; Godet, J.; Dusanter-Fourt, I.; Morle, F. Spi-1/PU.1 is a positive regulator of the Fli-1 gene involved in inhibition of erythroid differentiation in Friend erythroleukemic cell lines. Mol Cell Biol 1999, 19, 121-35.
  10. Rimmele, P.; Kosmider, O.; Mayeux, P.; Moreau-Gachelin, F.; Guillouf, C. Spi-1/PU.1 participates in erythroleukemogenesis by inhibiting apoptosis in cooperation with Epo signaling and by blocking erythroid differentiation. Blood 2007, 109, 3007-14.
  11. Papetti, M.; Skoultchi, A.I. Reprogramming leukemia cells to terminal differentiation and growth arrest by RNA interference of PU.1. Mol Cancer Res 2007, 5, 1053-62.
  12. Pereira, R.; Quang, C.T.; Lesault, I.; Dolznig, H.; Beug, H.; Ghysdael, J. Fli-1 inhibits differentiation and induces proliferation of primary erythroblasts. Oncogene 1999, 18, 1597-608.
  13. Ano, S.; Pereira, R.; Pironin, M.; Lesault, I.; Milley, C.; Lebigot, I.; Quang, C.T.; Ghysdael, J. Erythroblast transformation by Fli-1 depends upon its specific DNA binding and transcriptional activation properties. J Biol Chem 2004, 279, 2993-3002.
  14. Cui, J.W.; Vecchiarelli-Federico, L.M.; Li, Y.J.;Wang, G.J.; Ben-David, Y. Continuous Fli-1 expression plays an essential role in the proliferation and survival of F-MuLV-induced erythroleukemia and human erythroleukemia. Leukemia 2009, 23, 1311-9.
  15. Lee, C.R.; Cervi, D.; Truong, A.H.; Li, Y.J.; Sarkar, A.; Ben-David, Y. Friend virus-induced erythroleukemias: a unique and well-defined mouse model for the development of leukemia. AntiCancer Res 2003, 23, 2159-66.
  16. Kayali, S.; Giraud, G.; Morié, F.; Guyot, B. Spi-1, Fli-1 and Fli-3 (miR-17-92) oncogenes contribute to a single oncogenic network controlling cell proliferation in Friend erythroleukemia. PLOS ONE 2012, 7, e46799.
  17. Lesault, I.; Quang, C.T.; Frampton, J.; Ghysdael, J. Direct regulation of BCL-2 by FLI-1 is involved in the survival of FLI-1-transformed erythroblasts. Oncogene 1999, 18, 1597-608.
  18. Wong, K.S.; Li, Y.J.; Howard, J.; Ben-David, Y. Loss of p53 in F-MuLV induced-erythroleukemias accelerates the acquisition of mutational events that confers immortality and growth factor independence. Oncogene 1999, 18, 5525-34.
  19. Li, Y.J.; Higgins, R.R.; Pak, B.J.; Shivdasani, R.A.; Ney, P.A.; Archer, M.; Ben-David, Y. p45(NFE2) is a negative regulator of erythroid proliferation which contributes to the progression of Friend virus-induced erythroleukemias. Mol Cell Biol 2001, 21, 73-80.
  20. Tamir, A.; Howard, J.; Higgins, R.R.; Li, Y.J.; Berger, L.; Zacksenhaus, E.; Reis, M.; Ben-David, Y. Fli-1, an Ets-related transcription factor, regulates erythropoietin-induced erythroid proliferation and differentiation: evidence for direct transcriptional repression of the Rb gene during differentiation. Mol Cell Biol 1999,19, 4452-64.
  21. McDonald, J.; Beru, N.; Goldwasser, E. Rearrangement and expression of erythropoietin genes in transformed mouse cells. Mol Cell Biol 1987, 7, 365-70.
  22. Howard, J.C.; Berger, J.; Bani, M.R.; Hawley, R.G., Ben-David, Y. Activation of the erythropoietin gene in the majority of F-MuLV-induced erythroleukemias results in growth factor independence and enhanced tumorigenicity. Oncogene 1996, 12, 1405-15.
  23. Li, J.P.; D´Andrea, A.D.; Lodish, H.F.; Baltimore, D. Activation of cell growthby binding of Friend spleen focus-forming virus gp55 glycoprotein to the erythropoietin receptor. Nature 1990, 343, 762-4.
  24. D´Andrea A.D. The interaction of the erythropoietin receptor and gp55. Cancer Surv 1992, 15, 19-36.
  25. Quang, C.T.; Wessely, O.; Pironin, M.; Beug, H.; Ghysdael, J. Cooperation of Spi-1/PU.1 with an activated erythropoietin receptor inhibits apoptosis and Epo-dependent differentiation in primary and induces their Kit ligand-dependent proliferation. EMBO J 1997, 16, 5639-5653.
  26. Zochodne, B.; Truong, A.H.L., Stetler, K.; Higgins, R.R.; Howard, J.; Dumont, D.; Berger, S.A.; Ben-David, Y. Epo regulates erythroid proliferation and differentiation through distinct signaling pathways: implication for erythropoiesis and Friend virus-induced erythroleukemia. Oncogene 2000, 19, 2296-304.
  27. Cui, J.W.; Li, Y.J.; Sarkar, A.; Brown, J.; Tan, Y.H.; Premyslova, M.; Michaud, C.; Iscove, N.; Wang, G.J.; Ben-David, Y. Retroviral insertional activation of the Fli-3 locus in erythroleukemias encoding a cluster of microRNAs that convert Epo-induced differentiation to proliferation. Blood 2007, 110, 2631-40.
  28. Folpe, A.L.; Chand, E.M.; Goldblum, J.R.; Weiss, S.W. Expression of Fli-1, a nuclear transcription factor, distinguishes vascular neoplasms from potential mimics. Am J Surg Pathol 2001, 25, 1061-6.
  29. Mhawech-Fauceglia, P.; Herrmann, F.R.; Bshara, W.; Odunsi, K.; Terracciano, L.; Sauter, G.; Cheney, R.T.; Groth, J.; Penetrante, R. Friend leukemia integration-1 expression in malignant and benign tumors: a multiple tumor tissue microarray analysis using polyclonal antibody. J Clin Pathol 2007, 60, 694-700.
  30. Sakurai, T.; Kondoh, N.; Arai, M.; Hamada, J.; Yamada, T.; Kihara-Negishi, F.; Izawa, T.; Ohno, H.; Yamaqmoto, M.; Oikawa, T. Functional roles of Fli-1, a member of the Ets family of transcription factors, in human breast malignancy. Cancer Sci 2007, 98, 1775-84.
  31. Toriakovic, E.E.; Slipcevic, A.; Florenes, V.A.; Chibbar, R.; DeCoteau, J.F.; Bilalovic, N. Fli-1 expression in malignant melanoma. Histol Histopathol 2008, 23, 1309-14.
  32. Lin, O.; Filippa, D.A.; Teruya-Feldstein, J. Immunohistochemical evaluation of FLI-1 in acute lymphoblastic lymphoma (ALL): a potential diagnostic pitfall. Appl Immunohistochem Mol Morphol. 2009, 17, 409-12.
  33. Kornblau, S.M.; Qiu, Y.H.; Zhang, N.; Singh, N.; Faderi, S.; Ferrajoli, A.; York, H.; Qutub, A.A.; Coombes, K.R.; Watson, D.K. Abnormal expression of FLI1 protein is an adverse prognostic factor in acute myeloid leukemia. Blood 2011, 118, 5604-12.
  34. Bonetti, P.; Testoni, M.; Scandurra, M.; Ponzoni, M.; Piva, R.; Mensah, A.A.; Rinaldi, A.; Kwee, I.;+ Tibiletti, M.G.; Iqbal, J.; Greiner, T.C.; Chan, W.C.; Gaidano, G.; Piris, M.A.; Cavalli, F.; Zucca, E.; Ighirami, G.; Bertoni, F. Deregulation of ETS1 and FLI1 contributes to the pathogenesis of diffuse large B-cell lymphoma. Blood 2013, 122, 2233-41.
  35. Smeets, M.F.; Chan, A.C.; Dagger, S.; Bradley, C.K.; Wei, A.; Izon, D.J. Fli-1 overexpression in hematopoietic progenitors deregulates T-cell development and induces pe-T cell lymphoblastic leukemia/lymphoma. PLOS ONE 2013, 8.
  36. Song, W.; Hu, L.; Li, W.; Wang, G.; Li, Y.; Yan, L.; Li, A.; Cui, J. Oncogenic Fli-1 is a potential prognostic marker for the progression of epithelial ovarian cancer. BMC Cancer 2014, 14, 424.
  37. Liang, X.; Shi, D.; Yun, J.; Mao, Y.; Quyang, P.; Su, Z.; Fu, J.; Hou, J.; Deng, W.; Xie, F. Friend leukemia virus integration 1 expression has prognostic significance in nasopharyngeal carcinoma. Transl Oncol 2014, 7, 493-502.
  38. Song, W.; Zhang, T.; Li, W.; Mu, R.; Zhang, L.; Li, Y.; Jin, B.; Wang, N.; Li, A.; Cui, J. Overexpression of Fli-1 is associated with adverse prognosis of endometrial cancer. Cancer Invest 2015, 33, 469-75.
  39. Lin, S.F.; Wu, C.C.; Chai, C.Y. Increased FLI-1 expression is associated with poor prognosis in non-small cell lung cancers. Appl Immunohistochem Mol Morphol 2016, 24, 556-61.
  40. Ota, A.; Tagawa, H.; Kaman, S.; Tsuzuki, S.; Karpas, A.; Kira, S.; Yoshida, Y.; Seto, M. Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Res 2004, 64, 3087-95.
  41. Hayashita, Y.; Osada, H.; Tatematsu, Y.; Yamada, H.; Yanagisawa, K.; Tomida, S., Yatabe, Y.; Kawahara, K, Sekido, Y.; Takahashi, T. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Casncer Res 2005, 21, 9628-32.
  42. Diosdado, B.; van de Wiel, M.A.; Terhaar Sive Droste, J.S.; Mongera, S.; Postma, C.; Meijerink, W.J.H.J.; Carvalho, B.; Meijer, G.A. MiR-17-92 cluster is associated with 13q gain and c-myc expression during colorectal adenoma to adenocarcinoma progression. Br J Cancer 2009, 101, 707-14.
  43. Kantadalam, M.M.; Beta, M.; Maheswari, U.K.; Swaminathan, S.; Krishnakumar, S. Oncogenic microRNA 17-92 cluster is regulated by epithelial cell adhesion molecule and could be a potential therapeutic target in retinoblastoma. Mol Vision 2012, 18, 2279-87.
  44. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: a comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ 2013, 20, 1603-14.
  45. Roisman, A.; Garaicoa, F.H.; Metrebian, F.; Narbaitz, M.; Kohan, D.; Rivello, H.G.; Fernandez, I.; Pavlovsky, A.; Pavlovsky, M.; Hrenandéz, L.; Slavutsky, I. SOXC and miR17-92 gene expression profiling defines two subgroups with different clinical outcome in maqntle cell lymphoma. Gen Chrom Cancer 2016, 55, 531-40.
  46. Athanasiou, M.; Clausen, P.A.; Mavrothalassitis, G.J.; Zhang, X.K.; Watson, D.K.; Blair, D.G. Increased expression of the ETS-related transcription factor FLI-1/ERGB correlates with and can induce the megakaryocytic phenotype. Cell Growth Differ 1996, 7, 1525-34.
  47. Bastian, L.S.; Kwiatkowski, B.A.; Breininger, J.; Danner, S.; Roth, G. Regulation of the megakaryocytic glycoprotein IX promoter by the oncogenic Ets transcription factor Fli-1. Blood 1999, 93, 2637-44.
  48. Eisbacher, M.; Holmes, M.L.; Newton, A.; Hogg, P.J.; Khachigian, L.M.; Crossley, M.; Chong, B.H. Protein-protein interaction between Fli-1 and GATA-1 mediates synergistic expression of megakaryocyte-specific genes through cooperative DNA binding. Mol Cell Biol 2003, 23, 3427-41.
  49. Jackers, P.; Szalai, G.; Moussa, O.; Watson, D.K. Ets-dependent regulation of target gene expression during megakaryopoiesis. J Biol Chem 2004, 50, 52183-90.
  50. Hu, W.; Philips, A.S.; Kwok, J.C.; Eisbacher, M.; Chong, B.H. Identification of nuclear import and export signals within Fli-1: roles of the nuclear import signals in Fli-1-dependent activation of megakaryocyte-specific promoters. Mol Cell Biol 2005, 254, 3087-108.
  51. Pang, L.; Weiss, M.J.; Poncz, M. Megakaryocyte biology and related disorders. J Clin Invest 2005, 115, 3332-8.
  52. Pang, L.; Xue, H.H.; Szalai, G.; Wang, X.; Wang, Y.; Watson, D.K.; Leonard, W.J.; Blobel, G.A.; Poncz, M. Maturation stage-specific regulation of megakaryopoiesis by pointed-domain Ets proteins. Blood 2006, 108, 2198-206.
  53. Moussa, O.; LaRue, A.C.; Abangan, R.S.Jr.; Williams, C.R.; Zhang, X.K.; Masuya, M.; Gong, Y.Z.; Spyropoulos, D.D.; Ogawa, M.; Gilkeson, G.; Watson, D.K. Thrombocytopenia in mice lacking the carboxy-terminal regulatory domain of the Ets transcription factor Fli1. Mol Cell Biol 2010, 30, 5194-206.
  54. Doré, L.C.; Crispino, J.D. Transcription factor networks in erythroid cell and megakaryocyte development. Blood 2011, 118, 231-9.
  55. Neuwirtova, R.; Fuchs, O.; Holicka, M.; Vostry, M.; Kostecka, A.; Hajkova, H.; Jonasova, A.; Cermak, J.; Cmejla, R.; Pospisilova, D.; Belickova, M.; Siskova, M.; Hochova, I.; Vondrakova, J.; Sponerova, D.; Kadlckova, E.; Novakova, L.; Brezinova, J.; Michalova, K. Transcription factors Fli1 and EKLF in the differentiation of megakaryocytic and erythroid progenitor in 5q-syndrome and in Diamond-Blackfan anemia. Ann Hematol 2013, 92, 11-8.
  56. Tijssen, M.R.; Ghevaert, C. Transcription factors in late megakaryopoiesis and related platelet disorders. J Thromb Haemost 2013, 11, 593-604.
  57. Pimkin, M.; Kossenkov, A.V.; Mishra, T.; Morrissey, C.S.; Wu, W.; Keller, C.A,; Blobel, G.A.; Lee, D.; Beer, M.A.; Hardison, R.C.; Weiss, M.J. Divergent functions of hematopoietic transcription factors in lineage priming and differentiation during erythro-megakaryopoiesis. Genome Res 2014, 24, 1932-44.
  58. Zang, C.; Luyten, A.; Chen, J.; Liu, X.S.; Shivdasani, R.A. NF-E2, FLI-1 and RUNX1 collaborate at areas of dynamic chromatin to activate transcription in mature mouse megakaryocytes. Sci Rep 2016, 6, 30255.
  59. Hart, A.; Melet, F.; Grossfeld, P.; Chien, K.; Jones, C.; Tunnacliffe, A.; Favier, R.; Bernstein, A. Fli-1 is required for murine vascular and megakaryocytic development and is hemizygously deleted in patients with thrombocytopenia. Immunity 2000, 13, 167-77.
  60. Spyropoulos, D.D.; Pharr, P.N.; Lavenburg, K.R.; Jackers, P.; Papas, T.S.; Ogawa, M.; Watson, D.K. Hemorrhage, impaired hematopoiesis, and lethality in mouse embryos carrying a targeted disruption of the Fli-1 transcription factor. Mol Cell Biol 2000, 20, 5643-52.
  61. Liu, F.; Walmsley, M.; Rodaway, A.; Patient, R. Fli1 acts at the top of the transcriptional network driving blood and endothelial development. Curr Biol 2008, 18, 1234-40.
  62. Pimanda, J.E.; Ottersbach, K.; Knezevic, K.; Kinston, S.; Chan, W.Y.; Wilson, N.K.; Landry, J.R.; Wood, A.D.; Kolb-Kokocinski, A.; Green, A.R.; Tannahill, D.; Lacaud, G.; Kouskoff, V.; Göttgens, B. Gata2, Fli1, and Scl form a recursively wired gene-regulatory circuit during early hematopoietic development. Proc Natl Acad Sci U S A 2007, 104, 17692-7.
  63. Stachura, D.L.; Chou, S.T.; Weiss, M.J. Early block to erythromegakaryocytic development conferred by loss of transcription factor GATA-1. Blood 2006, 107, 87–97.
  64. Wang, X.; Crispino, J.D.; Letting, D.L.; Nakazawa, M.; Poncz, M.; Blobel, G.A. Control of megakaryocyte-specific gene expression by GATA-1 and FOG-1: role of Ets transcription factors. EMBO J 2002, 21, 5225–34.
  65. Walsh, J.C.; DeKoter, R.P.; Lee, H.J.; Smith, E.D.; Lancki, D.W.; Gurish, M.F.; Friend, D.S.; Stevens, R.L.; Anastasi, J.; Singh, H. Cooperative and antagonistic interplay between PU.1 and GATA-2 in the specification of myeloid cell fates. Immunity 2002, 17, 665–76.
  66. Tripic, T.; Deng, W.; Cheng, Y.; Zhang, Y.; Vakoc, C.R.; Gregory, G.D.; Hardison, R.C.; Blobel, G.A. SCL and associated proteins distinguish active from repressive GATA transcription factor complexes. Blood 2009, 113, 2191–201.
  67. Suzuki, M.; Kobayashi-Osaki, M.; Tsutsumi, S.; Pan, X.; Ohmori, S.; Takai, J.; Moriguchi, T.; Ohneda, O.; Ohneda, K.; Shimizu, R.; Kanki, Y.; Kodama, T.; Aburatani, H.; Yamamoto, M. GATA factor switching from GATA2 to GATA1 contributes to erythroid differentiation. Genes Cells 2013, 18, 921-33.
  68. Duncan, M.T.; Shin, S.; Wu, J.J.; Mays, Z.; Weng, S.; Bagheri, N.; Miller, W.M.; Shea, L.D. Dynamic transcription factor activity profiles reveal key regulatory interactions during megakaryocytic and erythroid differentiation. Biotechnol Bioeng 2014, 111, 2082-94.
  69. Starck, J.; Weiss-Gayet, M.; Gonnet, C.; Guyot, B.; Vicat, J.M.; Morlé, F. Inducible Fli-1 gene deletion in adult mice modifies several myeloid lineage commitment decisions and accelerates proliferation arrest and terminal erythrocytic differentiation. Blood 2010, 116, 4795-805.
  70. Starck, J.; Cohet, N.; Gonnet, C.; Sarrazin, S.; Doubeikovskaia, Z.; Doubeikovski, A.; Verger, A.; Duterque-Coquillaud, M.; Morle, F. Functional cross-antagonism between transcription factors FLI-1 and EKLF. Mol Cell Biol 2003, 23, 1390-402.
  71. Bouilloux, F.; Juban, G.; Cohet, N.; Buet, D.; Guyot, B.; Vainchenker, W.; Louache, F.; Morlé, F. EKLF restricts megakaryocytic differentiation at the benefit of erythrocytic differentiation. Blood 2008, 112, 576-84.
  72. Hoang, T.; Lambert, J.A.; Martin, R. SCL/TAL1 in Hematopoiesis and Cellular Reprogramming. Curr Top Dev Biol 2016, 118, 163-204.
  73. Li, L.; Freudenberg, J.; Cui, K.; Dale, R.; Song, S.H.; Dean, A.; Zhao, K.; Jothi, R.; Love, P.E. Ldb1-nucleated transcription complexes function as primary mediators of global erythroid gene activation. Blood 2013, 121, 4575-85.
  74. Love, P.E.; Warzecha, C.; Li, L. Ldb1 complexes: the new master regulators of erythroid gene transcription. Trends Genet 2014, 30, 1-9.
  75. Lu, J.; Guo, S.; Ebert, B.L.; Zhang, H.; Peng, X.; Bosco, J.; Pretz, J.; Schlanger, R.; Wang, J.Y.; Mak, R.H.; Dombkowski, D.M.; Preffer, F.I.; Scadden, D.T.; Golub, T.R. MicroRNA-mediated control of cell fate in megakaryocyte-erythrocyte progenitors. Dev Cell 2008, 14, 843–53.
  76. Pase, L.; Layton, J.E.; Kloosterman, W.P.; Carradice, D.; Waterhouse, P.M.; Lieschke, G.J. miR-451 regulates zebrafish erythroid maturation in vivo via its target gata2. Blood 2009, 113, 1794-804.
  77. Obeidi, N.; Pourfathollah, A.A.; Soleimani, M.; Nikougoftar Zarif, M.; Kouhkan, F. The Effect of Mir-451 Upregulation on Erythroid Lineage Differentiation of Murine Embryonic Stem Cells. Cell J 2016, 18, 165-78.
  78. Woolthuis, C.M.; Park, C.Y. Hematopoietic stem/progenitor cell commitment to the megakaryocyte lineage. Blood 2016, 127, 1242-8.
  79. Siripin, D.; Kheolamai, P.; U-Pratya, Y.; Supokawej, A.; Wattanapanitch, M.; Klincumhom, N.; Laowtammathron, C.; Issaragrisil, S. Transdifferentiation of erythroblasts to megakaryocytes using FLI1 and ERG transcription factors. Thromb Haemost 2015, 114, 593-602.
  80. Truong, A.H.; Ben-David, Y. The role of Fli-1 in normal cell function and malignant transformation. Oncogene 2000 Dec 18;19(55):6482-9.
  81. Watson, D.K.; Smyth, F.E.; Thompson, D.M.; Cheng, J.Q.; Testa, J.R.; Papas, T.S.; Seth, A. The ERGB/Fli-1 gene: isolation and characterization of a new member of the family of human ETS transcription factors. Cell Growth Differ 1992, 3, 705-13.
  82. Sarrazin, S.; Starck, J.; Gonnet, C.; Doubeikovski, A.; Melet, F.; Morle, F. Negative and translation termination-dependent positive control of FLI-1 protein synthesis by conserved overlapping 5' upstream open reading frames in Fli-1 mRNA. Mol Cell Biol 2000, 20, 2959-69.
  83. Dhulipala, P.D.; Lee, L.; Rao, V.N.; Reddy, E.S. Fli-1b is generated by usage of differential splicing and alternative promoter. Oncogene 1998, 17, 1149-57.
  84. Barat, C.; Barbeau, B.; Delattre, O.; Rassart, E. Presence of new alternative exons in human and mouse Fli-1 genes. Biochim Biophys Acta 2000, 1517, 164-70.
  85. Li, Y.; Luo, H.; Liu, T.; Zacksenhaus, E.; Ben-David, Y. The ets transcription factor Fli-1 in development, cancer and disease. Oncogene 2015, 34, 2022-31.
  86. Nowling, T.K.; Fulton, J.D.; Chike-Harris, K.; Gilkeson, G.S. Ets factors and a newly identified polymorphism regulate Fli1 promoter activity in lymphocytes. Mol Immunol 2008, 45, 1-12.
  87. Svenson, J.L.; Chike-Harris, K.; Amria, M.Y.; Nowling, T.K. The mouse and human Fli1 genes are similarly regulated by Ets factors in T cells. Genes Immun 2010, 11, 161-72.
  88. Morris, E.E.; Amria, M.Y.; Kistner-Griffin, E.; Svenson, J.L.; Kamen, D.L.; Gilkeson, G.S.; Nowling, T.K. A GA microsatellite in the Fli1 promoter modulates gene expression and is associated with systemic lupus erythematosus patients without nephritis. Arthritis Res Ther 2010, 12, R212.
  89. Rao, V.N.; Ohno, T.; Prasad, D.B.; Bhattacharya, G.; Reddy, E.S. Analysis of the Dna-binding and transcriptional activation functions of human Fli-1 protein. Oncogene 1993, 8, 2167-73.
  90. Moussa, O.; LaRue, A.C.; Abangan, R.S.Jr.; Williams, C.R.; Zhang, X.K.; Masuya, M.; Gong, Y.Z.; Spyropoulos, D.D.; Ogawa, M.; Gilkeson, G.; Watson, D.K. Thrombocytopenia in mice lacking the carboxy-terminal regulatory domain of the Ets transcription factor Fli1. Mol Cell Biol 2010, 30, 5194-206.
  91. Zhang, X.K.; Watson, D.K. The FLI-1 transcription factor is a short-lived phosphoprotein in T cells. J Biochem 2005, 137, 297-302.
  92. Asano, Y.; Trojanowska, M. Phosphorylation of Fli1 at threonine 312 by protein kinase C delta promotes its interaction with p300/CREB- binding protein-associated factor and subsequent acetylation in response to transforming growth factor beta. Mol Cell Biol 2009, 29, 1882-94.
  93. Bujor, A.M.; Asano, Y.; Haines, P.; Lafyatis, R.; Trojanowska, M. The c-Abl tyrosine kinase controls protein kinase Cδ-induced Fli-1 phosphorylation in human dermal fibroblasts. Arthritis Rheum 2011, 63, 1729-37.
  94. Truong, A.H.; Cervi, D.; Lee, J.; Ben-David, Y. Direct transcriptional regulation of MDM2 by Fli-1. Oncogene 2005, 24, 962-9.
  95. Athanasiou, M.; Mavrothalassitis, G.; Sun-Hoffman, L.; Blair, D.G. FLI-1 is a suppressor of erythroid differentiation in human hematopoietic cells. Leukemia 2000, 14, 439-45.
  96. Lakhanpal, G.K.; Vecchiarelli-Federico, L.M.; Li, Y.J.; Cui, J.W.; Bailey, M.L.; Spaner, D.E.; Dumont, D.J.; Barber, D.L.; Ben-David, Y. The inositol phosphatase SHIP-1 is negatively regulated by Fli-1 and its loss accelerates leukemogenesis. Blood 2010, 116, 428-36.
  97. Juban, G.; Giraud, G.; Guyot, B.; Belin, S.; Diaz, J.J.; Starck, J.; Guillouf, C.; Moreau-Gachelin, F.; Morlé, F. Spi-1 and Fli-1 directly activate common target genes involved in ribosome biogenesis in Friend erythroleukemic cells. Mol Cell Biol 2009, 29, 2852-64.
  98. Smeets, M.F.; Chan, A.C.; Dagger, S.; Bradley, C.K.; Wei, A.; Izon, D.J. Fli-1 overexpression in hematopoietic progenitors deregulates T cell development and induces pre-T cell lymphoblastic leukaemia/lymphoma. PLoS One 2013, 8, e62346.
  99. Göttgens, B.; Nastos, A.; Kinston, S.; Piltz, S.; Delabesse, E.C.; Stanley, M.; Sanchez, M.J.; Ciau-Uitz, A.; Patient, R.; Green, A.R. Establishing the transcriptional programme for blood: the SCL stem cell enhancer is regulated by a multiprotein complex containing Ets and GATA factors. EMBO J 2002, 21, 3039-50.
  100. Göttgens, B.; Broccardo, C.; Sanchez, M.J.; Deveaux, S.; Murphy, G.; Göthert, J.R.; Kotsopoulou, E.; Kinston, S.; Delaney L, Piltz S, Barton LM, Knezevic K, Erber WN, Begley CG, Frampton J, Green AR. The scl +18/19 stem cell enhancer is not required for hematopoiesis: identification of a 5' bifunctional hematopoietic-endothelial enhancer bound by Fli-1 and Elf-1. Mol Cell Biol 2004, 24, 1870-83.
  101. Seth, A.; Robinson, L.; Thompson, D.M.; Watson, D.K.; Papas, T.S. Transactivation of GATA-1 promoter with ETS1, ETS2 and ERGB/Hu-FLI-1 proteins: stabilization of the ETS1 protein binding on GATA-1 promoter sequences by monoclonal antibody. Oncogene 1993, 8, 1783-90.
  102. Lemarchandel, V.; Ghysdael, J.; Mignotte, V.; Rahuel, C.; Roméo, P.H. GATA and Ets cis-acting sequences mediate megakaryocyte-specific expression. Mol Cell Biol 1993, 13, 668-76.
  103. Deveaux, S.; Filipe, A.; Lemarchandel, V.; Ghysdael, J.; Roméo, P.H.; Mignotte, V. Analysis of the thrombopoietin receptor (MPL) promoter implicates GATA and Ets proteins in the coregulation of megakaryocyte-specific genes. Blood 1996, 87, 4678-85.
  104. Holmes, M.L.; Bartle, N.; Eisbacher, M.; Chong, B.H. Cloning and analysis of the thrombopoietin-induced megakaryocyte-specific glycoprotein VI promoter and its regulation by GATA-1, Fli-1, and Sp1. J Biol Chem 2002, 277, 48333-41.
  105. Okada, Y.; Watanabe, M.; Nakai, T.; Kamikawa, Y.; Shimizu, M.; Fukuhara, Y.; Yonekura, M.; Matsuura, E.; Hoshika, Y.; Nagai, R.; Aird, W.C.; Doi, T. RUNX1, but not its familial platelet disorder mutants, synergistically activates PF4 gene expression in combination with ETS family proteins. J Thromb Haemost 2013, 11, 1742-50.
  106. Gosiengfiao, Y.; Horvat, R.; Thompson, A. Transcription factors GATA-1 and Fli-1 regulate human HOXA10 expression in megakaryocytic cells. DNA Cell Biol 2007, 26, 577-87.
  107. Suzuki, E.; Karam, E.; Williams, S.; Watson, D.K.; Gilkeson, G.; Zhang, X.K. Fli-1 transcription factor affects glomerulonephritis development by regulating expression of monocyte chemoattractant protein-1 in endothelial cells in the kidney. Clin Immunol 2012, 145, 201-8.
  108. Lennard Richard, M.L.; Nowling, T.K.; Brandon, D.; Watson, D.K.; Zhang, X.K. Fli-1 controls transcription from the MCP-1 gene promoter, which may provide a novel mechanism for chemokine and cytokine activation. Mol Immunol 2015, 63, 566-73.
  109. Liu, F.; Patient, R. Genome-wide analysis of the zebrafish ETS family identifies three genes required for hemangioblast differentiation or angiogenesis. Circ Res 2008, 103, 1147-54
  110. Soncin, F.; Mattot, V.; Lionneton, F.; Spruyt, N.; Lepretre, F.; Begue, A.; Stehelin, D. VE-statin, an endothelial repressor of smooth muscle cell migration. EMBO J 2003, 22, 5700-11.
  111. Lou, N.; Lennard Richard, M.L.; Yu, J.; Kindy, M,; Zhang, X.K. The Fli-1 transcription factor is a critical regulator for controlling the expression of chemokine C-X-C motif ligand 2 (CXCL2). Mol Immunol 2017, 81, 59-66.
  112. Lennard Richard, M.L.; Sato, S.; Suzuki, E.; Williams, S.; Nowling, T.K.; Zhang, X.K. The Fli-1 transcription factor regulates the expression of CCL5/RANTES. J Immunol 2014, 193, 2661-8.
  113. Sundararaj, K.P.; Thiyagarajan, T.; Molano, I.; Basher, F.; Powers, T.W.; Drake, R.R.; Nowling, T.K. FLI1 Levels Impact CXCR3 Expression and Renal Infiltration of T Cells and Renal Glycosphingolipid Metabolism in the MRL/lpr Lupus Mouse Strain. J Immunol 2015, 195, 5551-60.
  114. Lennard Richard, M.L.; Brandon, D.; Lou, N.; Sato, S.; Caldwell, T.; Nowling, T.K.; Gilkeson, G.; Zhang, X.K. Acetylation impacts Fli-1-driven regulation of granulocyte colony stimulating factor. Eur J Immunol 2016, 46, 2322-32.
  115. Sato, S.; Lennard Richard, M.; Brandon, D.; Jones Buie, J.N.; Oates, J.C.; Gilkeson, G.S.; Zhang, X.K. A critical role of the transcription factor fli-1 in murine lupus development by regulation of interleukin-6 expression. Arthritis Rheumatol 2014, 66, 3436-44.
  116. Saigusa, R.; Asano, Y.; Yamashita, T.; Taniguchi, T.; Takahashi, T.; Ichimura, Y.; Toyama, T.; Yoshizaki, A.; Miyagaki, T.; Sugaya, M.; Sato, S. Fli1 deficiency contributes to the downregulation of endothelial protein C receptor in systemic sclerosis: a possible role in prothrombotic conditions. Br J Dermatol 2016, 174, 338-47.
  117. Asano, Y.; Trojanowska, M. Fli1 represses transcription of the human α2(I) collagen gene by recruitment of the HDAC1/p300 complex. PLoS One 2013, 8, e74930.
  118. Mora-Garcia, P.; Wei, J.; Sakamoto, K.M. G-CSF induces stabilization of ETS protein Fli-1 during myeloid cell development. Pediatr Res 2005, 57, 63-6.
  119. Hou, C.; Tsodikov, O.V. Structural basis for dimerization and DNA binding of transcriptio factor FLI1. Biochemistry 2015, 54, 7365-74.
  120. Song, W.; Li, W.; Li, L.; Zhang, S.; Yan, X.; Wen, X.; Zhang, X.; Tian, H.; Li, A.; Hu, J.F.; Cui, J. Friend leukemia virus integration 1 activates the Rho GTPase pathway and is associated with metastasis in breast cancer. Oncotarget 2015, 6, 23764-75.
  121. Scheiber, M.N.; Watson, P.M.; Rumboldt, T.; Stanley, C.; Wilson, R.C.; Findlay, V.J.; Anderson, P.E.; Watson, D.K. FLI1 expression is correlated with breast cancer cellular growth, migration, and invasion and altered gene expression. Neoplasia 2014, 16, 801-13.
  122. Lessnick, S.L.; Ladanyi, M.Molecular pathogenesis of Ewing sarcoma: new therapeutic and transcriptional targets. Annu Rev Pathol 2012, 7, 145-59.
  123. Jully, B.; Vijayalakshmi, R.; Gopal, G.; Sabitha, K.; Rajkumar, T. Junction region of EWS-FLI1 fusion protein has a dominant negative effect in Ewing's sarcoma in vitro. BMC Cancer 2012, 12, 513.
  124. Sankar, S.; Bell, R.; Stephens, B.; Zhuo, R.; Sharma, S.; Bearss, D.J.; Lessnick, S.L. Mechanism and relevance of EWS/FLI-mediated transcriptional repression in Ewing sarcoma. Front Oncol 2015, 5, 162.
  125. Sankar, S.; Bell, R.; Stephens, B.; Zhuo, R.; Sharma, S.; Bearss, D.J.; Lessnick, S.L. Mechanism and relevance of EWS/FLI-mediated transcriptional repression in Ewing sarcoma. Oncogene 2016, 35, 6155-6.
  126. Hensel, T.; Giorgi, C.; Schmidt, O.; Calzada-Wack, J.; Neff, F.; Buch, T.; Niggli, F.K.; Schäfer, B.W.; Burdach, S.; Richter, G.H. Targeting the EWS-ETS transcriptional program by BET bromodomain inhibition in Ewing sarcoma. Oncotarget 2016, 7, 1451-63.
  127. Jacques, C.; Lamoureux, F.; Baud'huin, M.; Rodriguez Calleja, L.; Quillard, T.; Amiaud, J.; Tirode, F.; Rédini, F.; Bradner, J.E.; Heymann, D.; Ory, B. Targeting the epigenetic readers in Ewing sarcoma inhibits the oncogenic transcription factor EWS/Fli1. Oncotarget 2016, 7, 24125-40
  128. Paronetto, M.P. Ewing sarcoma protein: a key player in human cancer. Int J Cell Biol 2013, 2013, 642853.
  129. Selvanathan, S.P.; Graham, G.T.; Erkizan, H.V.; Dirksen, U.; Natarajan, T.G.; Dakic, A.; Yu, S.; Liu, X.; Paulsen, M.T.; Ljungman, M.E.; Wu, C.H.; Lawlor, E.R.; Üren, A.; Toretsky, J.A. Oncogenic fusion protein EWS-FLI1 is a network hub that regulates alternative splicing. Proc Natl Acad Sci U S A 2015, 112, E1307-16.
  130. Cidre-Aranaz, F.; Alonso, J. EWS/FLI1 Target Genes and Therapeutic Opportunities in Ewing sarcoma. Front Oncol 2015, 5, 162.
  131. Loganathan, S.N.; Tang, N.; Fleming, J.T.; Ma, Y.; Guo, Y.; Borinstein, S.C.; Chiang, C.; Wang, J. BET bromodomain inhibitors suppress EWS-FLI1-dependent transcription and the IGF1 autocrine mechanism in Ewing sarcoma. Oncotarget 2016, 7, 43504-17.
  132. Ferri, E.; Petosa, C.; McKenna, C.E. Bromodomains: Structure, function and pharmacology of inhibition. Biochem Pharmacol 2016, 106, 1-18.
  133. Maiques-Diaz, A.; Somervaille, T.C. LSD1: biologic roles and therapeutic targeting. Epigenomics 2016, 8, 1103-16.
  134. Paulo, P.; Barros-Silva, J.D.; Ribeiro, F.R.; Ramalho-Carvalho, J.; Jerónimo, C.; Henrique, R.; Lind, G.E.; Skotheim, R.I.; Lothe, R.A.; Teixeira, M.R. FLI1 is a novel ETS transcription factor involved in gene fusions in prostate cancer. Gen Chrom Cancer 2012, 51, 240-9.
  135. Li, Y.J.; Zhao, X.; Vecchiarelli-Federico, L.M.; Li, Y.; Datti, A.; Cheng, Y.; Ben-David, Y. Drug-mediated inhibition of Fli-1 for the treatment of leukemia. Blood Cancer J 2012, 2, e54.
  136. Cesari, M.; Righi, A.; Cevolani, L.; Palmerini, E.; Vanel, D.; Donati, D.M.; Cammelli, S.; Gambarotti, M.; Ferrari, C.; Paioli, A.; Longhi, A.; Abate, M.E.; Picci, P.; Ferrari, S. Ewing sarcoma in patients over 40 years of age: a prospective analysis of 31 patients treated at a single institution. Tumori 2016, 102, 481-7.
  137. Gaspar, N.; Hawkins, D.S.; Dirksen, U.; Lewis, I.J.; Ferrari, S.; Le Deley, M.C.; Kovar, H.; Grimer, R.; Whelan, J.; Claude, L.; Delattre, O.; Paulussen, M.; Picci, P.; Sundby Hall, K.; van den Berg, H.; Ladenstein, R.; Michon, J.; Hjorth, L.; Judson, I.; Luksch, R.; Bernstein, M.L.; Marec-Bérard, P.; Brennan, B.; Craft, A.W.; Womer, R.B.; Juergens, H.; Oberlin, O. Ewing Sarcoma: Current Management and Future Approaches Through Collaboration. J Clin Oncol 2015, 33, 3036-46.
  138. Le Deley, M.C.; Paulussen, M.; Lewis, I.; Brennan, B.; Ranft, A.; Whelan, J.; Le Teuff, G.; Michon, J.; Ladenstein, R.; Marec-Bérard, P.; van den Berg, H.; Hjorth, L.; Wheatley, K.; Judson, I.; Juergens, H.; Craft, A.; Oberlin, O.; Dirksen, U. Cyclophosphamide compared with ifosfamide in consolidation treatment of standard-risk Ewing sarcoma: results of the randomized noninferiority Euro-EWING99-R1 trial. J Clin Oncol 2014, 32, 2440-8.
  139. Calderón-Montaño, J.M.; Burgos-Morón, E.; Orta, M.L.; Maldonado-Navas, D.; García-Domínguez, I.; López-Lázaro, M. Evaluating the cancer therapeutic potential of cardiac glycosides. Biomed Res Int 2014, 2014, 794930.
  140. Weidenbach, S.; Hou, C.; Chen, J.M.; Tsodikov, O.V.; Rohr, J. Dimerization and DNA recognition rules of mithramycin and its analogues. J Inorg Biochem 2016, 156, 40-7.
  141. Hou, C.; Weidenbach, S.; Cano, K.E.; Wang, Z.; Mitra, P.; Ivanov, D.N.; Rohr, J.; Tsodikov, O.V. Structures of mithramycin analogues bound to DNA and implications for targeting transcription factor FLI1. Nucleic Acids Res 2016, 44, 8990-9004.
  142. Weidenbach, S.; Hou, C.; Chen, J.M.; Tsodikov, O.V.; Rohr, J. Towards understanding the mechanism of action of mithramycin and its analogues: dimerization and DNA binding studies. Planta Med 2016, 81(S 01), S1-S381.
  143. Fuchs, O. Important genes in the pathogenesis of 5q- syndrome and their connection with ribosomal stress and the innate immune system pathway. Leuk Res Treatment 2012, 2012, 179402.
  144. Boultwood, J.; Fidler, C.; Strickson, A.J.; Watkins, F.; Gama, S.; Kearney, L.; Tosi, S.; Kasprzyk, A.; Cheng, J.F.; Jaju, R.J.; Wainscoat, J.S. Narrowing and genomic annotation of the commonly deleted region of the 5q- syndrome. Blood 2002, 99, 4638-41.
  145. Boultwood, J.; Pellagatti, A.; Cattan, H.; Lawrie, C.H.; Giagounidis, A.; Malcovati, L.; Della Porta, M.G.; Jädersten, M.; Killick, S.; Fidler, C.; Cazzola, M.; Hellström-Lindberg, E.; Wainscoat, J.S. Gene expression profiling of CD34+ cells in patients with the 5q-syndrome. Br J Haematol 2007, 139, 578-89.
  146. Kumar, M.S.; Narla, A.; Nonami, A.; Mullally, A.; Dimitrova, N.; Ball, B.; McAuley, J.R.; Poveromo, L.; Kutok, J.L.; Galili, N.; Raza, A.; Attar, E.; Gilliland, D.G.; Jacks, T.; Ebert, B.L. Coordinate loss of a microRNA and protein-coding gene cooperate in the pathogenesis of 5q- syndrome. Blood 2011, 118, 4666-73.
  147. Zhang, J.; Guo, H.; Zhang, H.; Wang, H.; Qian, G.; Fan, X.; Hoffman, A.R.; Hu, J.F.; Ge, S. Putative tumor suppressor miR-145 inhibits colon cancer cell growth by targeting oncogene Friend leukemia virus integration 1 gene. Cancer 2011, 117, 86-95.
  148. Ban, J.; Jug, G.; Mestdagh, P.; Schwentner, R.; Kauer, M.; Aryee, D.N.; Schaefer, K.L.; Nakatani, F.; Scotlandi, K.; Reiter, M.; Strunk, D.; Speleman, F.; Vandesompele, J.; Kovar, H. Hsa-mir-145 is the top EWS-FLI1-repressed microRNA involved in a positive feedback loop in Ewing's sarcoma. Oncogene 2011, 30, 2173-80.
  149. Starczynowski, D.T.; Kuchenbauer, F.; Argiropoulos, B.; Sung, S.; Morin, R.; Muranyi, A.; Hirst, M.; Hogge, D.; Marra, M.; Wells, R.A.; Buckstein, R.; Lam, W.; Humphries, R.K.; Karsan, A. Identification of miR-145 and miR-146a as mediators of the 5q-syndrome phenotype. Nat Med 2010, 16, 49-58.
  150. Imai, T.; Koike, K.; Kubo, T.; Kikuchi, T.; Amano, Y.; Takagi, M.; Okumura, N.; Nakahata, T. Interleukin-6 supports human megakaryocytic proliferation and differentiation in vitro. Blood 1991, 78, 1969-74.
  151. Vo, K.K.; Jarocha, D.J.; Lyde, R.B.; Hayes, V.; Thorn, C.S.; Sullivan, S.K.; French, D.L.; Poncz, M. FLI1 level during megakaryopoiesis affects thrombopoiesis and platelet biology. Blood 2017, 129, 3486-94.
  152. Vecchiarelli-Federico, L.M.; Liu, T.; Yao, Y.; Gao, Y.; Li, Y.; Li, Y.J.; Ben-David, Y. Fli-1 overexpression in erythroleukemic cells promotes erythroid de-differentiation while Spi-1/PU.1 exerts the opposite effect. Int J Oncol 2017, 51, 456-66.
  153. Ramani, N.; Aung, P.P.; Hwu, W.J.; Nagarajan, P.; Tetzlaff, M.T.; Curry, J.L.; Ivan, D.; Prieto, V.G.; Torres-Cabala, C.A. Aberrant expression of Fli-1 in melanoma. J Cutan Pathol 2017, 44, 790-3.
  154. Tsai, H.P.; Tsai, T.H.; Hsieh, Y.J.; Chen, Y.T.; Lee, C.L.; Tsai, Y.C.; She, T.C.; Liu, C.L.; Chai, C.Y.; Kwan, A.L. Overexpression of Fli- 1 in astrocytoma is associated with poor prognosis. Oncotarget 2017, 8, 29174-86.
  155. Li, L.; Song, W.; Yan, X.; Li, A.; Zhang, X.; Li, W.; Wen, X.; Zhou, L.; Yu, D.; Hu, J.F.; Cui, J. Friend leukemia virus integration 1 promotes tumorigenesis of small cell lung cancer cells by activating the miR-17-92 pathway. Oncotarget 2017, 8, 41975-87.
  156. Wang, H.; Ou, Y.; Ou, J.; Jian, Z. Fli-1 promotes metastasis by regulating MMP2 signaling in hepatocellular carcinoma. Mol Med Rep 2018, 17, 1986-92.