scholarly journals CEBPG promotes acute myeloid leukemia progression by enhancing EIF4EBP1

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
You Jiang ◽  
Shui-Yan Wu ◽  
Yan-Ling Chen ◽  
Zi-Mu Zhang ◽  
Yan-Fang Tao ◽  
...  

Abstract Background Acute myeloid leukemia (AML) is a myeloid neoplasm accounts for 7.6% of hematopoietic malignancies. AML is a complex disease, and understanding its pathophysiology is contributing to the improvement in the treatment and prognosis of AML. In this study, we assessed the expression profile and molecular functions of CCAAT enhancer binding protein gamma (CEBPG), a gene implicated in myeloid differentiation and AML progression. Methods shRNA mediated gene interference was used to down-regulate the expression of CEBPG in AML cell lines, and knockdown efficiency was detected by RT-qPCR and western blotting. The effect of knockdown on the growth of AML cell lines was evaluated by CCK-8. Western blotting was used to detect PARP cleavage, and flow cytometry were used to determine the effect of knockdown on apoptosis of AML cells. Genes and pathways affected by knockdown of CEBPG were identified by gene expression analysis using RNA-seq. One of the genes affected by knockdown of CEBPG was Eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1), a known repressor of translation. Knockdown of EIF4EBP1 was used to assess its potential role in AML progression downstream of CEBPG. Results We explored the ChIP-Seq data of AML cell lines and non-AML hematopoietic cells, and found CEBPG was activated through its distal enhancer in AML cell lines. Using the public transcriptomic dataset, the Cancer Cell Line Encyclopedia (CCLE) and western blotting, we also found CEBPG was overexpressed in AML. Moreover, we observed that CEBPG promotes AML cell proliferation by activating EIF4EBP1, thus contributing to the progression of AML. These findings indicate that CEBPG could act as a potential therapeutic target for AML patients. Conclusion In summary, we systematically explored the molecular characteristics of CEBPG in AML and identified CEBPG as a potential therapeutic target for AML patients. Our findings provide novel insights into the pathophysiology of AML and indicate a key role for CEBPG in promoting AML progression.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1648-1648 ◽  
Author(s):  
Yaser Heshmati ◽  
Gözde Turköz ◽  
Aditya Harisankar ◽  
Sten Linnarsson ◽  
Marios Dimitriou ◽  
...  

Abstract Acute myeloid leukemia (AML) is characterized by impaired myeloid differentiation of hematopoietic progenitors, causing uncontrolled proliferation and accumulation of immature myeloid cells in the bone marrow. Rearrangements of the mixed lineage leukemia (MLL) gene are common aberrations in acute leukemia and occur in over 70% in childhood leukemia and 5-10% in leukemia of adults. MLL rearrangements encode a fusion oncogenic H3K4 methytransferase protein, which is sufficient to transform hematopoietic cells and give rise to an aggressive subtype of AML. Leukemia where the MLL fusion oncogene is expressed is characterized by dismal prognosis and 30-60% of 5-years overall survival rate. The current standard treatment for AML is chemotherapy and in certain cases bone marrow transplantation. However, chemotherapy causes severe side effects on normal cells and an increased risk of relapse. Consequently, discovery of novel drug targets with better efficacy and low toxicity are needed to improve treatment of AML. In this study, we aimed to identify genes that are required for growth of AML cells and that encode proteins that potentially could be used as therapeutic targets. To do this, we performed high-throughput RNAi screening covering all annotated human genes and the homologous genes in mice, using barcoded lentiviral-based shRNA vectors. Stable loss-of-function screening was done in three AML cell lines (two human and one murine AML cell lines) as well as in a non-transformed hematopoietic control cell line. The candidate genes were selected based on that shRNA-mediated knockdown caused at least a 5-fold growth inhibition of leukemic cells and that the individual candidates were targeted by multiple shRNAs. The chromodomain Helicase DNA binding protein 4 (CHD4), a chromatin remodeler ATPase, displayed the most significant effect in reduced AML cell proliferation upon inhibition among the overlapping candidate genes in all three AML cell lines. CHD4 is a main subunit of the Nucleosome Remodeling Deacetylase (NuRD) complex and has been associated with epigenetic transcriptional repression. A recent study has shown that inhibition of CHD4 sensitized AML cells to genotoxic drugs by chromatin relaxation, which increases rate of double-stranded break (DSB) in leukemic cells. To verify whether CHD4 is exclusively essential for AML with MLL rearrangements, we inhibited CHD4 expression with two independent shRNAs in various AML cell lines with and without MLL translocations. In vitro monitoring of growth and viability indicated that knockdown of CHD4 efficiently suppressed growth in all tested cell lines, suggesting that CHD4 is required in general for growth of leukemic cells. To test the effect of CHD4 inhibition in normal hematopoiesis, we pursued knockdown of CHD4 and monitored effects in hematopoiesis using colony formation assays of human CD34+ cells. The results demonstrated that CHD4 knockdown had minor effects in colony formation as well as growth and survival of normal hematopoietic cells. Furthermore, to explore whether inhibition of CHD4 can prevent AML tumor growth and disease progression in vivo, we have generated a mouse model for AML. By transplanting AML cells transduced with shRNA against CHD4 into recipient mice, we showed that shRNA-mediated targeting of CHD4 not only significantly prolonged survival of AML transplanted mice but also in some cases completely rescued some mice from development of the disease. Collectively, these data suggested that CHD4 is required for AML maintenance in vivo. Next, to determine whether suppression of CHD4 can inhibit cell growth of different subpopulations and subtypes of AML, we performed loss of function studies of CHD4 on patient-derived AML cells ex vivo. Loss of CHD4 expression significantly decreased the frequency of leukemic initiating cells in different subtypes AML patient samples. In further in vivo studies using a xeno-tranplantation model for AML, we demonstrated that shRNA-mediated inhibition of CHD4 significantly reduced the frequency of leukemic cells in the marrow 6 weeks after transplantation. Taken together our results demonstrated the critical and selective role of CHD4 in propagation of patient-derived AML cells as well as in disease progression in mouse models for AML. We believe that CHD4 represents a novel potential therapeutic target that can be used to battle AML. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 17 (6) ◽  
pp. 1241-1252 ◽  
Author(s):  
Lisa E. Richter ◽  
Yiqian Wang ◽  
Michelle E. Becker ◽  
Rachel A. Coburn ◽  
Jacob T. Williams ◽  
...  

2020 ◽  
Author(s):  
Junyan Gao ◽  
Xinran Chu ◽  
Shan He ◽  
Li Gao ◽  
Hui Hou ◽  
...  

Abstract Background: Glycolysis, a multi-step enzymatic reaction, is considered to be the root of cancer development and progression. The aim of this study is to figure out the glycolytic enzyme, phosphoglycerate kinase 1 (PGK1) whether participate in the progression of acute myeloid leukemia (AML) and its possible mechanisms. Methods: Four datasets (GSE106096, GSE75086, GSE107968 and GSE106748) containing 30 leukemic blast cell samples of AML at diagnosis, 17 leukemic blast cell samples of AML relapse and 3 bone marrow CD34+ cell samples of healthy donors were downloaded from Gene Expression Omnibus (GEO) database and PGK1 was screened out as a potential survival biomarker in AML. Then we did a series of clinical sample verifications and gene set enrichment analysis (GSEA) focusing on PGK1. We further knocked down expression of PGK1 in myelogenous leukemia cell lines and explored its potential effects. Results: PGK1 expression was up-regulated among AML at diagnosis versus healthy control, AML relapse versus AML at diagnosis and AML relapse versus healthy control datasets. Through a serial of bioinformatic analyses (differentially expressed genes [DEGs] selection, function and pathway enrichments and protein-protein interaction [PPI] network establishment), PGK1 was identified as the most meaningful gene in AML progression. Furthermore, the generally high expression of PGK1 was confirmed in AML samples comparing with healthy controls in our single center and the high-expression PGK1 was associated with a comparatively low complete remission (CR) rate, a significantly high 5-year cumulative incidence of relapse (CIR), a poor 5-year event-free survival (EFS) rate, and a poor 5-year overall survival (OS) rate. The GSEA revealed that high-expression PGK1 in AML was associated with many pathways including cytosolic DNA sensing, pentose phosphate, base excision repair and DNA replication. In vitro, the transfected U937 and K562 cells with PGK1 knock-down showed decreased cell viability and increased apoptotic rate. PGK1 inhibition could greatly decrease the half maximal inhibitory concentrations (IC50) of cytarabine (Ara-C) and daunorubicin (DNR) in U937 and K562 cell lines.Conclusions: High-expression PGK1 was associated with poor prognosis in AML. PGK1 may serve to predict the AML progression and provide a novel therapeutic target for AML.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2772-2772
Author(s):  
M. Gorre ◽  
I. Jilani ◽  
R. Chang ◽  
J. Bareng ◽  
H. Chan ◽  
...  

Abstract The mammalian target of rapamycin (mTOR) is a serine/threonine kinase involved in the regulation of cell growth and proliferation. Once activated, mTOR can phosphorylate its downstream targets. One of these targets is the 4E-binding protein 1 (4E-BP1), which is phosphorylated and inactivated by mTOR in response to a growth signal. Phospho-4E-BP1 dissociates from the eukaryotic initiation factor 4E (eIF-4E), a translation initiation factor that subsequently binds the cap structure of 5′ mRNAs and initiates the translation of transcripts encoding genes involved in cell cycle control. Rapamycin and its analogs are immunosuppressant drugs that exert their activity by specific inhibition of mTOR. mTOR inhibition induces cell cycle arrest not only in normal lymphocytes but also in malignant cells. Using flow cytometry, we quantified the levels of 4E-BP1 and phosopho-4E-BP1 in CD34+ and CD3+ cells from bone marrow samples collected from patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). We then measured the antibody binding capacity of 100 CD34+ or CD3+ cells using QuantiBRITE and PE (phycoerythrin)-labeled antibodies in a 1:1 ratio. We demonstrated that CD34+ cells express significantly higher levels of 4E-BP1 (P=0.005) and phosphor-4E-BP1 (P=0.0001) as compared with CD3+ cells in patients with AML (n=49). In contrast, there was no significant difference in the levels of 4E-BP1 or phospho-4E-BP1 between CD34+ and CD3+ cells in patients with MDS (n=15). More importantly, in patients with AML, high expression of 4E-BP1 in CD34+ cells was associated with shorter survival (P=0.003) as well as shorter complete remission duration (CRD) (P=0.03). This association between survival and levels of 4E-BP1 was independent of cytogenetic abnormalities in this group of patients. This data not only suggests that the 4E-BP1 level, as measured in the CD34+ cells, can be an important prognostic indicator in AML but also suggests that 4E-BP1 plays a role in the biology of AML. Furthermore, targeting 4E-BP1 by mTOR inhibitors, or other means of down modulating 4E-BP1 levels, is a rational therapeutic approach in AML. Figure Figure


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2116-2116 ◽  
Author(s):  
Bjoern Hackanson ◽  
Kristi L. Bennett ◽  
Romulo M. Brena ◽  
Jinmai Jiang ◽  
Katie Maharry ◽  
...  

Abstract CCAAT/enhancer binding protein α (C/EBPα) is a master regulator for myeloid differentiation. As a potential tumor suppressor, its loss of function through mutations and posttranslational mechanisms has been thoroughly investigated in acute myeloid leukemia (AML). Recently, we have demonstrated the importance of epigenetic alterations in deregulating C/EBPα expression patterns in lung cancer and head and neck squamous cell carcinoma (HNSCC). In this study we investigated the role of DNA methylation and other epigenetic factors in the regulation of C/EBPα in AML using 94 patient samples and 7 leukemia cell lines. A comprehensive and quantitative DNA methylation analysis of C/EBPα’s large CpG island using MassARRAY (Sequenom©) technology and BioCOBRA identified a distinct and densely methylated upstream promoter region (−1423 bp to −1121 bp in relation to the transcription start site) in 20% (19 of 94) of AML patient samples and in five of seven leukemia cell lines, while the core promoter remained unmethylated. This aberrant DNA methylation pattern was associated with two generally prognostically favorable cytogenetic subgroups: inv(16)(p13q22) and t(15;17)(q22;q21). While DNA methylation levels in normal bone marrow samples (NBM) were very low (median: 0%, range: 0%–5%) when compared to NBM, we observed significantly higher DNA methylation levels in the inv(16) and t(15;17) cytogenetic subgroups, with median DNA methylation levels of 29% (range: 1% to 81%) and 5% (range: 1% to 94%), respectively (p<0.05). However, using semi-quantitative RT-PCR, we could not detect a significant correlation between DNA methylation of C/EBPα and its expression in the inv(16) subgroup. Interestingly, while epigenetic-targeting compounds induced up-regulation of C/EBPα mRNA in vitro, C/EBPα protein was diminished. This unexpected scenario was postulated to be compatible with the presence of a microRNA targeting C/EBPα mRNA. Using a computational microRNA prediction approach and functional studies, we show that C/EBPα mRNA is a target for microRNA-124a. We additionally demonstrate that microRNA-124a is frequently silenced by epigenetic mechanisms in AML, becomes upregulated following epigenetic treatment and targets the C/EBPα 3′-untranslated region (3′UTR). In this way, C/EBPα protein expression is reduced in a posttranscriptional manner. Our results indicate that epigenetic alterations of C/EBPα are a frequent event in AML and that epigenetic treatment can result in down-regulation of a key hematopoietic transcription factor.


Leukemia ◽  
2019 ◽  
Vol 33 (12) ◽  
pp. 2830-2841 ◽  
Author(s):  
Linus Angenendt ◽  
Eike Bormann ◽  
Caroline Pabst ◽  
Vijay Alla ◽  
Dennis Görlich ◽  
...  

Oncotarget ◽  
2013 ◽  
Vol 4 (9) ◽  
pp. 1438-1448 ◽  
Author(s):  
Audrey Astori ◽  
Hanne Fredly ◽  
Thomas Aquinas Aloysius ◽  
Lars Bullinger ◽  
Véronique Mansat-De Mas ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document