scholarly journals Homozygous Loss of a Critical CTCF Recognition Sequence in the HOXA gene Cluster Eliminates CTCF Binding but Does Not Appreciably Reduce HOXA9 expression in NPM1 Mutated AML Cells

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2613-2613
Author(s):  
Reza Ghasemi ◽  
Struthers B Heidi ◽  
David H Spencer

Abstract HOX genes encode homeodomain transcription factors that play a central role in hematopoietic stem/progenitor cell (HSPCs) self-renewal and the development of acute myeloid leukemia (AML). HOXA and HOXB genes display canonical expression patterns in primary AML samples that correlate with specific recurrent mutations, including expression of only HOXA genes in AMLs with MLL rearrangements and both HOXA and HOXB gene expression in normal karyotype AMLs with mutations in NPM1 (i.e., the NPMc mutation). Although regulation of HOXA genes in MLL-rearranged AML cells has been studied extensively, the regulatory mechanisms that govern HOXA expression in other AML types remains unclear. The chromatin organizing factor CTCF contributes to HOXA gene regulation by controlling chromatin looping and three-dimensional structure in a variety of cell types, including AML cells, and a recent study demonstrated that expression of HOXA9 in MLL-rearranged AML cell lines is dependent on a critical CTCF-binding site between HOXA7 and HOXA9 (CBS7/9). However, whether this site is required for HOXA gene expression in AMLs with NPMc remains unknown. The AML cell line OCI-AML3 contains the NPMc mutation (in addition to a DNMT3AR882C mutant allele) and expresses both HOXA and HOXB genes. We therefore used this cell line to investigate the role of CTCF in regulating HOXA expression in AML cells with this canonical stem cell expression pattern. ChIP-seq for CTCF via 'ChIPmentation' identified eight CTCF binding sites across the HOXA cluster in OCI-AML3 cells, including CBS7/9, which is consistent with previous data in AML cells. These sites were identical to those identified in MLL-rearranged AML cell lines, suggesting that CTCF binding is conserved between these AML types. Similar to previous work, ChIP-seq for histone methylation marks associated with active (H3K4 trimethylation) and repressed (H3K27 trimeylation) chromatin demonstrated distinct chromatin domains at the HOXA locus on either side of CBS7/9, indicating that CTCF acts as a barrier between these domains. Using a computational prediction tool for CTCF binding, we identified two potential CTCF-binding sequences at the CBS7/9 locus, including one bidirectional major site (CBS7/9-A; 20 bp) with a high binding score, separated by only 6 bp from a secondary unidirectional site (CBS7/9-B; 9 bp). To define the precise sequence important for CTCF binding in OCI-AML3 cells, we used CRISPR-Cas9 to generate a library of mutant cells with deletions of different sizes at the CBS7/9 locus, and then performed ChIP-seq on the mutant cells to measure CTCF binding. We found that mutants with deletions at CBS7/9-A were depleted relative to wild-type cells. Further analysis demonstrated that deletions as small as 10 bp affecting the CBS7/9-A site were sufficient to reduce CTCF binding to background levels. CRISPR-Cas9 generated deletions in the MLL-rearranged THP-1 cell line that spanned the same sequence also eliminated CTCF ChIP-seq signal, indicating that it is equally important for CTCF binding in MLL-rearranged and NPM1-mutant AML cells. Our collection of mutant OCI-AML3 cell lines included five with biallelic deletions of the CBS7/9-A site, which completely eliminated all CTCF binding at this locus, but did not affect cellular proliferation or survival. Surprisingly, preliminary analysis of expression in the five homozygous mutant OCI-AML3 cells demonstrated that HOXA9 gene expression by RT-qPCR remained largely intact compared to wild-type OCI-AML3 cells, and was not statistically different from three OCI-AML3 cell lines with a heterozygous deletion of the CBS7/9 site. In summary, we have mapped the precise recognition sequence for a critical CTCF binding site in the HOXA locus in human AML cells. Deletion of both alleles of this regulatory sequence in NPM1-mutated OCI-AML3 cells does not affect cell viability and appears to have only modest effects on expression of the adjacent HOXA9 gene. This is in contrast to a recent report suggesting that homozygous deletion of this CTCF binding site is critical for maintaining HOXA gene expression and may be required for cell viability in MLL-rearranged AML cell lines. These differences suggest that maintenance of HOXA expression in NPM1-mutated vs. MLL-rearranged AML cells may occur through distinct regulatory mechanisms. Disclosures No relevant conflicts of interest to declare.

2019 ◽  
Vol 16 (11) ◽  
pp. 1194-1201 ◽  
Author(s):  
Farhad Saravani ◽  
Ebrahim Saeedian Moghadam ◽  
Hafezeh Salehabadi ◽  
Seyednasser Ostad ◽  
Morteza Pirali Hamedani ◽  
...  

Background: The role of microtubules in cell division and signaling, intercellular transport, and mitosis has been well known. Hence, they have been targeted for several anti-cancer drugs. Methods: A series of 3-(alkylthio)-5,6-diphenyl-1,2,4-triazines were prepared and evaluated for their cytotoxic activities in vitro against three human cancer cell lines; human colon carcinoma cells HT-29, human breast adenocarcinoma cell line MCF-7, human Caucasian gastric adenocarcinoma cell line AGS as well as fibroblast cell line NIH-3T3 by MTT assay. Docking simulation was performed to insert these compounds into the crystal structure of tubulin at the colchicine binding site to determine a probable binding model. Compound 5d as the most active compound was selected for studying of microtubule disruption. Results: Compound 5d showed potent cytotoxic activity against all cell lines. The molecular modeling study revealed that some derivatives of triazine strongly bind to colchicine binding site. The tubulin polymerization assay kit showed that the cytotoxic activity of 5d may be related to inhibition of tubulin polymerization. Conclusion: The cytotoxicity and molecular modeling study of the synthesized compounds with their inhibition activity in tubulin polymerization demonstrate the potential of triazine derivatives for development of new anti-cancer agents.


Animals ◽  
2021 ◽  
Vol 11 (2) ◽  
pp. 502
Author(s):  
Filipe Almeida ◽  
Andreia Gameiro ◽  
Jorge Correia ◽  
Fernando Ferreira

Feline mammary carcinoma (FMC) is the third most common type of neoplasia in cats, sharing similar epidemiological features with human breast cancer. In humans, histone deacetylases (HDACs) play an important role in the regulation of gene expression, with HDAC inhibitors (HDACis) disrupting gene expression and leading to cell death. In parallel, microtubules inhibitors (MTIs) interfere with the polymerization of microtubules, leading to cell cycle arrest and apoptosis. Although HDACis and MTIs are used in human cancer patients, in cats, data is scarce. In this study, we evaluated the antitumor properties of six HDACis (CI-994, panobinostat, SAHA, SBHA, scriptaid, and trichostatin A) and four MTIs (colchicine, nocodazole, paclitaxel, and vinblastine) using three FMC cell lines (CAT-MT, FMCp, and FMCm), and compared with the human breast cancer cell line (SK-BR-3). HDACis and MTIs exhibited dose-dependent antitumor effects in FMC cell lines, and for all inhibitors, the IC50 values were determined, with one feline cell line showing reduced susceptibility (FMCm). Immunoblot analysis confirmed an increase in the acetylation status of core histone protein HDAC3 and flow cytometry showed that HDACis and MTIs lead to cellular apoptosis. Overall, our study uncovers HDACis and MTIs as promising anti-cancer agents to treat FMCs.


2019 ◽  
Vol 4 ◽  
pp. 150 ◽  
Author(s):  
Antje K. Grotz ◽  
Fernando Abaitua ◽  
Elena Navarro-Guerrero ◽  
Benoit Hastoy ◽  
Daniel Ebner ◽  
...  

Type 2 diabetes (T2D) is a global pandemic with a strong genetic component, but most causal genes influencing the disease risk remain unknown. It is clear, however, that the pancreatic beta cell is central to T2D pathogenesis. In vitro gene-knockout (KO) models to study T2D risk genes have so far focused on rodent beta cells. However, there are important structural and functional differences between rodent and human beta cell lines. With that in mind, we have developed a robust pipeline to create a stable CRISPR/Cas9 KO in an authentic human beta cell line (EndoC-βH1). The KO pipeline consists of a dual lentiviral sgRNA strategy and we targeted three genes (INS, IDE, PAM) as a proof of concept. We achieved a significant reduction in mRNA levels and complete protein depletion of all target genes. Using this dual sgRNA strategy, up to 94 kb DNA were cut out of the target genes and the editing efficiency of each sgRNA exceeded >87.5%. Sequencing of off-targets showed no unspecific editing. Most importantly, the pipeline did not affect the glucose-responsive insulin secretion of the cells. Interestingly, comparison of KO cell lines for NEUROD1 and SLC30A8 with siRNA-mediated knockdown (KD) approaches demonstrate phenotypic differences. NEUROD1-KO cells were not viable and displayed elevated markers for ER stress and apoptosis. NEUROD1-KD, however, only had a modest elevation, by 34%, in the pro-apoptotic transcription factor CHOP and a gene expression profile indicative of chronic ER stress without evidence of elevated cell death. On the other hand, SLC30A8-KO cells demonstrated no reduction in KATP channel gene expression in contrast to siRNA silencing. Overall, this strategy to efficiently create stable KO in the human beta cell line EndoC-βH1 will allow for a better understanding of genes involved in beta cell dysfunction, their underlying functional mechanisms and T2D pathogenesis.


2021 ◽  
Author(s):  
Pamela Himadewi ◽  
Xue Qing David Wang ◽  
Fan Feng ◽  
Haley Gore ◽  
Yushuai Liu ◽  
...  

Mutations in the adult β-globin gene can lead to a variety of hemoglobinopathies, including sickle cell disease and β-thalassemia. An increase in fetal hemoglobin expression throughout adulthood, a condition named Hereditary Persistence of Fetal Hemoglobin (HPFH), has been found to ameliorate hemoglobinopathies. Deletional HPFH occurs through the excision of a significant portion of the 3 prime end of the β-globin locus, including a CTCF binding site termed 3'HS1. Here, we show that the deletion of this CTCF site alone induces fetal hemoglobin expression in both adult CD34+ hematopoietic stem and progenitor cells and HUDEP-2 erythroid progenitor cells. This induction is driven by the ectopic access of a previously postulated distal enhancer located in the OR52A1 gene downstream of the locus, which can also be insulated by the inversion of the 3'HS1 CTCF site. This suggests that genetic editing of this binding site can have therapeutic implications to treat hemoglobinopathies.


2018 ◽  
Vol 500 (4) ◽  
pp. 852-859 ◽  
Author(s):  
Feng Wang ◽  
Zhongqiong Tang ◽  
Honglian Shao ◽  
Jun Guo ◽  
Tao Tan ◽  
...  

1993 ◽  
Vol 13 (12) ◽  
pp. 7612-7624
Author(s):  
E M Klenova ◽  
R H Nicolas ◽  
H F Paterson ◽  
A F Carne ◽  
C M Heath ◽  
...  

A novel sequence-specific DNA-binding protein, CTCF, which interacts with the chicken c-myc gene promoter, has been identified and partially characterized (V. V. Lobanenkov, R. H. Nicolas, V. V. Adler, H. Paterson, E. M. Klenova, A. V. Polotskaja, and G. H. Goodwin, Oncogene 5:1743-1753, 1990). In order to test directly whether binding of CTCF to one specific DNA region of the c-myc promoter is important for chicken c-myc transcription, we have determined which nucleotides within this GC-rich region are responsible for recognition of overlapping sites by CTCF and Sp1-like proteins. Using missing-contact analysis of all four nucleotides in both DNA strands and homogeneous CTCF protein purified by sequence-specific chromatography, we have identified three sets of nucleotides which contact either CTCF or two Sp1-like proteins binding within the same DNA region. Specific mutations of 3 of 15 purines required for CTCF binding were designed to eliminate binding of CTCF without altering the binding of other proteins. Electrophoretic mobility shift assay of nuclear extracts showed that the mutant DNA sequence did not bind CTCF but did bind two Sp1-like proteins. When introduced into a 3.3-kbp-long 5'-flanking noncoding c-myc sequence fused to a reporter CAT gene, the same mutation of the CTCF binding site resulted in 10- and 3-fold reductions, respectively, of transcription in two different (erythroid and myeloid) stably transfected chicken cell lines. Isolation and analysis of the CTCF cDNA encoding an 82-kDa form of CTCF protein shows that DNA-binding domain of CTCF is composed of 11 Zn fingers: 10 are of C2H2 class, and 1 is of C2HC class. CTCF was found to be abundant and conserved in cells of vertebrate species. We detected six major nuclear forms of CTCF protein differentially expressed in different chicken cell lines and tissues. We conclude that isoforms of 11-Zn-finger factor CTCF which are present in chicken hematopoietic HD3 and BM2 cells can act as a positive regulator of the chicken c-myc gene transcription. Possible functions of other CTCF forms are discussed.


BMC Cancer ◽  
2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Pushpaja Dodla ◽  
Vanitha Bhoopalan ◽  
Sok Kean Khoo ◽  
Cindy Miranti ◽  
Suganthi Sridhar

Abstract Background Tetraspanin CD82 is a tumor metastasis suppressor that is known to down regulate in various metastatic cancers. However, the exact mechanism by which CD82 prevents cancer metastasis is unclear. This study aims to identify genes that are regulated by CD82 in human prostate cell lines. Methods We used whole human genome microarray to obtain gene expression profiles in a normal prostate epithelial cell line that expressed CD82 (PrEC-31) and a metastatic prostate cell line that does not express CD82 (PC3). Then, siRNA silencing was used to knock down CD82 expression in PrEC-31 while CD82 was re-expressed in PC3 to acquire differentially-expressed genes in the respective cell line. Results Differentially-expressed genes with a P < 0.05 were identified in 3 data sets: PrEC-31 (+CD82) vs PrEC-31(−CD82), PC3–57 (+CD82) vs. PC3-5 V (−CD82), and PC3–29 (+CD82) vs. PC3-5 V (−CD82). Top 25 gene lists did not show overlap within the data sets, except (CALB1) the calcium binding protein calbindin 1 which was significantly up-regulated (2.8 log fold change) in PrEC-31 and PC3–29 cells that expressed CD82. Other most significantly up-regulated genes included serine peptidase inhibitor kazal type 1 (SPINK1) and polypeptide N-acetyl galactosaminyl transferase 14 (GALNT14) and most down-regulated genes included C-X-C motif chemokine ligand 14 (CXCL14), urotensin 2 (UTS2D), and fibroblast growth factor 13 (FGF13). Pathways related with cell proliferation and angiogenesis, migration and invasion, cell death, cell cycle, signal transduction, and metabolism were highly enriched in cells that lack CD82 expression. Expression of two mutually inclusive genes in top 100 gene lists of all data sets, runt-related transcription factor (RUNX3) and trefoil factor 3 (TFF3), could be validated with qRT-PCR. Conclusion Identification of genes and pathways regulated by CD82 in this study may provide additional insights into the role that CD82 plays in prostate tumor progression and metastasis, as well as identify potential targets for therapeutic intervention.


PLoS ONE ◽  
2016 ◽  
Vol 11 (2) ◽  
pp. e0148892 ◽  
Author(s):  
Claudia Stein ◽  
René Reiner Nötzold ◽  
Stefanie Riedl ◽  
Caroline Bouchard ◽  
Uta-Maria Bauer

2019 ◽  
Vol 50 (2) ◽  
pp. 184-196.e4 ◽  
Author(s):  
Claudia Gentile ◽  
Soizik Berlivet ◽  
Alexandre Mayran ◽  
Denis Paquette ◽  
Fanny Guerard-Millet ◽  
...  

2010 ◽  
Vol 55 (6) ◽  
pp. 1072-1082 ◽  
Author(s):  
Julia Starkova ◽  
Blanka Zamostna ◽  
Ester Mejstrikova ◽  
Roman Krejci ◽  
Harry A. Drabkin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document