New insights from Whole Genome Sequencing: BCLAF1 deletion as a structural variant that predisposes cells towards cellular transformation

  • Authors:
    • Lamech M. Mwapagha
    • Vimbaishe Chibanga
    • Hendrina Shipanga
    • M. Iqbal Parker
  • View Affiliations

  • Published online on: September 6, 2021     https://doi.org/10.3892/or.2021.8180
  • Article Number: 229
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer arises from a multi‑step cellular transformation process where some mutations may be inherited, while others are acquired during the process of malignant transformation. Aberrations in the BCL2 associated transcription factor 1 (BCLAF1) gene have previously been identified in patients with cancer and the aim of the present study was to identify structural variants (SVs) and the effects of BCLAF1 gene silencing on cell transformation. Whole‑genome sequencing was performed on DNA isolated from tumour biopsies with a histologically confirmed diagnosis of oesophageal squamous cell carcinoma (OSCC). Paired‑end sequencing was performed on the Illumina HiSeq2000, with 300 bp reads. Reads were aligned to the Homo sapiens reference genome (NCBI37) using ELAND and CASAVA software. SVs reported from the alignment were collated with gene loci, using the variant effect predictor of Ensembl. The affected genes were subsequently cross‑checked against the Genetic Association Database for disease and cancer associations. BCLAF1 deletion was identified as a noteworthy SV that could be associated with OSCC. Transient small interfering RNA‑mediated knockdown of BCLAF1 resulted in the altered expression of several downstream genes, including downregulation of the proapoptotic genes Caspase‑3 and BAX and the DNA damage repair genes exonuclease 1, ATR‑interacting protein and transcription regulator protein BACH1. BCLAF1 deficiency also attenuated P53 gene expression. Inhibition of BCLAF1 expression also resulted in increased colony formation. These results provide evidence that the abrogation of BCLAF1 expression results in the dysregulation of several cancer signalling pathways and abnormal cell proliferation.

Introduction

Cancers arise as a result of genetic changes in the genome such that cancer genomes contain inherited and sporadic nucleotide sequence changes. In addition to hereditary predisposition, direct mutational events and/or epigenetic modifications initiate changes in signalling pathways that can result in cellular transformation (1). At a molecular level, cancer development is a multistep process with alterations in the expression of three gene classes; namely oncogenes, tumour suppressor genes and stability genes (2,3). Overexpression of oncogenes and loss of tumour suppressor gene functions result in constitutive activation of pathways under conditions in which the wild-type gene is not active (4,5). By contrast, mutations in stability genes increase genetic instability, resulting in inefficient DNA repair mechanisms and cell cycle control (6,7).

The DNA sequence changes may include insertions and deletions (InDels), gene duplication, genomic rearrangements, loss of heterozygosity in somatic cells or inherited mutations (8). Given that InDels are more likely to be driver gene mutations, a substantial number of InDels in the coding DNA sequence is not surprising, since InDels are often deleterious as evidenced by their frequent association with human disease (9,10). InDels have also been shown to be components of gene and pseudogene evolution that are important in the long-term evolution of genome size (11). Thus, the abundance of InDels, which have a comparatively greater influence than single nucleotide polymorphisms (SNPs), is consistent with the abnormal function and devastating characteristics of cancer (10).

Our previous Whole Genome Sequencing (WGS) study in patients with oesophageal squamous cell carcinoma (OSCC) revealed large scale deletions of the BCL2 associated transcription factor 1 (BCLAF1) gene (12). BCLAF1 encodes a multifunctional protein with four splice variants that perform a wide range of seemingly unrelated functions (13). Some of these functions include tumour suppression (through promoting apoptosis, senescence, DNA repair and autophagy), embryonic development, T cell maturation and mRNA processing, as illustrated in Fig. 1.

BCLAF1 was originally identified in a yeast protein screen for E1B 19K interacting factors. E1B 19K is a homolog of anti-apoptotic BCL-2 family members and inhibits cell death signal transduction cascades activated by adenoviral proteins (13,14). Overexpression of BCLAF1 induces apoptosis, suggesting that BCLAF1 is a pro-apoptotic factor with potential tumour suppressor activity (13). However, the role of BCLAF1 as a tumour suppressor gene that promotes apoptosis and autophagy is debatable due to conflicting lines of evidence (15,16). Despite this, BCLAF1 mutations and loss of gene expression have been implicated in a range of cancers, including oesophageal cancer, non-Hodgkin's lymphoma, Burkitt's lymphoma, colon cancer, bladder cancer and rectal cancer (17). Understanding of the genomic abnormalities in OSCC is very limited, hence the need to identify genomic abnormalities underlying OSCC and to elucidate the molecular basis of the disease in order to guide the development of effective targeted therapies (18,19).

Although, BCLAF1 deletions have been observed in a number of tumour types, very little is known concerning BCLAF1 expression patterns in human cancers and the effects of its deficiency on gene expression and tumourigenesis (13,20). The present study aimed to identify structural variants (SVs), primarily large InDels, and the downstream molecular mechanisms involved in cellular transformation following the loss of BCLAF1 gene expression in a cultured cell line model.

Materials and methods

Sample collection

Patients were recruited at Groote Schuur Hospital, University of Cape Town (Cape Town, South Africa) in the gastrointestinal clinic between January 2010 and December 2021. Written informed consent was obtained from all patients before recruitment into the study. Patient ages ranged between 40 and 80 years, and the ratio of female to male patients was 60:40. Tumours were identified by endoscopic examination and biopsy after staining with acetic iodide solution. Normal biopsies were taken at least 10 cm from the tumour. Patients who had previously received any form of chemo- or radiotherapy were excluded from the study (21). Ethics approval was obtained from the University of Cape Town/Groote Schuur Hospital Human Research Ethics Committee (Cape Town, South Africa; approval no. HEC040/2005), and all methods were performed in accordance with the Declaration of Helsinki.

DNA extraction from tissue biopsies

Frozen tissue was thawed on ice and placed into a sterile 1.5 ml microcentrifuge tube containing 0.5 ml QIAzol Lysis Reagent™ (Qiagen, Inc.). The tip of a disposable probe was then placed in the tube and homogenised at full speed until the tissue lysate was uniformly homogeneous (1–5 min). An additional 0.5 ml QIAzol Lysis Reagent was added and mixed by vortexing for 30 sec, after which the samples were incubated for 5 min at room temperature (20–25°C) to permit the complete dissociation of nucleoprotein complexes. A total of 0.2 ml chloroform per 1 ml QIAzol Lysis Reagent was then added to each tube, capped and shaken vigorously by hand for 15 sec and incubated at room temperature for 2–3 min. The samples were centrifuged at 12,000 × g for 15 min at 4°C to separate the mixture into three phases: A colourless upper aqueous phase containing RNA, a white middle interphase containing DNA and a lower organic phenol phase containing protein. The upper aqueous phase was transferred to a new tube and stored at 4°C for later RNA extraction. A total of 0.3 ml 100% ethanol was then added to the interphase and mixed by inversion followed by incubation on ice for 30 min. The samples were then centrifuged at 14,000 × g for 5 min at 4°C to pellet the DNA, the ethanol was discarded and the pellet was rinsed in 1 ml 75% ethanol. The samples were centrifuged at 14,000 × g for 5 min at 4°C, and the supernatant was carefully removed and the DNA pellet was air dried for 5–15 min. The DNA pellet was dissolved in 200 µl 10 mM Tris/1 mM EDTA and quantitated on a NanoDrop 2000/2000c Spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.). Samples were stored at −20°C until required.

Variant calling

Four OSCC DNA biopsy samples were submitted to the Centre for Proteomic and Genomic Research (Cape Town, South Africa) for paired-end sequencing on the Illumina HiSeq2000 (Illumina, Inc.) using TruSeq SBS chemistry v3. or WGS. Quality control (QC) measures were carried out to validate the integrity of the samples for WGS with the Quant-iT™ PicoGreen™ dsDNA kit (cat. no. P7589; Invitrogen; Thermo Fisher Scientific, Inc.). Paired-end sequencing was performed on the Illumina HiSeq2000, with 2×100 bp reads. Reads were aligned to the NCBI37 Homo sapiens reference genome using ELAND and CASAVA version 1.8.2 software (22). Since matched normal samples were not included, variants with global allele frequency >10% in either the Exome Variant Server or 1000 Genomes Project were filtered out to reduce background noise. SVs reported from the alignment were collated with gene loci, (upstream, downstream and overlapping) using the Variant Effect Predictor of Ensembl (23). The samples were genotyped on an Illumina HumanOmni2.5–8 array (Illumina, Inc.) and the affected genes were subsequently cross-checked against the Genetic Association Database for disease and cancer associations (24). The deleted genes common to all the four tumour samples were described in HUGO Gene Nomenclature Committee (HGNC) (25) and Medical Subject Headings (MeSH) gene and disease terms, respectively (Courtesy of the U.S. National Library of Medicine).

WGS

DNA isolated from tumour biopsies obtained from patients with oesophageal squamous cell carcinoma (OSCC) were sent to the Centre for Proteomic and Genomic Research (CPGR), Cape Town, South Africa for WGS. The samples were taken from the Black and Mixed Ancestry population groups since these are the population groups most affected by OSCC in South Africa. The Quant-iT™ PicoGreen™ dsDNA kit (cat. no. P7589; Invitrogen; Thermo Fisher Scientific, Inc.) was used to determine DNA integrity and 10 nM DNA sample was used for sequencing. Paired-end sequencing was performed on the Illumina HiSeq2000, with 2×100 bp reads. PCR-free paired end libraries were manually generated from 500 ng 1 µg genomic DNA using a modified version of TruSeq DNA v2 Sample Preparation kits (cat. no. FC-121-2001; Illumina, Inc.). Fragmentation was performed using Covaris LE220 (Covaris, Inc.). After fragmentation and end repair, libraries were manually size-selected using bead-based size selection targeting 300 bp inserts (Agencourt AMPure XP A63881; Thermo Fisher Scientific, Inc.). Following size selection, libraries were A-tailed, and ligated before proceeding to library QC. Final Libraries were quality checked on a Caliper GX (PerkinElmer, Inc.) and quantified by reverse transcription-quantitative (RT-q)PCR on the Roche LightCycler 480 (Roche Diagnostics). Samples were clustered on three lanes with the Illumina v3 flowcells (Illumina, Inc.) using the Illumina cBot (Illumina, Inc.) with the TruSeq Paired End Cluster Kit (v3). Flowcells were sequenced as 100 bp-end reads on the Illumina HiSeq2000 using TruSeq SBS chemistry v3. DNA sequences were compared to the human reference genome (NCBI37), to perform a preliminary screen for InDels.

Maintenance of cells in culture

CT1 cells were obtained from Dr Namba and cultured as previously described (26) were cultured in complete medium [Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich; Merck KGaA) supplemented with 10% heat inactivated foetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA) and 100 U/ml penicillin and 100 mg/ml streptomycin] at 37°C in a humidified incubator with 5% CO2. Every 2 days, the medium was changed and the cells were sub-cultured when they reached 90% confluency, by detaching the cells with 0.05% trypsin-EDTA. Once detached, the trypsin-EDTA was inactivated by the addition of 5 ml complete DMEM. Cells were collected by centrifugation at 3,000 × g at 4°C for 5 min, suspended in complete DMEM and re-plated at a ratio of 1:3. Cells were regularly checked for mycoplasma contamination as previously described (27).

Small interfering (si)RNA transfection

Transient transfection with siRNA was used to knock down the expression of BCLAF1. Briefly, 2×105 cells were plated in a six-well dish (35 mm per well) with complete medium and incubated overnight at 37°C, in a humidified 5% CO2 incubator. The following day, the cells were washed once with 2 ml DMEM, then transfected using TransFectin™ Lipid Reagent (Bio-Rad Laboratories, Inc.), according to the manufacturer's instructions. Two transfection mixtures were prepared separately for each siRNA; the first mixture contained 1.5 µl (75 pmoles) siRNA in 50 µl DMEM and the second mixture had 10 µl TransFectin Lipid Reagent in 45 µl DMEM. After combining the mixtures and incubating them at room temperature for 45 min, the combined mixture was pipetted drop-wise into each well. The plates were incubated at 37°C in a humidified atmosphere of 5% CO2 for 5–7 h, after which, 1.750 ml DMEM containing 10% FBS, 50 U/ml penicillin and 50 µg/ml streptomycin was added and the cells incubated for an additional 48 h. Untransfected cells and a non-targeting siRNA were used as controls. Each experiment was conducted in triplicate. The siRNA duplex sequences are listed in Table SI.

RNA preparation and RT-qPCR

Total RNA was extracted as previously described using TRIzol® reagent (Thermo Fisher Scientific, Inc.) (27). cDNA was prepared from 2 µg total RNA using the ImProm-II™ Reverse Transcription System (Promega Corporation), according to the manufacturers' instructions. RT-qPCR assays were performed using SYBR® FAST qPCR kit (Roche Diagnostics) in a reaction volume of 20 µl. All amplifications were performed as follows: Initial denaturation at 95°C for 5 min, followed by 40 cycles at 95°C for 30 sec, 60°C for 20 sec and 72°C for 5 sec. Analysed genes were amplified in triplicate using the LightCycler® 480II (Roche Diagnostics) and normalized to GAPDH expression in the same sample using the 2−ΔΔCq method (28). A list of the primers used and their annealing temperatures are shown in Table SII.

Western blot analysis

Control cells and cells transfected with siRNA were lysed in RIPA buffer [10 mM Tris-HCl pH 7.6, 10 mM NaCl, 3 mM MgCl2 and 1% (v/v) Nonidet P-40 containing 50 µg/ml each of pepstatin, leupeptin and aprotinin]. Total protein concentration was determined using the Bradford assay (Bio-Rad Laboratories, Inc.). Total cell lysates (50 µg protein) were separated by electrophoresis on 12% SDS-PAGE gels under reducing conditions (50 mM β-mercaptoethanol). After electrophoresis, proteins were transferred to nitrocellulose membranes and blocked in 5% fat-free milk in Tris-buffered saline (TBS) containing 0.1% Tween-20 with shaking for 1 h at room temperature. The membranes were incubated overnight at 4°C with one of the following primary antibodies (1:1,000; all purchased from Abcam): Rabbit polyclonal anti-BCL2 associated transcription factor 1 (BTF; cat. no. ab107177), rabbit polyclonal anti-caspase-3 (phospho S150; cat. no. ab59425), rabbit polyclonal anti-Caspase-9 (cat. no. ab209495), rabbit monoclonal anti-BAX (cat. no. ab182733), rabbit polyclonal anti-Bcl-2 (cat. no. ab115807), rabbit polyclonal anti-Exonuclease 1 (EXO1; cat. no. ab106303), rabbit polyclonal anti-γ H2AX (phospho S139; cat. no. ab11174), rabbit polyclonal anti-p53 (cat. no. ab17990) and rabbit polyclonal anti-p21 (cat. no. ab219811). After several washes in TBS with 0.1% Tween-20 buffer, the nitrocellulose membranes were incubated by shaking at room temperature for 1 h with the required horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary antibody (1:3,000; cat. no.1706515; Bio-Rad Laboratories, Inc.) and detected using LumiGLO® substrate (KPL, Inc.).

Soft agar assay

The anchorage dependence and colony formation ability of the cells was assessed using a soft agar assay. Cells (2×105) were transfected with 75 ρmoles siRNA. Following transfection, cells were washed with 2 ml PBS and trypsinised with 500 µl 0.5% trypsin-EDTA. Then, 500 µl DMEM was added to inactivate trypsin. The cells were centrifuged at 3,000 × g at 4°C for 5 min and suspended in 1 ml DMEM. A base agar was prepared by mixing 1% agarose gel at 40°C with 2X DMEM (20% FBS and 2% P/S). Following which, 1 ml base agar was added to each well of a 6-well plate and gently swirled to cover the surface. The agar was allowed to set for 30 min at room temperature. Top agar was prepared by dissolving 0.7% agarose in a microwave, then cooling it to 40°C. A suspension of 2,500 cells in 0.1 ml was mixed with 3 ml 0.7% agar and 3 ml 2X DMEM. Then, 1 ml cell suspension was pipetted onto the base agar in triplicate. The cells were incubated at 37°C in a humidified 5% CO2 incubator for 21 days. The cells were fed twice a week with 1 ml complete medium. After 21 days, 0.5 ml 0.005% crystal violet (Sigma-Aldrich; Merck KGaA) was added for 1 h and pink stained colonies were counted in 15 different fields using an Olympus CKY41 light microscope (Olympus Corporation) at ×100 magnification. ImageJ (version 1.53i) software (National Institutes of Health) was used for semi-quantification of the number of colonies >150 µm in diameter.

Statistical analysis

The data are presented as the mean ± SEM. All experiments were performed in triplicate unless stated otherwise. Statistical analysis of the data was performed using GraphPad Prism 5 (GraphPad Software, Inc.). A two tailed unpaired Student's t-test was performed. P<0.05 was considered to indicate a statistically significant difference.

Results

SVs in OSCC

WGS data generated 4.27 billion reads of 100 bp covering at least 95% of the human reference genome. QC indicators, such as the duplication rate, coverage and mean depth of coverage, indicated that the experiments were successful in acquiring high-quality sequence reads (Table SIII). The minimum depth of coverage was 33.7, and the library preparation was successful across all samples without being compromised by PCR amplification bias after having obtained a 98% unique read proportion (Non-N reference) to the human reference genome (NCBI37).

The samples were genotyped on an Illumina HumanOmni2.5–8 array, and had a high quality score with a percentage base calling of >Q30 indicating a smaller probability of error. Fig. S1A shows that sample B381 had 89.0% of filtered and aligned reads with a mean depth of coverage of 40.7, while Fig. S1B shows that sample B450 had 90.7% of filtered and aligned reads with a mean depth of coverage of 35.0. Sample M456 had 89.3% of filtered and aligned reads with a mean depth of coverage of 33.6 (Fig. S1C). Whereas, sample M478 had 88.3% of filtered and aligned reads with a mean depth of coverage of 34.2 (Fig. S1D). Thus, the sequencing phase of this experiment achieved high quality reads that allowed for greater confidence in the variant calling procedure and results.

Alignment and variant calling

Illumina's proprietary alignment algorithm ELAND and CASAVA 1.8.2 software were used to align the reads to the NCBI37 human reference genome (22). As matched normal samples were not included, variants with global allele frequency >10% in either the Exome Variant Server or 1000 Genomes Project were filtered out to reduce background noise (29,30). SVs reported from the alignment were collated with gene loci (upstream, downstream and overlapping) using the variant effect predictor of Ensembl (23). The affected genes were subsequently cross-checked against the Genetic Association Database for disease and cancer associations (24). A large number of SNP effects from all four samples were observed (Table I), having 98% mean array agreement between the samples and the reference genome NCBI37 of which 38% were associated with Genes.

Table I.

Summary of SNP consequences across all four tumour samples.

Table I.

Summary of SNP consequences across all four tumour samples.

Tumour samples

Variant effectsB381B450B456M478
Array agreement, %98.599.297.699.2
SNP total, n4,919,4465,334,1734,185,8085,018,129
Genes, %38.938.938.839.1
Exons, %1.61.61.61.6
Ti/Tv, n2.12.12.12.1
Novel, n591,283617,474476,394656,181
Ti/Tv novel, n1.51.61.41.7
Synonymous, n14,26615,31011,75014,562
Missense, n13,34914,55611,24313,660
Nonsense, n140144118135
InDel total, n1,308,5221,358,2181,153,9011,303,928
Frameshift InDel, n417431378446
Splice regiona, n1,8191,8921,4501,857

a <=5b from splice site; InDels, insertions and deletions; SNPs, single nucleotide polymorphisms; Ti/Tv transition/transversion. Samples were aligned to the NCBI37 Homo sapiens reference genome using ELAND and CASAVA, and the SNP consequences reported from the alignment were collated with gene loci using the variant effect predictor of Ensembl. In all four samples there was a 98% mean array agreement between the genotyping and sequencing SNPs to the reference genome NCBI37 of which 38% were associated with Genes and the Ti/Tv ratio was 2.1, further validating the accuracy of SNP calling. The affected genes were subsequently cross-checked against the genetic association database for disease and cancer associations.

The transition/transversion (Ti/Tv) ratio has been used in multiple studies for assessing the specificity of SNP calls, with an empirical Ti/Tv ratio of ~2.1 for genome-wide variants (3133). Ti/Tv ratio is computed as the number of transition SNPs divided by the number of transversion SNPs (34). Typically, the Ti/Tv ratio is lower in novel SNPs than in known SNPs because of residual false positives and a relative deficit of transitions due to sequencing context bias (31). All the samples in the present study had a Ti/Tv ratio of 2.1, thus further validating the accuracy of SNP calling. The SNP sequence variation in the chromosome showed 90% call accuracy to the reference genome (NCBI37) across all the four samples, which was replicated with the autosomal, mitochondrial and the X-specific sites (data not shown).

The most prevalent SVs were insertions, deletions and copy number variation. Very few insertions were observed in all the four samples compared to deletions and copy number variations, which were common (data not shown). This study concentrated on identifying large SVs (>300 bp) and the role that they could play in OSCC. Since no common insertions were identified in all four samples, the evaluation of insertions was not explored further in this study.

Effects associated with deletions

A total of 697 deletions were observed in all four samples of which 349 within genes and 127 were associated with various cancers (Table II). Only 12 genes were shown to have deletions in the coding region or were associated with OSCC (Table III). Among the twelve genes; alcohol dehydrogenase 4 (ADH4), EGFR, muts homolog 3 (MSH3) and BCLAF1 have previously been associated with cancer (35,36), but only BCLAF1 was identified in the present study as a possible noteworthy SV owing to its novel association with OSCC.

Table II.

Summary of deletions common to all four tumour samples that shows the breakdown of the total number of deletions described in the samples.

Table II.

Summary of deletions common to all four tumour samples that shows the breakdown of the total number of deletions described in the samples.

DeletionsCommon in all four samples, n
Number of common deletions697
Deletions associated with genes349
Deletions associated with cancer127

[i] Deletions associated with genes were cross-checked against the genetic association database for disease and cancer associations.

Table III.

Description of gene deletions common to all four tumour samples.

Table III.

Description of gene deletions common to all four tumour samples.

Genes (HGNC)LocationVariant consequenceMeSH disease termsPercentage protein coding overlap, %
NXPE1Chr.11Non-coding exon variantUlcerative colitis; inflammatory bowel disease0.66
DLEU1Chr.13Non-coding exon variantChronic lymphocytic leukaemia0.32
TMBIM4Chr.123′UTR variantVenezuelan haemorrhagic fever; Bolivian haemorrhagic fever0.73
ADH4Chr.4Upstream gene variantAdenocarcinoma; oesophageal neoplasm; Parkinson's disease0.12
HYDINChr.16Coding sequence variantPrimary ciliary dyskinesia 5; Hydin-related primary ciliary dyskinesia0.49
KCNJ12Chr.17Frameshift variant 3′UTRHypokalemic periodic paralysis3.53
BCLAF1Chr.6Coding sequence variantNon-Hodgkin's lymphoma; colorectal cancer53.20
CACNA1BChr.9Coding sequence variantPulmonary aspergilloma0.34
EGFRChr.7Intron variantOesophageal neoplasms; colonic neoplasms; cervical squamous cell carcinoma0.24
FAM118AChr.22Coding sequence variantCardiovascular diseases; Diabetes Mellitus, Type 21.70
OR6K5PChr.1Non-coding exon variantNeuronitis25.35
MSH3Chr.5Intron variantAdenocarcinoma; oesophageal neoplasm; colonic neoplasms0.31

[i] HGNC, HUGO Gene Nomenclature Committee; MeSH, Medical Subject Headings. Percentage overlap of the protein coding region was reported from the alignment to the human reference genome (NCBI37), and collated with gene loci using the Variant Effect Predictor of Ensembl.

The BCLAF1 deletion had previously been shown to be associated with Non-Hodgkin's lymphoma and colorectal cancer (13,17). In the current study, the BCLAF1 deletion spanned 53.20% of the protein coding region as opposed to the other SVs and was proposed to have significant implications for the progression of OSCC.

siRNA-mediated BCLAF1 silencing

siRNA-mediated knockdown of BCLAF1 was determined at both the protein and mRNA levels. A decreased expression of BCLAF1 expression at both the mRNA and protein levels was observed after transfection with BCLAF1-siRNA (Figs. 2A and B and S2A).

Effect of BCLAF1 silencing on cellular gene expression

To assess whether the expression of genes downstream of BCLAF1 was altered by the targeted disruption of BCLAF1 expression, RNA and protein extracts from cells transfected with either non-specific scrambled control siRNA, BCLAF1 siRNA and untreated cells were collected 24 h post-transfection. RT-qPCR and western blot analyses were performed to determine the effects of BCLAF1 deficiency on pro-apoptotic, anti-apoptotic, DNA damage response (DDR) genes and cell cycle regulators. BCLAF1 knockdown resulted in a significant decrease in the expression of the apoptotic genes, such as BAX and Caspase-3, gene expression (Figs. 3A-D and S2B-D and Table SIV). Analysis of both the protein and mRNA levels showed no significant effect on the expression of the anti-apoptotic gene BCL2, but a significant effect was observed on the expression of cell cycle regulators such as P53 (Figs. 4A-D and S2E and H and Table SIV). Depletion of BCLAF1 also resulted in reduced expression levels of EXO1, ATRIP and BACH1 mRNA and the slight attenuation of EXO1 and H2AX protein levels (Figs. 5A-F and S2F and G and Table SIV).

Effect of BCLAF1 silencing on anchorage-dependent growth

To determine the effect of BCLAF1 on anchorage-dependent growth and cellular transformation, soft agar colony forming assays were performed. Normal cells are anchorage dependent and undergo apoptosis in the absence of a binding substrate. Transformed cells on the other hand, proliferate to form colonies (27). After incubation for 21 days, colonies were stained with 0.005% crystal violet, visualised and counted. There was a notable increase in size and number of colonies following BCLAF1 silencing compared with the controls (Fig. 6A-D).

Discussion

Cancer-derived cell lines are generally used as models in the study of cancer, but there is no perfect cell line model due to a number of well-known limitations (3742). Although cancer research is commonly conducted in epithelial cells, tumours are heterogeneous and the tumour microenvironment aids carcinogenesis (43). Due to the ability of immortalized cells to surpass replicative barriers in culture, they constitute a robust model system that can be used to study human cell transformation (43,44). CT1 fibroblasts were used as a model cell line in the present study to validate the effects of BCLAF1 knockdown on cellular transformation. The CT1 cells used in this study did not contain any p53 mutations and were thus suitable for studying the relationship between cell immortalization and changes in p53 gene expression (26,45) as opposed to human cell lines that are immortalized by treatment with oncogenic DNA viruses, such as SV40, papilloma and adenoviruses, since these viruses bind to and inactivate the proteins, such as p53 and Rb (46,47).

In the present study, four OSCC samples were subjected to WGS to identify large SVs (InDels) that may be associated with OSCC. A total of 4.27 billion reads of 300 bp fragments were generated covering 95% of the human reference genome (NCBI37) at a mean read depth of 33.7. This allowed for the reliable calling of SNPs and SVs across the human reference genome (NCBI37) and agreed with previous studies that also showed that an average read depth that exceeded 30X was the de facto standard for achieving reliable variant calling across 95% of the reference genome (48,49).

The Ti/Tv ratio is also a critical metric for assessing the specificity of SNP calling (41) and has been used as a quality control parameter for assessing the overall SNP quality in multiple studies with the expected Ti/Tv ratios in WGS being 2.1, generally indicating higher accuracy in variant calling (31,33). The present results showed a Ti/Tv ratio of 2.1 across all four samples, further confirming the reliability of these SNPs and SV calling.

SVs in the human genome play an important role in the genetics of complex diseases (1,48). Although the analysis of the present study reduced the number of variants of specific interest from the pool of SVs to a select few, large numbers of potentially functional alterations that could contribute to OSCC susceptibility were not investigated. From the 12 potential genes that were common in all four samples, only three genes have been previously shown to be associated with OSCC; ADH4, EGFR, MSH3 (17,34,35,5052) and one novel gene BCLAF1 that has not been reported in OSCC to date.

A reduction in ADH4 expression in poorly differentiated OSCC contributes to the maintenance of the tumour state by inhibiting the retinoic acid signalling pathway (53), while mutations in EGFR have also been shown to occur in primary OSCC (54,55). Other studies by Vogelsang et al (36,56) also showed that MSH3 polymorphisms, together with exposure to tobacco smoking increased the risk of oesophageal cancer. Although these genes (ADH4, EGFR and MSH3) had previously been associated with OSCC, we did not analyse them further for functional significance in the current study.

The BCLAF1 deletion on the other hand, is a notable SV due to its novel association with OSCC. Although the exact molecular role of BCLAF1 remains to be elucidated, BCLAF1 has been shown to have functional connections and mutation patterns consistent with the known hallmarks of cancer (20,57) suggesting BCLAF1 as a contributor to the progression of OSCC. Although there are currently no extensive studies depicting the role of BCLAF1 deletion in OSCC, previous studies have shown a role for BCLAF1 deletion in a number of cancers other than OSCC. Ungerbäck et al (51) identified BCLAF1 deletion as among the tumour suppressor candidate genes that may be responsible for or a contributing factor to colorectal cancer. Other cancers also linked with the loss of BCLAF1 include hepatocellular carcinoma (58), leukaemia (59), lung cancer (60), bladder cancer (15) and colon cancer (20,61).

BCLAF1 has also been shown to directly interact with and activate P53 gene transcription and that silencing of BCLAF1 reduces P53-dependent apoptosis, thus implicating P53 as a downstream target of BCLAF1 (62). This is also supported by other studies that identified P53 as among the significantly mutated genes in OSCC (63,64). DDR pathways protect cells from extensive DNA damage and cellular transformation (51,57,65). The network of DDR pathways consists of homologous recombination, mismatch repair, non-homologous end joining and double strand break repair (66). Since there is redundancy in these repair mechanisms and the genes are expressed at low levels in the absence of external DNA damaging agents (5,67), the expression levels of genes that are essential to these processes, including H2AX, BACH1, Ku70, EXO1 and ATRIP, were investigated.

BCLAF1 silencing reduced ATRIP expression in the present study, this was expected due to decreased BCLAF1/BRCA1 complex formation (65). BCLAF1 silencing had no significant impact on EXO1 expression, but then transcription factors such as MYC and E2F1 also positively regulate the EXO1 promoter (68). Additionally, the RAS/PI3K/AKT signalling pathways induce EXO1 gene expression (69). The activation of these pathways in the transformed cells could compensate for the decreased BCLAF1 levels. Thus, the BCLAF1 knockdown was insufficient to cause a significant change in EXO1 expression.

The siRNA-mediated silencing of BCLAF1 also altered the expression of important DNA damage response regulators, such as ATRIP, H2AX and EXO1. In the event of replicative stress, cells activate several genes to halt cell cycle progression and proliferation. P53 and P21 are potent inhibitors of G1/S phase progression through their interaction with CDK2 and CDK4/6 (70,71). In the G1 phase cell cycle arrest, P53 triggers transcription of P21 (cip1/waf1) a key component in cell cycle regulation (72). The present study confirmed that silencing BCLAF1 significantly attenuated P53 expression, which was in agreement with the results on BCLAF1 deficiency and downregulation of Caspase-3 and BAX protein levels.

Caspases regulate the intrinsic and extrinsic apoptosis pathways, resulting in DNA fragmentation, cytoskeletal breakdown and protein degradation (73,74). This current study showed that the expression of Caspase 3 was decreased after knockdown of BCLAF1 expression. This is likely mediated via the resulting increase in anti-apoptotic Ku70-Bax complexes that causes inhibition of Caspase-3 expression and activation (16). Thus, apoptosis resistance and cell survival are enhanced in siBCLAF1 treated cells. The BCL-2 family of genes are essential in apoptosis regulation and are either pro-apoptotic (Bax, Bad, Bak) or anti-apoptotic (BCL-2, BCL-XL) (75). The specific underlying mechanisms regarding the induction of apoptosis by BCLAF1 repression in transformed cells is still largely unknown. Although the transformed cells undergo apoptotic cell death following siBCLAF1 treatment, they still formed colonies on soft agar.

Although the present study showed that the abrogation of BCLAF1 expression in CT1 fibroblasts resulted in the disruption of several signalling pathways, the fact that a fibroblast cell line rather than an oesophageal cell line for the functional analysis is a limitation in this study.

The current study demonstrated that BCLAF1 deletion may be an important determinant in the progression of OSCC. To the best of our knowledge this is the first study linking BCLAF1 deletion in OSCC. It is possible that the link between BCLAF1 deletion and the progression of OSCC is via the downregulation of P53 and the deregulation of cellular processes involved in carcinogenesis. This hypothesis is also supported by studies that showed the role of BCLAF1 as a tumour suppressor and to promote the stability of genes required for DNA repair and maintenance of genomic stability (13,65).

To conclude, this study contributes towards our understanding of the underlying mechanisms involved in genomic alterations. It also provides evidence that abrogation of BCLAF1 expression results in the dysregulation of several cancer signalling pathways leading to cellular transformation. This in turn presents an opportunity for the design of novel therapeutic protocols based on the genomic traits of tumours.

Supplementary Material

Supporting Data

Acknowledgements

The present study forms part of the PhD Thesis ‘The Role of Viral Sequences in Genetic Aberrations and Malignant Transformation’ by LM submitted to the Faculty of Health Sciences, University of Cape Town, September 2014.

Funding

This study was supported by funds from the National Department of Health and the Medical Research Council (UK) with funds from the UK Government's Newton Fund and Glaxo Smith Klein (grant no. 046). This work was also supported by the National Research Foundation and the University of Cape Town.

Availability of data and materials

All data generated or analysed during the current study are available in the NLM NCBI dbVar repository (accession no. nstd195; http://www.ncbi.nlm.nih.gov/dbvar/studies/nstd195/).

Authors' contributions

LMM and MIP conceived and designed the experiments. LMM, VC and HS performed the experiments. LM, VC, HS and MIP analysed the data. MIP contributed reagents, materials and analysis tools. LMM and MIP wrote the manuscript. LMM and MIP confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Ethics approval was obtained from the University of Cape Town/Groote Schuur Hospital Human Research Ethics Committee (Cape Town, South Africa; approval no. HEC040/2005), and all methods were performed in accordance with the Declaration of Helsinki. Written informed consent was obtained from all patients before recruitment into the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ADH4

alcohol dehydrogenase 4

BCLAF1

BCL2 associated transcription factor 1

DDR

DNA damage response

DMEM

Dulbecco's modified Eagle's medium

DSB

double strand break

InDels

insertions and deletions

MSH3

muts homolog 3

OSCC

oesophageal squamous cell carcinoma

SNP

single nucleotide polymorphisms

WGS

Whole Genome Sequencing

References

1 

Feuk L, Carson AR and Scherer SW: Structural variation in the human genome. Nat Rev Genet. 7:85–97. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Vogelstein B and Kinzler KW: Cancer genes and the pathways they control. Nat Med. 10:789–799. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Fearon ER and Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 61:759–767. 1990. View Article : Google Scholar : PubMed/NCBI

4 

Knudson AG: Cancer genetics. Am J Med Genet. 111:96–102. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Hoeijmakers JH: Genome maintenance mechanisms for preventing cancer. Nature. 411:366–374. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Li GM: Mechanisms and functions of DNA mismatch repair. Cell Res. 18:85–98. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Motoyama N and Naka K: DNA damage tumor suppressor genes and genomic instability. Curr Opin Genet Dev. 14:11–16. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Popova T, Manié E, Stoppa-Lyonnet D, Rigaill G, Barillot E and Stern MH: Genome Alteration Print (GAP): A tool to visualize and mine complex cancer genomic profiles obtained by SNP arrays. Genome Biol. 10:R1282009. View Article : Google Scholar : PubMed/NCBI

9 

Kondrashov AS and Rogozin IB: Context of deletions and insertions in human coding sequences. Hum Mutat. 23:177–185. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Yang H, Zhong Y, Peng C, Chen JQ and Tian D: Important role of indels in somatic mutations of human cancer genes. BMC Med Genet. 11:1282010. View Article : Google Scholar : PubMed/NCBI

11 

Gregory TR: Insertion-deletion biases and the evolution of genome size. Gene. 324:15–34. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Mwapagha LM: The role of viral sequences in genetic aberrations and malignant transformation. University of Cape Town, 2014. http://hdl.handle.net/11427/12870

13 

Kasof GM, Goyal L and White E: Btf, a novel death-promoting transcriptional repressor that interacts with Bcl-2-related proteins. Mol Cell Biol. 19:4390–4404. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Fulda S, Gorman AM, Hori O and Samali A: Cellular stress responses: Cell survival and cell death. Int J Cell Biol. 2010:2140742010. View Article : Google Scholar : PubMed/NCBI

15 

Shen B, Tan M, Mu X, Qin Y, Zhang F, Liu Y and Fan Y: Upregulated SMYD3 promotes bladder cancer progression by targeting BCLAF1 and activating autophagy. Tumor Biol. 37:7371–7381. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Lee YY, Yu YB, Gunawardena HP, Xie L and Chen X: BCLAF1 is a radiation-induced H2AX-interacting partner involved in γH2AX-mediated regulation of apoptosis and DNA repair. Cell Death Dis. 3:e3592012. View Article : Google Scholar : PubMed/NCBI

17 

Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES and Getz G: Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 505:495–501. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, et al: The landscape of somatic copy-number alteration across human cancers. Nature. 463:899–905. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Hu N, Clifford RJ, Yang HH, Wang C, Goldstein AM, Ding T, Taylor PR and Lee MP: Genome wide analysis of DNA copy number neutral loss of heterozygosity (CNNLOH) and its relation to gene expression in esophageal squamous cell carcinoma. BMC Genomics. 11:5762010. View Article : Google Scholar : PubMed/NCBI

20 

Zhou X, Li X, Cheng Y, Wu W, Xie Z, Xi Q, Han J, Wu G, Fang J and Feng Y: BCLAF1 and its splicing regulator SRSF10 regulate the tumorigenic potential of colon cancer cells. Nat Commun. 5:45812014. View Article : Google Scholar : PubMed/NCBI

21 

Matejcic M, Mathew CG and Parker MI: The relationship between environmental exposure and genetic architecture of the 2q33 Locus with esophageal cancer in South Africa. Front Genet. 10:4062019. View Article : Google Scholar : PubMed/NCBI

22 

Kidd JM, Graves T, Newman TL, Fulton R, Hayden HS, Malig M, Kallicki J, Kaul R, Wilson RK and Eichler EE: A human genome structural variation sequencing resource reveals insights into mutational mechanisms. Cell. 143:837–847. 2010. View Article : Google Scholar : PubMed/NCBI

23 

McLaren W, Pritchard B, Rios D, Chen Y, Flicek P and Cunningham F: Deriving the consequences of genomic variants with the Ensembl API and SNP effect predictor. Bioinformatics. 26:2069–2070. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Becker KG, Barnes KC, Bright TJ and Wang SA: The genetic association database. Nat Genet. 36:431–432. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Tweedie S, Braschi B, Gray K, Jones TEM, Seal RL, Yates B and Bruford EA: Genenames. org: The HGNC and VGNC resources in 2021. Nucleic Acids Res. 49:D939–D946. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Namba M, Nishitani K and Kimoto T: Characteristics of WI-38 cells (WI-38 CT-1) transformed by treatment with Co-60 gamma rays. Gann. 71:300–307. 1980.PubMed/NCBI

27 

Mwapagha LM, Tiffin N and Parker MI: Delineation of the HPV11E6 and HPV18E6 pathways in initiating cellular transformation. Front Oncol. 7:2582017. View Article : Google Scholar : PubMed/NCBI

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Bailey JA, Yavor AM, Massa HF, Trask BJ and Eichler EE: Segmental duplications: Organization and impact within the current human genome project assembly. Genome Res. 11:1005–1017. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Bailey JA, Gu Z, Clark RA, Reinert K, Samonte RV, Schwartz S, Adams MD, Myers EW, Li PW and Eichler EE: Recent segmental duplications in the human genome. Science. 297:1003–1007. 2002. View Article : Google Scholar : PubMed/NCBI

31 

DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, Philippakis AA, del Angel G, Rivas MA, Hanna M, et al: A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 43:491–498. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Li H and Durbin R: Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 26:589–595. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Liu Q, Guo Y, Li J, Long J, Zhang B and Shyr Y: Steps to ensure accuracy in genotype and SNP calling from Illumina sequencing data. BMC Genomics. 13 (Suppl 8):S82012. View Article : Google Scholar : PubMed/NCBI

34 

Guo Y, Zhao S, Sheng Q, Ye F, Li J, Lehmann B, Pietenpol J, Samuels DC and Shyr Y: Multi-perspective quality control of Illumina exome sequencing data using QC3. Genomics. 103:323–328. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Kato H, Arao T, Matsumoto K, Fujita Y, Kimura H, Hayashi H, Nishiki K, Iwama M, Shiraishi O, Yasuda A, et al: Gene amplification of EGFR, HER2, FGFR2 and MET in esophageal squamous cell carcinoma. Int J Oncol. 42:1151–1158. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Vogelsang M, Paccez JD, Schäfer G, Dzobo K, Zerbini LF and Parker MI: Aberrant methylation of the MSH3 promoter and distal enhancer in esophageal cancer patients exposed to first-hand tobacco smoke. J Cancer Res Clin Oncol. 140:1825–1833. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Sharma SV, Haber DA and Settleman J: Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer. 10:241–253. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Mirabelli P, Coppola L and Salvatore M: Cancer cell lines are useful model systems for medical research. Cancers (Basel). 11:10982019. View Article : Google Scholar : PubMed/NCBI

39 

Borrell B: How accurate are cancer cell lines? Nature. 463:8582010. View Article : Google Scholar : PubMed/NCBI

40 

Weinstein JN: Cell lines battle cancer. Nature. 483:544–545. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Gillet JP, Varma S and Gottesman MM: The clinical relevance of cancer cell lines. J Natl Cancer Inst. 105:452–458. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Bhowmick NA, Neilson EG and Moses HL: Stromal fibroblasts in cancer initiation and progression. Nature. 432:332–337. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Boehm JS and Hahn WC: Immortalized cells as experimental models to study cancer. Cytotechnology. 45:47–59. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Namba M, Nishitani K and Kimoto T: Effects of theophylline on the cell growth of normal and malignant human cells transformed in culture. Gann. 71:621–627. 1980.PubMed/NCBI

46 

Itjima M, Mihara K, Kondo T, Tsuji T, Ishioka C and Namba M: Mutation in p53 and de-regulation of p53-related gene expression in three human cell lines immortalized with 4-nitroquinoline 1-oxide or 60Co gamma rays. Int J Cancer. 66:698–702. 1996. View Article : Google Scholar : PubMed/NCBI

47 

Rhim JS, Yoo JH, Park JH, Thraves P, Salehi Z and Dritschilo A: Evidence for the multistep nature of in vitro human epithelial cell carcinogenesis. Cancer Res. 50 (Suppl 17):5653S–5657S. 1990.PubMed/NCBI

48 

Yoon S, Xuan Z, Makarov V, Ye K and Sebat J: Sensitive and accurate detection of copy number variants using read depth of coverage. Genome Res. 19:1586–1592. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Sims D, Sudbery I, Ilott NE, Heger A and Ponting CP: Sequencing depth and coverage: Key considerations in genomic analyses. Nat Rev Genet. 15:121–132. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, Scherer SW and Lee C: Detection of large-scale variation in the human genome. Nat Genet. 36:949–951. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Ungerbäck J, Belenki D, Jawad ul-Hassan A, Fredrikson M, Fransén K, Elander N, Verma D and Söderkvist P: Genetic variation and alterations of genes involved in NFκB/TNFAIP3-and NLRP3-inflammasome signaling affect susceptibility and outcome of colorectal cancer. Carcinogenesis. 33:2126–2134. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Kuwano H, Kato H, Miyazaki T, Fukuchi M, Masuda N, Nakajima M, Fukai Y, Sohda M, Kimura H and Faried A: Genetic alterations in esophageal cancer. Surg Today. 35:7–18. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Han CL, Liao CS, Wu CW, Hwong CL, Lee AR and Yin SJ: Contribution to first-pass metabolism of ethanol and inhibition by ethanol for retinol oxidation in human alcohol dehydrogenase family. Eur J Biochem. 254:25–31. 1998. View Article : Google Scholar : PubMed/NCBI

54 

Hanawa M, Suzuki S, Dobashi Y, Yamane T, Kono K, Enomoto N and Ooi A: EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus. Int J Cancer. 118:1173–1180. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Guo M, Liu S, Herman JG, Zhuang H and Lu F: Brief communication Gefitinib-sensitizing mutation in esophageal carcinoma cell line Kyse450. Cancer Biol Ther. 5:152–155. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Vogelsang M, Wang Y, Veber N, Mwapagha LM and Parker MI: The cumulative effects of polymorphisms in the DNA mismatch repair genes and tobacco smoking in oesophageal cancer risk. PLoS One. 7:e369622012. View Article : Google Scholar : PubMed/NCBI

57 

Shao AW, Sun H, Geng Y, Peng Q, Wang P, Chen J, Xiong T, Cao R and Tang J: Bclaf1 is an important NF-κB signaling transducer and C/EBPβ regulator in DNA damage-induced senescence. Cell Death Differ. 23:865–875. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Wen Y, Zhou X, Lu M, He M, Tian Y, Liu L, Wang M, Tan W, Deng Y, Yang X, et al: Bclaf1 promotes angiogenesis by regulating HIF-1α transcription in hepatocellular carcinoma. Oncogene. 38:1845–1859. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Dell'Aversana C, Giorgio C, D'amato L, Lania G, Matarese F, Saeed S, Di Costanzo A, Belsito Petrizzi V, Ingenito C, Martens JHA, et al: miR-194-5p/BCLAF1 deregulation in AML tumorigenesis. Leukemia. 31:2315–2325. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Jiang T, Liu B, Wu D and Zhang F: BCLAF1 induces cisplatin resistance in lung cancer cells. Oncol Lett. 20:2272020. View Article : Google Scholar : PubMed/NCBI

61 

Wang H, Liang L, Fang J-Y and Xu J: Somatic gene copy number alterations in colorectal cancer: New quest for cancer drivers and biomarkers. Oncogene. 35:2011–2019. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Liu H, Lu ZG, Miki Y and Yoshida K: Protein kinase C delta induces transcription of the TP53 tumor suppressor gene by controlling death-promoting factor Btf in the apoptotic response to DNA damage. Mol Cell Biol. 27:8480–8491. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Lin DC, Hao JJ, Nagata Y, Xu L, Shang L, Meng X, Sato Y, Okuno Y, Varela AM, Ding LW, et al: Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat Genet. 46:467–473. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Song Y, Li L, Ou Y, Gao Z, Li E, Li X, Zhang W, Wang J, Xu L, Zhou Y, et al: Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 509:91–95. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Savage KI, Gorski JJ, Barros EM, Irwin GW, Manti L, Powell AJ, Pellagatti A, Lukashchuk N, McCance DJ, McCluggage WG, et al: Identification of a BRCA1-mRNA splicing complex required for efficient DNA repair and maintenance of genomic stability. Mol Cell. 54:445–459. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Dietlein F, Thelen L and Reinhardt HC: Cancer-specific defects in DNA repair pathways as targets for personalized therapeutic approaches. Trends Genet. 30:326–339. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Friedberg EC: DNA damage and repair. Nature. 421:436–440. 2003. View Article : Google Scholar : PubMed/NCBI

68 

Leung J, Ehmann G, Giangrande P and Nevins J: A role for Myc in facilitating transcription activation by E2F1. Oncogene. 27:4172–4179. 2008. View Article : Google Scholar : PubMed/NCBI

69 

Muthuswami M, Ramesh V, Banerjee S, Viveka Thangaraj S, Periasamy J, Bhaskar Rao D, Barnabas GD, Raghavan S and Ganesan K: Breast tumors with elevated expression of 1q candidate genes confer poor clinical outcome and sensitivity to Ras/PI3K inhibition. PLoS One. 8:e775532013. View Article : Google Scholar : PubMed/NCBI

70 

Vermeulen K, Van Bockstaele DR and Berneman ZN: The cell cycle: A review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif. 36:131–149. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Bertoli C, Skotheim JM and De Bruin RA: Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 14:518–828. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Kruse JP and Gu W: Modes of p53 regulation. Cell. 137:609–622. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Riedl SJ and Shi Y: Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 5:897–904. 2004. View Article : Google Scholar : PubMed/NCBI

74 

Thomadaki H and Scorilas A: BCL2 family of apoptosis-related genes: Functions and clinical implications in cancer. Crit Rev Clin Lab Sci. 43:1–67. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Cory S and Adams JM: The Bcl2 family: Regulators of the cellular life-or-death switch. Nat Rev Cancer. 2:647–656. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2021
Volume 46 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mwapagha LM, Chibanga V, Shipanga H and Parker MI: New insights from Whole Genome Sequencing: BCLAF1 deletion as a structural variant that predisposes cells towards cellular transformation. Oncol Rep 46: 229, 2021
APA
Mwapagha, L.M., Chibanga, V., Shipanga, H., & Parker, M.I. (2021). New insights from Whole Genome Sequencing: BCLAF1 deletion as a structural variant that predisposes cells towards cellular transformation. Oncology Reports, 46, 229. https://doi.org/10.3892/or.2021.8180
MLA
Mwapagha, L. M., Chibanga, V., Shipanga, H., Parker, M. I."New insights from Whole Genome Sequencing: BCLAF1 deletion as a structural variant that predisposes cells towards cellular transformation". Oncology Reports 46.4 (2021): 229.
Chicago
Mwapagha, L. M., Chibanga, V., Shipanga, H., Parker, M. I."New insights from Whole Genome Sequencing: BCLAF1 deletion as a structural variant that predisposes cells towards cellular transformation". Oncology Reports 46, no. 4 (2021): 229. https://doi.org/10.3892/or.2021.8180