GET THE APP

Gene Expression Meta-Analysis of Colon Rectal Cancer Tumour Cells Reveals Genes in Association with Tumorigenesis
..

Cancer Science & Therapy

ISSN: 1948-5956

Open Access

Research Article - (2022) Volume 14, Issue 5

Gene Expression Meta-Analysis of Colon Rectal Cancer Tumour Cells Reveals Genes in Association with Tumorigenesis

Rutvi Vaja*
*Correspondence: Rutvi Vaja, Department of Biomedical Sciences and Research, School of Science, Navrachana University, Vadodara-390021, Gujarat, India, Email:
Department of Biomedical Sciences and Research, School of Science, Navrachana University, Vadodara-390021, Gujarat, India

Received: 11-May-2022, Manuscript No. jcst-22-63901; Editor assigned: 12-May-2022, Pre QC No. P-63901; Reviewed: 21-May-2022, QC No. Q-63901; Revised: 24-May-2022, Manuscript No. R-63901; Published: 31-May-2022 , DOI: 10.37421/1948-5956.22.14.531
Citation: Vaja, Rutvi. “Gene Expression Meta-Analysis of Colon Rectal Cancer Tumour Cells Reveals Genes in Association with Tumorigenesis.” J Cancer Sci Ther 14 (2022): 531.
Copyright: © 2022 Vaja R. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Background: Every year, more than 12 million people are diagnosed with colorectal cancer (CRC), and more than 600,000 people die from it, making it second most deadly form of cancer. This work analyzes differential gene expression across CRC and other glandular tumour samples to identify expression changes potentially contributing to the development of CRC tumourigenesis.

Methods: This work defines 13 gene signatures representing four CRC tumour and 10 other glandular tumours that are colonic by origin. Gene Set Enrichment Analysis (GSEA) is used to define positive and negative CRC gene panels from GSEA-identified leading-edge genes using two CRC signatures. GSEA then is used to verify enrichment and leading-edge gene membership of CRC panels in two independent CRC gene signatures. Analysis is then extended to four individual and 10 glandular tumour signatures. Genes most associated with CRC tumourigenesis are predicted by intersecting membership of GSEA-identified leading-edges across signatures.

Results: Significant enrichment is observed between CRC gene identification signatures, from which the positive (55 genes) and negative (77 genes) CRC panels are defined. Non-random significant enrichment is observed between CRC gene panels and verification signatures, from which 54 over- and 72 under-expressed genes are shared across leading-edges. Considering other glandular tumour samples individually and in combination with CRC, significant non-random enrichment is observed across these signatures. Eight solute carrier family genes such as (SLC25A32, SLC22A3, SLC25A20, SLC36A1, SLC26A3, SLC9A2, SLC4A4 and SLC26A2) from the CRC panel were shared commonly across all the gene signatures leading-edges, regardless of the colonic tumour type.

Conclusion: This meta-analysis identifies gene expression changes associated with the process of CRC tumourigenesis. These changes may contribute to developing therapeutic treatments available for CRC patients.

Keywords

Glandular tumours • CRC • GSEA • Meta-analysis • Gene expression

Introduction

As per the statistics of World Health Organization (WHO), Colorectal Cancer (CRC) is the third most prevalent cancer in the world along with fourth leading cause of cancer related deaths [1]. Colon and rectal cancers account for most of the glandular malignancies with the incidences increasing with age [1]. Highly penetrant, autosomal dominantly or recessively inherited tumour predispositions cause about 5% of all colorectal cancers [2]. More than 945 000 people are diagnosed with colorectal cancer each year, with roughly 492 000 patients dying [3]. This type of cancer occurs infrequently, often as a result of genetic cancer syndromes or inflammatory bowel illnesses [3]. As per the GLOBOCAN database documented 1.8 million newly diagnosed cases of CRC and 861,600 cases of CRC-related mortality over the world [4]. CRC is a highly diverse disease caused mostly by interactions between genetic changes and environmental variables [5]. Despite advances in diagnosis and treatment, the survival rate of CRC patients has remained unchanged over the previous two decades, with more than half of patients having regional or distant metastasis at the time of diagnosis [6-8]. Several genes and cellular signalling pathways, including RACK1 (receptor for activated C kinase 1) and long non- coding RNA breast cancer anti-estrogen resistance 4 (lncRNA BCAR4), have been implicated in the formation and progression of CRC [6]. RACK1 expression, for example, has been shown to be considerably upregulated in CRC tissues when compared to adjacent normal tissues [6]. Despite this comprehensive and meticulous research to find novel targets for CRC management, a comprehensive description of the critical key genes and signalling pathways implicated in CRC is lacking, to the best of our knowledge [4-6].

A high-throughput method for detecting mRNA expression in tissues, gene microarray profile analysis, is becoming a more promising tool in medical oncology. An enhanced understanding of the molecular pathogenesis of many cancer types can be gained by analysing differential gene expression between tumour tissues and normal control tissues, allowing for the identification of prospective target genes and signalling pathways for precision medicine [7]. In earlier decades, microarray technology was employed in various researches on gene expression profiles in cancer, but only one study focused on CRC [8]. Apart from these studies, the comparative analysis of Differentially Expressed Genes (DEGs) remains relatively limited [9]. Furthermore, more research is needed to identify meaningful leading edge gene profiles for distinguishing CRC from normal tissues. In addition, the relationships among the DEGs should be clarified, along with the interaction networks and critical biological signalling pathways affected.

Many data mining analyses of mRNA, microRNA, long non-coding RNA, and DNA methylation have been performed on human cancers, particularly colon cancer, over the last few decades. A complete understanding about the molecular changes associated with CRC Tumor formation might help in assisting the development of new therapeutic interventions. Several studies have been done to elucidate molecular changes associated with CRC by examining gene expression changes in CRC- tumour forming cell cultures [10- 12]. There are some studies which provide a clinical reference for predicting the survival probability of patients with different clinical subtypes [13]. Other studies generated Protein-protein Interaction (PPI) networks, and centrality analysis as well was performed in order to identify the crucial genes that were potentially involved in the development of CRC [11]. Several bioinformatics tools and strategies have aided in the exploration of molecular mechanisms of tumour pathogenesis and provided clues for better knowledge of related malignancies by identifying early biomarkers and potential therapeutic targets of tumours. Colon cancer is a multifactorial disease caused by a variety of factors including genetic, environmental, and lifestyle influences, but the pathogenesis of the disease is yet unknown. Exploring and analysing colon cancer's molecular basis and important genes is critical to improve colon cancer prevention and treatment.

Previous work using a GSEA-based meta-analysis approach successfully identified known and novel genes associated with severe acute respiratory syndrome (SARS) infection through differential gene expression comparison between mRNA expression datasets. Therefore, this paper applied the same GSEA-based approach to analyze mRNA expression data of tumor and normal CRC tissue derived from human colonic biopsy samples by defining and comparing gene expression signatures (i.e., gene lists ranked by differential expression) [14,15].

In this particular study, we have used expression profiling array datasets to identify and verify gene expression changes associated with CRC tumour formation to elucidate molecular changes associated with the process of tumourogenesis in CRC pathogenesis. Gene expression changes associated with CRC tumour formation then were compared to changes resulting from other colonic tumour forming diseases like Ulcerative colitis, adenoma dysplasia and Hyperplastic Polyp, to examine the common leading edge genes in CRC that invoked changes and played a potential role in CRC tumour formation. Finally we verified the gene signatures in several other colonic tumour forming diseases such as Ulcerative coilitis, adenoma dyslapsia and Hyperplastic Polyp. A meta-analysis was performed lastly to find the common shared leading edge genes across all different conditions of CRC. The gene expression changes identified with across these several conditions holds the potential to actually improve the treatments targeted for CRC and its therapeutic intervention.

Methods

mRNA expression resources

To identify gene expression changes associated with CRC tumour forming cells, the Gene Expression Omnibus (GEO) repository was searched to find datasets for use in this study (Table 1) [16]. The six independent data series GSE44861, GSE113513, GSE44076 GSE10714, GSE32323 and GSE24514 had CRC tumour and normal tissue samples and hence we started our analysis from here. GSE44861 is an Affymetrix expression data collected from colon cancer patient tissues in which RNA from fresh frozen colon tissues were extracted using Trizol and hybridized to Affymetrix U113A arrays [17]. The platform used for GSE44861 was GPL3921 [HT_HG-U133A] Affymetrix HT Human Genome U133A Array. GSE113513 has samples from 14 colorectal cancer patients who had undergone surgical resection of colorectal cancer where Trizol (Thermo Fisher Scientific) extraction of total RNA was performed [18]. The platform used for GSE113513 was GPL15207 [PrimeView] Affymetrix Human Gene Expression Array. GSE44076 contained Gene expression profiles of paired normal adjacent mucosa and tumor samples from 98 individuals and 50 healthy colon mucosae, which were obtained through Affymetrix Human Genome U219 Arrays [19]. The platform used for GSE44076 was GPL13667 [HG-U219] Affymetrix Human Genome U219 Array. GSE10714 had expression data from human colonic biopsy samples on which Qiagen RN easy Mini extraction of total RNA was performed [20]. The platform used for GSE10714 was GPL570 [HG-U133_Plus_2] Affymetrix

Table 1: Datasets utilized for this study

Data-Set Description Platform Probes Genes
GSE44861 Gene expression profiling of 111 colon tissues from tumors and adjacent noncancerous tissues. RNA from fresh frozen colon tissues were extracted using Trizol and hybridized to Affymetrix U113A arrays GPL3927 22277 21248
GSE113513 Samples from fourteen colorectal cancer patients who had undergone surgical resection of colorectal cancer. Trizol (Thermo Fisher Scientific) extraction of total RNA was performed on the samples GPL15207 49395 48872
GSE44076 Paired normal adjacent mucosa and tumor samples from 98 individuals and 50 healthy colon mucosae. GPL13667 49386 48785
GSE10714 Expression data from human colonic biopsy sample. Total RNA was extracted from colonic biosy samples CRC and hybridized on Affymetrix HGU133 Plus 2.0 microarrays GPL570 54675 45782
GSE32323 Gene expression profiles for 17 pairs of cancer and non-cancerous tissues from colorectal cancer patients were measured by Affymetrix HG-U133 Plus 2.0 arrays. GPL570 54675 45782
GSE24514 Expression profiling of 34 MSI colorectal cancers and 15 normal colonic mucosas. Samples had Comparison of malignant and healthy tissue. GPL96 22283 21225

Human Genome U133 Plus 2.0 Array. GSE32323 consisted of gene expression profiles for 17 pairs of cancer and non-cancerous tissues from colorectal cancer patients were measured by Affymetrix HG- U133 plus 2.0 arrays. Here, total RNA was extracted from tissue specimens using Reasy kit (Qiagen, Hilden, Germany) [21]. The platform used for GSE32323 was GPL570 [HG-U133_Plus_2].

Affymetrix Human Genome U133 Plus 2.0 Array. Lastly, GSE24514 had expression data from human MSI colorectal cancer and normal colonic mucosa in which RNA rom fresh frozen tissues was extracted with Trizol reagent (Invitrogen) [22]. The platform used for GSE24514 was GPL96 [HGU133A] Affymetrix Human Genome U133A Array].

The expression data provided by GEO for all datasets were z-scored normalized across all samples within the dataset regardless of tumour cell or treatment type prior to use for analysis. The expression data was cleaned by removing probe identifiers given by GEO where 1) all samples having gene expression z-score of 0, or 2) duplicate identifiers/symbols were identified so only the identifier with the highest coefficient of variation were retained. The soft family data consisted of Entrez Gene ids, Gene symbols and Probe identifiers as well. If a dataset’s GEO-provided platform contained both Ensemble gene IDs and gene symbols for a probe, the GEO-provided platform files to convert between these two prove identifiers were used. For this particular study, we have converted the probe identifiers to gene symbols using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) v6.8 gene conversion tool in order to maintain uniformity [23]. In order to obtain other meta- data information like sample characteristics, sample source, Ensemble ids and GSM numbers, GEO- provided platform files were used.

Defining gene signatures

In order to examine the gene expression changes associated with tumour formation in CRC differential gene expression was measured for samples of interest from each dataset using Welch’s two-sample T test score of cleaned and normalized values. (For eg: tumour samples and normal tissue sample from one dataset formed one signature). Samples with the same origin, regardless of tumour stage, cancer type (metastasis or benign) were combined to form one signature. The resulting lists of genes along with their T-test scores were used to define 13 gene signatures. These signatures are formed from the gene lists ranked from high to low differential gene expression between tumors vs. normal tissue samples. The signatures derived from the same dataset used the same control samples. The gene location where T-score becomes negative (i.e., T-score = 0) and the T- score range for each signature are found in Table 2.

Table 2: Signatures defined in this Study.

Dataset Group 1
(No. of Samples)
Group 2 ( N o. of Samples
)
Gene signature Us e High Low
GSE44861 CRC tumour (56) Normal Tissue (5 5) Tumour vs. Normal I 10.77 -11.89
GSE113513 CRC tumour (14) Normal Tissue (1 4) Tumour vs. Normal I 12.95 -14.02
GSE44076 CRC tumour (50) Normal Tissue (9 8) Tumou vs. Normal V 35.69 -39.04
GSE10714 Adenoma low-grade dyslapsia (5) Normal Tissue (3) Adenoma vs. Normal C    
Hyperplastic polyp (11) Normal tissue (3) HyperplasticPolyp vs. Normal C
Ulcerative coilitis (3) Normal tissue (3) UC vs. Normal C
CRC-Beningn (3) Normal tissue (3) CRCB vs. Normal C
CRC-CD (4) Normal tissue (3) CRCD vs. Normal C
GSE32323 CRC Tumour (10) Normal tissue (1 0) Tumour vs. Normal V    
Tumour metastasis (3) Normal tissue (3) TumourM vs. NormalM C
Tu m o u r M e t a s t a s i s recurrence (4) Normal tissue (4) TumourMR vs. Normal MR C
GSE24514 Microsatellite instability tumour (34) Normal tissue (1 5) MSIT vs. Normal C    

Identification of genes associated with CRC tumour tissues

To identify gene expression changes associated with CRC Tumour tissues, two CRC tumour gene panels were generated (Figure 1). In order to do this, 500 genes from the positive and negative tails from both the GSE44861- derived Tumour vs. Normal and GSE113513-derived Tumour vs. Normal gene signatures were selected and then were used to form four individual query gene sets. GSEA compared each query gene set to the both the entire GSE44861-derived Tumour vs. Normal and GSE113513-derived Tumour vs. Normal gene signatures (reference). For our identification stage, we used the datasets GSE44861 as query first against GSE113513 as reference followed by GSE113513 as query dataset against the whole of GSE44861 Tumour vs. Normal signature. Leading- edge (LE) genes from each of these analyses were examined and shared leading-edge genes were used to define two CRC Tumour vs. Normal gene panels, one panel per tail(Positive CRC and Negative CRC Panels). Pathway enrichment analysis was performed on both CRC tumour gene panels using DAVID.

cancer-science-therapy-schematic

Figure 1. Schematic overview of gene panel identification.

Verification of CRC tumour gene panels

To verify the CRC Tumour gene panels, GSEA between CRC Tumour gene panels (Pos. CRC and Neg. CRC) and GSE44076-derived and GSE10714-derived Tumour vs. Normal signatures was performed. To assess if results generated from GSEA could be achieved randomly, 1000 gene panels consisting of 175- genes to match the average number of genes in the positive and negative CRC Tumour panels (query signature) were randomly selected from the GPL3927 platform used to define the GSE44861 gene signatures used for gene identification for GSEA against GSE44076-derived and GSE32323- derived Tumour vs. Normal signatures (reference signature). These analysis generated a null distribution of NES(Normalized enrichment score) to which were compared the NES achieved by CRC tumour gene panels for each reference gene signature and count the number of equal or better NES to estimate significance (i.e., null distribution p-value). The bar and whiskers Plot was created and calculated using Excel. Heat maps were generated by a user-friendly, web-based program maps. https://software.broadinstitute.org/ morpheus. Morpheus is software created by Broad Institute Software.

Comparison of CRC tumour gene panels to other types of tumour that are glandular in origin

GSEA was used to compare the identified CRC gene panels and gene signatures derived from samples of one of the six glandular tumour (Ulcerative Colitis, Hyperplastic Polyp, Adenoma with low grade dysplasia, Colorectal cancer – Benign, Colorectal cancer with a chronic disease and colorectal Tumours with microsatellite instability), compared to normal tissue samples was performed to compare gene expression changes across CRC tumour tissues and several other glandular tumours. Following this, random modelling as stated earlier was used to assess if results generated from GSEA could be achieved randomly. Leading-edge genes from each statistically significant (GSEA P- value <0.05), non-random (null distribution P-value < 0.05) GSEA were examined and analysed for common genes.

Gene signature approach identified gene expression changes associated with colorectal cancer tumor tissues in vitro

GSE44861-derived and GSE113513-derived Tumour vs. Normal gene signatures were defined to identify genes associated with response to Tumour tissue of human CRC colonic biopsy samples (Table 2). From both these two Tumour vs. Normal identification signatures, two gene sets were generated containing the 500 most differentially expressed genes from the positive and negative tails of each signature, capturing maximum coverage of the signature that was allowable by GSEA [24]. The T-score values for GSE44861 derived Tumour vs. Normal signature was >10.77 and <-11.89. For GSE113513 derived Tumour vs. Normal gene signature was >12.95 and <-14.0282. To find the similarity between these two signatures, enrichment was first calculated using GSEA between GSE113513- derived Tumour vs. Normal positive or negative tail gene sets (individual query set) and the GSE44861- derived Tumour vs. Normal(reference set) and achieved NES = 3.54 and NES = -3.64 for positive and negative tail query gene sets, respectively, both with a GSEA p-value <0.001. Similarly enrichment was calculated using GSEA between GSE44861- derived Tumour vs. Normal positive or negative tail gene sets (individual query set) and the GSE113513- derived Tumour vs. Normal (reference set) and achieved NES = 3.38 and NES = -4.14 for positive and negative tail query gene sets, respectively, both with a GSEA p-value <0.001. The identified leadingedge genes from GSEA are listed in Supplementary Table 1.

No genes in the positive CRC panel were mentioned in the published reports for GSE44861 and GSE113513, though some panel genes like Kruppel like factor-7(ubiquitous) (KLF-7) and RNA terminal phosphate cyclaselike 1(RCL-1) have reported connections with CRC tumour formation in the literature [25-28]. In the negative CRC gene panel, Keratin 20(ALOX) was found to have previous associations with CRC tumour formation [29]. Taking all the results together, this demonstrated the detection ability of using a GSEAbased approach to gene identification. The rest of the genes in the negative panobinostat panel had no prior association with CRC tumour formation from the published reports for GSE44861 and GSE113513. It can be speculated that genes lacking previously reported associations with CRC tumour formation that were identified here also are associated with human colonic biopsy samples.

To expand our analysis, the cellular roles of CRC gene panels were examined using DAVID to calculate enrichment between each CRC gene panel and pathways in popular known knowledgebase. It was noticed that, when compared to other databases, the GO BP database returned the most significantly enriched pathways (data not mentioned here) hence this discussion was concentrated on GO-BP data to prevent confusion caused by other overlapping pathway and gene inclusion differences across other multiple known knowledgebase.

DAVID identified nine significant GO-BP pathways (EASE score P-value < 0.05) from the positive CRC gene panel and 10 significant pathways from the negative CRC gene panel (Supplementary Table 2). Some significantly enriched pathways have experimentally established associations with CRC tumour formation, such as RNA processing pathway (GO: 0006396, P-value = 0.02), demonstrating the ability of our gene signature approach to detect pathways associated with CRC Tumour formation [25]. Other identified pathways, such as lipid catabolic process (GO: 0016042, P- value=0.04), have no prior associations to CRC Tumour formation. Therefore, it can be speculated that pathways that came out as a result without prior association to CRC Tumours identified here also were involved in CRC.

Enrichment of CRC gene panels and specific CRC panel genes verified in independent datasets

To verify our CRC gene panels, GSEA was used to calculate enrichment between our CRC panels (individual queries) and two verification gene signatures (individual references): GSE32323-derived Tumour vs. Normal and GSE44076-derived Tumour vs. Normal (Table 2). Significant similarity between positive and negative CRC panels and GSE32323-derived Tumour vs. Normal (NES = 2.26 for the positive CRC panel, Figure 2A, and NES = -2.56 for the negative CRC panel, Figure 2B, both GSEA P- value < 0.001) was found. To determine how likely the NES achieved for CRC gene panels would be achieved by random chance, 1000 randomly selected 175-gene panels were generated from the GSE44861-derived Tumour vs. Normal gene signature to match the average size and potential composition of our CRC gene panels. GSEA was then repeated using these randomly generated gene panels (individual queries) and the GSE44076-derived Tumour vs. Normal (reference) to generate a null distribution of NES achieved via a random chance.

cancer-science-therapy-identification

Figure 2. Schematic overview of gene panel identification.

From this, random NES ranged from 1.47 to -1.5 was found (data not shown), illustrating that NES achieved by our CRC panels are non-random (null distribution P-value <0.001). Taken together, these results demonstrate that the enrichment achieved from our panobinostat panels was true.

To determine which of our CRC panel genes were verified across all signatures, leading-edge genes identified by GSEA for each verification signature were examined. Leading-edge genes for GSE44076-derived and GSE32323-derived Tumour vs. Normal signatures are listed in Supplementary Tables 3-10. 51 genes from the positive CRC panel and 75 genes from the negative CRC panel were shared between verification signatures. These data together verify our shared leading-edge genes are associated with CRC tumour tissues in human colonic biopsy samples and support the hypothesis that identified genes without previously reported associations are also associated with CRC tumour tissues in human colonic biopsy samples.

Non-random enrichment of CRC tumour gene panels found in other glandular tumours

To expand this study, gene expression changes associated with CRC Tumour were compared to changes observed in other glandular tumours included in GEO Series previously used to examine the common shared leading edge genes between CRC tumour tissues and other glandular tumours. The following eight signatures were examined: Adenoma vs. Normal, Hyperplastic Polyp vs. Normal, Uc vs. Normal, CRCB vs. Normal, CRCD vs. Normal, Tumour M vs. Normal M, Tumour MR vs. Normal MR and MSIT vs. Normal (Table 2). For Adenoma vs. Normal gene signature vs. the positive panel NES = 1.785 and for Negative panel is NES = -2.00 (GSEA P. value <0.001). For Hyperplastic Poly vs. Normal gene signature, when running a query against the Positive panel NES = 1.86 and for negative panel NES = -2.022 m (GSEA P. value <0.001). For Uc vs. Normal gene signature, NES for positive panel = 1.54 and NES for negative panel is -1.87 m (GSEA P. value <0.001). For CRCB vs. Normal gene signature against positive panel query, NES = 0.9 and for negative panel query, NES = -1.7(GSEA P. value <0.001). For CRCD vs. Normal gene signature, the query against Positive panel yielded a NES of 1.866 whereas the NES of negative panel was -2.00 (GSEA P. value <0.001). The Tumour M vs. Normal M gene signature yielded NES of 2.39 for the positive gene panel as the query set and NES of -2.47 (GSEA P. value <0.001). For Tumour MR vs. Normal MR the NES for the positive panel was 2.33 and for the negative panel NES = -2.5 (GSEA P. value <0.001). For gene signature of MSIT vs. Normal the positive panel’s NES score is 2.307 and negative panel’s NES score is -3.12 (GSEA P. value <0.001). Both the positive and negative CRC panel showed significant enrichment score (GSEA P. value <0.001) in all the gene signatures with and were also non-random (null distribution P- values <0.001).

Leading-edge genes found amongst the other glandular tumours

Finally, leading-edge genes from each GSEA across other glandular tumours were examined to identify genes of potential interest. Supplemental Material S Tables 3 through 9 contains leading-edge genes from GSEA with each CRC gene panel across 8 comparison gene signatures (Table 2). Out of the 54 common shared genes in the Positive CRC panel across all the signatures, 48 genes stood out having a statistical significance of (P value <0.001). Whereas out of the 75 common shared genes in the negative CRC panel across all the signatures, 72 genes were of highly statistical significance of (P value <0.001).9 solute carrier family genes solute carrier family 25, member 32 /// solute carrier family 25 member 32(SLC25A32), solute carrier family 22 (extraneuronal monoamine transporter) member 3 (SLC 22 A 3), solute carrier family 25 (carnitine/acylcarnitine translocase) member 20 (SLC25A20),solute carrier family 36 (proton/amino acid symporter) member 1 (SLC36A1), solute carrier family 17 (sodium phosphate) member 4 (SLC17A4),solute carrier family 26 member 3 (SLC26A3), solute carrier family 9 (sodium/hydrogen exchanger)member 2 (SLC9A2), solute carrier family 4 sodium bicarbonate cotransporter member 4 (SLC4A4),and solute carrier family 26 (sulfate transporter), member 2(SLC26A2) from the CRC panel were shared commonly across all the gene signatures. Group of carbonic anhydrase protein coding genes from CRC gene Panel were also commonly shared across all gene signature’s namely carbonic anhydrase XII (CA12), carbonic anhydrase II (CA2), carbonic anhydrase IV (CA4) and carbonic anhydrase I (CA1) (Figure 3).

cancer-science-therapy-glandular

Figure 3. Enrichment of CRC gene panels and random models across other glandular tumours.

Table 3: Positive CRC gene panel defined in this study.

Entrez ID Gene Symbol Description
9603 NFE2L3 Nuclear factor (erythroid-derived 2)-like 3
7004 TEAD4 TEA domain family member 4
81034 SLC25A32 Solute carrier family 25, member 32 /// solute carrier family 25, member 32
9569 GTF2IRD1 GTF2I repeat domain containing 1
1876 E2F6 E2F transcription factor 6
57460 PPM1H Protein phosphatase 1H (PP2C domain containing)
8886 DDX18 DEAD (Asp-Glu-Ala-Asp) box polypeptide 18
5471 PPAT Phosphoribosyl pyrophosphate amido transferase
6581 SLC22A3 Solute carrier family 22 (extraneuronal monoamine transporter), member 3
11260 XPOT Exportin, tRNA (nuclear export receptor for tRNAs)
54517 PUS7 Pseudouridylate synthase 7 homolog (S. cerevisiae)
54529 ASNSD1 Asparagine synthetase domain containing 1
1312 COMT Catechol-O-methyltransferase
595 CCND1 Cyclin D1
60496 AASDHPPT Aminoadipate-semialdehyde dehydrogenase- phosphopantetheinyl transferase
1662 DDX10 DEAD (Asp-Glu-Ala-Asp) box polypeptide 10
6059 ABCE1 ATP-binding cassette, sub-family E (OABP), member 1
10807 SDCCAG3 Serologically defined colon cancer antigen 3
52 ACP1 Acid phosphatase 1, soluble
55795 PCID2 PCI domain containing 2
10196 PRMT3 Protein arginine methyltransferase 3
63875 MRPL17 Mitochondrial ribosomal protein L17
79728 PALB2 Partner and localizer of BRCA2
1615 DARS Aspartyl-tRNA synthetase
5361 PLXNA1 Plexin A1
26586 CKAP2 Cytoskeleton associated protein 2
79074 C2orf49 Chromosome 2 open reading frame 49
57122 NUP107 Nucleoporin 107kDa
8609 KLF7 Kruppel-like factor 7 (ubiquitous)
11146 GLMN Glomulin, FKBP associated protein
2118 ETV4 ETS variant gene 4 (E1A enhancer binding protein, E1AF) /// ets variant gene 4 (E1A enhancer binding protein, E1AF)
6282 S100A11 S100 calcium binding protein A11
23560 GTPBP4 GTP binding protein 4
8833 GMPS Guanine monphosphate synthetase
10171 RCL1 RNA terminal phosphate cyclase-like 1
3614 IMPDH1 IMP (inosine monophosphate) dehydrogenase 1
54881 TEX10 Testis expressed sequence 10
26009 ZZZ3 Zinc finger, ZZ-type containing 3
26031 OSBPL3 Oxysterol binding protein-like 3
51776 ZAK Sterile alpha motif and leucine zipper containing kinase AZK
26135 SERBP1 SERPINE1 mRNA binding protein 1
4233 MET Met proto-oncogene (hepatocyte growth factor receptor)
79084 WDR77 WD repeat domain 77
9128 PRPF4 PRP4 pre-mRNA processing factor 4 homolog (yeast)
10527 IPO7 Importin 7
6741 SSB Sjogren syndrome antigen B (autoantigen La)
22880 MORC2 MORC family CW-type zinc finger 2
9221 NOLC1 Nucleolar and coiled-body phosphoprotein 1
1736 DKC1 Dyskeratosis congenita 1, dyskerin

To rank shared leading-edge genes, T-test P value analysis using Excel- STAT software was used. Out of the 129 Leading edge genes (54 from positive gene panel and 75 from negative gene panel) we wanted to find out the significant leading edge genes. Hence using the parameter: P. Value < 0.001 we applied a P- value T-test in Excel to calculate the P-value. A total of 120 genes (48-Over-expressed and 72 under-expressed) out of the 129 were significantly valid leading edge genes. The volcano plot for the significant leading-edge genes is shown in (Figure 4 and Table 4).

cancer-science-therapy-edge

Figure 4. Volcano plot showing over-expressed and Under-expressed leading edge genes.

Table 4: Negative CRC gene panel defined in this study

Entrez Id Gene symbol Description
25840 METTL7A Methyltransferase like 7A
51196 PLCE1 Phospholipase C, epsilon 1
11099 PTPN21 Protein tyrosine phosphatase, non-receptor type 21
1908 EDN3 Endothelin 3
23228 PLCL2 Phospholipase C-like 2
51228 GLTP Glycolipid transfer protein
2110 ETFDH Electron-transferring-flavoprotein dehydrogenase
55743 CHFR Checkpoint with forkhead and ring finger domains
10223 GPA33 Glycoprotein A33 (transmembrane)
788 SLC25A20 Solute carrier family 25 (carnitine/acylcarnitine translocase), member 20
54884 RETSAT Retinol saturase (all-trans-retinol 13,14-reductase)
5873 RAB27A RAB27A, member RAS oncogene family
171586 ABHD3 Abhydrolase domain containing 3
55359 STYK1 Serine/threonine/tyrosine kinase 1
54474 ALOX Keratin 20
6414 SEPP1 Selenoprotein P, plasma, 1
3960 LGALS4 Lectin, galactoside-binding, soluble, 4 (galectin 4)
51411 BIN2 Bridging integrator 2
140803 TRPM6 Transient receptor potential cation channel, subfamily M, member 6
125 ADH1B Alcohol dehydrogenase IB (class I), beta polypeptide
80031 SEMA6D Sema domain, transmembrane domain (TM), and cytoplasmic domain, (semaphorin) 6D
5567 PRKACB Protein kinase, cAMP-dependent, catalytic, beta
5827 PXMP2 Peroxisomal membrane protein 2, 22kDa
64922 LRRC19 Leucine rich repeat containing 19
2168 FABP1 Fatty acid binding protein 1, liver
9218 VAPA VAMP (vesicle-associated membrane protein)-associated protein A, 33kDa
23171 GPD1L Glycerol-3-phosphate dehydrogenase 1-like
55620 STAP2 Signal-transducing adaptor protein-2
7263 TST Thiosulfate sulfurtransferase (rhodanese)
206358 SLC36A1 Solute carrier family 36 (proton/amino acid symporter), member 1
4128 MAOA Monoamine oxidase A
8857 FCGBP Fc fragment of IgG binding protein
51316 PLAC8 Placenta-specific 8
10351 ABCA8 ATP-binding cassette, sub-family A (ABC1), member 8
9060 PAPSS2 3'-phosphoadenosine 5'-phosphosulfate synthase 2
10891 PPARGC1A Peroxisome proliferator-activated receptor gamma, coactivator 1 alpha
55286 C4orf19 Chromosome 4 open reading frame 19
25994 HIGD1A HIG1 domain family, member 1A
10924 SMPDL3A Sphingomyelin phosphodiesterase, acid-like 3A
1087 CEACAM7 Carcinoembryonic antigen-related cell adhesion molecule 7
2517 FUCA1 Fucosidase, alpha-L- 1, tissue
2647 BLOC1S1 Biogenesis of lysosome-related organelles complex-1, subunit 1
771 CA12 Carbonic anhydrase XII
58472 SQRDL Sulfide quinone reductase-like (yeast)
760 CA2 Carbonic anhydrase II
3957 LGALS2 Lectin, galactoside-binding, soluble, 2 (galectin 2) /// lectin, galactoside-binding, soluble, 2 (galectin 2)
7102 TSPAN7 Tetraspanin 7
11148 HHLA2 HERV-H LTR-associating 2
4306 NR3C2 Nuclear receptor subfamily 3, group C, member 2
9314 KLF4 Kruppel-like factor 4 (gut)
10050 SLC17A4 Solute carrier family 17 (sodium phosphate), member 4
81618 ITM2C Integral membrane protein 2C /// integral membrane protein 2C
2494 NR5A2 Nuclear receptor subfamily 5, group A, member 2
35 ACADS Acyl-Coenzyme A dehydrogenase, C-2 to C-3 short chain
1811 SLC26A3 Solute carrier family 26, member 3
6549 SLC9A2 Solute carrier family 9 (sodium/hydrogen exchanger), member 2
957 ENTPD5 Ectonucleoside triphosphate diphosphohydrolase 5
2767 GNA11 Guanine nucleotide binding protein (G protein), alpha 11 (Gq class)
608 TNFRSF17 Tumor necrosis factor receptor superfamily, member 17
10917 BTNL3 Butyrophilin-like 3
3291 HSD11B2 Hydroxysteroid (11-beta) dehydrogenase 2
7358 UGDH UDP-glucose dehydrogenase
8671 SLC4A4 Solute carrier family 4, sodium bicarbonate cotransporter, member 4
123887 ZG16 Zymogen granule protein 16
5794 PTPRH Protein tyrosine phosphatase, receptor type, H
5333 PLCD1 Phospholipase C, delta 1
79148 MMP28 Matrix metallopeptidase 28
10590 SCGN Secretagogin, EF-hand calcium binding protein
23140 ZZEF1 Zinc finger, ZZ-type with EF-hand domain 1
1836 SLC26A2 Solute carrier family 26 (sulfate transporter), member 2
11240 PADI2 Peptidyl arginine deiminase, type II
27250 PDCD4 Programmed cell death 4 (neoplastic transformation inhibitor)
762 CA4 Carbonic anhydrase IV
759 CA1 Carbonic anhydrase I
1113 CHGA Chromogranin A (parathyroid secretory protein 1)
2980 GUCA2A Guanylate cyclase activator 2A (guanylin)

Results and Discussion

Colorectal cancer still stands as a predominant cancer and is the secondand third-most common cancer in women and men, respectively [26]. Despite considerable breakthroughs in treatment, colorectal cancer mortality remains high, with 40–50 percent of patients dying as a result of their illness. Identification of differentially expressed genes associated with CRC itself can contribute to the overall understanding of the molecular changes that drive cellular and molecular changes in CRC tumour development. This improved understanding can potentially contribute to the development of new therapeutic options to improve the prognosis for CRC patients. This work conducted a metaanalysis of gene expression signatures generated from mRNA expression data across CRC tumours and eight other glandular tumours which are colonic in nature, to identify differentially expressed genes associated with CRC tumour formation. Genes that change in response to treatment may contribute to developing treatment resistance long-term.

In this study, out of the 55 genes from the positive CRC gene panel, 54 of them were continuously constant throughout all the gene signatures and for the negative gene CRC panel which had 77 genes, 75 of them were uniform across all the other gene signatures. To find out whether, these genes are statistically significant or not we did a T-test for calculating the p value in excel. Among the genes identified in this study, from the positive CRC gene panel, 54 common shared genes in the Positive CRC panel across all the signatures, 48 genes stood out having a statistical significance of (P value < 0.001). Whereas out of the 75 common shared genes in the negative CRC panel across all the signatures, 72 genes were of highly statistical significance of (P value <0.001). Among the genes identified, studies have shown that nuclear factor (erythroidderived 2)-like 3(NFE2L3) decreases colon cancer cell proliferation in vitro and tumor growth in vivo [30]. Interestingly, sterile alpha motif and leucine zipper containing kinase AZK (ZAK) gene has involved functionalities in lung cancer tumourigenesis process and JNK pathway activation [31]. Methyltransferase like 7A (METTL7A) have shown previous associations with thyroid cancer but not in lung, uterine, ovarian, gastric, esophagus, pancreatic, liver, or colorectal cancers via bioinformatic analysis [32]. Aken together, these results suggested that the GSEA-based meta-analysis approach used here was successful in identifying cancer-related genes with and without CRC tumourigenesis associations.

This work had observable gene detection limitations that may have biological implications. For example, gene expression changes commonly associated with CRC tumourigenesis in genes like MSH2 and MSH6 both on chromosome 2 and MLH1, on chromosome 3 were not found in this study [4,30,33]. Platform variations, both in gene inclusion and primer nucleotide sequence, can substantially impact results generated from this, or any, bioinformatics analysis. While MSH2 and MSH6 were included in the GSE113513 identification dataset, their T-scores were insufficient to make the 500 gene cut-off required of GSEA to maintain statistical accuracy. However, this is an inherent limitation with the usage of GSEA, based approach which can only be overcomes by switching to a non-GSEA based approach. However, if the desired outcome is a prioritized list of potential gene candidates for further laboratory or clinical examination, the GSEA-based approach used here suffices (Tables 3 and 4).

A lack of direct experimental or clinical evidence substantially limited the conclusions drawn from this study of purely bioinformatics comprised work. Follow-up experiments must be done using laboratory techniques, such as Western blotting or qRT-PCR, to confirm top gene candidate predictions would support these conclusions drawn exclusively from mRNA expression data using GSEA based approach. Further analysis examining gene expression data from colonic biopsy samples undergoing any type of treatment for CRC that mimics clinical samples is needed to assess the prediction of results portrayed here. Further, gene expression data directly from CRC patients would be of particular interest to further explore the results generated here. Such an examination of gene expression data from treated and untreated human biopsy colonic CRC tissue samples would be limited due to challenges acquiring samples from tumor location.

Conclusion

This work used mRNA micro-array expression data to predict genes potentially involved in developing CRC tumours examining gene signatures. Through a GSEA-based meta-analysis approach, 54 over- expressed genes, most important ones being SLC25A32, SLC22A3, CA12, CA4, CA1 and SLC4A4, were identified amongst the 54 over-expressed genes as being most associated with CRC tumourigenesis regardless of its association with any other glandular tumour in origin. Overall, this work demonstrated the usefulness of a meta-analysis approach used previously to detect genes associated with SARS infection in identifying genes associated with DIPG treatment through application on mRNA expression data. Also, further laboratory, wet lab experiments and clinical examination into the role these identified gene

expression changes play in developing CRC tumours might improve treatment options available and life expectancy for CRC patients.

Declaration of Interests

The authors declare no conflicts of interest.

References

  1. Haraldsdottir, Sigurdis, Hulda M. Einarsdottir, Agnes Smaradottir and Adalsteinn Gunnlaugsson, et al. "Colorectal cancer-review." Laeknabladid 100 (2014): 75-82.
  2. Google Scholar, Crossref

  3. Heinimann, Karl. "Hereditary colorectal cancer: Clinics, diagnostics and management." Therapeutische Umschau. Revue Therapeutique 75 (2018): 601-606.
  4. Indexed at, Google Scholar, Crossref

  5. Weitz, J., M. Koch, J. Debus and T. Höhler, et al. Colorectal cancer. Lancet 365 (2005):153-165.
  6. Crossref

  7. Bray, Freddie, Jacques Ferlay, Isabelle Soerjomataram and Rebecca L. Siegel, et al. "Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries." CA: A Cancer J Clin 68 (2018): 394-424.
  8. Indexed at, Google Scholar, Crossref

  9. Andersen, Stig E., Ida B. Andersen, Benny V. Jensen and Per Pfeiffer, et al. "A systematic review of observational studies of trifluridine/tipiracil (TAS-102) for metastatic colorectal cancer." Acta Oncologica 58 (2019): 1149-1157.
  10. Indexed at, Google Scholar, Crossref

  11. Jin, Shaoju, Yishuang Mu, Xiaodong Wang and Zhenru Liu, et al. "Overexpressed RACK1 is positively correlated with malignant degree of human colorectal carcinoma." Mol Biol Rep 41 (2014): 3393-3399.
  12. Indexed at, Google Scholar, Crossref

  13. Shangguan, H, S.Y. Tan and J.R. Zhang. "Bioinformatics analysis of gene expression profiles in hepatocellular carcinoma." Eur Rev Med Pharmacol Sci 19 (2015): 2054-2061.
  14. Indexed at, Google Scholar

  15. Yu, Jiekai, Xiaofen Li, Chenhan Zhong and Dan Li, et al. "High-throughput proteomics integrated with gene microarray for discovery of colorectal cancer potential biomarkers." Oncotarget 7 (2016): 75279.
  16. Indexed at, Google Scholar, Crossref

  17. Shen, Xingjie, Meng Yue, Fansheng Meng and Jingyu Zhu, et al. "Microarray analysis of differentially-expressed genes and linker genes associated with the molecular mechanism of colorectal cancer." Oncol Lett 12 (2016): 3250-3258.
  18. Indexed at, Google Scholar, Crossref

  19. Behrens, J. "The role of the Wnt signalling pathway in colorectal tumorigenesis." Biochem Soc Transac 33 (2005): 672-675.
  20. Google Scholar, Crossref

  21. Worthley, Daniel L. and Barbara A. Leggett. "Colorectal cancer: Molecular features and clinical opportunities." Clin Biochem Rev 31 (2010): 31.
  22. Indexed at, Google Scholar

  23. Grady, William M. "Genomic instability and colon cancer." Cancer Metas Rev 23 (2004): 11-27.
  24. Indexed at, Google Scholar, Crossref

  25. Fang, Zhengyu, Sumei Xu, Yiwen Xie and Wenxi Yan. "Identification of a prognostic gene signature of colon cancer using integrated bioinformatics analysis." World J Surg Oncol 19 (2021): 1-14.
  26. Indexed at, Google Scholar, Crossref

  27. Aran, Veronica, Ana Paula Victorino, Luiz Claudio Thuler and Carlos Gil Ferreira. "Colorectal cancer: Epidemiology, disease mechanisms and interventions to reduce onset and mortality." Clin Colorectal Cancer 15 (2016): 195-203.
  28. Indexed at, Google Scholar, Crossref

  29. Park, Amber and Laura K. Harris. "Gene expression meta-analysis reveals interferon-induced genes Associated with SARS infection in lungs." Front Immunol 2 (2021): 2924.
  30. Indexed at, Google Scholar, Crossref

  31. Clough, Emily and Tanya Barrett. "The gene expression omnibus database." In StatisticalGenomics, Humana Press, New York, United States.
  32. Indexed at, Google Scholar, Crossref

  33. Ryan, Bríd M., Krista A. Zanetti, Ana I. Robles and Aaron J. Schetter, et al. "Germline variation in NCF4, an innate immunity gene, is associated with an increased risk of colorectal cancer." Int J Cancer 134 (2014): 1399-1407.
  34. Indexed at, Google Scholar, Crossref

  35. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi
  36. Berdiel-Acer, Mireia, Rebeca Sanz-Pamplona, Alexandre Calon and Daniel Cuadras, et al. "Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information." Mol Oncol 8 (2014): 1290-1305.
  37. Indexed at, Google Scholar, Crossref

  38. Leiszter, Katalin, Ferenc Sipos, Orsolya Galamb and Tibor Krenacs, et al. "Promoter hypermethylation-related reduced somatostatin production promotes uncontrolled cell proliferation in colorectal cancer." PLoS One 10 (2015): e0118332.
  39. Indexed at, Google Scholar, Crossref

  40. Galamb, O, S. Spisák, F. Sipos and K. Tóth, et al. "Reversal of gene expression changes in the colorectal normal-adenoma pathway by NS398 selective COX2 inhibitor." British J Cancer 102 (2010): 765-773.
  41. Indexed at, Google Scholar, Crossref

  42. Alhopuro, Pia, Heli Sammalkorpi, Iina Niittymäki and Mia Biström, et al. "Candidate driver genes in microsatellite‐unstable colorectal cancer." Int J Cancer 130 (2012): 1558-1566.
  43. Indexed at, Google Scholar, Crossref

  44. Huang, Da Wei, Brad T. Sherman, Xin Zheng and Jun Yang, et al. "Extracting biological meaning from large gene lists with DAVID." Curr Protocols Bioinformat 27 (2009): 13-11.
  45. Indexed at, Google Scholar, Crossref

  46. Subramanian, Aravind, Pablo Tamayo, Vamsi K. Mootha and Sayan Mukherjee, et al. "Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles." Proceed Nat Acad Sci 102 (2005): 15545-15550.
  47. Indexed at, Google Scholar, Crossref

  48. Oosterwijk, E. and R.J. Gillies. "Targeting ion transport in cancer." Philosoph Transact Royal Soc 369 (2014): 20130107.
  49. Indexed at, Google Scholar, Crossref

  50. Kuipers, E.J, W.M. Grady, D. Lieberman and T. Seufferlein, et al. "Colorectal cancer.” Nat Rev Disease Primers 1 (2015): 15065.
  51. Google Scholar, Crossref

  52. Bureau, Christophe, Naima Hanoun, Jérôme Torrisani and Jean-Pierre Vinel, et al. "Expression and function of Kruppel like-factors (KLF) in carcinogenesis." Curr Genomics 10 (2009): 353-360.
  53. Indexed at, Google Scholar, Crossref

  54. Boland, C. Richard and Ajay Goel. "Microsatellite instability in colorectal cancer." Gastroenterol 138 (2010): 2073-2087.
  55. Indexed at, Google Scholar, Crossref

  56. Goodman, Julie E., Elise D. Bowman, Stephen J. Chanock and Anthony J. Alberg, et al. "Arachidonate lipoxygenase (ALOX) and cyclooxygenase (COX) polymorphisms and colon cancer risk." Carcinogenesis 25 (2004): 2467-2472.
  57. Indexed at, Google Scholar, Crossref

  58. Bury, Marina, Benjamin Le Calve, Frédéric Lessard and Thomas Dal Maso, et al. "NFE2L3 controls colon cancer cell growth through regulation of DUX4, a CDK1 inhibitor." Cell Rep 29 (2019): 1469-1481.
  59. Indexed at, Google Scholar, Crossref

  60. Yang, Jaw‐Ji, Yi‐Ju Lee, Hsin‐Hung Hung and Wei‐Pu Tseng, et al. "ZAK inhibits human lung cancer cell growth via ERK and JNK activation in an AP‐1‐dependent manner." Cancer Sci 101 (2010): 1374-1381.
  61. Indexed at, Google Scholar, Crossref

  62. Zhou, Shasha, Yihang Shen, Min Zheng and Linlin Wang, et al. "DNA methylation of METTL7A gene body regulates its transcriptional level in thyroid cancer." Oncotarget 8 (2017): 34652.
  63. Indexed at, Google Scholar, Crossref

  64. https://www.ncbi.nlm.nih.gov/books/NBK22218/
Google Scholar citation report
Citations: 3968

Cancer Science & Therapy received 3968 citations as per Google Scholar report

Cancer Science & Therapy peer review process verified at publons

Indexed In

arrow_upward arrow_upward