Open Access

Copy number alterations and epithelial‑mesenchymal transition genes in diffuse and intestinal gastric cancers in Mexican patients

  • Authors:
    • Violeta Larios-Serrato
    • José-Darío Martínez‑Ezquerro
    • Hilda-Alicia Valdez‑Salazar
    • Javier Torres
    • Margarita Camorlinga‑Ponce
    • Patricia Piña‑Sánchez
    • Martha-Eugenia Ruiz‑Tachiquín
  • View Affiliations

  • Published online on: March 31, 2022     https://doi.org/10.3892/mmr.2022.12707
  • Article Number: 191
  • Copyright: © Larios-Serrato et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is a common malignancy with the highest mortality rate among diseases of the digestive system, worldwide. The present study of GC alterations is crucial to the understanding of tumor biology and the establishment of important aspects of cancer prognosis and treatment response. In the present study, DNA from Mexican patients with diffuse GC (DGC), intestinal GC (IGC) or non‑atrophic gastritis (NAG; control) was purified and whole‑genome analysis was performed with high‑density arrays. Shared and unique copy number alterations (CNA) were identified between the different tissues involving key genes and signaling pathways associated with cancer. This led to the molecular distinction and identification of the most relevant molecular functions to be identified. A more detailed bioinformatics analysis of epithelial‑mesenchymal transition (EMT) genes revealed that the altered network associated with chromosomal alterations included 11 genes that were shared between DGC, IGC and NAG, as well as 19 DGC‑ and 7 IGC‑exclusive genes. Furthermore, the main molecular functions included adhesion, angiogenesis, migration, metastasis, morphogenesis, proliferation and survival. The present study provided the first whole‑genome high‑density array analysis in Mexican patients with GC and revealed shared and exclusive CNA‑associated genes in DGC and IGC. In addition, a bioinformatics‑predicted network was generated, focusing on CNA‑altered genes associated with EMT and the hallmarks of cancer, as well as precancerous alterations that may lead to GC. Molecular signatures of diffuse and intestinal GC, predicted bioinformatically, involve common and distinct CNA‑EMT genes related to the hallmarks of cancer that are potential candidates for screening biomarkers of GC, including early stages.

Introduction

According to the Global Cancer Observatory statistics, cancer is the leading cause of death in the world, with 9.9 million deaths in 2020; the incidence rate of cancer was 20% in the Caribbean and South America, with high mortality rates (14%). Worldwide, gastric cancer (GC) is estimated to be the fifth most common cancer type in both sexes, ranking sixth for new cases, with over one million cases per year and third in mortality (1).

In Mexico, according to statistics from the National Institute of Statistics and Geography, three out of 10 cancer-associated deaths among patients aged 30–59 years were due to cancer of the digestive system. From 2011 to 2016, four out of 10 and three out of 10 cancer-associated deaths respectively occurred in females and males aged >60 years and resulted from tumors in digestive organs (2).

GC refers to any malignancy originating in the region between the gastroesophageal junction and the pylorus. The World Health Organization and the Lauren classification system (3) have classified GC into two types: Intestinal GC (IGC) and diffuse GC (DGC). Intestinal or differentiated gastric cancer is characterized by localized and expansive growth, while DGC has an infiltrating growth pattern, is an undifferentiated adenocarcinoma and features dispersed cells with individual or group invasive capacity (4). The development of IGC is preceded by a precancerous process of several years and stages: Active chronic gastritis, multifocal atrophic gastritis, complete intestinal metaplasia, incomplete intestinal metaplasia, dysplasia and adenocarcinoma (5). GC has a multifactorial origin: Diet, lifestyle, genetics and socioeconomic factors, and it has been observed that 80% of cases of IGC are associated with previous Helicobacter pylori (H. pylori) infection (6,7). GC is characterized by a complex etiology with a set of factors, including genetic alterations and external factors. However, it has been reported that <3% of GC is due to heredity and includes hereditary DGC, proximal polyposis of the stomach and hereditary colorectal cancer not associated with polyposis (6). With respect to molecular pathogenesis, chromosomal instability (aneuploidy, chromosomal translocation, amplification, deletions and loss of heterozygosity), gene fusion and microsatellite instability (hypermethylation of gene repair promoters) are involved (7).

Copy number alterations (CNA) represent a class of genetic variation that involve cumulative somatic variations. CNA are defined as non-inherited genetic alterations that occur in somatic cells (8). These unbalanced structural variants usually contain gains or losses. Their interpretation and the CNA report continue to be a topic of interest in health and have an important role in GC (9,10).

The majority of gastric adenocarcinomas, similar to numerous other types of solid tumor, exhibit defects in the maintenance of genome stability, resulting in DNA CNA that may be analyzed using comparative genomic hybridization (CGH) (11). This is a widespread and common phenomenon among humans and several studies have focused on understanding these genomic alterations that are responsible for cancer and may be used for its diagnosis and prognosis (12).

At present, there are few published studies involving genotyping of GC samples using high-density microarrays (1315); however, in those altered chromosomes, gains and losses have a phenotypic impact and different signaling pathways are involved. The presence of CNA changes the genetic dose and would modify several molecular mechanisms, such as epithelial-mesenchymal transition (EMT), which is the transformation of epithelial cells to mesenchymal cells and is a critical stage for the transition to metastasis (16). There are currently >1,184 genes in the EMT Gene Database (dbEMT 2.0), which are involved in other cancer-related processes, such as proliferative signaling, evasion of growth suppressors, avoidance of immune destruction, inactivation of replicative immortality, tumor-promoting inflammation, induction of angiogenesis, genomic instability, mutation, resting cell death, deregulation of cellular energetic activity, invasion and cell plasticity (17). EMT includes activation of transcription factors, expression of specific cell-surface proteins, reorganization and expression of cytoskeletal proteins and the production of extracellular matrix-degrading enzymes (18). EMT has been associated with the progression of cancer and increased stemness of tumors (18,19), and was observed to be involved in the formation, invasion and metastasis of GC (20,21). In addition, an association has been established between the presence of CNA and its effect on the expression level of EMT-associated genes in different cancer cell lines (22). Studies on CNA events involving Latin American populations are limited (23,24). In fact, at present, only a small number of studies have performed GC genotyping using whole-genome high-density microarrays in Mexican patients with GC (14,15). Therefore, the present study aimed to determine CNA in DGC and IGC to identify, through bioinformatics analyses, the main genes and signaling pathways involving EMT-associated genes.

Materials and methods

Samples

Institutional Review Board approval was obtained for the present study (approval no. 2008-785-001). The samples were collected at the Regional General Hospital No. 1 ‘Dr. Carlos MacGregor Sánchez Navarro’, Specialty Hospital ‘Dr. Bernardo Sepúlveda’ and Oncology Hospital from IMSS in Mexico City (Mexico). Clinical data and patient samples were processed following obtainment of written informed consent. All of the samples were collected over three years (April 2010 to May 2013) following standardized endoscopy preservation protocols (25). Histological assessment of the biopsies was performed by two trained pathologists independently. They assigned the phenotypic diagnosis of diffuse or intestinal tumors and non-atrophic gastritis (NAG) samples. Only samples with the same diagnosis (‘identical results’) by two independent expert pathologists were included in the analysis.

A total of 21 patients (5 females and 16 males) with tissue samples that met the criteria for DGC (n=7) and IGC (n=7) diagnoses, as well as subjects with NAG (n=7) as controls, were included. In the absence of an established measurement (gold standard), a value was arbitrarily determined to provide guidance to investigate relevant alterations. To identify the most relevant alterations for GC, the present analysis focused on alterations present in at least three patients (cut-off, ≥3 patients; ≥40% samples).

DNA extraction

DNA extraction was performed using a commercial kit (QIAamp® DNA Micro Kit; Qiagen GmbH) according to the manufacturer's protocol. The extraction was modified to include an initial incubation at 95°C for 15 min, followed by a 5-min incubation at room temperature, prior to digestion with proteinase K (Qiagen GmbH) for three days at 56°C in a water bath, and fresh enzyme was added at 24 h intervals, as described previously (26).

DNA quality assessment and preparation

The extracted DNA was quantified using spectrophotometry (Nanodrop 2000; Thermo Fisher Scientific, Inc.). Multiplex PCR was performed to assess the quality of DNA (Multiplex PCR kit; Qiagen GmbH) with a set of primers to amplify various regions of the GAPDH gene (27). Products were visualized using 1% agarose gel electrophoresis (RedGel® Nucleic acid gel stain; Biotium) and documented under an ultraviolet light transilluminator system (Syngene).

High-density whole-genome microarray analysis

The samples were analyzed using the Affymetrix® CytoScan™ microarray (Affymetrix; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol and with 250 ng DNA, except for the addition of five PCR cycles to increase the DNA sample. The PCR products (90 µg) were fragmented and labeled using additional PCR (https://assets.thermofisher.com/TFS-Assets/LSG/manuals/703038_cytoscan_assay_UG.pdf).

Copy number processing

The raw intensity files (.CEL), retrieved from the commercial platform, were analyzed using their proprietary software, Chromosome Analysis Suite (ChAS) v3.2 and NetAff 33 Libraries, based on the construction of the hg19 genome (February 2009) as a reference model.

Data processing was based on the segmentation algorithm, where the Log2 ratio for each marker was calculated relative to the reference signal profile. To calculate the copy number variation (CNV), the data were normalized to baseline reference intensities using the reference model (provided by ChAS), including 270 HapMap samples and 96 healthy individuals. The Hidden Markov Model, available in ChAS, was used to determine the CN state and their breakpoints. The customized high-resolution condition was used as a filter for the determination of CNV: CN gains with a 50-marker count and 400 Kb, and CN losses with a 50-marker count and 100 Kb. The median absolute pairwise difference (MAPD) and the single nucleotide polymorphism quality control (SNP QC) score were used as the quality control parameters. Only samples with values of MAPD >0.25 and SNP QC <15 were included in the further analysis.

Bioinformatics analysis

A Perl script was developed to load the CNV segment data files generated by ChAS for each sample to compare the files to generate a list of genes that contained event types (gains or losses), frequencies of altered regions, including chromosomes and cytogenetic bands and Online Mendelian Inheritance in Man information, and to incorporate additional information from different databases (haploinsufficiency information from the DECIPHER database of genomic variation, genes reported at dbEMT 2.0 and genes affected in gastric adenocarcinoma from Harmonized Cancer Datasets; Table SI).

The genes altered in at least three patients (cut-off, ≥3) with DGC, IGC or NAG were included for analysis and visualizations were performed using R v4.0.2 and Bioconductor v3.12 packages (Table SII). The karyotype was created with KaryoploteR and the Bioconductor software annotation package (BSgenome.Hsapiens.UCSC.hg19 v1.4.0). The comparison among samples was performed by generating Venn diagrams with the jvenn server and a heatmap with gplots. Gene Ontology (GO) analyses were performed with the ClusterProfiler v3.16.1 packages (org.Hs.eg.db v3.11.4, enrich plot v1.8.1 and GOplot v1.0.2), with the support of functional enrichment analysis using the database for annotation, visualization and integrated discovery (DAVID) v6.8 resource (Table SII). The profile of altered molecular function (MF) terms in GC was summarized according to the proportion of CNA-associated genes and the MF GO terms from the DAVID database, adjusted by the false discovery rate. Dot plots, heatmaps and chord plots were utilized to visualize the GC CNA profiles for DGC, IGC and NAG.

To identify the main genes and signaling pathways involving CNA EMT-associated genes, GC CNA-associated genes (cut-off, ≥3) were analyzed and compared according to those previously reported in the dbEMT 2.0, accessed on 12th October, 2020.

Finally, to establish the profile-associated hallmarks of cancer involving DGC, IGC and NAG EMT-associated genes, an interaction network was generated using CNA type (gains and losses) based on genetic and physical interactions and biological pathways. Furthermore, associations were determined using the GeneMANIA prediction server and Cytoscape v.3.8.2, including the manual annotation of their corresponding cancer hallmarks [adhesion, angiogenesis, inflammation, migration, metastasis, morphogenesis, proliferation and survival (28)], with punctual scrutiny and assistance from databases, such as The Human Protein Atlas. Table SII provides information on the databases, protocols, software and specific packages used.

Results

Sample characteristics

Samples from 21 patients with GC from Mexico (third-generation Mexicans) between 35 and 91 years of age (mean ± SD, 59.61 ± 15.94 years), without any previous cancer treatment (naïve) were included in the present study. The samples included seven cases who had DGC, seven who had IGC and seven who had NAG (control samples). The raw data were deposited in the NCBI Gene Expression Omnibus database (ID no. GSE117093). There are seven adjacent tissue files (.CEL); however, these files were not included in the data analysis, as certain adjacent tissues were contaminated with cancer cells or these were not of the quality required for subsequent analyses.

Table I presents the ID and the percentage of neoplastic cells for tumor tissues ranging between 50 and 70%. Blood agar culture indicated that one patient with IGC and three patients with NAG were positive for H. pylori (data obtained from our biobank database). The patient data are also presented in Table I (2931).

Table I.

Characteristics of GC and NAG cases analyzed in the present study (n=21).

Table I.

Characteristics of GC and NAG cases analyzed in the present study (n=21).

IDAge, yearsSexCancer type% CCH. pyloriTNMTreatment
3CG-00872MIntestinal70PositiveIB T1 N1 M0Naïve
3CG-12680MIntestinal60NegativeIIA T4 N0 M0Naïve
3CG-12891MIntestinal70NegativeIIA T3 N2 M0Naïve
3CG-04652FIntestinal60NegativeIV T4 N2 M0Naïve
3CG-09959MIntestinal50NegativeII T3 N0 M0Naïve
3CG-14671MIntestinal60NegativeIIB T3 N2 M0Naïve
3CG-10469MIntestinal60NegativeIII A T4 N0 M0Naïve
3CG-04758MDiffuse70NegativeIV T4 N3 M0Naïve
3CG-17376MDiffuse70NegativeIII A T2 N3 M0Naïve
8CG-00476MDiffuse70NegativeII T1 N0 M0Naïve
1CG-00145MDiffuse60NegativeIV T4N2M1Naïve
3CG-03555MDiffuse60NegativeIV T4N2M0Naïve
3CG-04264MDiffuse50NegativeIV T4, N2 M0Naïve
3CG-06438MDiffuse50NegativeIV T4 N2 M0Naïve
4GB-00164MNAG0NegativeNANA
4GB-03162MNAG0NegativeNANA
4GB-01535FNAG0NegativeNANA
4GB-02539MNAG0PositiveNANA
4GB-03376FNAG0PositiveNANA
4GB-03638FNAG0PositiveNANA
4GB-04277FNAG0NegativeNANA

[i] ID, identification code; CC, cancer cells; M, male; F, female; NA, not applicable; TNM-T, extent of the primary tumor; TNM-N, absence or presence and extent of regional lymph node involvement; TNM-M, absence or presence of distant metastasis; NAG, non-atrophic gastritis.

Genomic detection of CNA

The total number of CNAs was obtained and they were classified as either gains or losses for each chromosome in the GC and NAG samples. From the total CNAs, DGC had more CNAs compared to IGC (3,505 and 2,781, respectively), while there were 828 events in the NAG samples. With respect to the tissue, more gains than losses were observed in both cancer types, DGC (2,310 and 1,195, respectively) and IGC (1,550 and 1,231, respectively), but the opposite was observed in NAG (375 and 453, respectively) (Table SIII).

To identify the most relevant CNA in GC and NAG, alterations occurring in at least three patients (cut-off, ≥3) were analyzed. This comparison indicated a similar pattern for total CNA, with more events in DGC (n=710) than in IGC (n=590) or in NAG (n=332). In addition, more gains than losses were observed in DGC (516 and 194, respectively), IGC (314 and 276, respectively) and even in NAG (196 and 136, respectively), which was different when all of the patients were included. Furthermore, DGC had the highest number of gains and IGC had the highest number of losses (Table SIII). Table II lists chromosomes and sizes with gain and loss numbers, representative and summarized.

Table II.

Principal affected chromosomes by CNA cumulative length in DGC, IGC and NAG.

Table II.

Principal affected chromosomes by CNA cumulative length in DGC, IGC and NAG.

Type/chromosomeGainsLossesLength, Mb-cl
DGC
  1327-117.9
  4-15540.8
  5-14874.23
IGC
  1-14833.78
  8365-139.8
  X-66167.1
NAG
  6-280.40
  721-0.20
  1410-3.02
  17-151.86
  X-870.47
  X207-1.20

[i] The locations of copy number alterations are presented in Table SIV. DGC, diffuse gastric cancer; IGC, intestinal gastric cancer; NAG, non-atrophic gastritis; Mb-cl, Megabases cumulative length.

To visualize the distribution of DGC and IGC chromosome gains and losses, the identified CNA present in a karyogram (cut-off, ≥3) was plotted, which displayed alterations according to the coordinates of the Human genome hg19 (Fig. 1). The top five altered cytobands are provided in Tables III and SIV.

Table III.

Top five altered cytobands in DGC, IGC and NAG.

Table III.

Top five altered cytobands in DGC, IGC and NAG.

Type/cytobandGainsLength, Mb-clNumber of patients
DGC
  Xq283711.56
  8q24.222515.85
  1q32.12612.165
  8q24.32517.466
  1q23.32215.266
IGC
  8q24.32412.6624
  13q34184.90184
  8q24.21186.26194
  8q24.22166.48564
  8q12.1165.85234
NAG
  Xq26.22434.0266
  Xq21.22233.5867
  Xp22.3319167.5047
  Xq231663.796
  Xq26.313217.7747

[i] DGC, diffuse gastric cancer; IGC, intestinal gastric cancer; NAG, non-atrophic gastritis; Mb-cl, Megabases cumulative length.

Of note, in DGC and IGC, the most frequent CNA lengths were between 100 and 200 Kb, while lengths of 1–50 Kb were more common in NAG, with respect to gains and losses (Table SV).

GC genes associated with CNA

Overall, 2,441 CNA-associated genes were identified in DGC, IGC and NAG. GC had 2,420 affected genes (99%), while only 108 genes (4%) were affected in NAG; of these alterations, certain candidates were shared between GC and NAG. There were 1,317 unique CNA-associated genes in DGC, 596 in IGC and 21 in NAG. Furthermore, both cancer types shared 420 genes, while 60 genes were shared between GC and NAG. In addition, 19 genes in NAG were shared with DGC and eight genes with IGC (Fig. 2A; Table SVI).

To identify the possible emerging patterns among the samples, hierarchical clustering heatmaps were generated (Fig. 2B). The results provided the molecular signature and hierarchical clustering of samples according to the 2,441 genes. The emerging pattern of altered genes affected by CNA distinguishes DGC and IGC from NAG.

GO analysis of GC

The functional profile for GC was generated through enrichment and GO analysis of the 2,420 GC-altered genes; 1,317 genes were only altered in DGC and 596 only altered in IGC. To identify the principal MFs altered in GC, these CNA-associated genes were categorized, independently of the GC type, into two groups: Gains and losses (Fig. 3A and B). The top 10 MFs associated with CNA gains or losses revealed that transcription activator, tyrosine kinase activity, growth factors and hormone binding, as well as intracellular signal transduction genes were enriched in GC. Gene losses mainly involved transcription coactivator and serine/threonine kinase activity, as well as several receptors binding to hormone, steroid hormone, nuclear receptor, β-catenin, intermediate filament and mitogen-activated protein kinase binding genes. In addition, the principal CNA-associated genes affecting the MF by GC type (DGC and IGC) were identified (Figs. 3C and D and S1).

CNA-EMT genes in DGC and IGC

To identify the main genes and signaling pathways involving the CNA-EMT genes in GC and NAG, GC CNA-associated genes were compared against a comprehensive and annotated database of EMT genes (dbEMT 2.0). A total of 551 CNA-EMT genes were found in DGC, 619 in IGC and 28 in NAG. Using the cut-off ≥3, 112 genes in DGC, 66 in IGC and 5 in NAG were obtained. The complete data of EMT-associated genes for DGC, IGC and NAG, with chromosome and cytoband locations, CNA type (gain or loss), and the P-values are provided in Table SI.

GO analysis of the EMT-associated genes

GO enrichment analysis was performed to determine the MF of the main CNA-EMT genes affected in DGC, IGC and NAG (Fig. 4). The results indicated that gains in the CNA-EMT genes in DGC were associated with transmembrane receptor tyrosine kinase, DNA and RNA binding and receptor binding for insulin, growth factors, Toll-like receptors, hormone, as well as SMAD, p53, chromatin, calcium ion binding and microtubule binding (Fig. 4A). Losses in CNA-EMT genes included associations with DNA and chromatin binding, nuclear hormone receptor binding, β-catenin, steroid hormone, mitogen-activated protein binding, intermediate filament binding, p53 binding and RNA polymerase II-specific DNA binding (Fig. 4B). Furthermore, gains in CNA-EMT genes in IGC were associated with insulin receptor substrate and phosphatase binding, kinase regulation, neurotrophin receptor binding, 1-phosphatidylinositol-3-kinase activity, transmembrane receptor protein tyrosine kinase adaptor activity and VEGF receptor binding, while losses in CNA-EMT genes in IGC were only associated with coenzyme binding and transcription coactivator activity (Fig. 4C). On the other hand, the main MF for gains in the CNA-EMT genes in NAG included transcription regulatory region DNA, transcriptional activator activity RNA, armadillo repeat and C2H2 zinc finger domain binding, γ- and β-catenin binding, as well as cysteine-type endopeptidase inhibitor activity involved in apoptotic process and estrogen receptor, as well as steroid hormone receptor activity, while losses in CNA-EMT genes in NAG were associated with damaged DNA, WW domain, p53 binding and DNA-binding transcription activator activity (Fig. 4D).

CNA-EMT genes associated with the hallmarks of cancer

Based on the main molecular profile of altered CNA-EMT genes in GC and NAG, the functional network between 39 previously selected unique CNA-EMT genes (19 genes for DGC, 7 for IGC, 11 common to GC and two for NAG; cut-off, ≥3 patients) was generated. Gained genes, with the highest degree and at least four interactions per gene, were EGFR, MICAL2, MYC, NDRG and PIK3R1, while lost genes included GLI2, EP300 and PTPN11. The principal functions associated with these CNA-EMT genes have been previously associated with several hallmarks of cancer: Adhesion, angiogenesis, inflammation, migration, metastasis, morphogenesis, proliferation and survival (Fig. 5).

Discussion

To the best of our knowledge, the present study was the first whole-genome high-density array study on GC in Mexican patients with DGC and IGC, as well as NAG as non-cancerous controls. Using this experimental strategy, it was possible to generate a karyogram and obtain molecular signatures for DGC and IGC, and their association with CNA-EMT genes, independent of age, sex, percentage of cancer cells, presence/absence of H. pylori infection, TNM and treatment (naïve samples in the present study). In addition, the genomic analysis was focused on the molecular profile of GC, particularly involving alterations of EMT-associated genes, given their role in cancer progression, as epithelial cell transformation to mesenchymal cells is fundamental to metastasis (32) and chemoresistance (33,34). The results of the present study are consistent with those previously reported in the literature (detailed above), which provides validity and robustness to the results and enables the reporting of novel data or data not yet investigated to identify potential diagnostic, prognostic and treatment response markers.

Globally, the alteration profile in GC was dominated by gains. This phenomenon, where gains are more abundant than losses, has been previously reported in different tumor cell lines, including gastric cancer cell lines (35). Chromosomal gains in cancer may result in increased gene functions, providing cancer cells with a competitive advantage for the development of metastasis (36), while chromosomal losses may involve the downregulation of tumor suppressor genes (37), disrupting homeostasis and accelerating cancer progression. The most affected CNA chromosomes for DGC were 1, 4 and 5; for IGC 1, 8 and X, and for NAG 6, 7, 14, 17 and X. The altered cytobands associated with GC observed in the present study are in agreement with previous studies. For instance, 8q24 has been associated with the development of different types of tumor (38). The highest frequencies of gains in advanced GC were found at 8q24.21 (65%) and 8q24.3 (60%), and the pattern of CNA in advanced GC was different from that in early GC. This increase in CNA numbers is associated with disease progression from early to advanced GC (39). The 8q24 cytoband has also been reported in Latin American countries, such as Brazil (40) and Venezuela (41), as well as in Asian countries, including Korea (42).

Of note, the most frequent CNA length in GC was 100–200 Kb, in both DGC and IGC compared with 1–50 Kb in NAG. The biological implications of this difference in length in GC compared with non-cancerous tissues, such as NAG, is yet to be determined. Furthermore, it is important to highlight that a resolution of 100–200 Kb versus Mb is an advantage of molecular resolution approaches over classical cytogenetics (CGH and fluorescence in situ hybridization) to discover ‘small’ potentially important alterations in cancer samples.

The cumulative length averages (Megabases, Mb) of these alterations were 183.44 Mb for DGC, 113.56 Mb for IGC and 1.19 Mb for NAG. These lengths, whether gained or lost, describe the magnitude of global alterations per tissue; however, their relevance lies in the MFs, biological processes and interaction networks in which they participate.

In the present study, a molecular profile that distinguishes GC from NAG was identified based on 2,441 genes affected by CNA. They are associated with GC, as well as the differences and similarities among histological subtypes (undifferentiated DGC and well-differentiated IGC) compared with that in non-cancerous tissue, such as NAG (43). Of note, 60 affected genes shared between GC and NAG were identified; 19 genes were shared exclusively with DGC, while only eight were shared with IGC. This emerging pattern of shared altered genes between cancerous and non-cancerous tissues should be further studied to identify possible CNA-dependent oncogenic pathways and progression trajectories from NAG to either GC subtype, particularly in conjunction with environmental factors, such as H. pylori infection, diet and lifestyle, that may be associated with the spread patterns affecting patient survival (43).

In the heatmap, a separation between NAG and GC was observed, exhibiting clusters based on the molecular profiles of CNA-associated genes. There is a greater heterogeneity among the IGC samples in clusters but there were more genes affected in DGC. The front-line tool for IGC distinction has been based on different criteria, such as the Lauren histopathological classification system. However, due to challenges, including disagreements in the correct assignment, diagnosis and treatment, new criteria have been proposed, such as molecular characterization according to The Cancer Genome Atlas (TCGA) Research Network, which divides GC into four subtypes (44). The results of the present study agree with the requirement for new proposals for the classification of GC, which includes defined subgroups, with the integration of several genomic and genetic parameters where CNA are present.

In the present study, the MF profile of GC CNA-associated genes was analyzed and determined. With respect to gains, there were increased alterations involving transcription, signaling, tyrosine kinases, growth factors, hormones and insulin, while with respect to losses, molecules involved in transcription, serine/threonine and MAP kinases, steroid hormones, β-catenin binding and filament binding were decreased. These gene sets are important in GC biology. There were 13 CNA genes in IGC (including CDH1, LAST1, ROCK1 and WWOX) and 49 CNA genes in DGC (including CRIM1, EGFR, MIR9-1, MUC1, MYC, NDRG1, SCRIB, SNAI2, VEGF and ZEB2); therefore, these genes were further analyzed with the intention of comparing the results of the present study with those of others and organizing the data in a biologically coherent context. For instance, CDH1 codes for E-cadherin and, from a simplified viewpoint, E-cadherin maintains the epithelial phenotype; if CDH1 is lost, this promotes the mesenchymal phenotype, i.e., it favors loss of adhesion and metastasis (32).

In the present study, an enrichment analysis of unique CNA-associated genes for all tissues was performed and several shared MFs, such as protein binding, were obtained. Several gained-genes that encode for RNA-binding proteins (45) have diverse targets and participate in tumor progression by regulating homeostasis and changing expression patterns. Chromatin binding is another altered function in GC that participates in regulating eukaryotic gene expression, methylation profile modulation, and genome stability maintenance (46). EMT is a process that involves changes in histone modification, DNA methylation and chromatin accessibility. These changes may be promoted through transcription, allowing the cell to have an identity or to have a mesenchymal-epithelial transition-EMT conversion (47). The kinase function in DGC and IGC gains have recently been considered key regulators in the development of cancer (48). Numerous kinases were associated with the initiation and progression of carcinogenesis and are one of the main therapeutic targets for the development of inhibitors in the clinical field. Kinases are able to promote EMT and enhance invasion, migration and evasion of apoptosis (49). In the present study, PIK3R1 and PIK3CA were associated with IGC. The PI3K pathway is a key regulatory hub for cell growth, survival and metabolism (50). Activation of PI3K is a frequent hallmark of cancer, highlighted by the prevalence of somatic mutations in genes encoding key components of this pathway (51). These enzymes are responsible for transferring a phosphate group; however, the reverse process is performed by phosphatases, which are also affected in IGC. PIK3R1 is a gene frequently affected by mutations or copy numbers in various types of cancer, according to the TCGA project. These genes converge with the PI3K/AKT/mTOR pathway, which is involved in the regulation of several processes (51).

To date, the differences between DGC and IGC have been insufficiently investigated and understood; differences in etiology, location, incidence and genetic profiles have been observed (52). In the present study, a CNA-EMT network for GC was generated with relevant genes according to different criteria: Frequency among patients, genetic connections, reported pathways and experimental associations with several databases [dbEMT 2.0, The Human Protein Atlas, COSMIC (53)] and Cancer Hallmark Genes (54). Shared and exclusively altered genes were observed for each tissue type. The common CNA-EMT genes between DGC and IGC include GLI2, which has been associated with proliferation (55); EP300, with multiple functions as an inhibitor of antitumor immune response via metabolic modulation (56); PTPN11, associated with GC progression; and NDRG1, associated with metastasis and poor prognosis in GC (57). Another relevant gene in DGC is EGFR, which, due to its association with CNV GC, is now the target for the development of anti-GC therapies (58). IGC-associated EMT genes include MICAL2, MYC and PIK3R1. MICAL2, a destabilizing F-actin in cytoskeletal dynamics, has been associated with poor prognosis in GC (59). MYC gains have also been reported in several GC studies, as expected for a common oncogenic gene (60) associated with proliferation, differentiation and apoptosis (61). PIK3R1 participates in the PI3K/AKT signaling pathway, with roles in apoptosis and cell survival, as well as chemotherapy resistance in GC (62).

A large amount of data remains to be analyzed, including loss of heterozygosity, mosaicism and other gene sets that participate in different hallmarks of cancer. Another limitation of the present study was the absence of a transcriptomic analysis to validate the GC EMT signature, particularly for DGC and IGC. Yet, the concordance of CNA with expression alterations in EMT-associated genes is plausible, as previously observed for multiple types of cancer from TCGA (35). In addition, further inclusion of precancerous stages would allow further analysis of the ‘profile’ of IGC progression. The results of the present genomic approach coincide with those already reported in the literature, which provides validity and solidity to the results. After all, this strategy allowed us to report novel or thus far scarce data, or those not previously investigated, to identify differential GC CNA, identify associations with relevant MFs associated with the hallmarks of cancer and predict the EMT signature for DGC and IGC. It may be hypothesized that these networks will potentially provide treatment targets, as well as diagnostic and prognostic markers. In addition, the use of NAG as a non-malignant control allowed for investigation of the molecular and cellular events of GC and the identification of potential biomarkers for the ‘early’ stages of GC.

Supplementary Material

Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Ms. Irma P. Ramos-Vega, Infectious and Parasitic Diseases Medical Research Unit (UIMEIP), High Specialty Medical Unit (UMAE)-Pediatrics Hospital ‘Dr. Silvestre Frenk Freund’, XXI Century National Medical Center, IMSS; Mr. Brian-Alexander Cruz-Ramírez and Ms. Alejandra García-Bejarano, Oncological Diseases Medical Research Unit (UIMEO), UMAE-Oncology Hospital, XXI Century National Medical Center, IMSS for their technical assistance.

Funding

The present study was supported by the Fondo de Investigación en Salud-Instituto Mexicano del Seguro Social (grant nos. FIS/IMSS/PROT/G16/1573 and FIS/IMSS/PROT/PRIO/13/027).

Availability of data and materials

All datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request. The data have been deposited in the Gene Expression Omnibus database (GEO; http://www.ncbi.nlm.nih.gov/geo/) under accession no. GSE117093. Pre-print: doi: https://doi.org/10.1101/2021.11.22.469612.

Authors' contributions

VLS, HAVS and JDME performed the molecular experiments, participated in data analysis to provide the results and prepared, wrote and discussed the manuscript. JT, MCP and PPS were responsible for clinical aspects and recruited the patients, discussed the data and revised the manuscript. MERT contributed to the design of the study, supervised the study and critically reviewed, revised and wrote the manuscript. JT and MERT were responsible for acquiring financial support. VLS and MERT confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Institutional Review Board approval was obtained for the study (approval no. 2008-785-001). Clinical data and patient samples were processed following written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

INEGI-Social Communication, . Statistics on World Cancer Day (February 4)-National Data. INEGI. 2018.

3 

Lauren P: The two histological main types of gastric carcinoma: Diffuse and so-called intestinal-type carcinoma: An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 64:31–49. 1965. View Article : Google Scholar : PubMed/NCBI

4 

Espejo Romero H and Navarrete Siancas J: Classification of stomach adenocarcinomas. Rev Gastroenterol Peru. 23:199–212. 2003.(In Spanish). PubMed/NCBI

5 

Piazuelo MB, Epplein M and Correa P: Gastric cancer: An infectious disease. Infect Dis Clin North Am. 24853–869. (VII)2010. View Article : Google Scholar : PubMed/NCBI

6 

Nagini S: Carcinoma of the stomach: A review of epidemiology, pathogenesis, molecular genetics and chemoprevention. World J Gastrointest Oncol. 4:156–169. 2012. View Article : Google Scholar : PubMed/NCBI

7 

McLean MH and El-Omar EM: Genetics of gastric cancer. Nat Rev Gastroenterol Hepatol. 11:664–674. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Mikhail FM, Biegel JA, Cooley LD, Dubuc AM, Hirsch B, Horner VL, Newman S, Shao L, Wolff DJ and Raca G: Technical laboratory standards for interpretation and reporting of acquired copy-number abnormalities and copy-neutral loss of heterozygosity in neoplastic disorders: A joint consensus recommendation from the American College of Medical Genetics and Genomics (ACMG) and the Cancer Genomics Consortium (CGC). Genet Med. 21:1903–1916. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Wallander K, Eisfeldt J, Lindblad M, Nilsson D, Billiau K, Foroughi H, Nordenskjöld M, Liedén A and Tham E: Cell-free tumour DNA analysis detects copy number alterations in gastro-oesophageal cancer patients. PLoS One. 16:e02454882021. View Article : Google Scholar : PubMed/NCBI

10 

Han B, Ren D, Mao B, Song X, Yang W, Zhang H and Gao F: Tumor copy number alteration (CNA) burden as a prognostic factor for overall survival in Chinese gastric cancers. J Clin Oncol. 37 (Suppl 15):e155552019. View Article : Google Scholar

11 

Milne AN: Early-onset gastric cancer: Learning lessons from the young. World J Gastrointest Oncol. 2:59–64. 2010. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Seabra AD, Araújo TM, Mello Junior FA, Di Felipe Ávila Alcântara D, De Barros AP, De Assumpção PP, Montenegro RC, Guimarães AC, Demachki S, Burbano RM and Khayat AS: High-density array comparative genomic hybridization detects novel copy number alterations in gastric adenocarcinoma. Anticancer Res. 34:6405–6415. 2014.PubMed/NCBI

14 

Morales-Guerrero SE, Rivas-Ortiz CI, Ponce de León-Rosales S, Gamboa-Domínguez A, Rangel-Escareño C, Uscanga-Domínguez LF, Aguilar-Gutiérrez GR, Kershenobich-Stalnikowitz D, Castillo-Rojas G and López-Vidal Y: Translation of gastric disease progression at gene level expression. J Cancer. 11:520–532. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Nadauld LD, Garcia S, Natsoulis G, Bell JM, Miotke L, Hopmans ES, Xu H, Pai RK, Palm C, Regan JF, et al: Metastatic tumor evolution and organoid modeling implicate TGFBR2 as a cancer driver in diffuse gastric cancer. Genome Biol. 15:4282014. View Article : Google Scholar : PubMed/NCBI

16 

Vasaikar SV, Deshmukh AP, den Hollander P, Addanki S, Kuburich NA, Kudaravalli S, Joseph R, Chang JT, Soundararajan R and Mani SA: EMTome: A resource for pan-cancer analysis of epithelial-mesenchymal transition genes and signatures. Br J Cancer. 124:259–269. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Varga J and Greten FR: Cell plasticity in epithelial homeostasis and tumorigenesis. Nat Cell Biol. 19:1133–1141. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Christofori G: New signals from the invasive front. Nature. 441:444–450. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Murai T, Yamada S, Fuchs BC, Fujii T, Nakayama G, Sugimoto H, Koike M, Fujiwara M, Tanabe KK and Kodera Y: Epithelial-to-mesenchymal transition predicts prognosis in clinical gastric cancer: EMT in clinical gastric cancer. J Surg Oncol. 109:684–689. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Xia P and Xu XY: Epithelial-mesenchymal transition and gastric cancer stem cell. Tumour Biol. 39:10104283176983732017. View Article : Google Scholar : PubMed/NCBI

22 

Wang G and Anastassiou D: Pan-cancer driver copy number alterations identified by joint expression/CNA data analysis. Sci Rep. 10:171992020. View Article : Google Scholar : PubMed/NCBI

23 

Owen GI, Pinto MP, Retamal IN, Fernádez MF, Cisternas B, Mondaca S, Sanchez C, Galindo H, Nervi B, Ibañez C, et al: Chilean gastric cancer task force: A study protocol to obtain a clinical and molecular classification of a cohort of gastric cancer patients. Medicine (Baltimore). 97:e04192018. View Article : Google Scholar : PubMed/NCBI

24 

Araújo TM, Seabra AD, Lima EM, Assumpção PP, Montenegro RC, Demachki S, Burbano RM and Khayat AS: Recurrent amplification of RTEL1 and ABCA13 and its synergistic effect associated with clinicopathological data of gastric adenocarcinoma. Mol Cytogenet. 9:522016. View Article : Google Scholar : PubMed/NCBI

25 

Eltoum I, Fredenburgh J, Myers RB and Grizzle WE: Introduction to the theory and practice of fixation of tissues. J Histotechnol. 24:173–190. 2001. View Article : Google Scholar

26 

Fischer I, Cunliffe C, Bollo RJ, Weiner HL, Devinsky O, Ruiz-Tachiquin ME, Venuto T, Pearlman A, Chiriboga L, Schneider RJ, et al: Glioma-like proliferation within tissues excised as tubers in patients with tuberous sclerosis complex. Acta Neuropathol. 116:67–77. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Utrera-Barillas D, Valdez-Salazar HA, Gómez-Rangel D, Alvarado-Cabrero I, Aguilera P, Gómez-Delgado A and Ruiz-Tachiquin ME: Is human cytomegalovirus associated with breast cancer progression? Infect Agent Cancer. 8:122013. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Jeon J and Cheong JH: Clinical implementation of precision medicine in gastric cancer. J Gastric Cancer. 19:235–253. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Zubarayev M, Min EK and Son T: Clinical and molecular prognostic markers of survival after surgery for gastric cancer: Tumor-node-metastasis staging system and beyond. Transl Gastroenterol Hepatol. 4:592019. View Article : Google Scholar : PubMed/NCBI

31 

Brierley JD, Gospodarowicz MK and Wittekind C: TNM Classification of Malignant Tumours: Edition 8. John Wiley & Sons; New Jersey: pp. 1–16. 2016, PubMed/NCBI

32 

Bure IV, Nemtsova MV and Zaletaev DV: Roles of E-cadherin and noncoding RNAs in the epithelial-mesenchymal transition and progression in gastric cancer. Int J Mol Sci. 20:28702019. View Article : Google Scholar : PubMed/NCBI

33 

Marin JJG, Perez-Silva L, Macias RIR, Asensio M, Peleteiro-Vigil A, Sanchez-Martin A, Cives-Losada C, Sanchon-Sanchez P, Sanchez De Blas B, Herraez E, et al: Molecular bases of mechanisms accounting for drug resistance in gastric adenocarcinoma. Cancers (Basel). 12:21162020. View Article : Google Scholar : PubMed/NCBI

34 

De Las Rivas J, Brozovic A, Izraely S, Casas-Pais A, Witz IP and Figueroa A: Cancer drug resistance induced by EMT: Novel therapeutic strategies. Arch Toxicol. 95:2279–2297. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Zhao M, Liu Y and Qu H: Expression of epithelial-mesenchymal transition-related genes increases with copy number in multiple cancer types. Oncotarget. 7:24688–24699. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Wee Y, Wang T, Liu Y, Li X and Zhao M: A pan-cancer study of copy number gain and up-regulation in human oncogenes. Life Sci. 211:206–214. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Zhao M and Zhao Z: Concordance of copy number loss and down-regulation of tumor suppressor genes: A pan-cancer study. BMC Genomics. 17 Suppl 7(Suppl 7):5322016. View Article : Google Scholar : PubMed/NCBI

38 

Tong Y, Tang Y, Li S, Zhao F, Ying J, Qu Y, Niu X and Mu D: Cumulative evidence of relationships between multiple variants in 8q24 region and cancer incidence. Medicine (Baltimore). 99:e207162020. View Article : Google Scholar : PubMed/NCBI

39 

Arakawa N, Sugai T, Habano W, Eizuka M, Sugimoto R, Akasaka R, Toya Y, Yamamoto E, Koeda K, Sasaki A, et al: Genome-wide analysis of DNA copy number alterations in early and advanced gastric cancers. Mol Carcinog. 56:527–537. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Anauate AC, Leal MF, Wisnieski F, Santos LC, Gigek CO, Chen ES, Calcagno DQ, Assumpção PP, Demachki S, Arasaki CH, et al: Analysis of 8q24.21 miRNA cluster expression and copy number variation in gastric cancer. Future Med Chem. 11:947–958. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Labrador L, Torres K, Camargo M, Santiago L, Valderrama E and Chiurillo MA: Association of common variants on chromosome 8q24 with gastric cancer in Venezuelan patients. Gene. 566:120–124. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Jin DH, Park SE, Lee J, Kim KM, Kim S, Kim DH and Park J: Copy number gains at 8q24 and 20q11-q13 in gastric cancer are more common in intestinal-type than diffuse-type. PLoS One. 10:e01376572015. View Article : Google Scholar : PubMed/NCBI

43 

Korivi BR, Faria S, Aly A, Sun J, Patnana M, Jensen CT, Wagner-Bartak N and Bhosale PR: Intestinal and diffuse gastric cancer: A retrospective study comparing primary sites. Clin Imaging. 56:33–40. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Cancer Genome Atlas Research Network, . Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Qin H, Ni H, Liu Y, Yuan Y, Xi T, Li X and Zheng L: RNA-binding proteins in tumor progression. J Hematol Oncol. 13:902020. View Article : Google Scholar : PubMed/NCBI

46 

Morgan MA and Shilatifard A: Chromatin signatures of cancer. Genes Dev. 29:238–249. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Brabletz T, Kalluri R, Nieto MA and Weinberg RA: EMT in cancer. Nat Rev Cancer. 18:128–134. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Bhullar KS, Lagarón NO, McGowan EM, Parmar I, Jha A, Hubbard BP and Rupasinghe HPV: Kinase-targeted cancer therapies: Progress, challenges and future directions. Mol Cancer. 17:482018. View Article : Google Scholar : PubMed/NCBI

49 

Olea-Flores M, Zuñiga-Eulogio MD, Mendoza-Catalán MA, Rodríguez-Ruiz HA, Castañeda-Saucedo E, Ortuño-Pineda C, Padilla-Benavides T and Navarro-Tito N: Extracellular-signal regulated kinase: A central molecule driving epithelial-mesenchymal transition in cancer. Int J Mol Sci. 20:28852019. View Article : Google Scholar : PubMed/NCBI

50 

Castel P, Toska E, Engelman JA and Scaltriti M: The present and future of PI3K inhibitors for cancer therapy. Nat Cancer. 2:587–597. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Zhang Y, Kwok-Shing Ng P, Kucherlapati M, Chen F, Liu Y, Tsang YH, de Velasco G, Jeong KJ, Akbani R, Hadjipanayis A, et al: A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell. 31:820–832.e3. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Assumpção PP, Barra WF, Ishak G, Coelho LGV, Coimbra FJF, Freitas HC, Dias-Neto E, Camargo MC and Szklo M: The diffuse-type gastric cancer epidemiology enigma. BMC Gastroenterol. 20:2232020. View Article : Google Scholar : PubMed/NCBI

53 

Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I and Forbes SA: The COSMIC cancer gene census: Describing genetic dysfunction across all human cancers. Nat Rev Cancer. 18:696–705. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Zhang D, Huo D, Xie H, Wu L, Zhang J, Liu L, Jin Q and Chen X: CHG: A systematically integrated database of cancer Hallmark genes. Front Genet. 11:292020. View Article : Google Scholar : PubMed/NCBI

55 

Wan J, Zhou J, Zhao H, Wang M, Wei Z, Gao H, Wang Y and Cui H: Sonic hedgehog pathway contributes to gastric cancer cell growth and proliferation. Biores Open Access. 3:53–59. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Krupar R, Watermann C, Idel C, Ribbat-Idel J, Offermann A, Pasternack H, Kirfel J, Sikora AG and Perner S: In silico analysis reveals EP300 as a panCancer inhibitor of anti-tumor immune response via metabolic modulation. Sci Rep. 10:93892020. View Article : Google Scholar : PubMed/NCBI

57 

Dong X, Hong Y, Sun H, Chen C, Zhao X and Sun B: NDRG1 suppresses vasculogenic mimicry and tumor aggressiveness in gastric carcinoma. Oncol Lett. 18:3003–3016. 2019.PubMed/NCBI

58 

Liang L, Fang JY and Xu J: Gastric cancer and gene copy number variation: Emerging cancer drivers for targeted therapy. Oncogene. 35:1475–1482. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Mariotti S, Barravecchia I, Vindigni C, Pucci A, Balsamo M, Libro R, Senchenko V, Dmitriev A, Jacchetti E, Cecchini M, et al: MICAL2 is a novel human cancer gene controlling mesenchymal to epithelial transition involved in cancer growth and invasion. Oncotarget. 7:1808–1825. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Herold S, Herkert B and Eilers M: Facilitating replication under stress: An oncogenic function of MYC? Nat Rev Cancer. 9:441–444. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Zhang L, Hou Y, Ashktorab H, Gao L, Xu Y, Wu K, Zhai J and Zhang L: The impact of C-MYC gene expression on gastric cancer cell. Mol Cell Biochem. 344:125–135. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Huang X, Li Z, Zhang Q, Wang W, Li B, Wang L, Xu Z, Zeng A, Zhang X, Zhang X, et al: Circular RNA AKT3 upregulates PIK3R1 to enhance cisplatin resistance in gastric cancer via miR-198 suppression. Mol Cancer. 18:712019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 25 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Larios-Serrato V, Martínez‑Ezquerro J, Valdez‑Salazar H, Torres J, Camorlinga‑Ponce M, Piña‑Sánchez P and Ruiz‑Tachiquín M: Copy number alterations and epithelial‑mesenchymal transition genes in diffuse and intestinal gastric cancers in Mexican patients. Mol Med Rep 25: 191, 2022
APA
Larios-Serrato, V., Martínez‑Ezquerro, J., Valdez‑Salazar, H., Torres, J., Camorlinga‑Ponce, M., Piña‑Sánchez, P., & Ruiz‑Tachiquín, M. (2022). Copy number alterations and epithelial‑mesenchymal transition genes in diffuse and intestinal gastric cancers in Mexican patients. Molecular Medicine Reports, 25, 191. https://doi.org/10.3892/mmr.2022.12707
MLA
Larios-Serrato, V., Martínez‑Ezquerro, J., Valdez‑Salazar, H., Torres, J., Camorlinga‑Ponce, M., Piña‑Sánchez, P., Ruiz‑Tachiquín, M."Copy number alterations and epithelial‑mesenchymal transition genes in diffuse and intestinal gastric cancers in Mexican patients". Molecular Medicine Reports 25.5 (2022): 191.
Chicago
Larios-Serrato, V., Martínez‑Ezquerro, J., Valdez‑Salazar, H., Torres, J., Camorlinga‑Ponce, M., Piña‑Sánchez, P., Ruiz‑Tachiquín, M."Copy number alterations and epithelial‑mesenchymal transition genes in diffuse and intestinal gastric cancers in Mexican patients". Molecular Medicine Reports 25, no. 5 (2022): 191. https://doi.org/10.3892/mmr.2022.12707