Open Access

Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis

  • Authors:
    • Ru Huang
    • Wenchao Gu
    • Bin Sun
    • Lei Gao
  • View Affiliations

  • Published online on: March 1, 2018     https://doi.org/10.3892/mmr.2018.8664
  • Pages: 6387-6396
  • Copyright: © Huang 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

Trastuzumab, the first targeted antibody against human epidermal growth factor receptor 2 (HER2), has been used to treat gastric cancer patients with HER2 overexpression. However, trastuzumab resistance often occurs following an initial period of benefits, and the underlying mechanisms remain largely unclear. The present study revealed that collagen type IV α1 chain (COL4A1), whose expression is upregulated in gastric cancer tissues and trastuzumab‑resistant gastric cancer cells, may potentially confer trastuzumab resistance in gastric cancer. By performing bioinformatics analysis of 2 microarray datasets, the present study initially identified COL4A1, overexpressed in gastric cancer tissues and trastuzumab‑resistant gastric cancer cells, as a potential candidate for inducing trastuzumab resistance. The drug resistance function of COL4A1 in gastric cancer was then validated by performing protein/gene interactions and biological process annotation analyses, and further validated by analyzing the functionality of microRNAs that target COL4A1 mRNA. Collectively, these data indicated that COL4A1 may confer trastuzumab resistance in gastric cancer.

Introduction

Gastric cancer is the third most frequently diagnosed cancer type, and has been the leading cause of cancer-related death in less-developed countries (1). Although advances in therapeutic strategies such as surgery and systemic chemotherapy, have improved the clinical outcomes for gastric cancer patients, the prognosis of these patients remains poor because of frequent cancer recurrence (2). Nevertheless, due to the encouraging development in newly targeted therapies, such a disappointing clinical condition has been improving. Trastuzumab, the first targeted antibody against human epidermal growth factor receptor 2 (HER2), has been approved for the treatment of patients with HER2-positive metastatic gastric cancer (3). However, even with trastuzumab resistance, gastric cancer almost inevitably progresses, as the tumors become resistant to trastuzumab after an initial period of clinical benefits (4). Mechanisms leading to trastuzumab resistance in breast cancer, such as cross-talk between HER2 and other intracellular kinase receptors, have been described recently (5), however, the underlying mechanisms of trastuzumab resistance in gastric cancer remain largely unknown.

Cancer cells are able to activate alternative survival pathways yielding drug resistance in response to chemotherapy, and thereby leads to the chemotherapeutic treatment failure (6). Since drug resistance is a common cause limiting the efficacy of cancer treatment, several mechanisms are elucidated to be responsible for the resistance development, such as increased rates of drug efflux, apoptosis resistance, and microRNAs (miRNAs) mediated overexpression of many drug resistance-related genes (7). Microarray technology, a high-throughput platform that analyzes gene expression, in combination with bioinformatics analysis has been widely used as a promising tool to acquire gene signature during tumorigenesis or drug resistance, and identify prognostic biomarkers in cancer patients (810). Abnormal expression patterns of drug resistance-related genes commonly play important roles in drug resistance (11), thus, exploring and identifying the critical drug resistance-related genes based on microarray analysis would have a significant impact.

Therefore, in the present study, we sought to identify the potential genes that promote trastuzumab resistance in gastric cancer through retrieving microarray data from public databases and comprehensive bioinformatics analysis.

Materials and methods

Microarray data

The gene expression profiles of GSE26899, GSE77346, GSE54129, and GSE65801 were obtained from the Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo). In detail, GSE26899 dataset is consisted of 96 clinical gastric tumor tissues and 12 adjacent normal tissues; GSE77346 dataset is consisted of 1 trastuzumab-sensitive cell line and 4 trastuzumab-resistant cell lines (12); GSE54129 includes 111 human gastric cancer tissues and 21 non-cancerous tissues; GSE65801 contains 32 gastric cancer tissues and 32 paired non-cancerous tissues (13).

Processing of microarray data

The raw microarray data files of the datasets downloaded from the GEO website were subsequently analyzed via using the GEO2R (www.ncbi.nlm.nih.gov/geo/geo2r/), an online tool comparing two or more groups of samples in the same experimental setting (14). False Discovery Rate (FDR) of P-value adjusted (adj. P) to 0.05 and |logFC|>1 were set as the cut-off criteria.

Functional and pathway enrichment analyses

Gene ontology (GO) analysis is a commonly used approach for functional studies with three ontologies including biological process, molecular function, and cellular component (15), while Kyoto Encyclopedia of Genes and Genomes (KEGG) is a knowledge base for the systematic study of gene functions (16). To study the functional annotations of differentially expressed genes (DEGs), we next employed Database for Annotation, Visualization and Integrated Discovery (DAVID, david.abcc.ncifcrf.gov/,) to process the GO and KEGG analyses of DEGs identified in gastric cancer samples. P<0.05 was set as the threshold.

Protein-protein interaction (PPI) network construction and module analysis

The Search Tool for the Retrieval of Interacting Genes (STRING), an online database (string-db.org) designed to evaluate PPI information, covers 9,643,763 proteins from more than 2,000 organisms, which was used to construct the PPI. To evaluate the interactive associations of DEGs identified from GSE26899, we mapped these DEGs to the STRING (version 10.5) database. Confidence score >0.4 was selected as significant. PPI networks were constructed by STRING and visualized by Cytoscape. Subsequently, the plug-in Molecular Complex Detection (MCODE) was employed to screen the modules of PPI networks in Cytoscape with the threshold set as follows: MCODE scores >10.

Survival analysis of collagen type IV α1 chain (COL4A1)

To evaluate the association between COL4A1 level and its clinical outcomes, Kaplan-Meier plotter (KM plotter; www.kmplot.com), an online survival analysis tool, was performed. KM plotter is capable of assessing the effect of 54,675 genes on overall survival via using 10,188 cancer samples including 4,142 breast, 1,648 ovarian, 2,437 lung, and 1,065 gastric cancer patients (17). Patients with gastric cancer were separated into high- and low-expression groups according to the level of COL4A1, and the overall survival was then analyzed. The hazard ratio (HR) with 95% confidence intervals and log rank P-value were calculated.

Analysis of COL4A1 by geneMANIA and coremine

GeneMANIA, an online tool (www.genemania.org/), can be used to generate hypotheses of gene function, analyze gene lists, and prioritize genes for functional assays (18). After selecting Homo sapiens from the nine optional organisms, COL4A1 was entered into the search bar and the results were then collected. Annotation of biological processes involving COL4A1 was performed by consulting the Coremine Medical online database (www.coremine.com/medical/).

Prediction of miRNAs

To predict the miRNAs targeting the mRNA of COL4A1, miRWalk (version 2.0, zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/), an online platform supplying information about predicted and experimentally validated miRNA-target interactions, was then employed (19). Herein, nine prediction programs (miRWalk, miRanda, miRDB, miRNAMap, Pictar2, PITA, RNA22, RNAhybrid and Targetscan) were selected. These predicted miRNAs were then overlapped by at least seven programs, and selected for further analysis. Pathway enrichment analysis of these miRNAs was performed by using the DIANA-mirPath web server (snf-515788.vm.okeanos.grnet.gr/index.php?r=mirpath) (20).

Statistical analysis

SPSS 22.0 software (IBM Corp., Armonk, NY, USA) was used to analyze data. Two tailed Student's t-test was used to compare the two groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Module acquisition based on the DEGs identified in gastric cancer tissues

Drug resistance-related genes identified from in vitro drug-induced resistant models may simply represent transcriptional changes, thus, complementary agents targeting these genes are usually failure to translate into clinical practice (21). Because drug resistance acquisition can arise before the malignant transformation stage (22), the genes playing important roles in tumorigenesis and drug resistance is thereby more likely to be critical for resistance occurrence. Therefore, we first identified 509 DEGs in gastric cancer tissues from the GSE26899 dataset using a 2-fold-change and adj. P<0.05 as the threshold cutoff. Among these DEGs, the expression of these 172 genes was significantly upregulated, while that of 337 genes was significantly downregulated in cancer tissues (Fig. 1A). To explore the potential roles of these significantly upregulated DEGs, GO and KEGG pathway analyses, which can provide valuable insights regarding protein function, were performed (23). As shown in Fig. 1B, GO results showed that these significantly upregulated DEGs were largely associated with extracellular region part, collagen, extracellular matrix, and cell or biological adhesion processes. KEGG results also consistently revealed that these DEGs were highly enriched in extracellular matrix-receptor interaction and focal adhesion pathways (Fig. 1C). Based on further DEGs analysis with the STRING database, the PPI network of these DEGs, which contained 505 nodes and 1,207 edges, was subsequently constructed. Using the MCODE plug-in in Cytoscape, we obtained the module with the highest score (Fig. 2A), and also performed a cluster analysis of these genes in the module (Fig. 2B). The module with the highest score was selected based on these DEGs identified from gastric cancer tissues via bioinformatics methods.

Expression of COL4A1, overexpressed in gastric cancer tissues, is also upregulated in trastuzumab-resistant gastric cancer cells

To further explore the potential genes contributing to trastuzumab resistance, we retrieved GSE77346 data, a microarray dataset consisted of one trastuzumab-sensitive gastric cancer cell line and four trastuzumab-resistant gastric cancer cell lines (12). After screening the significantly upregulated DEGs in trastuzumab-resistant cancer cells, COL4A1, one of hub genes in the selected module (Fig. 2A), was also found to be significantly upregulated in trastuzumab-resistant cells (Fig. 3A), suggesting COL4A1 might be important in both tumorigenesis and trastuzumab resistance. Since the gene expression is not always consistent with its protein amount (24), further validation of COL4A1 protein level in clinical gastric cancer tissues is quite necessary. By employing the Human Protein Atlas database, an online tool analyzing protein level from clinical specimens, we observed that COL4A1 was positively expressed in gastric cancer tissue, but negatively expressed in normal gastric tissue (Fig. 3B). Two other datasets, including GSE54129 and GSE65801 were also used to validate the mRNA level of COL4A1, which was indeed significantly upregulated in clinical gastric cancer samples (Fig. 3C and D). To investigate potential regulation mechanisms of COL4A1 in gastric cancer, the genomic alteration of COL4A1 in The Cancer Genome Atlas (TCGA) cohort was analyzed using the cBioPortal (www.cbioportal.org/) (25). As shown in Fig. 3E, the amplification, missense mutation, truncating mutation, and deletion of COL4A1 accounted for 4.3, 4.8, 2.0, and 0.5% of stomach adenocarcinoma cases, respectively. The prognostic value of COL4A1 in patients with gastric cancer was analyzed using the KM plotter according to the low and high expression of COL4A1. As shown in Fig. 3F, the high mRNA level of COL4A1 [HR 1.56 (1.31–1.86)] was associated with poor overall survival of gastric cancer patients. Altogether, these data suggest that COL4A1 is a potential gene candidate that promotes the development of gastric cancer and subsequent trastuzumab resistance.

Drug resistance function of COL4A1 is validated by protein/gene interactions and biological process annotation analyses

As a user-friendly web interface for functional prediction of genes, GeneMANIA has been widely used as an effective tool to predict and explore drug resistance-related genes (18). Accordingly, COL4A1 showed interactions with 20 proteins/genes; among these 6 genes were involved in conferring drug resistance in cancer (Fig. 4A). The mRNA expression of these 6 genes in gastric cancer cells and trastuzumab-resistant gastric cancer cells were also evaluated using the microarray data of GSE77346. As shown in Fig. 4B, the mRNA level of angiopoietin 2 (ANGPT2), COL3A1, COL15A1, and secreted protein acidic and cysteine rich (SPARC) were upregulated in trastuzumab-resistant gastric cancer cells. Among these genes, COL4A1 co-expressed and co-localized with ANGPT2, collagen type VI α 3 chain (COL6A3), and SPARC, respectively. It has been reported bevacizumab resistance in glioblastoma and doxorubin resistance in liver cancer are largely mediated by an enforcement of ANGPT2/TIE2 signaling (26,27). Additionally, overexpression of COL6A3, one of the most highly upregulated genes in oxaliplatin and cisplatin resistant ovarian cancer cells, has also been recognized to confer cisplatin resistance in sensitive cancer cells (28). Moreover, the accumulation of intracellular SPARC can drive imatinib resistance in chronic myelogenous leukemia cells (29), and also modulate cisplatin resistance via modulating the Let-7f-1 miRNA/HMGB1 signaling in medulloblastoma cells (30). COL3A1 and COL15A1, two types of fibrillar collagen highly expressed in chemotherapeutic drugs resistant ovarian cancer cells (31), were also exhibited to co-express, share protein domains, and co-localize with COL4A1 (Fig. 4A). Additionally, COL4A1 also was co-expressed, shared pathways, and had physical interactions with platelet derived growth factor subunit B (PDGFB). Akt/PDGF-B signaling can regulate Akt, and thus confer the hypoxia-induced cisplatin resistance in liver cancer cells (32). PDGFB may contribute to the resistant phenotype and sustain signaling through MAPK and Akt in breast cancer cells (33). The Coremine Medical database is a freely available online tool for obtaining information on health, medicine, and biology (33); thus, we used this database to annotate the biological process of COL4A1. As shown in Fig. 4C, 12 biological processes significantly associated with COL4A1, gastric cancer, and drug resistance (P<0.01) were annotated. Considering that the close relationships of COL4A1 with these processes and the close relationships of the 12 processes with gastric cancer and drug resistance, COL4A1 might be involved in the development of drug resistance in gastric cancer via its effect on these biological processes. In detail, cell growth related biological processes (including 4 cell growth, cell proliferation, and apoptotic process), gene expression regulation-related (including gene expression, RNA interference, and reverse transcription), and especially, the epithelial-mesenchymal transition (EMT) biological process were identified to be closely associated with the development of COL4A1 in the gastric cancer drug resistance. Therefore, these results suggest that COL4A1 may confer drug resistance in gastric cancer via regulating cell growth, gene expression, and in particular, epithelial mesenchymal transition (EMT) processes.

Drug resistance function of COL4A1 is further validated by analyzing functionality of miRNAs that target COL4A1 mRNA

Post-transcriptional gene expression regulated by miRNAs is important for multiple cellular processes during development and pathogenesis (34). Amplification and overexpression of tumor-promoting miRNAs or genetic loss of tumor-suppressing miRNAs are tightly correlated with the development of cancer and chemotherapeutic resistance (35). Therefore, studying target genes of miRNAs is a focus of interest due to the diagnostic and therapeutic relevance, and the gene functions can also be predicted according to functionality of these miRNAs targeting the gene (36). To obtain the miRNAs targeting COL4A1 mRNA, the miRNA-mRNA interaction analysis was performed by using the miRWalk. We identified 553 miRNAs predicted to transcriptionally target COL4A1 mRNA, indicating the regulation of COL4A1 by miRNAs. These miRNAs predicted by at least 8 of 9 prediction tools were then submitted to pathway enrichment using DIANA miRPath (20). As shown in Table I, the top 5 highly associated pathway were ECM-receptor interaction, TGF-β signaling pathway, viral carcinogenesis, proteoglycans in cancer, and Hippo signaling pathway, which are almost reported to be related with drug resistance (3741). It has been established TGFβ signaling can activate autophagy process, and thereby lead to the oxaliplatin resistance in colorectal cancer (38). In addition, the activation of Hippo signaling also contributes to the drug resistance and cancer relapse (42). YAP, an effector of hippo signaling, has also been reported to alter clinical response of EGFR-tyrosine kinase inhibitors in lung cancer patients (43). Furthermore, it has also been reported that inactivation of Hippo pathway can restore gemcitabine sensitivity among a variety of cancers (44). In the present study, as shown in Table II, 9 of top 10 miRNAs that target COL4A1 mRNA were tightly associated with drug resistance in cancers (4551). For instance, loss of intracellular miR-29b promotes cisplatin resistance in gastric cancer. Consistently, ectopic overexpression of miR-29b in cholangiocarcinoma cells can confer gemcitabine sensitivity to HuH28 cells (45). MiR-506 overexpression can confer hydroxycamptothecin resistance in colon cancer cells by inhibiting PPARα expression (50). Collectively, these data provide further supports for the drug resistance function of COL4A1 in gastric cancer.

Table I.

Top 8 enriched pathways regulated by microRNAs that target collagen type IV α1 mRNA and their associations with drug resistance in gastric cancer.

Table I.

Top 8 enriched pathways regulated by microRNAs that target collagen type IV α1 mRNA and their associations with drug resistance in gastric cancer.

Author, yearmiRNAs (hsa-miR-)KEGG pathwayP-valueRegulation of drug resistance in cancers(Refs.)
Wu et al, 201729b-3p, 124-3p, 148a-3p, 29a-3p, 152-3p, 148b-3p, 506-3p, 628-5p, 29c-3p, 767-5p, 637, 33a-5p, 33b-5p, 203a, 374a-5p, 300, 106a-5p, 98-5p, 381-3p, let-7b-5p, 7c-5p, 7b-3p, 7a-5pECM-receptor interaction 1.14×10‒15Yes(37)
Sun et al, 2017 TGF-β signaling pathway 6.58×10‒11Yes(38)
Viral carcinogenesis 9.72×10‒11None
Lanzi et al, 2017 Proteoglycans in cancer 3.94×10‒10Yes(39)
Gujral et al, 2017 Hippo signaling pathway 6.66×10‒10Yes(44)
Lee et al, 2015 Cell cycle 1.01×10‒9Yes(40)
Wallerand et al, 2010 Adherens junction 2.99×10‒8Yes(41)
Pathways in cancer 3.46×10‒8Cancer pathway

[i] The miRNAs predicted by at least 7 of the 9 prediction tools were submitted to DIANA-miRPath version 3.0 to perform KEGG analysis. miRNA/miR, microRNA; KEGG, Kyoto Encyclopedia of Genes and Genomes; ECM, extracellular matrix; TGF, transforming growth factor.

Table II.

Top 11 microRNAs targeting collagen type IV α1 mRNA predicted by microRNA-mRNA interactions, and their drug resistance-associated functions in cancer.

Table II.

Top 11 microRNAs targeting collagen type IV α1 mRNA predicted by microRNA-mRNA interactions, and their drug resistance-associated functions in cancer.

miRNA-mRNA prediction tools

Author, yearmiRNA (hsa-)ABCDEFGHIDrug resistance-related functions of miRNAs in cancers(Refs.)
Okamoto et al, 2013miR-29b-3p111111111Drug resistance-related(45)
Liu et al, 2016miR-124-3p111111111Drug resistance-related(46)
Chen et al, 2017miR-148a-3p111111111Drug resistance-related(47)
Zhong et al, 2013miR-29a-3p111111111Drug resistance-related(48)
Chen et al, 2017miR-152-3p111111111Drug resistance-related(47)
Sui et al, 2015miR-148b-3p111101111Drug resistance-related(49)
Tong et al, 2011miR-506-3p111111011Drug resistance-related(50)
miR-628-5p111011111Drug resistance-related
Zhang et al, 2013miR-29c-3p111111011Drug resistance-related(51)
miR-767-5p011111111Drug resistance-related
miR-637111101111Drug resistance-related

[i] A, miRWalk; B, miRanda; C, miRDB; D, miRNAMap; E, Pictar2; F, PITA; G, RNA22; H, RNAhybrid; I, TargetScan; miRNA/miR, microRNA.

Discussion

In the present study, based on the bioinformatics analysis of two microarray datasets including GSE26899 and GSE77346, COL4A1, which was overexpressed in gastric cancer tissues and trastuzumab-resistant gastric cancer cells, was identified as a potential gene that promotes gastric cancer and trastuzumab resistance. By combining the protein/gene interactions, biological process annotation, and miRNAs-mRNA interaction analyses, we showed that COL4A1 may confer trastuzumab resistance in gastric cancer.

Traditional chemotherapy and newly targeted therapy are two important methods of cancer treatment; however, the clinical efficacy of both is largely limited due to the occurrence of subsequent drug resistance (52). Mechanisms underlying drug resistance, such as alteration of drug targets or metabolism and genetic mutation (11) have been elucidated. Similar to that in breast cancer, the HER2-positivity rate in gastric cancer can range from 20 to 42% (53). However, although the benefits of trastuzumab against HER2-positive gastric cancer have been formally established (3,53), trastuzumab resistance is almost inevitable and eventually leads to the therapeutic failure (4). Interfering with the combination of trastuzumab and HER2 (54) and constitutive dimerization of the HER2 receptor (55) have been recognized as causes of trastuzumab resistance. Recently, abnormal expression of tumorigenesis or drug resistance-related genes, such as phosphatidylinositol 3-kinase/Akt signaling (56) and insulin like growth factor 1 receptor (57), has been also identified as the important mechanisms causing trastuzumab resistance. However, a comprehensive study of specific molecular mutation underlying trastuzumab resistance is still needed. Drug resistance-related genes identified from induced drug-resistant cancer cells may simply be a result of transcriptional changes that may be irrelevant to resistance mechanisms (58). Thus, targeting these genes usually fail to translate into clinical practice (21). It has been recently shown that the ability to acquire drug resistance can arise even before the malignant transformation stage. For instance, overexpression of telomerase or inactivation of p53 can contribute to the drug-resistant phenotype in pre-tumorigenic models (22). Insulin-like growth factor signaling has also been deemed a factor that promotes both the development of various tumors and their resistance to chemotherapy (59,60). This evidence thus reminds us that the genes playing a key role in both tumor development and subsequent drug resistance likely produce the essential molecule for drug resistance. Herein, through retrieving microarray datasets and employing subsequent bioinformatics analysis, COL4A1 was validated as an important gene that drives trastuzumab resistance in gastric cancer.

The heterotrimers formed by COL4A1 and COL4A2 presents in almost all the basement membranes, which are a specialized form of the extracellular matrix. Besides that the basement membranes can mediate tissue compartmentalization and transfer environmental signals to epithelial cells (61), it is also an important structural and functional component of blood vessels. Accordingly, mutations in COL4A1 are pleiotropic and contribute to many diseases, such as myopathy, hemorrhagic stroke, and tumor progression (62). Upregulated COL4A1 produced by bladder cancer cells plays pivotal roles in tumor invasion via induction of tumor budding, while overexpression of COL4A1 also contributes to breast cancer cells proliferation, which indicates targeting COL4A1 can be an attractive approach for cancer treatment (63,64). Besides, COL4A1 has also been identified as one of biomarkers for prognosis of intrahepatic cholangiocarcinoma (65). Given that an interaction between PDGFB and HIF-1α and an interplay between SPARC, BCL-2, and caspase-8 have been recognized to augment chemotherapy-induced apoptosis, and thereby inducing resistance (32,66). Herein, our results suggest COL4A1 can drive trastuzumab resistance in gastric cancer via multiple mechanisms, such as cell proliferation and miRNAs-mediated post-transcriptional modification. Consistent with our results, a previous study also reported the sustained EMT phenotype induced by prolonged trastuzumab treatment may lead to trastuzumab resistance in gastric cancer cells (67). Small molecules that targeting drug resistance-related genes have shown promising clinical efficacy in cancer treatment. For instance, erbB3 overexpression drives paclitaxel resistance in breast cancer, MM-121/SAR256212, an erbB3-targeted antibody, was thus designed and shows augmented effect on paclitaxel resistance (68). Activation of the PI3K pathway frequently occurs in cancers and leads to drug resistance, but clinical benefits of PI3K inhibitors have been modest to date. Fortunately, LEE011, a specific CDK 4/6 inhibitor currently under clinical development, has shown promising effects against PI3K inhibition resistance (69). HER2-positive patients receiving trastuzumab may inevitably develop resistance due to excessive activation of the PI3K/AKT pathway. A phase 1 clinical trial has been performed to evaluate the efficacy of the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors; interestingly, MK-2206 is safe, and reversed the trastuzumab resistance in HER2-overexpressing patients (70). Collectively, this evidence suggests that identification of drug-resistant genes through bioinformatics methods and subsequent design of small molecule drugs may have great potential.

In conclusion, our results show that COL4A1 may confer trastuzumab resistance in gastric cancer via multiple mechanisms based on bioinformatics analysis. However, further investigations elucidating the drug resistance function of COL4A1 in trastuzumab-resistant gastric cancer models are necessary.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LG conceived the project and designed the research plan. RH and WG performed the experiments and analyzed the data with input from BS who assisted in experimental design and data analyses. RH and LG wrote the manuscript, and all authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Corso S and Giordano S: How can gastric cancer molecular profiling guide future therapies? Trends Mol Med. 22:534–544. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 376:687–697. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Aprile G, Giampieri R, Bonotto M, Bittoni A, Ongaro E, Cardellino GG, Graziano F, Giuliani F, Fasola G, Cascinu S and Scartozzi M: The challenge of targeted therapies for gastric cancer patients: The beginning of a long journey. Exp Opin Investig Drugs. 23:925–942. 2014. View Article : Google Scholar

5 

Ritter CA, Perez-Torres M, Rinehart C, Guix M, Dugger T, Engelman JA and Arteaga CL: Human breast cancer cells selected for resistance to trastuzumab in vivo overexpress epidermal growth factor receptor and ErbB ligands and remain dependent on the ErbB receptor network. Clin Cancer Res. 13:4909–4919. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Gottesman MM, Lavi O, Hall MD and Gillet JP: Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol. 56:85–102. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Garofalo M and Croce CM: MicroRNAs as therapeutic targets in chemoresistance. Drug Resist Updat. 16:47–59. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Lu Y, Lemon W, Liu PY, Yi Y, Morrison C, Yang P, Sun Z, Szoke J, Gerald WL, Watson M, et al: A gene expression signature predicts survival of patients with stage I non-small cell lung cancer. PLoS Med. 3:e4672006. View Article : Google Scholar : PubMed/NCBI

9 

Chen F, Xiang CX, Zhou Y, Ao XS, Zhou DQ, Peng P, Zhang HQ, Liu HD and Huang X: Gene expression profile for predicting survival of patients with meningioma. Int J Oncol. 46:791–797. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Kulasingam V and Diamandis EP: Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat Clin Pract Oncol. 5:588–599. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Yin F, Liu X, Li D, Wang Q, Zhang W and Li L: Tumor suppressor genes associated with drug resistance in ovarian cancer (Review). Oncol Rep. 30:3–10. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Piro G, Carbone C, Cataldo I, Di Nicolantonio F, Giacopuzzi S, Aprile G, Simionato F, Boschi F, Zanotto M, Mina MM, et al: An FGFR3 autocrine loop sustains acquired resistance to trastuzumab in gastric cancer patients. Clin Cancer Res. 22:6164–6175. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Li H, Yu B, Li J, Su L, Yan M, Zhang J, Li C, Zhu Z and Liu B: Characterization of differentially expressed genes involved in pathways associated with gastric cancer. PLoS One. 10:e01250132015. View Article : Google Scholar : PubMed/NCBI

14 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41:D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Hulsegge I, Kommadath A and Smits MA: Globaltest and GOEAST: Two different approaches for Gene Ontology analysis. BMC Proc. 3 Suppl 4:pp. S102009; View Article : Google Scholar : PubMed/NCBI

16 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Gyorffy B, Lánczky A and Szállási Z: Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocr Relat Cancer. 19:197–208. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, Franz M, Grouios C, Kazi F, Lopes CT, et al: The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38:W214–W220. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

20 

Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, Dalamagas T and Hatzigeorgiou AG: DIANA-miRPath v3.0: Deciphering microRNA function with experimental support. Nucleic Acids Res. 43:W460–W466. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Borst P and Wessels L: Do predictive signatures really predict response to cancer chemotherapy? Cell cycle (Georgetown, Tex.). 9:4836–4840. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Yague E, Arance A, Kubitza L, O'Hare M, Jat P, Ogilvie CM, Hart IR, Higgins CF and Raguz S: Ability to acquire drug resistance arises early during the tumorigenesis process. Cancer Res. 67:1130–1137. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Xing Z, Chu C, Chen L and Kong X: The use of Gene Ontology terms and KEGG pathways for analysis and prediction of oncogenes. Biochim Biophys Acta. 1860:2725–2734. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Maier T, Guell M and Serrano L: Correlation of mRNA and protein in complex biological samples. FEBS Lett. 583:3966–3973. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

26 

Labussiere M, Cheneau C, Prahst C, Gállego Pérez-Larraya J, Farina P, Lombardi G, Mokhtari K, Rahimian A, Delattre JY, Eichmann A and Sanson M: Angiopoietin-2 May be involved in the resistance to bevacizumab in recurrent glioblastoma. Cancer Invest. 34:39–44. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Li T, Liu Z, Jiang K and Ruan Q: Angiopoietin2 enhances doxorubin resistance in HepG2 cells by upregulating survivin and Ref-1 via MSK1 activation. Cancer Lett. 337:276–284. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Sherman-Baust CA, Weeraratna AT, Rangel LB, Pizer ES, Cho KR, Schwartz DR, Shock T and Morin PJ: Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells. Cancer Cell. 3:377–386. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Fenouille N, Puissant A, Dufies M, Robert G, Jacquel A, Ohanna M, Deckert M, Pasquet JM, Mahon FX, Cassuto JP, et al: Persistent activation of the Fyn/ERK kinase signaling axis mediates imatinib resistance in chronic myelogenous leukemia cells through upregulation of intracellular SPARC. Cancer Res. 70:9659–9670. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Pannuru P, Dontula R, Khan AA, Herbert E, Ozer H, Chetty C and Lakka SS: miR-let-7f-1 regulates SPARC mediated cisplatin resistance in medulloblastoma cells. Cell Signal. 26:2193–2201. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Januchowski R, Zawierucha P, Rucinski M and Zabel M: Microarray-based detection and expression analysis of extracellular matrix proteins in drugresistant ovarian cancer cell lines. Oncol Rep. 32:1981–1990. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Lau CK, Yang ZF, Ho DW, Ng MN, Yeoh GC, Poon RT and Fan ST: An Akt/hypoxia-inducible factor-1alpha/platelet-derived growth factor-BB autocrine loop mediates hypoxia-induced chemoresistance in liver cancer cells and tumorigenic hepatic progenitor cells. Clin Cancer Res. 15:3462–3471. 2009. View Article : Google Scholar : PubMed/NCBI

33 

de Leeuw N, Dijkhuizen T, Hehir-Kwa JY, Carter NP, Feuk L, Firth HV, Kuhn RM, Ledbetter DH, Martin CL, van Ravenswaaij-Arts CM, et al: Diagnostic interpretation of array data using public databases and internet sources. Hum Mutat. 33:930–940. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Sayed D and Abdellatif M: MicroRNAs in development and disease. Physiol Rev. 91:827–887. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Lin S and Gregory RI: MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Blenkiron C and Miska EA: miRNAs in cancer: Approaches, aetiology, diagnostics and therapy. Hum Mol Genet. 16:R106–R113. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Wu X, Wang H, Lian Y, Chen L, Gu L, Wang J and Huang Y, Deng M, Gao Z and Huang Y: GTSE1 promotes cell migration and invasion by regulating EMT in hepatocellular carcinoma and is associated with poor prognosis. Sci Rep. 7:51292017. View Article : Google Scholar : PubMed/NCBI

38 

Sun C, Wang FJ, Zhang HG, Xu XZ, Jia RC, Yao L and Qiao PF: miR-34a mediates oxaliplatin resistance of colorectal cancer cells by inhibiting macroautophagy via transforming growth factor-beta/Smad4 pathway. World J Gastroenterol. 23:1816–1827. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Lanzi C, Zaffaroni N and Cassinelli G: Targeting heparan sulfate proteoglycans and their modifying enzymes to enhance anticancer chemotherapy efficacy and overcome drug resistance. Curr Med Chem. 24:2860–2886. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Lee B, Sandhu S and McArthur G: Cell cycle control as a promising target in melanoma. Curr Opin Oncol. 27:141–150. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Wallerand H, Robert G, Pasticier G, Ravaud A, Ballanger P, Reiter RE and Ferrière JM: The epithelial-mesenchymal transition-inducing factor TWIST is an attractive target in advanced and/or metastatic bladder and prostate cancers. Urol Oncol. 28:473–479. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Yu FX, Zhao B and Guan KL: Hippo pathway in organ size control, tissue homeostasis and cancer. Cell. 163:811–828. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Lee JE, Park HS, Lee D, Yoo G, Kim T, Jeon H, Yeo MK, Lee CS, Moon JY, Jung SS, et al: Hippo pathway effector YAP inhibition restores the sensitivity of EGFR-TKI in lung adenocarcinoma having primary or acquired EGFR-TKI resistance. Biochem Biophys Res Commun. 474:154–160. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Gujral TS and Kirschner MW: Hippo pathway mediates resistance to cytotoxic drugs. Proc Natl Acad Sci USA. 114:pp. E3729–E3738. 2017; View Article : Google Scholar : PubMed/NCBI

45 

Okamoto K, Miyoshi K and Murawaki Y: miR-29b, miR-205 and miR-221 enhance chemosensitivity to gemcitabine in HuH28 human cholangiocarcinoma cells. PLoS One. 8:e776232013. View Article : Google Scholar : PubMed/NCBI

46 

Liu YX, Wang L, Liu WJ, Zhang HT, Xue JH, Zhang ZW and Gao CJ: MiR-124-3p/B4GALT1 axis plays an important role in SOCS3-regulated growth and chemo-sensitivity of CML. J Hematol Oncol. 9:692016. View Article : Google Scholar : PubMed/NCBI

47 

Chen MJ, Cheng YM, Chen CC, Chen YC and Shen CJ: MiR-148a and miR-152 reduce tamoxifen resistance in ER+ breast cancer via downregulating ALCAM. Biochem Biophys Res Commun. 483:840–846. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Zhong S, Li W, Chen Z, Xu J and Zhao J: MiR-222 and miR-29a contribute to the drug-resistance of breast cancer cells. Gene. 531:8–14. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Sui C, Meng F, Li Y and Jiang Y: miR-148b reverses cisplatin-resistance in non-small cell cancer cells via negatively regulating DNA (cytosine-5)-methyltransferase 1 (DNMT1) expression. J Transl Med. 13:1322015. View Article : Google Scholar : PubMed/NCBI

50 

Tong JL, Zhang CP, Nie F, Xu XT, Zhu MM, Xiao SD and Ran ZH: MicroRNA 506 regulates expression of PPAR alpha in hydroxycamptothecin-resistant human colon cancer cells. FEBS Lett. 585:3560–3568. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Zhang JX, Qian D, Wang FW, Liao DZ, Wei JH, Tong ZT, Fu J, Huang XX, Liao YJ, Deng HX, et al: MicroRNA-29c enhances the sensitivities of human nasopharyngeal carcinoma to cisplatin-based chemotherapy and radiotherapy. Cancer Lett. 329:91–98. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Holohan C, Van Schaeybroeck S, Longley DB and Johnston PG: Cancer drug resistance: An evolving paradigm. Nat Rev Cancer. 13:714–726. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Albarello L, Pecciarini L and Doglioni C: HER2 testing in gastric cancer. Adv Anat Pathol. 18:53–59. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Price-Schiavi SA, Jepson S, Li P, Arango M, Rudland PS, Yee L and Carraway KL: Rat Muc4 (sialomucin complex) reduces binding of anti-ErbB2 antibodies to tumor cell surfaces, a potential mechanism for herceptin resistance. Int J Cancer. 99:783–791. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Castiglioni F, Tagliabue E, Campiglio M, Pupa SM, Balsari A and Menard S: Role of exon-16-deleted HER2 in breast carcinomas. Endocr Relat Cancer. 13:221–232. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Serra V, Markman B, Scaltriti M, Eichhorn PJ, Valero V, Guzman M, Botero ML, Llonch E, Atzori F, Di Cosimo S, et al: NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Res. 68:8022–8030. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Nahta R, Yuan LX, Zhang B, Kobayashi R and Esteva FJ: Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res. 65:11118–11128. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Boyer J, Allen WL, McLean EG, Wilson PM, McCulla A, Moore S, Longley DB, Caldas C and Johnston PG: Pharmacogenomic identification of novel determinants of response to chemotherapy in colon cancer. Cancer Res. 66:2765–2777. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Denduluri SK, Idowu O, Wang Z, Liao Z, Yan Z, Mohammed MK, Ye J, Wei Q, Wang J, Zhao L and Luu HH: Insulin-like growth factor (IGF) signaling in tumorigenesis and the development of cancer drug resistance. Genes Dis. 2:13–25. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Casa AJ, Dearth RK, Litzenburger BC, Lee AV and Cui X: The type I insulin-like growth factor receptor pathway: A key player in cancer therapeutic resistance. Front Biosci. 13:3273–3287. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Sado Y, Kagawa M, Kishiro Y, Sugihara K, Naito I, Seyer JM, Sugimoto M, Oohashi T and Ninomiya Y: Establishment by the rat lymph node method of epitope-defined monoclonal antibodies recognizing the six different alpha chains of human type IV collagen. Histochem Cell Biol. 104:267–275. 1995. View Article : Google Scholar : PubMed/NCBI

62 

Kalluri R: Basement membranes: Structure, assembly and role in tumour angiogenesis. Nat Rev Cancer. 3:422–433. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Miyake M, Hori S, Morizawa Y, Tatsumi Y, Toritsuka M, Ohnishi S, Shimada K, Furuya H, Khadka VS, Deng Y, et al: Collagen type IV alpha 1 (COL4A1) and collagen type XIII alpha 1 (COL13A1) produced in cancer cells promote tumor budding at the invasion front in human urothelial carcinoma of the bladder. Oncotarget. 8:36099–36114. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Jin R, Shen J, Zhang T, Liu Q, Liao C, Ma H, Li S and Yu Z: The highly expressed COL4A1 genes contributes to the proliferation and migration of the invasive ductal carcinomas. Oncotarget. 8:58172–58183. 2017.PubMed/NCBI

65 

Sulpice L, Rayar M, Desille M, Turlin B, Fautrel A, Boucher E, Llamas-Gutierrez F, Meunier B, Boudjema K, Clément B and Coulouarn C: Molecular profiling of stroma identifies osteopontin as an independent predictor of poor prognosis in intrahepatic cholangiocarcinoma. Hepatology. 58:1992–2000. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Rahman M, Chan AP, Tang MJ and Tai IT: Correction: A peptide of SPARC interferes with the interaction between caspase8 and Bcl2 to resensitize chemoresistant tumors and enhance their regression in vivo. PLoS One. 10:e01272262015. View Article : Google Scholar : PubMed/NCBI

67 

Yang Z, Guo L, Liu D, Sun L, Chen H, Deng Q, Liu Y, Yu M, Ma Y, Guo N and Shi M: Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget. 6:5072–5087. 2015.PubMed/NCBI

68 

Wang S, Huang J, Lyu H, Cai B, Yang X, Li F, Tan J, Edgerton SM, Thor AD, Lee CK and Liu B: Therapeutic targeting of erbB3 with MM-121/SAR256212 enhances antitumor activity of paclitaxel against erbB2-overexpressing breast cancer. Breast Cancer Res. 15:R1012013. View Article : Google Scholar : PubMed/NCBI

69 

Vora SR, Juric D, Kim N, Mino-Kenudson M, Huynh T, Costa C, Lockerman EL, Pollack SF, Liu M, Li X, et al: CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 26:136–149. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Hudis C, Swanton C, Janjigian YY, Mino-Kenudson M, Huynh T, Costa C, Lockerman EL, Pollack SF, Liu M, Li X, et al: A phase 1 study evaluating the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors. Breast Cancer Res. 15:R1102013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 17 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
Huang R, Gu W, Sun B and Gao L: Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis. Mol Med Rep 17: 6387-6396, 2018
APA
Huang, R., Gu, W., Sun, B., & Gao, L. (2018). Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis. Molecular Medicine Reports, 17, 6387-6396. https://doi.org/10.3892/mmr.2018.8664
MLA
Huang, R., Gu, W., Sun, B., Gao, L."Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis". Molecular Medicine Reports 17.5 (2018): 6387-6396.
Chicago
Huang, R., Gu, W., Sun, B., Gao, L."Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis". Molecular Medicine Reports 17, no. 5 (2018): 6387-6396. https://doi.org/10.3892/mmr.2018.8664