Open Access

Upregulated expression of SAC3D1 is associated with progression in gastric cancer

  • Authors:
    • An‑Gui Liu
    • Jin‑Cai Zhong
    • Gang Chen
    • Rong‑Quan He
    • Yi‑Qiang He
    • Jie Ma
    • Li‑Hua Yang
    • Xiao‑Jv Wu
    • Jun‑Tao Huang
    • Jian‑Jun Li
    • Wei‑Jia Mo
    • Xin‑Gan Qin
  • View Affiliations

  • Published online on: April 15, 2020     https://doi.org/10.3892/ijo.2020.5048
  • Pages: 122-138
  • Copyright: © Liu 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

SAC3 domain containing 1 (SAC3D1) has been reported to be involved in numerous types of cancer. However, the role of SAC3D1 in GC has not yet been elucidated. In the present study, the mRNA expression level of SAC3D1 between GC and normal tissues were assessed with a continuous variable meta‑analysis based on multiple datasets from public databases. The protein expression level of SAC3D1 in GC and normal tissues was assessed by an in‑house immunohistochemistry (IHC). The association between SAC3D1 expression and some clinical parameters was assessed based on the TCGA and IHC data. Survival analysis was performed to assess the association between SAC3D1 expression and the survival of GC patients. The co‑expressed genes of SAC3D1 were determined by integrating three online tools, and the enrichment analyses were performed to determine SAC3D1‑related pathways and hub co‑expressed genes. SAC3D1 was significantly upregulated in GC tumor tissues in comparison to normal tissues with the SMD being 0.45 (0.12, 0.79). The IHC results also indicated that SAC3D1 protein expression in GC tissues was markedly higher than in normal tissues. The SMD following the addition of the IHC data was 0.59 (0.11, 1.07). The protein levels of SAC3D1 were positively associated with the histological grade, T stage and N stage of GC (P<0.001). The TCGA data also revealed that the SAC3D1 mRNA level was significantly associated with the N stage (P<0.001). Moreover, prognosis analysis indicated that SAC3D1 was closely associated with the prognosis of patients with GC. Moreover, 410 co‑expressed genes of SAC3D1 were determined, and these genes were mainly enriched in the cell cycle. In total, 4 genes (CDK1, CCNB1, CCNB2 and CDC20) were considered key co‑expressed genes. On the whole, these findings demonstrate that SAC3D1 is highly expressed in GC and may be associated with the progression of GC.

Introduction

Gastric cancer (GC) is a common malignant tumor of the digestive system that originates in the gastric mucosal epithelium. GC is a frequently diagnosed type of cancer and is an important leading cause of cancer-related mortality according to the cancer statistics of 2019 (1). Currently, the majority of patients with early-stage GC have a relatively long-term survival time after selecting surgery as a principal treatment option (2-4). In recent years, a program combining immunotherapy, molecular targeted therapy and neoadjuvant chemoradiotherapy has been shown to be a promising treatment method for GC (5-9). However, the molecular mechanisms associated with the occurrence and progression of GC remain unclear. Therefore, the exploration of cancer-related genes and specific molecular targets for the effective treatment of GC is imperative.

SAC3 domain containing 1 (SAC3D1) is a protein-coding gene located on chromosome 11 and is widely found in the cytoplasm, cytoskeleton, microtubule tissue center, centrosome and spindle (10). SAC3D1 has been reported to be abnormally expressed in multiple types of cancer and may be associated with the occurrence or progression of numerous types of cancer. A previous study reported that SAC3D1 may serve as a prognostic biomarker in hepatocellular carcinoma by combining the data of Gene Expression Omnibus (GEO), The Cancer Genome Atlas and International Cancer Genome Consortium (11). The prognostic value of SAC3D1 has also been demonstrated in colon cancer (12). You et al reported that SAC3D1 was associated with SLC2A5-inhibited adjacent lung adenocarcinoma cytoplasmic pro-B cell progression (13). However, the role and molecular mechanisms of action of SAC3D1 in GC have not yet been reported. According to a preliminary calculation with TCGA RNA-seq data, SAC3D1 was found to be significantly abnormally expressed in GC. Thus, it was speculated that SAC3D1 may play a pivotal clinical role in GC.

In the present study, GC microarray data and RNA-seq data were integrated to assess the mRNA expression of SAC3D1 in GC, and an in-house immunohistochemistry (IHC) was performed to further validate the protein expression level of SAC3D1. The co-expressed genes of SAC3D1 in GC were also collected and the possible molecule molecular mechanisms of action of SAC3D1 were analyzed by bioinformatics methods (Fig. 1).

Materials and methods

Data sources and processing

GC microarray and RNA-seq data were screened from the Sequence Read Archive (SRA; https://www.ncbi.nlm.nih.gov/sra) (14), Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/) (15), ArrayExpress(http://www.ebi.ac.uk/arrayexpress/) (16) and Oncomine (https://www.oncomine.org/resource/main.html) (17) databases with the following keywords: ('gastric' OR 'stomach' OR 'gastrointestinal') AND ('cancer' OR 'carcinoma' OR 'tumor' OR 'adenocarcinoma'). The inclusion criteria were as follows: First, the experimental group and the control group should be human GC samples and healthy samples, respectively. Second, lymph node metastasis and distant metastasis tissues were also included in the present study. Third, the calculated mRNA expression data should be provided by all included datasets. The information of included GC microarray and RNA-seq data is presented in Table I. Besides, microarray and RNA-seq data with prognostic data were screened separately for prognostic-related analysis. The mRNA expression matrix data of each dataset were downloaded, and the mRNA expression data of SAC3D1 were extracted. The SAC3D1 expression data underwent a log2 transformation and were divided into cancer groups and normal groups. The GC RNA seq data of the TCGA database were downloaded from UCSC Xena (https://xena.ucsc.edu/), which included sequencing data of 373 GC and 32 normal tissues. The data were processed as microarray data. The GC-related clinical parameters, including sex, grade, age, TNM stage and survival data, were also acquired from UCSC Xena.

Table I

SAC3D1 expression profile based on immunohistochemistry data, GEO datasets and TCGA sequencing data.

Table I

SAC3D1 expression profile based on immunohistochemistry data, GEO datasets and TCGA sequencing data.

DatasetsCountryYearPlatformPatients
Normal
t-valueP-value
NumberMeanSDNumberMeanSD
GSE103236Romania2017GPL41331010.1270.7002199.31670.423-3.0080.008
GSE81948Italy2017GPL6244157.51010.1293757.54430.108220.530.603
GSE54129China2017GPL5701116.90170.51905216.90550.264620.050.96
GSE26942USA2016GPL69472058.94930.71607129.02240.377340.610.551
GSE84787China2016GPL17077109.7583.58558109.79342.841050.0240.981
GSE64951USA2015GPL570637.60951.74653317.19082.02533-1.0360.303
GSE63089China2014GPL5175457.11860.52955457.07020.53337-0.4320.667
GSE56807China2014GPL517557.05610.2471156.97740.32808-0.4280.68
GSE29272USA2013GPL961347.02230.524611346.38740.2917212.244<0.001
GSE38940Argentina2012GPL5936340.02240.31734310.07450.475330.5150.609
GSE33429China2012GPL5175, GPL9128
254.95220.14036255.01530.118721.7150.093
GSE20143India2010GPL93655-1.05850.603792-0.80160.230930.5590.601
GSE13911Italy2008GPL80389.30521.38313317.19421.57059-5.857<0.001
GSE2685Japan2005GPL571227.09030.1747387.00790.27967-0.9680.341
GSE109476China2018GPL24530511.51940.3444511.12030.52596-1.420.194
GSE112369Japan2018GPL15207379.00610.4449258.69540.40925-2.7840.007
GSE26899USA2016GPL6947969.40180.6073129.02240.377343.02720.007
GSE79973China2016GPL57089.35850.325198.57980.607773.2290.0056
TCGA---37317.39660.788273216.81330.34279-7.984<0.001
IHC---17910.18991.930741473.23812.7779326.57<0.001

[i] SAC3D1, SAC3 domain containing 1.

In-house IHC

The tissue array that included 179 cases of GC tissues and 147 normal tissues was purchased from Pantomics, Inc. and some clinical information for each sample, such as age, sex, tumor pathological grade and clinical stage, were also provided. In the IHC analysis, SAC3D1 was detected with anti-SAC3D1 antibody (at a 1/500 dilution; cat. no. ab122809, Abcam's RabMAb technology). The SAC3D1 expression intensity for each sample was evaluated based on the score, and the score was generated from the product of the proportion of stained cells among all cells (0, <5%; 1, 5-25%; 2, 25-50%; 3, 50-75%; 4, >75%) and the staining degree of the positive cells (0, no staining; 1, light yellow or yellow; 2, brown; 3, dark brown) (18). Images were captured using an optical microscope (Motic China Group Co., Ltd.). Moreover, to improve the accuracy of results, Image-Pro Plus version 6.0 software (Media Cybernetics, Inc.) was also used to evaluate the area and density of the dyed region and the integrated optical density (IOD) value of the IHC section. The mean densitometry of the digital image (magnification, ×400) was regarded as representative SAC3D1 staining intensity (indicating the relative SAC3D1 expression level). The IOD values of the tissue areas from 179 cases of gastric cancer tissues and 147 normal tissues randomly selected fields were calculated counted in a blinded manner and subjected to statistical analysis.

Mutations of the SAC3D1 in GC

Genetic alterations of SAC3D1 in GC were investigated based on high throughput data in cBio-Portal for Cancer Genomics (cBioportal) (http://cBioportal.org) and Catalogue Of Somatic Mutations In Cancer (COSMIC) (https://cancer.sanger.ac.uk/cosmic), including missense mutation, truncating mutation, deep deletion, and amplification.

Acquisition of co-expressed genes of SAC3D1 in GC

The co-expressed genes of SAC3D1 were obtained from the Multi Experiment Matrix (https://biit.cs.ut.ee/mem/index.cgi) (19) and COXPRESdb (http://coxpresdb.jp) (20). In the Multi Experiment Matrix, P<0.05 was regarded as statistically significant. In COXPRESdb, 2000 was set as the upper limit. In addition, GC-related differentially expressed genes were calculated with the edgeR package based on TCGA and GTEx data, and a log (fold change) equal to 1 and P<0.05 was defined as including condition. The overlapped genes of three parts were considered co-expressed genes of SAC3D1 in GC.

Enrichment and protein-protein interaction (PPI) analysis

The genes co-expressed with SAC3D1 were submitted to DAVID (https://david.ncifcrf.gov/) (21) for an enrichment analysis, including gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. STRING (https://string-db.org/) (22) was utilized to construct a PPI network, and based on the degree of nodes, hub co-expressed genes of SAC3D1 were identified.

Validation of hub co-expressed genes

The expression of hub co-expressed genes was further validated at the mRNA level based on the microarray and RNA-seq data via a meta-analysis and the protein expression levels of hub co-expressed genes were verified in Human Protein Atlas (HPA) (https://www.proteinatlas.org/) (23). The sensitivity and specificity of hub co-expressed genes on differentiating GC tissues and normal tissues were also calculated. Besides, genetic alterations of the hub co-expressed genes in GC were also investigated in cBioportal. A prognosis related meta-analysis was also conducted to assess the prognosis value of hub co-expressed gene, respectively. Moreover, the expression relationship between SAC3D1 and hub co-expressed genes was presented by correlation analysis.

Statistical analysis

Independent and paired sample t-tests were performed in SPSS 19.0 to calculate and evaluate the expression level of SAC3D1 in GC tissues and normal tissues based on the GC microarray data, RNA seq data and IHC data. Stata 12.0 was used to perform a continuous variable meta-analysis and calculate the value of SMD. One-way analysis of variance was used in the present study to compare the differences in the mean of three or more sets of data. Bonferroni and Tamhane's T2 were used as post hoc tests for equal variance assumed and equal variance not assumed, respectively. In addition, the sensitivity and specificity of SAC3D1 on differentiating GC tissues and normal tissues were evaluated by drawing ROC curves in GraphPad Prism5 based on microarray data, RNA seq data, and IHC data. Stata 12.0 was also used to integrate the results of each ROC with a summary ROC. Finally, a Spearman's correlation analysis was used to examine the relationship between the expression of SAC3D1 and core co-expressed genes.

Results

Expression and clinical value of SAC3D1 in GC based on chips and RNA-seq data

First, a total of 18 eligible GEO chips and a section of TCGA sequencing data were collected, including 1,241 gastric cancer samples and 452 normal samples, from which the expression data of SAC3D1 was extracted. The expression of SAC3D1 in each chip or section of TCGA sequencing data was clarified through independent or paired sample t-tests. For the GEO chips, 5 chips (GSE103236, GSE29272, GSE13911, GSE112369 and GSE26899) exhibited a significantly upregulated trend of SAC3D1 in GC. For the TCGA sequencing data, SAC3D1 was found to be upregulated in 373 gastric cancer tissues (17.3966±0.78827) compared to 32 normal tissues (16.8133±0.34279, P<0.001) (Table I and Fig. 2). To further improve the accuracy of the results, the results of t-tests based on 18 eligible GEO chips and a section of TCGA sequencing data were integrated by a continuous variable meta-analysis. The results indicated that SAC3D1 was clearly upregulated in GC tissues with the SMD of the random effect model being 0.45 (0.12, 0.79), and the funnel plot indicated that there was no publication bias (Fig. 3A and B). The ROC of all chips and RNA-seq data was calculated (Table II and Fig. 4), and the AUC of sROC was 0.71 (0.67, 0.75), with pooled sensitivity and specificity being 0.68 (0.61, 0.74) and 0.66 (0.60, 0.72) (Fig. 5A and B). The prognosis-related meta-analysis indicated that the overexpression of SAC3D1 was closely associated with the poor prognosis of patients with GC [HR, 2.83 (2.25, 3.57); P<0.001] (Fig. 3E).

Table II

Potential of SAC3D1 to serve as a bio-marker on identifying gastric cancer tissues and normal tissue.

Table II

Potential of SAC3D1 to serve as a bio-marker on identifying gastric cancer tissues and normal tissue.

DatasetsSensitivitySpecificityTPFPFNTN
GSE10323680.00%77.80%8227
GSE8194853.33%60.00%8273
GSE5412953.15%61.90%5985213
GSE2694251.71%66.67%1064998
GSE8478760.00%70.00%6347
GSE6495158.73%54.84%37142617
GSE6308957.78%55.56%26201925
GSE5680780.00%60.00%4213
GSE2927282.09%74.63%1103424100
GSE3894064.71%51.61%22151216
GSE3342964.00%64.00%169916
GSE2014380.00%50.00%4111
GSE1391186.84%83.87%335526
GSE268563.64%62.50%14385
GSE10947680.00%80.00%4114
GSE11236962.16%68.00%2381417
GSE2689962.50%75.00%603369
GSE79973100.00%88.89%8108
TCGA72.39%65.63%2701110321
IHC96.65%86.39%173206127

[i] TP, true positive; FP, false positive; FN, false negative; TN, true negative; SAC3D1, SAC3 domain containing 1.

Expression and clinical value of SAC3D1 in GC based on chips, RNA seq data and IHC data

The protein expression of SAC3D1 was clearly high expressed in 179 GC tissues compared with 147 paracancerous tissues (Fig. 2T). The results of t-tests based on IHC data, 18 eligible GEO chips and a section of TCGA RNA-seq data were also merged by a meta-analysis. An upregulation of SAC3D1 was finally determined with the SMD of the random effect model being 0.59 (0.11, 1.07), and a corresponding funnel plot indicated that there was no publication bias (Fig. 3C and D). After constructing the sROC curve based on the IHC data, 18 eligible GEO chips and a section of TCGA RNA-seq data, it was found that SAC3D1 has a certain potential to be identified as a molecular indicator to identify GC tissues and normal tissues, and the sensitivity and specificity was 0.72 (0.63, 0.79) and 0.68 (0.62, 0.74), respectively (Fig. 5C and D). Moreover, it was found that the positive ratio of SAC3D1 staining was comparable with the original methods using Image-Pro Plus version 6.0 software (Fig. S1 and Table SI).

Association of SAC3D1 expression with clinical parameters

According to the IHC data, the upregulation of SAC3D1 was statistically associated with the histological grade, clinical stage, T stage and N stage of GC. In a more advanced stage of the disease, or histological grade, the protein expression intensity of SAC3D1 was stronger than that in low-stage or grade. Thus, it was speculated that SAC3D1 may be involved in the development and progression of GC (Fig. 6 and Table III). In addition, the association between SAC3D1 and some clinical parameters was also calculated using the TCGA data, and the results indicated that the expression of SAC3D1 was associated with the N stage (Table IV, F=7.596, P<0.001).

Table III

Association between SAC3D1 expression and some clinical pathological parameters based on immunohistochemistry data.

Table III

Association between SAC3D1 expression and some clinical pathological parameters based on immunohistochemistry data.

Clinicopathological parametersGroupSAC3D1 expression
t-valueP-value
CasesMean ± SD
TissueGC tissue179 10.1899±1.93074
Normal tissue1473.2381±2.7779326.57P<0.001
Age (years)≤5046 10.3043±2.22979
>50128 10.1484±1.836160.4660.642
SexMale12810.125±1.99606
Female4610.3696±1.79330.7310.466
TT1-T2549.1111±1.9683
T3-T412010.675±1.73041-5.029P<0.001
NN0659.1692±1.98879
N187 10.7356±1.69445
N222 11.0455±1.4301917.277P<0.001
StageIA-IB388.7105±1.99875
IIA-IIB117 10.5043±1.75491
IIIA19 11.2105±1.3572418.192P<0.001
Histological gradeI288.5714±2.1846
II5610.25±1.77098
III63 10.8125±1.62202F=15.261P<0.001

[i] SAC3D1, SAC3 domain containing 1.

Table IV

Association between SAC3D1 expression and some clinical pathological parameters based on TCGA data.

Table IV

Association between SAC3D1 expression and some clinical pathological parameters based on TCGA data.

Clinicopathological parametersSAC3D1 expression
t-valueP-value
nMean ± SD
Tissue
 Non-tumor32 16.8133±0.34279
 GC373 17.3966±0.78827-7.984<0.001
Sex
 Male25817.3224±0.7312
 Female143 17.4426±0.827681.5040.133
Age (years)
 <60124 17.3489±0.74119
 ≥60273 17.3752±0.78175-0.3160.752
Grade
 G11117.094±1.12374
 G214717.3488±0.7554
 G3235 17.3891±0.76078
 Gx8 17.3374±0.72202F=0.5570.644
TNM
 T1-T1b25 17.2418±0.75401
 T2-T2b88 17.3177±0.85172
 T317917.452±0.72991
 T4-T4b105 17.3393±0.71874F=1.1170.342
 N0121 17.4878±0.77501
 N1104 17.3645±0.74107
 N28517.361±0.63662
 N3-N3b74 17.3808±0.79604
 Nx16 16.3963±0.82501F=7.596<0.001
 M035217.393±0.73777
 M127 17.1293±1.08533
 Mx2217.2109±0.7571F=1.9570.143
 I-IB5917.397±0.84514
Stage
 II-IIB124 17.4667±0.69269
 III-IIIC156 17.4222±0.64408
 IV42 17.2256±0.98701F=1.1420.332

[i] SAC3D1, SAC3 domain containing 1.

Genetic alterations of the SAC3D1 in GC

From the online analysis of cBioPortal and COSMIC, it was found that SAC3D1 has a mutation in GC, although the genetic alteration rate was relatively low. Therefore, it was hypothesized that the role of highly expressed SAC3D1 in the development of GC may not be mutated, amplification-mediated (Fig. 7).

Enrichment and PPI analysis of co-expressed gene of SAC3D1

A total of 8,364 and 2,000 co-expressed genes of SAC3D1 were obtained in the Multi Experiment Matrix (https://biit.cs.ut.ee/mem/index.cgi) and COXPRESdb, respectively. In addition, 4,640 GC-related differentially expressed genes were acquired after TCGA and GTEx data calculations. Finally, 410 overlapping genes of 3 parts were considered co-expressed genes of SAC3D1 in GC (Fig. 8A). The GO-enriched analysis indicated that SAC3D1 and co-expressed genes were mainly enriched in mitotic sister chromatid segregation, nuclear chromosome and ATP binding (Table V and Fig. 8C-F). In the KEGG pathway analysis, the SAC3D1 and co-expressed genes were mainly enriched in DNA replication and the cell cycle (Table VI and Fig. 8B and G). The PPI network indicated that CDK1, CCNB1, CCNB2 and CDC20 were the hub co-expressed genes of SAC3D1 in GC (Fig. 9A and B).

Table V

The top 10 GO items associated with SAC3D1 and its co-expressed genes.

Table V

The top 10 GO items associated with SAC3D1 and its co-expressed genes.

CategoryIDTermCountP-value
BPGO:0051301Cell division696.42E-44
BPGO:0006260DNA replication485.22E-40
BPGO:0007067Mitotic nuclear division496.14E-31
BPGO:0000082G1/S transition of mitotic cell cycle334.98E-28
BPGO:0007062Sister chromatid cohesion321.46E-26
BPGO:0006270DNA replication initiation196.20E-22
BPGO:0006281DNA repair355.10E-18
BPGO:0000086G2/M transition of mitotic cell cycle266.69E-16
BPGO:0000070Mitotic sister chromatid segregation141.42E-15
BPGO:0000722Telomere maintenance via recombination148.38E-14
CCGO:0005654Nucleoplasm1897.71E-53
CCGO:0005634Nucleus2111.05E-22
CCGO:0000776Kinetochore231.67E-18
CCGO:0000777Condensed chromosome kinetochore239.02E-18
CCGO:0000922Spindle pole259.22E-18
CCGO:0000775Chromosome, centromeric region191.14E-16
CCGO:0005829Cytosol1412.46E-16
CCGO:0005813Centrosome416.89E-15
CCGO:0030496Midbody226.37E-13
CCGO:0005819Spindle211.70E-12
MFGO:0005515Protein binding3055.16E-29
MFGO:0005524ATP binding807.62E-13
MFGO:0003682Chromatin binding352.18E-11
MFGO:0019901Protein kinase binding343.39E-11
MFGO:0043142Single-stranded DNA-dependent
ATPase activity72.59E-08
MFGO:0008017Microtubule binding216.22E-08
MFGO:0003677DNA binding721.66E-07
MFGO:0003697Single-stranded DNA binding141.79E-07
MFGO:0003684Damaged DNA binding111.44E-06
MFGO:0003777Microtubule motor activity121.88E-06

[i] SAC3D1, SAC3 domain containing 1.

Table VI

The 10-most KEGG pathways associated with SAC3D1 and its co-expressed genes.

Table VI

The 10-most KEGG pathways associated with SAC3D1 and its co-expressed genes.

CategoryIDTermP-value
KEGGhsa04110Cell cycle1.05E-31
KEGGhsa03030DNA replication2.24E-19
KEGGhsa00240Pyrimidine metabolism1.25E-08
KEGGhsa03430Mismatch repair1.01E-07
KEGGhsa04115p53 signaling pathway1.78E-06
KEGGhsa04114Oocyte meiosis1.87E-06
KEGGhsa03460Fanconi anemia pathway1.20E-05
KEGGhsa03410Base excision repair2.55E-05
KEGGhsa03420Nucleotide excision repair2.71E-04
KEGGhsa05203Viral carcinogenesis5.13E-04

[i] SAC3D1, SAC3 domain containing 1.

Validation of hub co-expressed genes based on TCGA and HPA

Various types of mutations of the 4 hub co-expressed genes (CDK1, CCNB1, CCNB2 and CDC20) were observed in GC (Fig. 9C). CDK1, CCNB1, CCNB2 and CDC20 were evidently highly expressed in GC based on the microarray and RNA-seq data mRNA expression data (Fig. 10A-D) and CDK1, CCNB1, CCNB2 and CDC20 may also serve as biomarkers differentiating GC tissues and normal tissues with a relative high sensitivity and specificity (Fig. 10E-H). The high expression trends of these 4 genes were also observed in protein expression data based on the HPA database (Fig. 11). These genes were risk factors affecting the prognosis of gastric cancer (Fig. 12A-D). Moreover, Spearman's correlation analysis indicated that there were significant positive correlations between SAC3D1 and these core co-expressed genes (Fig. 12E-H).

Discussion

In the present study, the expression of SAC3D1 in GC was determined by integrated and thoroughly re-processed 18 GEO chips, TCGA RNA-seq data and IHC data, which included 1,420 GC tissues and 599 normal tissues. Notably, both SAC3D1 mRNA and protein levels were observed to be upregulated in GC tissues. The overexpression SAC3D1 was associated with the histological grade, clinical stage, T stage and N stage of GC, revealing that SAC3D1 may be involved in the development and progression of GC. Enrichment analysis revealed that SAC3D1 and 4 other SAC3D1-related genes (CDK1, CCNB1, CCNB2 and CDC20) are important for GC development via the cell cycle pathway.

Numerous studies have reported the overexpression of SAC3D1 in several types of cancer, including hepatocellular carcinoma (11), colon cancer (12) and lung adenocarcinoma (13). Recent studies have assessed the prognostic value of SAC3D1 using GEO, the Cancer Genome Atlas and International Cancer Genome Consortium and suggested that SAC3D1 may be a credible prognosis-related biomarker for hepatocellular carcinoma (11). In colon cancer, the upregulation of SAC3D1 was confirmed by a quantitative PCR (12). In lung adenocarcinoma, SAC3D1 may be involved in the inhibition of cytoplasmic pro-B cell developmental mechanisms in paracancerous tissue of lung adenocarcinoma by low glucose transporter SLC2A5 (13). However, to the best of our knowledge, no studies to date have clarified the expression of SAC3D1 in GC, and the expression of SAC3D1 in other cancers was only validated based on small sample sizes or a single method, which may decrease the reliability of their conclusion. Particularly, no research or clinical trials have specifically been done attempting to reveal the molecular mechanisms of SAC3D1 in cancers, including GC.

To explore the possible molecular mechanisms of actoin of SAC3D1 in GC, an enrichment analysis was performed for SAC3D1 and its co-expressed genes. The results indicated that SAC3D1 and co-expressed genes were positively associated with the cell cycle. Additionally, numerous studies have demonstrated that the cell cycle pathway plays an important role in cancer cells. Cao et al reported that the regulatory mechanism of BIRC5 and co-expressed genes in lung carcinoma may be closely related to the cell cycle (24). Liu et al reported that upregulated differentially expressed genes participated in regulating breast cancer cells by the cell cycle pathway (25). Moreover, Qiu et al revealed that the modules and central genes associated with the development of breast cancer were significantly enriched in the cell cycle pathway (26). Feng et al investigated poor prognosis-related genes of ovarian cancer by bioinformatics analysis and found that these genes were mainly enriched in the cell cycle pathway (27). It has also been reported that the cell cycle pathway is the key signaling pathway for 8 target therapy of neuroblastomas (28). Zhang et al reported that LncRNA CASC11 promoted the proliferation, migration, and invasion of GC cells in vitro via the cell cycle pathway (29). A number of studies have documented that the cell cycle pathway may play a role in the regulation of multiple types of cancer, including GC and enrichment analysis revealed that SAC3D1 and its co-expressed genes were involved in the cell cycle pathway. This prompted the hypothesis that SAC3D1 may be related to the occurrence and progression of GC. A total of 4 genes (CDK1, CCNB1, CCNB2 and CDC20) were determined as the core co-expressed genes of SAC3D1 in GC, and it was speculated that SAC3D1 may cooperate with these genes to promote the progression of GC. Further in vitro experimental analyses are still required to verify the findings of the present study, such as SAC3D1 overexpression or interference.

CDK1 is a cell cycle-related gene that can be regulated by KIAA0101 and is involved in the occurrence and development of GC (30). CDK1 can also be regulated by LncRNA CASC11 and then participate in the proliferation, migration, and invasion of GC cells (29). Guo et al demonstrated that rhCNB may decrease the expression of cell cycle B1 and CDK1 proteins and participate in the mechanism of cell cycle arrest (31). CCNB1 is a cell cycle-related gene that can be regulated by ISL1 to promote the proliferation and tumor growth of GC cells (32). CCNB1 can be used as a biomarker to monitor prognosis and hormone therapy in ER breast cancer (33). It has also been reported that the overexpression of CCNB1 induced by chk1 can promote the proliferation and tumor growth of human colorectal cancer cells and inhibit the induction of apoptosis in some colorectal cancer cells (34). CCNB1 could also activate FOXM1 and promote the proliferation of human hepatocel-lular carcinoma cells (35). CCNB1 may serve as a promising diagnostic tool for determining the high risk of recurrence in patients with non-myenteric invasive bladder cancer (36). CCNB2 is a cell cycle-related gene that can be regulated by ISL1 to promote the proliferation and tumor growth of GC cells (32). In addition, the overexpression of CCNB2 protein is related to the clinical progress and poor prognosis of non-small cell lung cancer, and over-expressed CCNB2 is a biomarker of poor prognosis in Chinese patients with non-small cell lung cancer (37). The increased expression of the cell cycle-related gene CCNB2 is related to the advanced growth of prostate cancer cell subsets (38). Kim et al reported that the expression of CDC20 in early GC was significantly higher than that in normal mucous membranes (39). The upregulation of CDC20 was associated with invasive progress and poor prognosis in GC, and it was identified as an independent marker for predicting clinical outcomes in patients with GC (40). It has also been reported that CDC20 expression can be used as a biomarker for tumor prognosis or as a therapeutic target for other human cancers (41). In addition, CDC20 can mediate docetaxel resistance to castrated prostate cancer (42).

Microarray and RNA-seq data were combined to evaluate the prognostic value of 4 hub co-expressed genes via a prognostic-related meta-analysis. It was found that the upregulation of these genes were closely related to the poor prognosis of patients with GC. From online analysis, it was found that the genetic alterations rate of SAC3D1 and its hub co-expressed genes in GC was relatively low. Therefore, it speculated that mutation and amplification may not be the main reasons for SAC3D1 to promote the development of GC. Further experimental analyses are warranted. In conclusion, the findings of the present study demonstrate that SAC3D1 is highly expressed in GC and may be associated with the progression of GC.

Supplementary Data

Funding

The present study was supported by the Guangxi medical and Health Appropriate Technology Development And Promotion Application Project (S201657), Guangxi Zhuang Autonomous Region Health Committee Self-financed Scientific Research Project (Z20190594), Guangxi Degree and Postgraduate Education Reform and Development Research Projects, China (JGY2019050), Future Academic Star of Guangxi Medical University (WLXSZX19077).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the TCGA (http://cancergenome.nih.gov/), the GEO (https://www.ncbi.nlm.nih.gov/geo/) and the SRA (https://www.ncbi.nlm.nih.gov/sra/) data portals. The in-house IHC data from the present study can be acquired from the correspondence author on reasonable request.

Authors' contributions

AGL and JCZ collected data from public datasets and analyzed the data and performed the statistical analysis. XGQ and WJM performed in-house IHC experiments. GC, RQH and JJL participated in the conception and design of the study and in language modification. AGL, JCZ and YQH drafted the manuscript and analyzed the GO and KEGG terms. JM, LHY, XJW and JTH conceived and designed the study and assisted in the drafting of the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

This research program was approved by the Ethics Committee of the First Affiliated Hospital of Guangxi Medical University. All participants signed informed consent forms as collected by Pantomics.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank all members of the Molecular Oncology Group of the First Affiliated Hospital of Guangxi Medical University (Nanning, Guangxi Zhuang Autonomous Region 530021, China) for their professional suggestions. At the same time, the authors would like to thank GEO, ArrayExpress, Oncomine, SRA, TCGA, Human Protein Atlas and other websites for providing valuable data.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Kim HH, Han SU, Kim MC, Kim W, Lee HJ, Ryu SW, Cho GS, Kim CY, Yang HK, Park DJ, et al: Korean Laparoendoscopic Gastrointestinal Surgery Study (KLASS) Group: Effect of laparo-scopic distal gastrectomy vs. open distal gastrectomy on long-term survival among patients with stage I gastric cancer: The KLASS-01 Randomized Clinical Trial. JAMA Oncol. 5:506–513. 2019. View Article : Google Scholar : PubMed/NCBI

3 

De Franco L, Marrelli D, Voglino C, Vindigni C, Ferrara F, Di Mare G, Iudici L, Marini M and Roviello F: Prognostic value of perineural invasion in resected gastric cancer patients according to Lauren histotype. Pathol Oncol Res. 24:393–400. 2018. View Article : Google Scholar

4 

Liu W, Quan H, Chen X, Ouyang Y and Xiao H: Clinicopathological features and prognosis of young gastric cancer patients following radical gastrectomy: A propensity score matching analysis. Sci Rep. 9:59432019. View Article : Google Scholar : PubMed/NCBI

5 

Huang T, Song C, Zheng L, Xia L, Li Y and Zhou Y: The roles of extracellular vesicles in gastric cancer development, micro-environment, anti-cancer drug resistance, and therapy. Mol Cancer. 18:622019. View Article : Google Scholar

6 

Ahn MJ, Lee K, Lee KH, Kim JW, Kim IY and Bae WK: Combination of anti-PD-1 therapy and stereotactic radiosurgery for a gastric cancer patient with brain metastasis. A case report BMC Cancer. 18:1732018. View Article : Google Scholar

7 

Zhao D, Zhang Y and Song L: MiR-16-1 targeted silences far upstream element binding protein 1 to advance the chemosensitivity to adriamycin in gastric cancer. Pathol Oncol Res. 24:483–488. 2018. View Article : Google Scholar

8 

Xu J, Zhu J and Wei Q: Adjuvant radiochemotherapy versus chemotherapy alone for gastric cancer: Implications for target definition. J Cancer. 10:458–466. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Charalampakis N, Economopoulou P, Kotsantis I, Tolia M, Schizas D, Liakakos T, Elimova E, Ajani JA and Psyrri A: Medical management of gastric cancer: A 2017 update. Cancer Med. 7:123–133. 2018. View Article : Google Scholar :

10 

Rappaport N, Fishilevich S, Nudel R, Twik M, Belinky F, Plaschkes I, Stein TI, Cohen D, Oz-Levi D, Safran M, et al: Rational confederation of genes and diseases: NGS interpretation via GeneCards, MalaCards and VarElect. Biomed Eng Online. 16(Suppl 1): 722017. View Article : Google Scholar : PubMed/NCBI

11 

Han ME, Kim JY, Kim GH, Park SY, Kim YH and Oh SO: SAC3D1: A novel prognostic marker in hepatocellular carcinoma. Sci Rep. 8:156082018. View Article : Google Scholar : PubMed/NCBI

12 

Fan J, Yan D, Teng M, Tang H, Zhou C, Wang X, Li D, Qiu G and Peng Z: Digital transcript profile analysis with aRNA-LongSAGE validates FERMT1 as a potential novel prognostic marker for colon cancer. Clin Cancer Res. 17:2908–2918. 2011. View Article : Google Scholar : PubMed/NCBI

13 

You J, Wang L, Huang J, Jiang M, Chen Q, Wang Y and Jiang Z: Low glucose transporter SLC2A5-inhibited human normal adjacent lung adenocarcinoma cytoplasmic pro-B cell development mechanism network. Mol Cell Biochem. 399:71–76. 2015. View Article : Google Scholar

14 

Tsui B, Dow M, Skola D and Carter H: Extracting allelic read counts from 250,000 human sequencing runs in sequence read archive. Pac Symp Biocomput. 24:196–207. 2019.PubMed/NCBI

15 

Gao L, Zhang LJ, Li SH, Wei LL, Luo B, He RQ and Xia S: Role of miR-452-5p in the tumorigenesis of prostate cancer: A study based on the Cancer Genome Atl (TCGA), Gene Expression Omnibus (GEO), and bioinformatics analysis. Pathol Res Pract. 214:732–749. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Guo YN, Luo B, Chen WJ, Chen X, Peng ZG, Wei KL and Chen G: Comprehensive clinical implications of homeobox A10 in 3,199 cases of non-small cell lung cancer tissue samples combining qRT-PCR, RNA sequencing and microarray data. Am J Transl Res. 11:45–66. 2019.PubMed/NCBI

17 

Li J, Su T, Yang L, Zhang C and He Y: High expression of MRE11 correlates with poor prognosis in gastric carcinoma. Diagn Pathol. 14:602019. View Article : Google Scholar : PubMed/NCBI

18 

Lin P, Xiong DD, Dang YW, Yang H, He Y, Wen DY, Qin XG and Chen G: The anticipating value of PLK1 for diagnosis, progress and prognosis and its prospective mechanism in gastric cancer: A comprehensive investigation based on high-throughput data and immunohistochemical validation. Oncotarget. 8:92497–92521. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Gan BL, He RQ, Zhang Y, Wei DM, Hu XH and Chen G: Downregulation of HOXA3 in lung adenocarcinoma and its relevant molecular mechanism analysed by RT-qPCR, TCGA and in silico analysis. Int J Oncol. 53:1557–1579. 2018.PubMed/NCBI

20 

Obayashi T, Kagaya Y, Aoki Y, Tadaka S and Kinoshita K: COXPRESdb v7: A gene coexpression database for 11 animal species supported by 23 coexpression platforms for technical evaluation and evolutionary inference. Nucleic Acids Res. 47:D55–D62. 2019. View Article : Google Scholar :

21 

Zhong X, Huang G, Ma Q, Liao H, Liu C, Pu W, Xu L, Cai Y and Guo X: Identification of crucial miRNAs and genes in esophageal squamous cell carcinoma by miRNA-mRNA integrated analysis. Medicine (Baltimore). 98:e162692019. View Article : Google Scholar

22 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2019. View Article : Google Scholar

23 

Colwill K, Gräslund S, Graslund S, Renewable Protein Binder and Working Group: A roadmap to generate renewable protein binders to the human proteome. Nat Methods. 8:551–558. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Cao Y, Zhu W, Chen W, Wu J, Hou G and Li Y: Prognostic value of BIRC5 in lung adenocarcinoma lacking EGFR, KRAS, and ALK mutations by integrated bioinformatics analysis. Dis Markers. 2019:54512902019. View Article : Google Scholar : PubMed/NCBI

25 

Liu F, Wu Y, Mi Y, Gu L, Sang M and Geng C: Identification of core genes and potential molecular mechanisms in breast cancer using bioinformatics analysis. Pathol Res Pract. 215:1524362019. View Article : Google Scholar : PubMed/NCBI

26 

Qiu J, Du Z, Wang Y, Zhou Y, Zhang Y, Xie Y and Lv Q: Weighted gene co-expression network analysis reveals modules and hub genes associated with the development of breast cancer. Medicine (Baltimore). 98:e143452019. View Article : Google Scholar

27 

Feng H, Gu ZY, Li Q, Liu QH, Yang XY and Zhang JJ: Identification of significant genes with poor prognosis in ovarian cancer via bioinformatical analysis. J Ovarian Res. 12:352019. View Article : Google Scholar : PubMed/NCBI

28 

Shaabanpour Aghamaleki F, Mollashahi B, Aghamohammadi N, Rostami N, Mazloumi Z, Mirzaei H, Moradi A, Sheikhpour M and Movafagh A: Bioinformatics analysis of key genes and pathways for medulloblastoma as a therapeutic target. Asian Pac J Cancer Prev. 20:221–227. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Zhang L, Kang W, Lu X, Ma S, Dong L and Zou B: LncRNA CASC11 promoted gastric cancer cell proliferation, migration and invasion in vitro by regulating cell cycle pathway. Cell Cycle. 17:1886–1900. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Wang Z, Dang C, Yan R, Zhang H, Yuan D and Li K: Screening of cell cycle-related genes regulated by KIAA0101 in gastric cancer. Nan Fang Yi Ke Da Xue Xue Bao. 38:1151–1158. 2018.In Chinese. PubMed/NCBI

31 

Guo Y, Huang Y, Tian S, Xie X, Xing G and Fu J: Genetically engineered drug rhCNB induces apoptosis and cell cycle arrest in both gastric cancer cells and hepatoma cells. Drug Des Devel Ther. 12:2567–2575. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Shi Q, Wang W, Jia Z, Chen P, Ma K and Zhou C: ISL1, a novel regulator of CCNB1, CCNB2 and c-MYC genes, promotes gastric cancer cell proliferation and tumor growth. Oncotarget. 7:36489–36500. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ding K, Li W, Zou Z, Zou X and Wang C: CCNB1 is a prognostic biomarker for ER+ breast cancer. Med Hypotheses. 83:359–364. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Fang Y, Yu H, Liang X, Xu J and Cai X: Chk1-induced CCNB1 overexpression promotes cell proliferation and tumor growth in human colorectal cancer. Cancer Biol Ther. 15:1268–1279. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Chai N, Xie HH, Yin JP, Sa KD, Guo Y, Wang M, Liu J, Zhang XF, Zhang X, Yin H, et al: FOXM1 promotes proliferation in human hepatocellular carcinoma cells by transcriptional activation of CCNB1. Biochem Biophys Res Commun. 500:924–929. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Kim SK, Roh YG, Park K, Kang TH, Kim WJ, Lee JS, Leem SH and Chu IS: Expression signature defined by FOXM1-CCNB1 activation predicts disease recurrence in non-muscle-invasive bladder cancer. Clin Cancer Res. 20:3233–3243. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Qian X, Song X, He Y, Yang Z, Sun T, Wang J, Zhu G, Xing W and You C: CCNB2 overexpression is a poor prognostic biomarker in Chinese NSCLC patients. Biomed Pharmacother. 74:222–227. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Horning AM, Wang Y, Lin CK, Louie AD, Jadhav RR, Hung CN, Wang CM, Lin CL, Kirma NB, Liss MA, et al: Single-cell RNA-seq reveals a subpopulation of prostate cancer cells with enhanced cell-cycle-related transcription and attenuated androgen response. Cancer Res. 78:853–864. 2018. View Article : Google Scholar

39 

Kim Y, Choi JW, Lee JH and Kim YS: Spindle assembly checkpoint MAD2 and CDC20 overexpressions and cell-in-cell formation in gastric cancer and its precursor lesions. Hum Pathol. 85:174–183. 2019. View Article : Google Scholar

40 

Ding ZY, Wu HR, Zhang JM, Huang GR and Ji DD: Expression characteristics of CDC20 in gastric cancer and its correlation with poor prognosis. Int J Clin Exp Pathol. 7:722–727. 2014.PubMed/NCBI

41 

Gayyed MF, El-Maqsoud NM, Tawfiek ER, El Gelany SA and Rahman MF: A comprehensive analysis of CDC20 overexpression in common malignant tumors from multiple organs: Its correlation with tumor grade and stage. Tumour Biol. 37:749–762. 2016. View Article : Google Scholar

42 

Wu F, Lin Y, Cui P, Li H, Zhang L, Sun Z, Huang S, Li S, Huang S, Zhao Q, et al: Cdc20/p55 mediates the resistance to docetaxel in castration-resistant prostate cancer in a Bim-dependent manner. Cancer Chemother Pharmacol. 81:999–1006. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 57 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu AG, Zhong JC, Chen G, He RQ, He YQ, Ma J, Yang LH, Wu XJ, Huang JT, Li JJ, Li JJ, et al: Upregulated expression of SAC3D1 is associated with progression in gastric cancer. Int J Oncol 57: 122-138, 2020
APA
Liu, A., Zhong, J., Chen, G., He, R., He, Y., Ma, J. ... Qin, X. (2020). Upregulated expression of SAC3D1 is associated with progression in gastric cancer. International Journal of Oncology, 57, 122-138. https://doi.org/10.3892/ijo.2020.5048
MLA
Liu, A., Zhong, J., Chen, G., He, R., He, Y., Ma, J., Yang, L., Wu, X., Huang, J., Li, J., Mo, W., Qin, X."Upregulated expression of SAC3D1 is associated with progression in gastric cancer". International Journal of Oncology 57.1 (2020): 122-138.
Chicago
Liu, A., Zhong, J., Chen, G., He, R., He, Y., Ma, J., Yang, L., Wu, X., Huang, J., Li, J., Mo, W., Qin, X."Upregulated expression of SAC3D1 is associated with progression in gastric cancer". International Journal of Oncology 57, no. 1 (2020): 122-138. https://doi.org/10.3892/ijo.2020.5048