Open Access

A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies

  • Authors:
    • Chunlin Ge
    • Xuan Zhu
    • Xing Niu
    • Bingye Zhang
    • Lijie Chen
  • View Affiliations

  • Published online on: November 3, 2020     https://doi.org/10.3892/mmr.2020.11663
  • Article Number: 25
  • Copyright: © Ge 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

The purpose of the present study was to identify aberrantly expressed genes for gallbladder cancer based on the annotation analysis of microarray studies and to explore their potential functions. Differential gene expression was investigated in cholesterol polyps, gallbladder adenoma and gallbladder cancer using microarrays. Subsequently, microarray results were comprehensively analyzed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to determine the affected biological processes or pathways. Differentially expressed genes (DEGs) of cholesterol polyps, gallbladder adenoma and gallbladder cancer were identified. Following comprehensive analysis, 14 genes were found to be differentially expressed in the gallbladder wall of both gallbladder cancer and gallbladder adenoma. The 20 most significantly upregulated genes were only upregulated in the gallbladder wall of gallbladder cancer, but not in the gallbladder wall of cholesterol polyps and gallbladder adenoma. In addition, 182 DEGs were upregulated in the gallbladder wall of gallbladder adenoma compared with the gallbladder wall of cholesterol polyps. A total of 20 most significant DEGs were found in both the tumor and gallbladder wall of gallbladder cancer. In addition, the most significant DEGs that were identified were only upregulated in the tumor of gallbladder cancer. GO and KEGG analysis indicated that the aforementioned DEGs could participate in numerous biological processes or pathways associated with the development of gallbladder cancer. The present findings will help improve the current understanding of tumorigenesis and the development of gallbladder cancer.

Introduction

Gallbladder cancer is the most common malignant tumor of the biliary tract and is the third most common gastrointestinal malignancy worldwide (1). Due to the vague clinical symptoms and signs of gallbladder cancer, most patients are diagnosed at an advanced stage (2). Since the etiology and pathogenesis of gallbladder cancer are still unclear, it is essential to research the molecular mechanism of the disease and explore novel potential biomarkers that may assist early diagnosis and treatment.

Gallbladder adenoma is a rare disease and rarely malignant, but transformation may occur (3). Previous evidence has proposed that adenomas are the premalignant lesions of gallbladder cancer (3,4). However, the genetic evidence is still poorly defined (5). Due to the poor prognosis of gallbladder cancer, it is crucial to distinguish benign and malignant gallbladder adenoma (6). There is a need for accurate diagnostic methods to distinguish between benign and malignant diseases. Currently, the size and number of gallbladder polyps, along with patient age, are typically used to assist with distinguishing benign and malignant diseases (7). For example, previous research has found that conjugated bile acids (glycochenodeoxycholic and taurochenodeoxycholic) could be identified as possible biomarkers for cholesterol polyps and adenomatous polyps, and the gallbladder bile acids glycochenodeoxycholic acid and taurochenodeoxycholic acid are highly expressed in cholesterol polyps (8). For patients with gallbladder carcinoma, compared with healthy individuals and patients with cholesterol polyps, serum vascular endothelial growth factors (SVEGF)-C are closely related with lymph node metastasis, distant metastasis and stage, in addition, SVEGF-D has a positive relationship with the tumor depth, lymph, distant metastasis and stage that could represent available biomarkers for the diagnosis of gallbladder carcinoma (6).

The present study aimed to comprehensively analyze a transcriptome profile and identify DEGs in gallbladder cancer based on annotation analysis of microarray studies.

Materials and methods

Patients and tissue samples

Gallbladder stones (two men and one woman; age range, 60–62 years), gallbladder adenoma (two men and one woman; age range, 60–62 years) and gallbladder carcinoma (two men and one woman; age range, 60–63 years) tissues (n=3 each) were obtained from the Department of Pancreaticobiliary Surgery, the First Affiliated Hospital of China Medical University between September 2018 and December 2019. All cases were reviewed by two or more independent pathologists. No patients received radiation or chemotherapy before surgery. During the surgery, fresh tumor tissues or gallbladder wall tissues were collected in the operating room and immediately frozen in liquid nitrogen within 15 min and then stored in RNA Fixer reagent (Thermo Fisher Scientific, Inc.) at −80°C for total RNA extraction. The present study was approved by the Ethics Committee of The First Affiliated Hospital of China Medical University (2018075). All patients who participated in the study signed written informed consent.

RNA extraction and transcript analysis

RNA extraction was performed using an RNeasy kit (Qiagen, Inc.) according to the manufacturer's protocol. Total RNA was quantified using a NanoDrop ND-2000 spectrophotometer (Thermo Fisher Scientific, Inc.) and the RNA integrity was assessed with an Agilent Bioanalyzer 2100 (Agilent Technologies, GmbH). Total RNA samples were analyzed on the Agilent Bioanalyzer 2100 and amplified RNA (aRNA) was prepared using the GeneChip 3′IVT Express kit (Affymetrix; Thermo Fisher Scientific, Inc.). Briefly, cDNA was synthesized by reverse transcription, and a double-stranded DNA template was then obtained by second-strand synthesis. Subsequently, an aRNA labeled with biotin was inverted in vitro utilizing GeneChip 3′IVT Express kit (Affymetrix; Thermo Fisher Scientific, Inc.) at 40°C for 16 h and stored at 4°C. The aRNA was purified, fragmented and hybridized with the chip probe (Beckman Coulter, Inc.). Following hybridization, the chip was automatically washed using a GeneChip Hybridization Wash and Stain kit (Affymetrix; Thermo Fisher Scientific, Inc.) and dyed using GeneChip Fluidics Station 450 instrument (Affymetrix; Thermo Fisher Scientific, Inc.). Finally, it was scanned to obtain the image and the Affymetrix microarray data using a GeneChip Scanner 3000 (Affymetrix; Thermo Fisher Scientific, Inc.).

To obtain the raw data, the Feature Extraction function in GeneSpring (version 10.5.1.1; Agilent Technologies, GmbH) was utilized to analyze the array image. Briefly, the raw data were normalized with the quantile algorithm. In the experiment, probe groups in the lowest 20% of the signal strength in the two sample groups were filtered as background noise. The coefficient of variation of the probe group was calculated in the sample group, and the probe group with a coefficient of variation >25% in both groups were also filtered out. Finally, DEG transcripts were identified.

To explore DEGs in different gallbladder diseases, further analysis was conducted, as shown in Table I. In the present study, gallstones served as normal samples compared with cholesterol polyps, gallbladder adenoma and gallbladder cancer.

Table I.

Six groups for differential expression analysis via microarray analysis.

Table I.

Six groups for differential expression analysis via microarray analysis.

GroupsTissue typeNumber of samplesComparison
Group IGallbladder wall3Cholesterol polyps vs. gallbladder stones
Group IIGallbladder wall3Gallbladder adenoma vs. gallbladder stones
Group IIIGallbladder wall3Gallbladder cancer vs. gallbladder stones
Group IVGallbladder wall3Gallbladder adenoma vs. cholesterol polyps
Group VGallbladder wall3Gallbladder cancer vs. gallbladder adenoma
Group VITumor tissue3Gallbladder cancer vs. gallbladder adenoma
Differential expression analysis

In the present study, linear models for microarray data (version 3.44.3; Bioconductor) were performed based on empirical Bayesian distribution to calculate the P-value (9). The screening criteria for DEGs was as follows: |Fold change (FC)|>1.5 and P-value <0.05. To probe out DEGs in cholesterol polyps, gallbladder adenoma and gallbladder cancer, differential expression analyses including scatter plot analysis, volcano plot analysis and hierarchical clustering analysis were performed using GraphPad Prism (version 7.0; GraphPad Software, Inc.).

Comparative analysis

To explore differentially expressed genes between different diseases a comparative analysis was undertaken, as shown in Table II and Fig. 1. In group II, the gallbladder wall of gallbladder adenoma and gallbladder wall of gallbladder stones were compared. In group III, the gallbladder wall of gallbladder cancer and gallbladder wall of gallbladder stone were compared. In group IV, comparative analysis was performed between gallbladder wall of gallbladder adenoma and gallbladder wall of cholesterol polyps. For group V, comparative analysis between gallbladder wall of gallbladder cancer and gallbladder wall of gallbladder adenoma was presented.

Table II.

Comprehensive analysis of DEGs in the six groups.

Table II.

Comprehensive analysis of DEGs in the six groups.

Comparative groupsGallbladder wall of gallstoneCholesterol polyps (gallbladder wall)Gallbladder adenoma (gallbladder wall)Gallbladder cancer (gallbladder wall)Gallbladder adenoma (tumor wall)Gallbladder cancer (tumor wall)
I
II
III
IV
V
VI
A
  a=I∩II∩III
  b=II∩III-a
  c=II-b-I∩II
  d=III-b-I∩III
B=IV
C=V∩VI
D=VI–V∩VI

[i] ∩ represents the common DEGs between two groups, √ and ○ represent different samples (gallbladder wall of gallstone, gallbladder wall of cholesterol polyps, gallbladder wall of gallbladder adenoma and tumor wall of gallbladder adenoma) used for intersection. DEGs, differentially expressed genes.

Functional enrichment analysis

To explore the biological processes or pathways involved in DEGs, Gene Ontology (GO; http://geneontology.org/) and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.kegg.jp/kegg/) pathway enrichment analyses were performed (10,11). GO terms include ‘biological process’ (BP), ‘molecular function’ (MF) and ‘cellular component’ (CC).

Validation of the differential expression and prognostic value of key genes using gene expression profiling interactive analysis (GEPIA)

Key genes were verified by GEPIA (http://gepia.cancer-pku.cn/) in The Cancer Genome Atlas (https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) and Genotype-Tissue Expression dataset (GTEx; http://commonfund.nih.gov/GTEx/) (12). Differential expression and overall survival (OS) analyses were performed.

Reverse transcription-quantitative PCR (RT-qPCR) assay

A total of 10 pairs of gallbladder cancer tissues and normal tissues (five men and five woman; age range, 55–65 years) were collected from the Department of Pancreaticobiliary Surgery, the First Affiliated Hospital of China Medical University between September 2018 and December 2019. All patients signed written informed consent. Total RNA was extracted from tissues using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). According to the manufacturer's instructions, reverse transcription was performed using a TaqMan Real-Time PCR kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). RT-qPCR was run on a CFX96 Real-Time PCR detection system (Bio-Rad Laboratories, Inc.). The following primer pairs were used for the qPCR: HLA class II histocompatibility antigen, DP a1 chain (HLA-DPB1) forward, 5′-ATGACACTCTTCTGAATTGACTG-3′ and reverse, 5′-GGTAATGATAAAACATGCTCTC-3′; nuclear receptor subfamily 4 group A member 2 (NR4A2) forward, 5′-TCATCTCCTCAGACTGGGGG-3′ and reverse, 5′-TGTACCAAATGCCCCTGTCC-3′; ephrin-B2 (EFNB2) forward, 5′-TATGCAGAACTGCGATTTCCAA-3′ and reverse, 5′-TGGGTATAGTACCAGTCCTTGTC-3′; four and a half LIM domains protein 1 forward (FHL1), 5′-AAATGCACAAAGTGTGCCCG-3′ and reverse, 5′-TCGTTTGGGACACTCAGCAC-3′; insulin-like growth factor-binding protein 7 (IGFBP7) forward, 5′-ACAGTGGTTGATGCCTTAC-3′ and reverse, 5′-CCCTTATGGGTTGCTAACTAC-3′; Rho Family GTPase (RND) forward, 5′-CTATGACCAGGGGGCAAATA-3′ and reverse, 5′-TCTTCGCTTTGTCCTTTCGT-3′; E3 ubiquitin-protein ligase NEURL1B (NEURL1B) forward, 5′-ACAGCAGCTTCCAAGACACA-3′ and reverse, 5′-GTTGGGCAGGCTGTAGTAGG-3′; and GAPDH forward, 5′-ACTCCCATTCTTCCACCTTTG-3′ and reverse, 5′-CCCTGTTGCTGTAGCCATATT-3′. GADPH served as an internal control. The relative expression levels were determined using the 2−ΔΔCq method (13).

Statistical analysis

All statistical analysis was conducted on R (14) or GraphPad Prism 7.0 (GraphPad Software, Inc.). Data were expressed as the mean ± SD. Each experiment was repeated at least three times. Comparisons between groups were analyzed by a Student's t-test. GO and KEGG annotation enrichment analyses were evaluated using a Fisher's exact test. For OS analysis, the samples were divided into high and low expression groups according to the median expression value of key genes. Differences between two groups were compared with Kaplan-Meier curves, followed by log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

Transcript analysis results

In the present study, the gene expression profiles in the compared groups were analyzed via microarray analysis. The differential expression analysis results were shown in Table III according to |FC|>1.5 and P-value <0.05. The DEGs were marked for further analysis (Tables SISVI listed all DEGs in the six compared groups). The scatter plots show the distribution of upregulated genes in the two groups (Fig. 2). The volcano plots were used to show the DEGs between different compared groups. In Fig. 3, the red and green dots represented DEGs with the criteria of | FC |>1.5 and P-value <0.05, and the gray dot indicates genes with no significant difference.

Table III.

Differentially expressed genes in six different comparative groups.

Table III.

Differentially expressed genes in six different comparative groups.

Comparative groupsTotal number of upregulated genesTotal number of downregulated genes
I198  43
II346271
III116  40
IV182502
V830533
VI540632

To effectively distinguish DEGs in the different comparison groups, unsupervised hierarchical clustering analysis was performed. This analysis could distinguish between the different samples in the six comparison groups (Figs. S1S6).

Comparative analysis

In comparison A (Groups I–III; Table II): i) A total of eight commonly upregulated genes [including Uncharacterized LOC101928168 (LOC101928168), 3-Hydroxy-3-Methylglutaryl-CoA Synthase 2 (HMGCS2), Secretagogin, EF-Hand Calcium Binding Protein (SCGN), Chimerin 2 (CHN2), X-Linked Kx Blood Group (XK), Mucin 6, Oligomeric Mucus/Gel-Forming (MUC6), Phospholipid Phosphatase 5 (PLPP5) and Heat Shock Protein Family H Member 1 (HSPH1)] and one downregulated gene [ST8 α-N-Acetyl-Neuraminide α-2,8-Sialyltransferase 4 (ST8SIA4)] were identified in cholesterol polyps, gallbladder adenoma and gallbladder cancer for gallbladder walls (data not shown); ii) A total of 14 common DEGs were found to overlap in the gallbladder wall of gallbladder cancer and gallbladder adenoma (Table IV); iii) A total of 273 differentially upregulated genes were only expressed in the gallbladder wall of gallbladder adenoma, of which the 20 most significantly DEGs, according to the FC, were selected to continue further analysis (Tables IV and V). A total of 85 upregulated genes were identified in the gallbladder wall of gallbladder cancer (Table VI). The 20 most significantly DEGs were selected, as shown in Table VI.

Table IV.

Common DEGs in the gallbladder wall of gallbladder cancer and gallbladder adenoma.

Table IV.

Common DEGs in the gallbladder wall of gallbladder cancer and gallbladder adenoma.

A, Upregulated genes

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
1906EDN1Endothelin 11.5496187690.0293906740.955609729
83661MS4A8Membrane-spanning 4-domains, subfamily A, member 82.0034548540.0169409210.955609729
213ALBAlbumin2.2468855920.0440861260.955609729
10232MSLNMesothelin3.5775534790.0349448820.955609729
1515CTSVCathepsin V1.8103600670.0184382630.955609729
573BAG1BCL2 associated athanogene 11.6199844890.0452945510.955609729
100507412LOC100507412Uncharacterized LOC1005074122.4055609160.005884840.955609729
55283MCOLN3Mucolipin 32.5979854180.0354364530.955609729
7586ZKSCAN1Zinc finger with KRAB and SCAN domains 12.3039882090.0107955010.955609729

B, Downregulated genes

Entrez accession no.Gene symbolGene name Fold-changeP-valueFalse discovery rate

6926TBX3T-box 3−1.973819410.0248869220.955609729
25987TSKUTsukushi, small leucine rich proteoglycan−1.5069344540.0495051430.955609729
6319SCDStearoyl-coa desaturase (delta-9-desaturase)−1.6924110380.0311495260.955609729
4857NOVA1Neuro-oncological ventral antigen 1−1.7541393160.0418346760.955609729
5209PFKFB3 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3−1.8595926990.0154506020.955609729

Table V.

Top 20 upregulated genes in the gallbladder wall of gallbladder adenoma.

Table V.

Top 20 upregulated genes in the gallbladder wall of gallbladder adenoma.

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
3115HLA-DPB1Major histocompatibility complex, class II, DP β114.392945870.0185427170.95561
10321CRISP3Cysteine-rich secretory protein 38.5960467760.0005695540.95561
9153SLC28A2Solute carrier family 28 (concentrative nucleoside transporter), member 27.9728747210.0020385870.95561
1733DIO1Deiodinase, iodothyronine, type I4.134023720.0001449840.95561
2568GABRPγ-aminobutyric acid (GABA) A receptor, pi4.118636110.0325413530.95561
6555SLC10A2Solute carrier family 10 (sodium/bile acid cotransporter), member 23.7297613795.99474E-050.95561
6819SULT1C2Sulfotransferase family 1C member 23.7166426970.0181046730.95561
4496MT1HMetallothionein 1H3.5279578720.0006683360.95561
4494MT1FMetallothionein 1F3.5056614220.0026754510.95561
54346UNC93AUnc-93 homolog A (C. Elegans)3.3551563560.0085163540.95561
148523CIARTCircadian associated repressor of transcription3.1362342780.0264669790.95561
388561ZNF761Zinc finger protein 7613.1111328470.0029498540.95561
100127888SLCO4A1-AS1SLCO4A1 antisense RNA 13.0724225990.0069322070.95561
4495MT1GMetallothionein 1G3.0501214020.000195330.95561
2069EREGEpiregulin3.0315031750.0487168060.95561
7364UGT2B7UDP glucuronosyltransferase 2 family, polypeptide B72.9711950840.0357300270.95561
3821KLRC1Killer cell lectin-like receptor subfamily C, member 12.9028030010.001491720.95561
3822KLRC2Killer cell lectin-like receptor subfamily C, member 22.9028030010.001491720.95561
10562OLFM4Olfactomedin 42.8995251790.034558140.95561
990CDC6Cell division cycle 62.7634745640.0013617240.95561

Table VI.

Top 20 upregulated genes in the gallbladder wall of gallbladder cancer.

Table VI.

Top 20 upregulated genes in the gallbladder wall of gallbladder cancer.

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
54474KRT20Keratin 20, type I6.0275290.0142630.99994
11075STMN2Stathmin 24.5455560.0210020.99994
22943DKK1Dickkopf WNT signaling pathway inhibitor 13.9838410.0347890.99994
3790KCNS3Potassium voltage-gated channel, modifier subfamily S, member 33.1932450.0220640.99994
3606IL18Interleukin 182.7406350.0293880.99994
8174MADCAM1Mucosal vascular addressin cell adhesion molecule 12.6573040.0388340.99994
1305COL13A1Collagen, type XIII, α12.5933260.0162790.99994
7171TPM4Tropomyosin 42.4927880.0120880.99994
84189SLITRK6SLIT and NTRK like family member 62.4199040.0079570.99994
79966SCD5Stearoyl-CoA desaturase 52.3999260.022290.99994
56892C8orf4Chromosome 8 open reading frame 42.3630660.0309480.99994
81671VMP1Vacuole membrane protein 12.3548910.0171920.99994
406991MIR21MicroRNA 212.3548910.0171920.99994
55612FERMT1Fermitin family member 12.2684970.0421150.99994
55816DOK5Docking protein 52.2544990.0093690.99994
24147FJX1Four jointed box 12.2414260.0277880.99994
2043EPHA4EPH receptor A42.1815550.0369220.99994
1908EDN3Endothelin 32.1747760.028920.99994
1316KLF6Kruppel-like factor 62.173530.0226110.99994
440712C1orf186Chromosome 1 open reading frame 1862.1655750.0345760.99994
5318PKP2Plakophilin 22.1347740.0438530.99994
63923TNNTenascin N2.0810140.0408740.99994
78989COLEC11Collectin subfamily member 112.0671310.0005180.99994
100131541LOC100131541Uncharacterized LOC1001315412.0328020.0350340.99994
5727PTCH1Patched 12.011070.0170850.99994
1359CPA3Carboxypeptidase A3 (mast cell)1.9924770.0220020.99994
3775KCNK1Potassium channel, two pore domain subfamily K, member 11.9758910.023280.99994
5732PTGER2Prostaglandin E receptor 21.950770.0064020.99994
51751HIGD1BHIG1 hypoxia inducible domain family member 1B1.92570.0251330.99994
23705CADM1Cell adhesion molecule 11.9078710.0447450.99994

In comparison B (Group IV; Table I), 684 DEGs in the gallbladder wall of gallbladder adenoma, of which 182 were upregulated and 502 were downregulated, shown in Table SIV. The top 20 DEGs are shown in Table VII. In comparison C, it was revealed that 177 DEGs were expressed both in the tumor tissue and gallbladder wall in gallbladder cancer. The 20 most significantly DEGs were selected according to the FC (Table VIII). In comparison D, 459 upregulated genes were found in the tumor of gallbladder cancer. The top 20 upregulated genes that were identified according to FC in Table IX.

Table VII.

Top 20 upregulated genes in the gallbladder wall of gallbladder adenoma compared with the gallbladder wall of cholesterol polyps.

Table VII.

Top 20 upregulated genes in the gallbladder wall of gallbladder adenoma compared with the gallbladder wall of cholesterol polyps.

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
8076MFAP5Microfibrillar associated protein 510.0558280.003131940.930859211
8483CILPCartilage intermediate layer protein5.75568110.011484260.930859211
8839WISP2WNT1 inducible signaling pathway protein 25.01268420.0018366360.930859211
4495MT1GMetallothionein 1G4.9589110.0240604910.930859211
4496MT1HMetallothionein 1H4.71192530.0182606890.930859211
2202EFEMP1EGF containing fibulin-like extracellular matrix protein 13.95361760.0196795550.930859211
7364UGT2B7UDP glucuronosyltransferase 2 family, polypeptide B73.95126150.0401658380.930859211
3489IGFBP6Insulin like growth factor binding protein 63.36726360.0350741660.930859211
10562OLFM4Olfactomedin 43.30457940.0185700130.930859211
4494MT1FMetallothionein 1F3.25101450.012642330.930859211
64167ERAP2Endoplasmic reticulum aminopeptidase 23.17516890.0392604570.930859211
1543CYP1A1Cytochrome P450, family 1, subfamily A, polypeptide 13.03490920.0413125150.930859211
388561ZNF761Zinc finger protein 7612.87430530.005505480.930859211
683BST1Bone marrow stromal cell antigen 12.83270430.007021930.930859211
55057AIM1LAbsent in melanoma 1-like2.82673510.0314810940.930859211
100127888SLCO4A1-AS1SLCO4A1 antisense RNA 12.6894540.010484060.930859211
30835CD209CD209 molecule2.60283410.0481635320.930859211
148523CIARTCircadian associated repressor of transcription2.59222330.0298373550.930859211
10720UGT2B11UDP glucuronosyltransferase 2 family, polypeptide B112.58711950.0057787140.930859211
2199FBLN2Fibulin 22.56600980.0243973180.930859211

Table VIII.

Top 20 upregulated genes both in the tumor and gallbladder wall in gallbladder cancer.

Table VIII.

Top 20 upregulated genes both in the tumor and gallbladder wall in gallbladder cancer.

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
1490CTGFConnective tissue growth factor12.7210.00490.663
3115HLA-DPB1Major histocompatibility complex, class II, DP β18.03890.00660.663
3400ID4Inhibitor of DNA binding 4, dominant negative helix-loop-helix protein6.9194E-060.044
3399ID3Inhibitor of DNA binding 3, dominant negative helix-loop-helix protein5.80990.00010.435
1282COL4A1Collagen, type IV, α15.36010.03980.663
4929NR4A2Nuclear receptor subfamily 4 group A member 25.00110.01130.663
2669GEMGTP binding protein overexpressed in skeletal muscle4.63920.00940.663
1948EFNB2Ephrin-B24.52160.00870.663
3397ID1Inhibitor of DNA binding 1, dominant negative helix-loop-helix protein4.36730.01210.663
2919CXCL1Chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, α)4.21910.04530.663
390RND3Rho family GTPase 34.16880.00170.663
2273FHL1Four and a half LIM domains 14.13660.0110.663
3490IGFBP7Insulin like growth factor binding protein 74.10390.03690.663
54492NEURL1BNeuralized E3 ubiquitin protein ligase 1B3.84870.00830.663
301ANXA1Annexin A13.8110.02420.663
10631POSTNPeriostin, osteoblast specific factor3.73390.02240.663
79772MCTP1Multiple C2 domains, transmembrane 13.67790.02470.663
3486IGFBP3Insulin like growth factor binding protein 33.45810.03040.663
5327PLATPlasminogen activator, tissue3.2880.00230.663
26353HSPB8Heat shock protein family B (small) member 83.20120.04160.663

Table IX.

Top 20 upregulated genes in the tumor of gallbladder cancer.

Table IX.

Top 20 upregulated genes in the tumor of gallbladder cancer.

Entrez accession no.Gene symbolGene nameFold-changeP-valueFalse discovery rate
1048CEACAM5Carcinoembryonic antigen-related cell adhesion molecule 5143.4154524 2.06062×10−50.0894515
10562OLFM4Olfactomedin 490.22299879 3.89271×10−50.0960529
7020TFAP2ATranscription factor AP-2 α (activating enhancer binding protein 2 α)18.24994233 3.9829×10−40.1609015
1015CDH17Cadherin 17, LI cadherin (liver-intestine)16.14818371 1.31492×10−30.1871078
10451VAV3VAV guanine nucleotide exchange factor 316.0872594 7.651281×10−30.2974407
1604CD55CD55 molecule, decay accelerating factor for complement (Cromer blood group)14.25260113 3.93845×10−60.0512905
1373CPS1Carbamoyl-phosphate synthase 114.14519343 2.998467×10−20.3979446
3872KRT17Keratin 17, type I11.21846461 2.5881794×10−20.3854011
5268SERPINB5Serpin peptidase inhibitor, clade B (ovalbumin), member 511.07304422 1.2500212×10−20.3344667
9843HEPHHephaestin11.07030706 8.947495×10−30.3090802
213ALBAlbumin10.912577540.0347300420.408424
3158HMGCS2 3-hydroxy-3-methylglutaryl-CoA synthase 2 (mitochondrial)10.74010853 6.900844×10−30.2884597
63928CHP2Calcineurin-like EF-hand protein 27.987754539 1.5774911×10−20.3509474
7031TFF1Trefoil factor 17.96013740.0178232770.3616271
11074TRIM31Tripartite motif containing 317.589614806 1.280721×10−30.1871078
23213SULF1Sulfatase 17.505284011 2.8219398×10−20.3929094
3216HOXB6Homeobox B67.355635549 1.101194×10−30.1861479
84419C15orf48Chromosome 15 open reading frame 487.266966891 3.1186816×10−20.4004668
2982GUCY1A3Guanylate cyclase 1, soluble, α37.041356237 1.9708261×10−20.3677601
8329HIST1H2AIHistone cluster 1, h2ai6.935644729 7.41761×10−40.1737322
Function enrichment analysis

To better understand the biological pathways that were affected in the gallbladder walls of cholesterol polyps, gallbladder adenoma and gallbladder cancer, GO analysis was conducted on the DEGs. Fig. 4A shows that the GO terms that experienced the most significant enrichment of comparison A was the BP ‘Immune system process’, MF ‘Integrin binding’ and the CC ‘Anchoring junction’. Fig. 4B shows that the DEGs of gallbladder walls in cholesterol polyps vs. gallbladder adenoma participate in a number of GO pathways, the number of DEGs with the highest count had roles in the BP ‘Tissue development’, the CC ‘Cell junction’ and the MF ‘Receptor binding’. Fig. 4C demonstrates that the number of DEGs of the tumor tissues in cholesterol polyps compared with gallbladder adenoma had the highest count in the BP ‘Positive regulation of gene expression’, the CC ‘Cytoskeleton’ and the MF ‘Enzyme binding’.

KEGG pathway enrichment analysis

The biological pathways that were enriched by DEGs were also analyzed with KEGG. Fig. 5A shows that the most significantly enriched pathways in the gallbladder walls of cholesterol polyps, gallbladder adenoma and gallbladder cancer compared with normal groups was ‘cell adhesion molecules (CAMs)’. Fig. 5B shows that the most significantly enriched pathway with DEGs in the gallbladder walls of gallbladder adenoma vs. gallbladder cancer was ‘Cell adhesion molecules (CAMs)’. Fig. 5C indicates that the most significantly enriched pathway in the tumor tissues of gallbladder adenoma vs. gallbladder cancer was the ‘Systemic lupus erythematosus’.

Validation of key genes in gallbladder cancer

Among the top 20 DEGs in the gallbladder wall and tumor of gallbladder cancer, seven novel DEGs, including HLA-DPB1 (Fig. 6A), NR4A2 (Fig. 6B), EFNB2 (Fig. 6C), FHL1 (Fig. 6D), IGFBP7 (Fig. 6E), RND3 (Fig. 6F) and NEURL1B (Fig. 6G) for gallbladder cancer were validated using GEPIA. HLA-DPB1, EFNB2, IGFBP7 and NEURL1B had a significantly higher expression in gallbladder cancer tissues compared with normal tissues. The prognostic value of these DEGs were further analyzed. There was no significant difference between low HLA-DPB1 expression and prognosis of gallbladder cancer (Fig. 7A). The low expression of NR4A2 (Fig. 7B) indicated poorer OS for patients with gallbladder cancer. No significant difference was found between patients with low EFNB2 expression and those with high EFNB2 expression (Fig. 7C). Low FHL1 expression predicted significantly less favorable OS (Fig. 7D). There was no significant difference observed for different expression levels of IGFBP7 (Fig. 7E), RND3 (Fig. 7F) and NEURL1B (Fig. 7G). Therefore, the data presented here indicated that only NR4A2 and FHL1 could represent potential prognostic markers for patients with gallbladder cancer. Following validation using RT-qPCR, HLA-DPB1 (Fig. 8A), NR4A2 (Fig. 8B) and EFNB2 (Fig. 8C) had significantly higher expression levels in gallbladder cancer tissues compared with normal tissues. However, there was no statistical difference in FHL1 expression between gallbladder cancer tissues and normal tissues (Fig. 8D). Moreover, high IGFBP7 expression was determined in gallbladder cancer tissues compared with normal tissues (Fig. 8E). As shown in Fig. 8F, RND3 mRNA expression was significantly decreased in gallbladder cancer tissues compared with normal tissues. A significantly higher expression level of NEURL1B was also detected in gallbladder cancer tissues compared with normal tissues (Fig. 8G).

Discussion

Gallbladder disease is one of the most common causes of upper abdominal pain (15). It is critical to focus on gallbladder diseases due to the potential for malignant degeneration of any gallbladder lesion (15). Gallbladder adenomas and primary adenocarcinomas have been identified as the most common benign and malignant tumors, respectively (16). Nevertheless, efforts have been put into elucidating the pathophysiological mechanisms leading to the development of gallbladder cancer, however, most of these mechanisms remain unknown. Therefore, it is crucial to disclose the molecular mechanisms of gallbladder cancer to promote the development of new cancer biomarkers and appropriate treatment strategies.

Different from other microarray studies, in the present study, the microarrays were comprehensively analyzed (1719). Firstly, eight differentially expressed upregulated genes were found, which included LOC101928168, HMGCS2, SCGN, CHN2, XK, MUC6, PLPP5 and HSPH1 both in cholesterol polyps and gallbladder adenoma from gallbladder walls. Secondly, 14 common DEGs were identified in the gallbladder walls of gallbladder cancer and gallbladder adenoma. It is important to distinguish benign and malignant gallbladder adenoma due to the poor diagnosis of gallbladder cancer. T-Box Transcription Factor 3, Tsukushi, Small Leucine Rich Proteoglycan, Stearoyl-CoA Desaturase (SCD), NOVA Alternative Splicing Regulator 1 and 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 3 were downregulated in the gallbladder wall of gallbladder cancer and gallbladder adenoma; EDN1, MS4A8, ALB, MSLN, CTSV, BAG1, LOC100507412, MCOLN3 and ZKSCAN1 were upregulated in the gallbladder wall of gallbladder cancer and gallbladder adenoma. Thirdly, 273 upregulated genes were expressed in the gallbladder wall of gallbladder adenoma. Fourthly, the 20 most significantly DEGs that were upregulated in the gallbladder wall of gallbladder cancer were identified including KRT20, STMN2, DKK1, KCNS3, IL18, MADCAM1, COL13A1, TPM4, SLITRK6, SCD5, C8orf4, VMP1, MIR21, FERMT1, DOK5, FJX1, EPHA4, EDN3, KLF6 and C1orf186. Among them, DKK1 is known to regulate tumor angiogenesis, which is essential for tumor invasive growth and metastasis (20). IL18 has been reported to be a candidate cytokine that may provide a new insight into the development of next generation cancer immunotherapy (21). Desaturated fatty acids are essential for tumor cell survival, and SCD5 may represent a viable target for the development of novel agents for cancer treatment (22), which could become a candidate for the treatment of gallbladder cancer. KLF6 is a member of the Kruppel-like family of zinc finger transcription factors, which has been identified as a mutated tumor inhibitor in selective human cancer types, but not gallbladder cancer (23).

A total of 182 upregulated DEGs in the gallbladder walls of gallbladder adenoma were obtained and compared with that of cholesterol polyps. The top 20 most significantly expressed genes included MFAP5, CILP, WISP2, MT1G, MT1H, EFEMP1, UGT2B7, IGFBP6, OLFM4, MT1F, ERAP2, CYP1A1, ZNF761, BST1, AIM1L, SLCO4A1-AS1, CD209, CIART, UGT2B11 and FBLN2. The overexpression of CCN5/WISP2 in adipose tissue has previously been secreted and circulated in the blood in a transgenic mouse model, which suggests that WISP2 could become a biomarker in blood for gallbladder adenoma and cholesterol polyps (24). The gene expression of MT1G, MT1H and MT1F in human peripheral blood lymphocytes can be used as potential biomarkers for cadmium exposure (25). Cadmium exposure could contribute to the development of gallbladder cancer (26). EFEMP1 expression accumulates angiogenesis and accelerates the growth of cervical cancer in vivo (27). Patients with UGT2B7*1/*2 genotypes, UGT2B7 genetic variation are at risk for suboptimal immune recovery due to significant long-term autologous induction (28). The expression of IGFBP-6 in vascular endothelial cells is upregulated by hypoxia and IGFBP-6 suppresses angiogenesis in vitro and in vivo (29), but this has not been reported in the context of gallbladder cancer. OLFM4 expression is associated with cancer differentiation, stage, metastasis and prognosis in a variety of cancer types, such as breast cancer, esophageal adenocarcinoma and gastrointestinal cancer, suggesting that it has underlying clinical value as an early cancer biomarker or therapeutic target (30). CYP1A1/1A2 isoenzymes are involved in EROD activity in blood lymphocytes (31); however, there is currently no previous report on the functions of this gene in gallbladder cancer. The production of extracellular cADPR, catalyzed by BST-1, followed by concentrating the uptake of cyclic nucleotides by hemopoietic progenitors, may be physiologically relevant in normal hematopoiesis (32), but its function in gallbladder cancer remains unknown. CD209 has been identified to present on monocyte-derived DCs, a cell adjuvant for cancer immunotherapy (33). FBLN2 is a novel gene associated with hypertension (34).

The top 20 upregulated genes were expressed both in tumors and gallbladder walls of gallbladder cancer, which included CTGF, HLA-DPB1, ID3, ID4, COL4A1, NR4A2, GEM, EFNB2, ID1, CXCL1, RND3, FHL1, IGFBP7, NEURL1B, ANXA1, POSTN, MCTP1, IGFBP3, PLAT and HSPB8. The study focused on whether these genes expression levels could be assessed using blood or bile. CTGF could play an important role in the inflammation of gallbladder cancer (35). Therefore, CTGF has the potential to become a future biomarker for gallbladder cancer, circulating in the blood and bile. It has been revealed that the dynamic changes of growth centers and plasma cell differentiation are determined by ID3 and E protein activity (35). Following validation using RT-qPCR, the genes HLA-DPB1, NR4A2, EFNB2, IGFBP7 and NEURL1B were found to be highly expressed in gallbladder cancer. RND3 was significantly decreased in gallbladder cancer. HLA-DPB1, NR4A2 and FHL1 could be underlying prognostic markers for gallbladder cancer.

In the present study, a transcriptome profile was comprehensively analyzed enabling the identification of DEGs in gallbladder cancer, based on an annotation analysis of microarray studies. The present findings could provide a novel understanding on the tumorigenesis and development of gallbladder cancer.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This work was funded by Natural Science Foundation of Liaoning Province (grant no. 2020-BS-283).

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

CG conceived and designed the study. XZ and XN conducted most of the experiments and data analysis and wrote the manuscript. BZ and LC participated in acquiring data and helped draft the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The First Affiliated Hospital of China Medical University (2018075). All patients who participated in the study signed written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

DEGs

differentially expressed genes

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

aRNA

amplified RNA

FC

fold-change

References

1 

Sicklick JK, Fanta PT, Shimabukuro K and Kurzrock R: Genomics of gallbladder cancer: The case for biomarker-driven clinical trial design. Cancer Metastasis Rev. 35:263–275. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Chandra V, Kim JJ, Mittal B and Rai R: MicroRNA aberrations: An emerging field for gallbladder cancer management. World J Gastroenterol. 22:1787–1799. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Kim YT, Kim J, Jang YH, Lee WJ, Ryu JK, Park YK, Kim SW, Kim WH, Yoon YB and Kim CY: Genetic alterations in gallbladder adenoma, dysplasia and carcinoma. Cancer Lett. 169:59–68. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Itoi T, Watanabe H, Ajioka Y, Oohashi Y, Takel K, Nishikura K, Nakamura Y, Horil A and Saito T: APC, K-ras codon 12 mutations and p53 gene expression in carcinoma and adenoma of the gall-bladder suggest two genetic pathways in gallbladder carcinogenesis. Pathol Int. 46:333–340. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Lee SH, Lee DS, You IY, Jeon WJ, Park SM, Youn SJ, Choi JW and Sung R: Histopathologic analysis of adenoma and adenoma-related lesions of the gallbladder. Korean J Gastroenterol. 55:119–126. 2010.(In Korean). View Article : Google Scholar : PubMed/NCBI

6 

Liu MC, Jiang L, Hong HJ, Meng ZW, Du Q, Zhou LY, She FF and Chen YL: Serum vascular endothelial growth factors C and D as forecast tools for patients with gallbladder carcinoma. Tumour Biol. 36:6305–6312. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Choi JH, Yun JW, Kim YS, Lee EA, Hwang ST, Cho YK, Kim HJ, Park JH, Park DI, Sohn CI, et al: Pre-operative predictive factors for gallbladder cholesterol polyps using conventional diagnostic imaging. World J Gastroenterol. 14:6831–6834. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Zhao MF, Huang P, Ge CL, Sun T, Ma ZG and Ye FF: Conjugated bile acids in gallbladder bile and serum as potential biomarkers for cholesterol polyps and adenomatous polyps. Int J Biol Markers. 31:e73–e79. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

10 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Kanehisa M, Goto S, Sato Y, Furumichi M and Tanabe M: KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40((Database Issue)): D109–D114. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45W:W98–W102. 2017. View Article : Google Scholar

13 

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

14 

R Core Team: R: A language and environment for statistical computing. R Foundation for Statistical Computing; Vienna, Austria: 2013, simplehttp://www.R-project.org/

15 

Rodriguez S, Gaunt TR, Guo Y, Zheng J, Barnes MR, Tang W, Danish F, Johnson A, Castillo BA, Li YR, et al: Lipids, obesity and gallbladder disease in women: Insights from genetic studies using the cardiovascular gene-centric 50K SNP array. Eur J Hum Genet. 24:106–112. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Mellnick VM, Menias CO, Sandrasegaran K, Hara AK, Kielar AZ, Brunt EM, Doyle MB, Dahiya N and Elsayes KM: Polypoid lesions of the gallbladder: Disease spectrum with pathologic correlation. Radiographics. 5:387–399. 2015. View Article : Google Scholar

17 

Wang F, Wang R, Li Q, Qu X, Hao Y, Yang J, Zhao H, Wang Q, Li G, Zhang F, et al: A transcriptome profile in hepatocellular carcinomas based on integrated analysis of microarray studies. Diagn Pathol. 12:42017. View Article : Google Scholar : PubMed/NCBI

18 

Meng LZ, Fang JG, Sun JW, Yang F and Wei YX: Aberrant expression profile of long noncoding RNA in human sinonasal squamous cell carcinoma by microarray analysis. Biomed Res Int. 2016:10957102016. View Article : Google Scholar : PubMed/NCBI

19 

Liu L, Liu Y, Liu C, Zhang Z, Du Y and Zhao H: Analysis of gene expression profile identifies potential biomarkers for atherosclerosis. Mol Med Rep. 14:3052–3058. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Park H, Jung HY, Choi HJ, Kim DY, Yoo JY, Yun CO, Min JK, Kim YM and Kwon YG: Distinct roles of DKK1 and DKK2 in tumor angiogenesis. Angiogenesis. 17:221–234. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Ma Z, Li W, Yoshiya S, Xu Y, Hata M, El-Darawish Y, Markova T, Yamanishi K, Yamanishi H, Tahara H, et al: Augmentation of immune checkpoint cancer immunotherapy with IL18. Clin Cancer Res. 22:2969–2980. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Roongta UV, Pabalan JG, Wang X, Ryseck RP, Fargnoli J, Henley BJ, Yang WP, Zhu J, Madireddi MT, Lawrence RM, et al: Cancer cell dependence on unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy. Mol Cancer Res. 9:1551–1561. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Yin D, Komatsu N, Miller CW, Chumakov AM, Marschesky A, McKenna R, Black KL and Koeffler HP: KLF6: mutational analysis and effect on cancer cell proliferation. Int J Oncol. 30:65–72. 2007.PubMed/NCBI

24 

Grünberg JR, Elvin J, Paul A, Hedjazifar S, Hammarstedt A and Smith U: CCN5/WISP2 and metabolic diseases. J Cell Commun Signal. 12:309–318. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Chang XL, Jin TY and Zhou YF: Metallothionein 1 isoform gene expression induced by cadmium in human peripheral blood lymphocytes. Biomed Environ Sci. 19:104–109. 2006.PubMed/NCBI

26 

Lee MH, Gao YT, Huang YH, McGee EE, Lam T, Wang B, Shen MC, Rashid A, Pfeiffer RM, Hsing AW and Koshiol J: A metallomic approach to assess associations of serum metal levels with gallstones and gallbladder cancer. Hepatology. 71:917–928. 2020. View Article : Google Scholar : PubMed/NCBI

27 

En-lin S, Sheng-guo C and Hua-qiao W: The expression of EFEMP1 in cervical carcinoma and its relationship with prognosis. Gynecol Oncol. 117:417–422. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Habtewold A, Amogne W, Makonnen E, Yimer G, Riedel KD, Ueda N, Worku A, Haefeli WE, Lindquist L, Aderaye G, et al: Long-term effect of efavirenz autoinduction on plasma/peripheral blood mononuclear cell drug exposure and CD4 count is influenced by UGT2B7 and CYP2B6 genotypes among HIV patients. J Antimicrob Chemother. 66:2350–2361. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Zhang C, Lu L, Li Y, Wang X, Zhou J, Liu Y, Fu P, Gallicchio MA, Bach LA and Duan C: IGF binding protein-6 expression in vascular endothelial cells is induced by hypoxia and plays a negative role in tumor angiogenesis. Int J Cancer. 130:2003–2012. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Liu W and Rodgers GP: Olfactomedin 4 expression and functions in innate immunity, inflammation, and cancer. Cancer Metastasis Rev. 35:201–212. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Dey A, Parmar D, Dayal M, Dhawan A and Seth PK: Cytochrome P450 1A1 (CYP1A1) in blood lymphocytes evidence for catalytic activity and mRNA expression. Life Sci. 69:383–393. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Podestà M, Benvenuto F, Pitto A, Figari O, Bacigalupo A, Bruzzone S, Guida L, Franco L, Paleari L, Bodrato N, et al: Concentrative uptake of cyclic ADP-ribose generated by BST-1+ stroma stimulates proliferation of human hematopoietic progenitors. J Biol Chem. 280:5343–5349. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Osugi Y, Vuckovic S and Hart DN: Myeloid blood CD11c(+) dendritic cells and monocyte-derived dendritic cells differ in their ability to stimulate T lymphocytes. Blood. 100:2858–2866. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Vallvé JC, Serra N, Zalba G, Fortuño A, Beloqui O, Ferre R, Ribalta J and Masana L: Two variants in the fibulin2 gene are associated with lower systolic blood pressure and decreased risk of hypertension. PLoS One. 7:e430512012. View Article : Google Scholar : PubMed/NCBI

35 

Gloury R, Zotos D, Zuidscherwoude M, Masson F, Liao Y, Hasbold J, Corcoran LM, Hodgkin PD, Belz GT, Shi W, et al: Dynamic changes in Id3 and E-protein activity orchestrate germinal center and plasma cell development. J Exp Med. 213:1095–1111. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 23 Issue 1

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
Ge C, Zhu X, Niu X, Zhang B and Chen L: A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies. Mol Med Rep 23: 25, 2021
APA
Ge, C., Zhu, X., Niu, X., Zhang, B., & Chen, L. (2021). A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies. Molecular Medicine Reports, 23, 25. https://doi.org/10.3892/mmr.2020.11663
MLA
Ge, C., Zhu, X., Niu, X., Zhang, B., Chen, L."A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies". Molecular Medicine Reports 23.1 (2021): 25.
Chicago
Ge, C., Zhu, X., Niu, X., Zhang, B., Chen, L."A transcriptome profile in gallbladder cancer based on annotation analysis of microarray studies". Molecular Medicine Reports 23, no. 1 (2021): 25. https://doi.org/10.3892/mmr.2020.11663