Open Access

Expression levels and prognostic values of annexins in liver cancer

  • Authors:
    • Chunbo Zhuang
    • Pei Wang
    • Ting Sun
    • Lei Zheng
    • Liang Ming
  • View Affiliations

  • Published online on: October 30, 2019     https://doi.org/10.3892/ol.2019.11025
  • Pages: 6657-6669
  • Copyright: © Zhuang 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

Annexins are a superfamily of calcium‑dependent phospholipid‑binding proteins that are implicated in a wide range of biological processes. The annexin superfamily comprises 13 members in humans (ANXAs), the majority of which are frequently dysregulated in cancer. However, the expression patterns and prognostic values of ANXAs in liver cancer are currently largely unknown. The present study aimed to analyze the expression levels of ANXAs and survival data in patients with liver cancer from the Oncomine, GEPIA, Kaplan‑Meier plotter and cBioPortal for Cancer Genomics databases. The results demonstrated that ANXA1, A2, A3, A4 and A5 were upregulated, whereas ANXA10 was downregulated in liver cancer compared with normal liver tissues. The expression of ANXA10 was associated with pathological stage. High expression levels of ANXA2 and A5 were significantly associated with poor overall survival (OS) rate whereas ANXA7 and A10 were associated with increased OS. The prognostic values of ANXAs in liver cancer were determined based on sex and clinical stage, which revealed that ANXA2, A5, A7 and A10 were associated with OS in male, but not in female patients. In addition, the potential biological functions of ANXAs were identified by Gene Ontology functional annotation and Kyoto Encyclopedia of Genes Genomes pathway analysis; the results demonstrated that ANXAs may serve a role in liver cancer through the neuroactive ligand‑receptor interaction pathway. In conclusion, the results of the present study suggested that ANXA1, A2, A3, A4, A5 and A10 may be potential therapeutic targets for liver cancer treatment, and that ANXA2, A5, A7 and A10 may be potential prognostic biomarkers of liver cancer.

Introduction

Liver cancer is the second leading cause of cancer-associated mortality worldwide, resulting in >500,000 deaths per year (1). Despite recent advances in surgical resection and liver transplantation, the 5-year survival rate of patients with liver cancer remains <17% (2). In the majority of cases, liver cancer is diagnosed at an advanced stage with limited therapeutic options. Only 10–20% of tumors are considered surgically resectable at the time of diagnosis (3), and the long-term survival of the patients remains unsatisfactory due to postsurgical recurrence. Thus, an improved understanding of the molecular mechanisms, as well as identification of prognostic biomarkers and potential therapeutic targets of liver cancer are desirable.

Annexins (ANX) are a superfamily of calcium-dependent phospholipid-binding proteins with a structural homology of 40–60% (4). In humans, annexins comprise 13 members (ANXA1-A11, A13 and A8L1; A12 is unassigned) (4), the nomenclature of which is summarized in Table I. Each ANXA is composed of two major domains; the amino-terminus allows interactions with cytoplasmic proteins, whereas the carboxyl-terminus contains the calcium- and membrane-binding sites (4). ANXAs are involved in a wide range of biological processes such as vesicle trafficking (5), anti-inflammation (6), anti-coagulation (7), calcium signaling (8), cell differentiation, apoptosis and proliferation (4). A number of studies have reported aberrant expression levels of ANXAs in various types of cancer, and that ANXAs may function as tumor suppressors or promoters depending on the cancer type. For example, ANXA1 serves a tumor-promoting role in colorectal and lung cancer (9,10), but functions as a tumor suppressor in esophageal and gastric cancer (11). The role of ANXA1 has also been investigated in hepatocellular carcinoma (HCC), but the results are controversial (12). In addition, other ANXAs, including ANXA2, A3, A4, A6, A7 and A10, have been reported to be dysregulated in liver cancer (1318). However, the majority of these studies only reported the expression levels of ANXAs and lacked any prognosis data. The expression patterns, prognostic roles and functions of ANXAs in liver cancer remain unclear.

Table I.

Nomenclature and characterization of the ANXAs.

Table I.

Nomenclature and characterization of the ANXAs.

ANXASymbolSynonymsChromosomal locationMass, Da (length, a.a.)
Annexin A1ANXA1ANX1, LPC19q21.1338,714 (346)
Annexin A2ANXA2ANX2, ANX2L4, CAL1H, LPC2, LIP215q22.238,694 (339)
Annexin A3ANXA3ANX34q21.2136,375 (323)
Annexin A4ANXA4ANX42p13.335,883 (319)
Annexin A5ANXA5ANX5, ENX24q2735,937 (320)
Annexin A6ANXA6ANX65q33.175,873 (673)
Annexin A7ANXA7ANX710q22.252,739 (488)
Annexin A8ANXA8ANX810q11.2230,693 (276)
Annexin A8 Like 1ANXA8L1ANXA8L210q11.2236,879 (327)
Annexin A9ANXA9ANX311q21.338,364 (345)
Annexin A10ANXA10ANX144q32.337,278 (324)
Annexin A11ANXA11ANX1110q22.354,390 (505)
Annexin A13ANXA13ANX138q24.1335,463 (316)

In the present study, the distinct expression patterns, prognostic values and potential functions of ANXAs in liver cancer were investigated by analyzing the gene expression, copy number variation and survival data published online.

Materials and methods

Ethics statement

The present study was approved by the Academic Committee of Zhengzhou University (Zhengzhou, Henan, China) and performed in accordance with the principles of the Declaration of Helsinki. Online databases were used to retrieve the datasets.

ANXA mRNA expression level evaluation

The Oncomine database (https://www.oncomine.org) was used to evaluate the mRNA expression levels of ANXAs in different types of cancer. The mRNA levels of ANXAs in cancer tissues and normal controls were compared using a Student's t-test, with P≤0.01 and fold change (FC)>2.

GEPIA (http://gepia.cancer-pku.cn) is an online database for analyzing tumor and normal sample RNA sequencing data from The Cancer Genome Atlas (TCGA; http://portal.gdc.cancer.gov/) and the Genotype-Tissue Expression (GTEx) projects (http://commonfund.nih.gov/GTEx/). In the present study, the GEPIA database was used to analyze the mRNA expression levels of ANXAs in liver cancer and normal liver samples. Each ANXA was entered into the database separately and analyzed with the settings |log2(FC)|≥1 and P≤0.01. The method for differential gene expression analysis is one-way ANOVA.

Kaplan-Meier survival analysis

The Kaplan-Meier (KM) plotter (http://kmplot.com) database was used to analyze the associations between ANXA mRNA expression levels and OS of patients with liver cancer and the log-rank test was used to obtain the P-values. Briefly, ANXAs were entered into the database and analyzed using different settings of clinical parameters [e.g. sex and pathological stage (19)]. The cases were divided into high or low expression groups based on the median expression level of each gene. Differences in OS were tested by Cox proportional hazards regression. KM survival plots were obtained with the number-at-risk, hazard ratio (HR), 95% confidence intervals (CI) and P-value displayed on the webpage. P<0.05 was considered to indicate a statistically significant difference.

cBioPortal for cancer genomics

The cBioPortal for Cancer Genomics (www.cbioportal.org) provides visualization, analysis and downloads of large-scale cancer genomics datasets (20). The liver HCC (TCGA, provisional) dataset (21), which contained 442 patients with HCC, was selected for analyses of ANXAs. The alterations of ANXAs, as well as the network between ANXAs and the 50 most frequently altered neighboring genes were obtained according to the instructions on the cBioPortal.

Gene function and pathway enrichment analysis

Gene Ontology (GO; http://geneontology.org/) analysis was used to predict the enriched biological functions of ANXAs and genes associated with ANXA alterations (neighboring genes). A Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.kegg.jp/) pathway analysis was used to determine the enriched biological pathways of ANXAs and their neighboring genes. The Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool (https://david.ncifcrf.gov/) was used to perform the GO functional annotation and KEGG pathway enrichment analysis.

Results

mRNA expression levels of ANXAs in patients with liver cancer

The mRNA expression levels of 13 ANXAs in liver cancer were compared with those in normal samples using the Oncomine database. As presented in (Fig. 1), ANXA1, A2, A3 and A4 were upregulated, whereas ANXA10 was downregulated in liver cancer compared with normal tissues. Among these, ANXA2 was upregulated in four datasets [Mas (22), Roessler2 (23), Roessler (23) and Wurmbach (24)] with fold-changes of 2.409, 3.506, 3.528 and 2.350, respectively (Table II). In addition, the Mas dataset exhibited higher mRNA levels of ANXA1, A3 and A4 in HCC compared with those in normal liver, with fold-changes of 5.649, 5.253 and 2.270, respectively. ANXA10 was the only downregulated ANXA gene in liver cancer, with fold-changes of −6.349, −5.586 and −10.711 in the Roessler, Roessler2 and Wurmbach datasets, respectively. However, for the transcription levels of ANXA5, A6, A7, A8, A8L1, A9, A11 and A13, no significant difference was identified between tumors and normal liver samples (data not shown).

Table II.

Significant changes of ANXA expression at the transcriptional level between liver cancer and normal liver tissues using the Oncomine database.

Table II.

Significant changes of ANXA expression at the transcriptional level between liver cancer and normal liver tissues using the Oncomine database.

ANXAComparisonFold changeP-valuet-testDatabase(Refs.)
ANXA1HCC vs. Normal5.649 1.60×10−139.880Mas(22)
ANXA2HCC vs. Normal3.506 1.10×10−6520.365Roessler2(23)
HCC vs. Normal2.409 1.28×10−108.629Mas(22)
HCC vs. Normal3.528 2.69×10−76.044Roessler(23)
HCC vs. Normal2.350 2.78×10−44.128Wurmbach(24)
ANXA3HCC vs. Normal5.253 2.18×10−2817.617Mas(22)
ANXA4HCC vs. Normal2.270 1.03×10−118.673Mas(22)
ANXA10HCC vs. Normal−10.711 1.36×10−10−8.183Wurmbach(24)
HCC vs. Normal−5.586 2.50×10−69−22.733Roessler2(23)
HCC vs. Normal−6.439 8.55×10−8−7.275Roessler(23)

[i] HCC, hepatocellular carcinoma.

ANXA mRNA levels are associated with clinicopathological parameters of patients with liver cancer

The mRNA expression levels of 13 ANXAs in liver cancer were accessed from the GEPIA database. As demonstrated in (Fig. 2), four dysregulated members were identified. The expression levels of ANXA2, A5 and A11 were significantly higher (P<0.01), whereas ANXA10 was significantly lower in liver cancer compared with those in normal liver tissues (P<0.01). In addition, the ANXA mRNA levels were compared between groups of patients with different pathological stages [classified according to American Joint Committee on Cancer TNM (19)] of liver cancer. The results revealed that the expression levels of ANXA3, A8, A8L1 and A10 were significantly different between patients with different pathological stages (Fig. 3). However, the results of ANXA3, A8 and 8L1 levels need to be interpreted with caution due to their very low expression levels.

Prognostic values of ANXAs in patients with liver cancer

The prognostic values of 13 ANXAs in liver cancer were determined using the KM plotter database. The results demonstrated that four members were significantly associated with prognosis. As presented in (Fig. 4), the survival curves revealed that high mRNA expression levels of ANXA2 and A5 were significantly associated with poor prognosis (HR, 1.45; 95% CI, 1.02–2.05; P=0.035 and HR, 1.67; 95% CI, 1.18–2.37; P=0.0035, respectively), whereas high mRNA expression levels of ANXA7 and A10 were significantly associated with improved prognosis compared with the respective low expression groups (HR, 0.56; 95% CI, 0.39–0.80; P=0.0012 and HR, 0.55;95% CI,0.39–0.78; P=0.00078, respectively), although the high expression of ANXA10 resulted in decreased survival time. The other ANXAs, including ANXA1 (HR, 0.84; 95% CI, 0.59–1.18; P=0.31), A3 (HR, 0.84; 95% CI, 0.59–1.19; P=0.33), A4 (HR, 0.89; 95% CI, 0.63–1.26; P=0.53), A6 (HR, 0.82; 95% CI, 0.58–1.16; P=0.26), A8 (HR, 1.30; 95% CI, 0.92–1.84; P=0.14), A8L1 (HR, 1.16; 95% CI, 0.82–1.64; P=0.39), A9 (HR, 1.29; 95% CI, 0.91–1.83; P=0.14), A11 (HR, 0.91; 95% CI, 0.64–1.28; P=0.59) and A13 (HR, 1.15; 95% CI, 0.81–1.62; P=0.43) were not associated with OS.

Prognostic values of ANXAs in patients with liver cancer according to clinicopathological features

The association between individual ANXAs and other clinicopathological features such as sex and clinical stages were further analyzed. As presented in Table III, in male patients with liver cancer, high mRNA expression levels of ANXA2 and A5 were significantly associated with poor OS (HR, 1.95; 95% CI, 1.23–3.18; P=0.036 and HR, 1.93;95% CI, 1.22–3.05; P=0.004, respectively), whereas high mRNA expression levels of ANXA7 and A10 were significantly associated with improved OS compared with the low expression group (HR, 0.53; 95% CI, 0.33–0.83; P=0.0051 and HR, 0.49; 95% CI, 0.31–0.79; P=0.0023, respectively), which was similar to that in all patients with liver cancer. None of the ANXAs were identified as associated with OS in female patients. High expression of ANXA5 was significantly associated with poor OS in patients with stage I (HR, 2.21; 95% CI, 1.17–4.18; P=0.012) and stage III (HR, 2.06; 95% CI, 1.10–3.84; P=0.021) liver cancer; high expression of ANXA7 (HR, 0.41; 95% CI, 0.22–0.78; P=0.0048) and A10 (HR, 0.37;95% CI, 0.20–0.70; P=0.0017) were significantly associated with improved OS in patients with stage III liver cancer (Table IV). The expression levels of other ANXAs were not significantly associated with OS in patients at different clinical stages, although the expression of ANXA2 (HR, 1.71; 95% CI, 0.92–3.18; P=0.088) was modestly associated with poor OS in patients with stage I liver cancer.

Table III.

Association between ANXAs and the sex of patients with liver cancer.

Table III.

Association between ANXAs and the sex of patients with liver cancer.

ANXAsSexCases, nHR (95% CI)P-value
ANXA1Male2460.72 (0.46–1.12)0.1390
Female1180.97 (0.56–1.68)0.8994
ANXA2Male2461.95 (1.23–3.18)0.0036
Female1181.28 (0.74–2.24)0.3754
ANXA3Male2460.81 (0.52–1.26)0.3489
Female1180.80 (0.45–1.41)0.4426
ANXA4Male2461.02 (0.66–1.59)0.9253
Female1180.93 (0.53–1.63)0.7996
ANXA5Male2461.93 (1.22–3.05)0.0040
Female1181.42 (0.81–2.47)0.2176
ANXA6Male2460.72 (0.46–1.12)0.1458
Female1180.88 (0.50–1.53)0.6474
ANXA7Male2460.53 (0.33–0.83)0.0051
Female1180.97 (0.55–1.69)0.9079
ANXA8Male2461.28 (0.82–1.99)0.2792
Female1181.33 (0.76–2.33)0.3218
ANXA8L1Male2461.30 (0.83–2.02)0.2463
Female118 0.96(0.55–1.67)0.8717
ANXA9Male2461.44 (0.92–2.26)0.1061
Female1181.15 (0.66–2.01)0.6123
ANXA10Male2460.49 (0.31–0.79)0.0023
Female1180.61 (0.35–1.07)0.0817
ANXA11Male2460.84 (0.54–1.31)0.4366
Female1180.88 (0.51–1.53)0.6490
ANXA13Male2461.50 (0.96–2.35)0.0748
Female1180.83 (0.48–1.45)0.5123

[i] P<0.05 was considered to indicate a statistically significant difference. HR, hazard ratio; CI, confidence interval.

Table IV.

Association between ANXAs and the clinical stage of patients with liver cancer.

Table IV.

Association between ANXAs and the clinical stage of patients with liver cancer.

ANXAsClinical stageaCases, nHR (95% CI)P-value
ANXA1I1700.82 (0.45–1.50)0.5175
II830.87 (0.4–1.90)0.7345
III830.74 (0.41–1.33)0.3048
ANXA2I1701.71 (0.92–3.18)0.0884
II830.99 (0.45–2.14)0.9710
III831.46 (0.81–2.64)0.2051
ANXA3I1700.93 (0.50–1.7)0.8034
II830.70 (0.32–1.56)0.3842
III830.6 (0.33–1.11)0.1002
ANXA4I1700.84 (0.46–1.54)0.5760
II830.74 (0.34–1.6)0.4393
III831.43 (0.78–2.62)0.2393
ANXA5I1702.21 (1.17–4.18)0.0122
II830.81 (0.37–1.76)0.5909
III832.06 (1.1–3.84)0.0209
ANXA6I1701.04 (0.57–1.91)0.8932
II830.94 (0.43–2.06)0.8786
III830.58 (0.31–1.07)0.0761
ANXA7I1700.60 (0.32–1.11)0.0988
II830.70 (0.31–1.56)0.3820
III830.41 (0.22–0.78)0.0048
ANXA8I1700.76 (0.41–1.41)0.3879
II831.87 (0.84–4.17)0.1216
III831.10 (0.61–2.01)0.7468
ANXA8L1I1700.98 (0.54–1.80)0.9551
II831.65 (0.74–3.68)0.2183
III830.65 (0.35–1.19)0.1583
ANXA9I1701.29 (0.70–2.39)0.4065
II831.78 (0.80–3.96)0.1532
III831.47 (0.81–2.68)0.2039
ANXA10I1700.60 (0.32–1.12)0.1019
II830.76 (0.35–1.65)0.4800
III830.37 (0.20–0.70)0.0017
ANXA11I1700.97 (0.53–1.77)0.9131
II830.48 (0.21–1.08)0.0689
III831.01 (0.56–1.81)0.9758
ANXA13I1701.17 (0.63–2.14)0.6189
II831.30 (0.59–2.84)0.5122
III831.59 (0.87–2.89)0.1273

a The data of stage IV was not shown as the sample number was too low for meaningful analysis. HR, hazard ratio; CI, confidence interval.

Predicted functions and pathways of ANXAs in liver cancer

Using the cBioPortal online tool, alterations (copy number variation, mutations and mRNA expression change of ANXAs) were identified in 205 out of 442 patients with liver cancer (46.4%) in the selected dataset (Fig. 5A). An interaction network for ANXAs and the 50 most frequently altered neighboring genes was constructed. The results demonstrated that signal transduction-associated genes, such as EGFR, PRKCA, HCRT, GNAI1 and GNBs, were associated with ANXA alterations (Fig. 5B).

In order to investigate the potential biological functions of ANXAs in liver cancer, GO functional annotation and KEGG pathway enrichment analyses were performed for ANXAs, and the 50 most frequently altered neighboring genes using the DAVID. The results of the GO analysis revealed that ANXAs and their neighboring genes were involved in several biological processes, such as ‘neuropeptide signaling pathway’, ‘phospholipase C-activating G-protein coupled receptor signaling pathway’, ‘positive regulation of cytosolic calcium ion concentration’, ‘G-protein coupled receptor signaling pathway’ and ‘signal transduction’ (Fig. 6A). The cellular component analysis revealed that ANXAs and their neighboring genes were primarily located in ‘plasma membrane’, ‘axon terminus’, ‘dendrite’ and ‘extracellular exosome’ (Fig. 6B). The molecular function analysis suggested that ANXAs and the neighboring genes were primarily enriched in ‘calcium-dependent phospholipid binding’, ‘calcium-dependent protein binding’, ‘calcium ion binding’, ‘G-protein coupled receptor binding’ and ‘neuropeptide receptor activity’ (Fig. 6C). In addition, the results of the KEGG pathway analysis demonstrated that ANXAs and the neighboring genes were primarily enriched in ‘neuroactive ligand-receptor interaction’, ‘GABAergic synapse’, ‘chemokine signaling pathway’, ‘calcium signaling pathway’ and ‘pathways in cancer’ (Fig. 6D).

Discussion

Aberrant expression of ANXAs is common in various types of cancer such as melanoma, pancreatic cancer, gastric cancer, lung cancer and breast cancer (8). ANXAs have been demonstrated to be involved in carcinogenesis and progression of liver cancer according to previous studies (1218). However, the complex roles of ANXAs in the carcinogenesis, progression and prognosis of liver cancer remain to be elucidated. In the present study, the mRNA expression and prognostic values of different ANXAs in liver cancer were investigated by bioinformatics analysis.

ANXA1 is the first known member of ANXAs, which has been reported to be involved in a wide range of cell signaling pathways including inflammatory, cell differentiation, proliferation and apoptosis (2527). ANXA1 enhances growth and migration in breast cancer by mediating alternative macrophage polarization in the tumor microenvironment (28). In addition, ANXA1 regulates TGF-β signaling and promotes metastasis of basal-like breast cancer cells (29). In liver cancer, the expression pattern of ANXA1 in previous studies is controversial. Suo et al (30) reported that ANXA1 was upregulated in liver cancer compared with nontumor tissues, and that high levels of ANXA1 expression were significantly associated with tumor grade. Lin et al (12) reported that high ANXA1 expression predicted poor prognosis and enhanced the malignant phenotype of tumor cells in liver cancer. However, Hongsrichan et al (31) identified no ANXA1 protein expression in liver cancer by immunohistochemistry, and Xue et al (32) reported decreased ANXA1 expression levels in liver cancer using tissue microarray analysis. In the present study, the expression of ANXA1 was increased in liver cancer compared with normal tissues, which was consistent with Suo's and Lin's studies. However, the present study did not observe a significant association between ANXA1 expression and OS of patients with liver cancer. The prognostic value of ANXA1 in liver cancer requires further investigation.

ANXA2 is one of the most abundant ANXAs, and is widely distributed in the nucleus, cytoplasm, endosomes and extracellular space. The potential role of ANXA2 in cancer has been widely investigated. Previous studies have reported that ANXA2 is upregulated in various types of cancer including lung (33), breast (34), gastric (35), pancreatic (36), colorectal (37) and liver (38) cancers. Increased expression of ANXA2 is associated with cancer development and poor prognosis (39,40). In addition, knockdown of ANXA2 effectively suppresses tumor progression in vitro and in vivo; studies that focused on the underlying molecular mechanisms have demonstrated that ANXA2 either promotes tumor cell invasion by forming a heterotetramer with S100A10 (41), contributing to heterotypic cell-cell interactions between tumor cells and microvascular endothelial cells, or facilitates tumor cell proliferation and chemoresistance by inhibiting p53 expression and activating the transcription factors STAT3 and NFκB (37,42,43). In liver cancer, protein and mRNA expression levels of ANXA2 were identified as upregulated and associated with poor prognosis (38,44). Knockdown of ANXA2 suppressed liver cancer cell migration and invasion by regulating the trafficking of CD147-harboring membrane microvesicles (45). In addition, the expression of ANXA2 was also elevated in the serum of patients with liver cancer compared with healthy controls (46). The results of the present study confirmed that ANXA2 was upregulated in liver cancer and that high expression of ANXA2 was significantly associated with poor OS.

ANXA3 has been demonstrated to function either as a tumor suppressor or promoter candidate in different types of cancer. Upregulation of ANXA3 enhances drug resistance and promotes tumor metastasis in breast cancer (47) and promotes tumor growth and predicts poor prognosis in gastric cancer (48), whereas in prostate cancer, ANXA3 protein expression is downregulated, which is associated with the tumor stage and Gleason score (49). In liver cancer, ANXA3 is upregulated and preferentially expressed in cancer stem cells. High levels of ANXA3 maintains cancer cell stemness via hypoxia-inducible factor α/Notch and JNK signaling pathways (50,51). ANXA3 promotes tumorigenesis and drug resistance in liver cancer, which makes it a potential therapeutic target (18). In addition, serum ANXA3 is also increased in liver cancer compared with normal liver tissues; therefore, ANXA3 is a promising biomarker for the diagnosis, prognosis and therapeutic response evaluation of liver cancer (52). In the present study, ANXA3 was upregulated in liver cancer compared with healthy tissues, but the mRNA level of ANXA3 was not associated with OS.

ANXA4 is primarily expressed in epithelial cells (53). Recent studies have demonstrated that ANXA4 is upregulated and acts as an oncogene in multiple types of cancer, including lung (54), colorectal (55), cervical (56) and gallbladder cancer (57). In liver cancer, ANXA4 has been reported as upregulated, particularly in patients with early recurrence or metastasis (58). High expression levels of ANXA4 predicted early recurrence or metastasis and poor OS of patients with liver cancer (58). In addition, inhibition of ANXA4 suppresses liver cancer cell proliferation, migration and invasion both in vivo and in vitro (15,58). Consistent with these results, the results of the present study demonstrated that the expression of ANXA4 was higher in liver cancer compared with normal tissues, but it was not associated with OS.

ANXA5 exhibits tumor promoter activity in the majority of different types of tumor, including liver cancer (59). Guo et al (60) analyzed protein expression in five pairs of matched primary tumor and tumor thrombus samples using two-dimensional gel electrophoresis, which revealed that ANXA5 was upregulated in tumor thrombus samples. Sun et al (61) reported that the expression of ANXA5 was positively associated with the progression and metastasis of liver cancer, and that ANXA5 promoted carcinogenesis via the integrin and mitogen-activated protein kinase kinase-extracellular signal-regulated kinase pathways. Consistent with these studies, the results of the present study demonstrated that ANXA5 expression was increased in liver cancer compared with normal liver tissues. Furthermore, to the best of our knowledge, the prognostic role of ANXA5 in liver cancer has not yet been reported. In the present study, it was revealed that high expression of ANXA5 was significantly associated with poor OS.

There are limited studies available that focus on ANXA6 in liver cancer. The protein level of ANXA6 is decreased, whereas the mRNA level is increased in liver cancer compared with non-tumorous tissues, suggesting post-transcriptional regulation of ANXA6 (16). To the best of our knowledge, there are currently no reports stating the prognostic value of ANXA6 in liver cancer. In the present study, no significant differences were observed in the mRNA level of ANXA6 between liver cancer and normal tissues, and ANXA6 expression was not associated with OS. In addition, the roles of ANXA8, A8L1, A9, A11 and A13 in liver cancer have rarely been reported. The present study demonstrated that these ANXAs were not associated with OS in patients with liver cancer.

ANXA7 has been reported to be upregulated in liver cancer and to promote tumor cell migration and invasion by interacting with galectin-3 or receptor of activated protein C kinase 1 (6264), whereas the inhibition of ANXA7 decreases tumor cell invasion and migration (65). Consistent with these studies, the present study revealed modestly increased expression levels of ANXA7 in liver cancer compared with normal liver tissues. In contrast, high expression levels of ANXA7 were associated with improved OS. This contradictory result was also reported by Wang et al (66), who demonstrated that upregulation of ANXA7 increased liver cancer cell migration in vitro, but decreased lymph node metastasis in vivo. In addition, the aforementioned study reported a dynamic change of ANXA7 expression during liver cancer progression (66). Therefore, the exact role of ANXA7 in liver cancer remains unclear and requires further investigation.

ANXA10 is the latest ANXA member to be identified (67). In previous studies, aberrant expression of ANXA10 was associated with carcinogenesis and progression of various types of cancer (6870), which suggested a possible tumor promoter or suppressor role, although its functional role remains to be clarified. In liver cancer, downregulation of ANXA10 is associated with the malignant phenotype of tumor cells and poor prognosis (14). Overexpression of ANXA10 inhibits proliferation and promotes apoptosis of HepG2 cells (71). Consistent with these studies, the results of the present study confirmed that ANXA10 was decreased and associated with clinical stage in patients with liver cancer. In addition, high expression of ANXA10 was associated with an improved prognosis compared with patients with low ANXA10 expression.

Previous studies have suggested that the risk of developing liver cancer in males is higher compared with that in females (7275). Irrespective of the etiology, the morbidity of liver cancer in males is 2–4-fold higher compared with that in females (3). A number of studies have suggested that the androgen receptor (AR) may be responsible for the sex disparity observed in liver cancer (76,77). Of note, a study has reported that the transcription and splicing of ANXA7 is regulated by AR-signaling in prostate cancer (78). However, to the best of our knowledge, the role of ANXAs in the sex disparity of liver cancer has rarely been studied. In the present study, the mRNA expression levels of ANXA2, A5, A7 and A10 were significantly associated with OS in male, but not in female patients, which suggested a possible role for these ANXAs in sex disparity.

The potential biological functions of ANXAs in liver cancer were also investigated in the present study through GO functional annotation and KEGG pathway enrichment analyses. The GO analysis revealed that ANXAs and their neighboring genes primarily participated in ‘neuropeptide signaling pathway’, ‘G-protein coupled receptor signaling pathway’, ‘positive regulation of cytosolic calcium ion concentration’ and ‘signal transduction’, which were consistent with previous studies reporting that ANXAs serve important roles in calcium signaling (7,79). In addition, the KEGG pathway analysis demonstrated that ANXAs and their neighboring genes were primarily involved in ‘neuroactive ligand-receptor interaction’, ‘GABAergic synapse’, ‘chemokine signaling pathway’, ‘calcium signaling pathway’ and ‘pathways in cancer’. Among them, the most significantly enriched pathway was the ‘neuroactive ligand-receptor interaction’, suggesting that ANXAs may function in liver cancer through the neuroactive ligand-receptor interaction pathway.

In the present study, the expression levels, prognostic roles and potential biological functions of 13 ANXAs in liver cancer were systemically investigated. The results indicated that ANXA1, A2, A3, A4, A5 and A10 may be potential therapeutic targets for liver cancer treatment, whereas ANXA2, A5, A7 and A10 may be potential prognostic biomarkers of liver cancer. However, functional experiments are required in order to confirm the role of ANXAs in liver cancer progression as well as their specificity and sensitivity as biomarkers. The results of the present study introduced ANXA2/5/10 as good candidates for future experimental works.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 81700514) and the Key Scientific Research Projects in Colleges and Universities of Henan Province (grant no. 19A320064).

Availability of data and materials

The datasets generated and/or analyzed during the present study are available in the (TCGA) and (Oncomine) repositories, (https://portal.gdc.cancer.gov/, http://www.oncomine.org/).

Authors' contributions

CZ and LM conceived and designed the study. CZ, PW, TS and LZ collected and analyzed the data. CZ and PW wrote the initial draft of the manuscript.. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Academic Committee of Zhengzhou University (Zhengzhou, Henan, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

3 

El-Serag HB: Hepatocellular carcinoma. N Engl J Med. 365:1118–1127. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Enrich C, Rentero C and Grewal T: Annexin A6 in the liver: From the endocytic compartment to cellular physiology. Biochim Biophys Acta Mol Cell Res. 1864:933–946. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Sugimoto MA, Vago JP, Teixeira MM and Sousa LP: Annexin A1 and the resolution of inflammation: Modulation of neutrophil recruitment, apoptosis, and clearance. J Immunol Res. 2016:82392582016. View Article : Google Scholar : PubMed/NCBI

6 

Rand JH, Wu XX, Quinn AS and Taatjes DJ: Resistance to annexin A5 anticoagulant activity: A thrombogenic mechanism for the antiphospholipid syndrome. Lupus. 17:922–930. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Gerke V, Creutz CE and Moss SE: Annexins: linking Ca2+ signalling to membrane dynamics. Nat Rev Mol Cell Biol. 6:449–461. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Mussunoor S and Murray GI: The role of annexins in tumour development and progression. J Pathol. 216:131–140. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Ydy LR, do Espirito Santo GF, de Menezes I, Martins MS, Ignotti E and Damazo AS: Study of the annexin A1 and its associations with carcinoembryonic antigen and mismatch repair proteins in colorectal cancer. J Gastrointest Cancer. 47:61–68. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Biaoxue R, Xiling J, Shuanying Y, Wei Z, Xiguang C, Jinsui W and Min Z: Upregulation of Hsp90-beta and annexin A1 correlates with poor survival and lymphatic metastasis in lung cancer patients. J Exp Clin Cancer Res. 31:702012. View Article : Google Scholar : PubMed/NCBI

11 

Gao Y, Chen Y, Xu D, Wang J and Yu G: Differential expression of ANXA1 in benign human gastrointestinal tissues and cancers. BMC Cancer. 14:5202014. View Article : Google Scholar : PubMed/NCBI

12 

Lin Y, Lin G, Fang W, Zhu H and Chu K: Increased expression of annexin A1 predicts poor prognosis in human hepatocellular carcinoma and enhances cell malignant phenotype. Med Oncol. 31:3272014. View Article : Google Scholar : PubMed/NCBI

13 

Ibrahim MM, Sun MZ, Huang Y, Jun M, Jin Y, Yue D, Jiasheng W, Zhang J, Qazi AS, Sagoe K and Tang J: Down-regulation of ANXA7 decreases metastatic potential of human hepatocellular carcinoma cells in vitro. Biomed Pharmacother. 67:285–291. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Liu SH, Lin CY, Peng SY, Jeng YM, Pan HW, Lai PL, Liu CL and Hsu HC: Down-regulation of annexin A10 in hepatocellular carcinoma is associated with vascular invasion, early recurrence, and poor prognosis in synergy with p53 mutation. Am J Pathol. 160:1831–1837. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Liu YY, Ge C, Tian H, Jiang JY, Zhao FY, Li H, Chen TY, Yao M and Li JJ: The transcription factor Ikaros inhibits cell proliferation by downregulating ANXA4 expression in hepatocellular carcinoma. Am J Cancer Res. 7:1285–1297. 2017.PubMed/NCBI

16 

Meier EM, Rein-Fischboeck L, Pohl R, Wanninger J, Hoy AJ, Grewal T, Eisinger K, Krautbauer S, Liebisch G, Weiss TS and Buechler C: Annexin A6 protein is downregulated in human hepatocellular carcinoma. Mol Cell Biochem. 418:81–90. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Mohammad HS, Kurokohchi K, Yoneyama H, Tokuda M, Morishita A, Jian G, Shi L, Murota M, Tani J, Kato K, et al: Annexin A2 expression and phosphorylation are up-regulated in hepatocellular carcinoma. Int J Oncol. 33:1157–1163. 2008.PubMed/NCBI

18 

Pan QZ, Pan K, Weng DS, Zhao JJ, Zhang XF, Wang DD, Lv L, Jiang SS, Zheng HX and Xia JC: Annexin A3 promotes tumorigenesis and resistance to chemotherapy in hepatocellular carcinoma. Mol Carcinog. 54:598–607. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The eighth edition AJCC cancer staging manual: Continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Cancer Genome Atlas Research Network. Electronic address, . simplewheeler@bcm.edu; Cancer Genome Atlas Research Network: Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 169:1327–1341.e23. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Mas VR, Maluf DG, Archer KJ, Yanek K, Kong X, Kulik L, Freise CE, Olthoff KM, Ghobrial RM, McIver P and Fisher R: Genes involved in viral carcinogenesis and tumor initiation in hepatitis C virus-induced hepatocellular carcinoma. Mol Med. 15:85–94. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Roessler S, Jia HL, Budhu A, Forgues M, Ye QH, Lee JS, Thorgeirsson SS, Sun Z, Tang ZY, Qin LX and Wang XW: A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 70:10202–10212. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Wurmbach E, Chen YB, Khitrov G, Zhang W, Roayaie S, Schwartz M, Fiel I, Thung S, Mazzaferro V, Bruix J, et al: Genome-wide molecular profiles of HCV-induced dysplasia and hepatocellular carcinoma. Hepatology. 45:938–947. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Sheikh MH and Solito E: Annexin A1: Uncovering the many talents of an old protein. Int J Mol Sci. 19(pii): E10452018. View Article : Google Scholar : PubMed/NCBI

26 

D'Acquisto F, Piras G and Rattazzi L: Pro-inflammatory and pathogenic properties of Annexin-A1: The whole is greater than the sum of its parts. Biochem Pharmacol. 85:1213–1218. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Locatelli I, Sutti S, Jindal A, Vacchiano M, Bozzola C, Reutelingsperger C, Kusters D, Bena S, Parola M, Paternostro C, et al: Endogenous annexin A1 is a novel protective determinant in nonalcoholic steatohepatitis in mice. Hepatology. 60:531–544. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Moraes LA, Kar S, Foo SL, Gu T, Toh YQ, Ampomah PB, Sachaphibulkij K, Yap G, Zharkova O, Lukman HM, et al: Annexin-A1 enhances breast cancer growth and migration by promoting alternative macrophage polarization in the tumour microenvironment. Sci Rep. 7:179252017. View Article : Google Scholar : PubMed/NCBI

29 

de Graauw M, van Miltenburg MH, Schmidt MK, Pont C, Lalai R, Kartopawiro J, Pardali E, Le Dévédec SE, Smit VT, van der Wal A, et al: Annexin A1 regulates TGF-beta signaling and promotes metastasis formation of basal-like breast cancer cells. Proc Natl Acad Sci USA. 107:6340–6345. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Suo A, Zhang M, Yao Y, Zhang L, Huang C, Nan K and Zhang W: Proteome analysis of the effects of sorafenib on human hepatocellular carcinoma cell line HepG2. Med Oncol. 29:1827–1836. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Hongsrichan N, Rucksaken R, Chamgramol Y, Pinlaor P, Techasen A, Yongvanit P, Khuntikeo N, Pairojkul C and Pinlaor S: Annexin A1: A new immunohistological marker of cholangiocarcinoma. World J Gastroenterol. 19:2456–2465. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Xue LY, Teng LH, Zou SM, Ren LQ, Zheng S, Luo W, Bi R and Lü N: Expression of annexin I in different histological types of carcinomas. Zhonghua Zhong Liu Za Zhi. 29:444–448. 2007.(In Chinese). PubMed/NCBI

33 

Luo CH, Liu QQ, Zhang PF, Li MY, Chen ZC and Liu YF: Prognostic significance of annexin II expression in non-small cell lung cancer. Clin Transl Oncol. 15:938–946. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Wang T, Yuan J, Zhang J, Tian R, Ji W, Zhou Y, Yang Y, Song W, Zhang F and Niu R: Anxa2 binds to STAT3 and promotes epithelial to mesenchymal transition in breast cancer cells. Oncotarget. 6:30975–30992. 2015.PubMed/NCBI

35 

Han Y, Ye J, Dong Y, Xu Z and Du Q: Expression and significance of annexin A2 in patients with gastric adenocarcinoma and the association with E-cadherin. Exp Ther Med. 10:549–554. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Huang YK, Liu H, Wang XZ and Zhu S: Annexin A2 and CD105 expression in pancreatic ductal adenocarcinoma is associated with tumor recurrence and prognosis. Asian Pac J Cancer Prev. 15:9921–9926. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Xiu D, Liu L, Qiao F, Yang H, Cui L and Liu G: Annexin A2 coordinates STAT3 to regulate the invasion and migration of colorectal cancer cells in vitro. Gastroenterol Res Pract. 2016:35214532016. View Article : Google Scholar : PubMed/NCBI

38 

Zhang H, Yao M, Wu W, Qiu L, Sai W, Yang J, Zheng W, Huang J and Yao D: Up-regulation of annexin A2 expression predicates advanced clinicopathological features and poor prognosis in hepatocellular carcinoma. Tumour Biol. 36:9373–9383. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Kling T, Ferrarese R, Ó hAilín D, Johansson P, Heiland DH, Dai F, Vasilikos I, Weyerbrock A, Jörnsten R, Carro MS and Nelander S: Integrative modeling reveals annexin A2-mediated epigenetic control of mesenchymal glioblastoma. EBioMedicine. 12:72–85. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Luo S, Xie C, Wu P, He J, Tang Y, Xu J and Zhao S: Annexin A2 is an independent prognostic biomarker for evaluating the malignant progression of laryngeal cancer. Exp Ther Med. 14:6113–6118. 2017.PubMed/NCBI

41 

Liu Y, Myrvang HK and Dekker LV: Annexin A2 complexes with S100 proteins: Structure, function and pharmacological manipulation. Br J Pharmacol. 172:1664–1676. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Wang CY, Chen CL, Tseng YL, Fang YT, Lin YS, Su WC, Chen CC, Chang KC, Wang YC and Lin CF: Annexin A2 silencing induces G2 arrest of non-small cell lung cancer cells through p53-dependent and -independent mechanisms. J Biol Chem. 287:32512–32524. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Jung H, Kim JS, Kim WK, Oh KJ, Kim JM, Lee HJ, Han BS, Kim DS, Seo YS, Lee SC, et al: Intracellular annexin A2 regulates NF-κB signaling by binding to the p50 subunit: Implications for gemcitabine resistance in pancreatic cancer. Cell Death Dis. 6:e16062015. View Article : Google Scholar : PubMed/NCBI

44 

Zhang HJ, Yao DF, Yao M, Huang H, Wu W, Yan MJ, Yan XD and Chen J: Expression characteristics and diagnostic value of annexin A2 in hepatocellular carcinoma. World J Gastroenterol. 18:5897–5904. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Zhang W, Zhao P, Xu XL, Cai L, Song ZS, Cao DY, Tao KS, Zhou WP, Chen ZN and Dou KF: Annexin A2 promotes the migration and invasion of human hepatocellular carcinoma cells in vitro by regulating the shedding of CD147-harboring microvesicles from tumor cells. PLoS One. 8:e672682013. View Article : Google Scholar : PubMed/NCBI

46 

El-Abd N, Fawzy A, Elbaz T and Hamdy S: Evaluation of annexin A2 and as potential biomarkers for hepatocellular carcinoma. Tumour Biol. 37:211–216. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Du R, Liu B, Zhou L, Wang D, He X, Xu X, Zhang L, Niu C and Liu S: Downregulation of annexin A3 inhibits tumor metastasis and decreases drug resistance in breast cancer. Cell Death Dis. 9:1262018. View Article : Google Scholar : PubMed/NCBI

48 

Wang K and Li J: Overexpression of ANXA3 is an independent prognostic indicator in gastric cancer and its depletion suppresses cell proliferation and tumor growth. Oncotarget. 7:86972–86984. 2016.PubMed/NCBI

49 

Köllermann J, Schlomm T, Bang H, Schwall GP, von Eichel-Streiber C, Simon R, Schostak M, Huland H, Berg W, Sauter G, et al: Expression and prognostic relevance of annexin A3 in prostate cancer. Eur Urol. 54:1314–1323. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Pan QZ, Pan K, Wang QJ, Weng DS, Zhao JJ, Zheng HX, Zhang XF, Jiang SS, Lv L, Tang Y, et al: Annexin A3 as a potential target for immunotherapy of liver cancer stem-like cells. Stem Cells. 33:354–366. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Tong M, Fung TM, Luk ST, Ng KY, Lee TK, Lin CH, Yam JW, Chan KW, Ng F, Zheng BJ, et al: ANXA3/JNK signaling promotes self-renewal and tumor growth, and its blockade provides a therapeutic target for hepatocellular carcinoma. Stem Cell Reports. 5:45–59. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Ma XL, Jiang M, Zhao Y, Wang BL, Shen MN, Zhou Y, Zhang CY, Sun YF, Chen JW, Hu B, et al: Application of serum annexin A3 in diagnosis, outcome prediction and therapeutic response evaluation for patients with hepatocellular carcinoma. Ann Surg Oncol. 25:1686–1694. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Dreier R, Schmid KW, Gerke V and Riehemann K: Differential expression of annexins I, II and IV in human tissues: An immunohistochemical study. Histochem Cell Biol. 110:137–148. 1998. View Article : Google Scholar : PubMed/NCBI

54 

Gaudio E, Paduano F, Ngankeu A, Ortuso F, Lovat F, Pinton S, D'Agostino S, Zanesi N, Aqeilan RI, Campiglia P, et al: A Fhit-mimetic peptide suppresses annexin A4-mediated chemoresistance to paclitaxel in lung cancer cells. Oncotarget. 7:29927–29936. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Duncan R, Carpenter B, Main LC, Telfer C and Murray GI: Characterisation and protein expression profiling of annexins in colorectal cancer. Br J Cancer. 98:426–433. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Choi CH, Chung JY, Chung EJ, Sears JD, Lee JW, Bae DS and Hewitt SM: Prognostic significance of annexin A2 and annexin A4 expression in patients with cervical cancer. BMC Cancer. 16:4482016. View Article : Google Scholar : PubMed/NCBI

57 

Yao HS, Sun C, Li XX, Wang Y, Jin KZ, Zhang XP and Hu ZQ: Annexin A4-nuclear factor-kappaB feedback circuit regulates cell malignant behavior and tumor growth in gallbladder cancer. Sci Rep. 6:310562016. View Article : Google Scholar : PubMed/NCBI

58 

Chen W, Chen L, Cai Z, Liang D, Zhao B, Zeng Y, Liu X and Liu J: Overexpression of annexin A4 indicates poor prognosis and promotes tumor metastasis of hepatocellular carcinoma. Tumour Biol. 37:9343–9355. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Peng B, Guo C, Guan H, Liu S and Sun MZ: Annexin A5 as a potential marker in tumors. Clin Chim Acta. 427:42–48. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Guo WX, Man XB, Yuan HX, Shi J, Xue J, Wu MC and Cheng SQ: Proteomic analysis on portal vein tumor thrombus-associated proteins for hepatocellular carcinoma. Zhonghua Yi Xue Za Zhi. 87:2094–2097. 2007.(In Chinese). PubMed/NCBI

61 

Sun X, Liu S, Wang J, Wei B, Guo C, Chen C and Sun MZ: Annexin A5 regulates hepatocarcinoma malignancy via CRKI/II-DOCK180-RAC1 integrin and MEK-ERK pathways. Cell Death Dis. 9:6372018. View Article : Google Scholar : PubMed/NCBI

62 

Srivastava M, Torosyan Y, Raffeld M, Eidelman O, Pollard HB and Bubendorf L: ANXA7 expression represents hormone-relevant tumor suppression in different cancers. Int J Cancer. 121:2628–2636. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Du Y, Meng J, Huang Y, Wu J, Wang B, Ibrahim MM and Tang J: Guanine nucleotide-binding protein subunit beta-2-like 1, a new Annexin A7 interacting protein. Biochem Biophys Res Commun. 445:58–63. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Song L, Mao J, Zhang J, Ibrahim MM, Li LH and Tang JW: Annexin A7 and its binding protein galectin-3 influence mouse hepatocellular carcinoma cell line in vitro. Biomed Pharmacother. 68:377–384. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Huang Y, Wang Q, Du Y, Bai L, Jin F, Zhang J, Fan S, Wang H, Song L, Gao Y, et al: Inhibition of annexin A7 gene and protein induces the apotosis and decreases the invasion, migration of the hepatocarcinoma cell line. Biomed Pharmacother. 68:819–824. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Wang XY, Gao F, Sun YR, Bai LL, Ibrahim MM, Wang B and Tang JW: In vivo and in vitro effect of hepatocarcinoma lymph node metastasis by upregulation of Annexin A7 and relevant mechanisms. Tumour Biol. 37:911–924. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Morgan RO, Jenkins NA, Gilbert DJ, Copeland NG, Balsara BR, Testa JR and Fernandez MP: Novel human and mouse annexin A10 are linked to the genome duplications during early chordate evolution. Genomics. 60:40–49. 1999. View Article : Google Scholar : PubMed/NCBI

68 

Kim J, Kim MA, Jee CD, Jung EJ and Kim WH: Reduced expression and homozygous deletion of annexin A10 in gastric carcinoma. Int J Cancer. 125:1842–1850. 2009. View Article : Google Scholar : PubMed/NCBI

69 

van der Heijden AG, Mengual L, Lozano JJ, Ingelmo-Torres M, Ribal MJ, Fernández PL, Oosterwijk E, Schalken JA, Alcaraz A and Witjes JA: A five-gene expression signature to predict progression in T1G3 bladder cancer. Eur J Cancer. 64:127–136. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Zhu J, Wu J, Pei X, Tan Z, Shi J and Lubman DM: Annexin A10 is a candidate marker associated with the progression of pancreatic precursor lesions to adenocarcinoma. PLoS One. 12:e01750392017. View Article : Google Scholar : PubMed/NCBI

71 

Liu X, Peng X, Hu Z, Zhao Q, He J, Li J and Zhong X: Effects of over-expression of ANXA10 gene on proliferation and apoptosis of hepatocellular carcinoma cell line HepG2. J Huazhong Univ Sci Technolog Med Sci. 32:669–674. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Li Y, Xu A, Jia S and Huang J: Recent advances in the molecular mechanism of sex disparity in hepatocellular carcinoma. Oncol Lett. 17:4222–4228. 2019.PubMed/NCBI

73 

Liu C, Ren YF, Dong J, Ke MY, Ma F, Monga SPS, Wu R, Lv Y and Zhang XF: Activation of SRY accounts for male-specific hepatocarcinogenesis: Implication in gender disparity of hepatocellular carcinoma. Cancer Lett. 410:20–31. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Zheng B, Zhu YJ, Wang HY and Chen L: Gender disparity in hepatocellular carcinoma (HCC): Multiple underlying mechanisms. Sci China Life Sci. 60:575–584. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM and Karin M: Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 317:121–124. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Kanda T, Takahashi K, Nakamura M, Nakamoto S, Wu S, Haga Y, Sasaki R, Jiang X and Yokosuka O: Androgen receptor could be a potential therapeutic target in patients with advanced hepatocellular carcinoma. Cancers (Basel). 9:pii432017. View Article : Google Scholar

77 

Yeh SH and Chen PJ: Gender disparity of hepatocellular carcinoma: The roles of sex hormones. Oncology. 78 (Suppl 1):S172–S179. 2010. View Article : Google Scholar

78 

Torosyan Y, Simakova O, Naga S, Mezhevaya K, Leighton X, Diaz J, Huang W, Pollard H and Srivastava M: Annexin-A7 protects normal prostate cells and induces distinct patterns of RB-associated cytotoxicity in androgen-sensitive and -resistant prostate cancer cells. Int J Cancer. 125:2528–2539. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Grewal T, Wason SJ, Enrich C and Rentero C: Annexins-insights from knockout mice. Biol Chem. 397:1031–1053. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhuang C, Wang P, Sun T, Zheng L and Ming L: Expression levels and prognostic values of annexins in liver cancer. Oncol Lett 18: 6657-6669, 2019
APA
Zhuang, C., Wang, P., Sun, T., Zheng, L., & Ming, L. (2019). Expression levels and prognostic values of annexins in liver cancer. Oncology Letters, 18, 6657-6669. https://doi.org/10.3892/ol.2019.11025
MLA
Zhuang, C., Wang, P., Sun, T., Zheng, L., Ming, L."Expression levels and prognostic values of annexins in liver cancer". Oncology Letters 18.6 (2019): 6657-6669.
Chicago
Zhuang, C., Wang, P., Sun, T., Zheng, L., Ming, L."Expression levels and prognostic values of annexins in liver cancer". Oncology Letters 18, no. 6 (2019): 6657-6669. https://doi.org/10.3892/ol.2019.11025