Open Access

Actin-binding protein anillin promotes the progression of hepatocellular carcinoma <em>in vitro</em> and in mice

Retraction in: /10.3892/etm.2023.12120

  • Authors:
    • Huanxia Jia
    • Zhenya Gao
    • Fang Yu
    • Hongfang Guo
    • Baoyu Li
  • View Affiliations

  • Published online on: March 1, 2021     https://doi.org/10.3892/etm.2021.9885
  • Article Number: 454
  • Copyright: © Jia 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

Hepatocellular carcinoma (HCC) is a common type of tumor with high mortality worldwide. Investigations associated with the molecular etiology of HCC and screening novel therapeutic targets are still urgently in need. Anillin (ANLN), as a type of evolutionarily conserved actin‑binding protein, is involved in multiple cellular processes. ANLN widely affected the progression and metastasis of several types of cancer, and its overexpression was frequently demonstrated in previous studies. The present study demonstrated high expression of ANLN in human HCC tissues, which was also associated the prognosis of patients with HCC. The associations between ANLN expression and the clinicopathological features were determined, including the number of tumor nodes (P=0.011) and tumor size (P=0.003) of patients with HCC. It was found that ANLN promoted cell proliferation, invasion and migration of HCC cells in vitro, and affected tumor growth in vivo. Therefore, ANLN is suggested as a promising therapeutic target for the treatment of HCC.

Introduction

Hepatocellular carcinoma (HCC) is a common type of malignant tumor and the most frequent mortality associated with cancer worldwide (1). The standard therapy of early stage HCC include radio frequency ablation, hepatic resection, and transplantation (2). Last decades have witnessed advances in targeted therapy for HCC. Despite the promising effects of targeted therapy drugs, such as sorafenib, on promoting survival among patients with advanced HCC, toxicity remain to be solved (3,4). Hence, it is of great importance to investigate the molecular etiology associated with HCC, which may offer new therapy and therapeutic targets for its treatment (4).

Anillin (ANLN), as a type of evolutionarily conserved actin-binding protein, is involved in multiple cellular processes, such as cytokinesis (5,6). ANLN contains actin and myosin binding domains in the conserved N-terminal, an anillin homology domain in C terminus and a PH domain binding to RhoA, Ect2, and Septins for the assembly and maintenance of the cleavage furrow (6,7). Previous studies indicated that the lack of ANLN was associated with the correct assembly of the cleavage furrow (8,9).

ANLN was widely involved in the progression and metastasis of multiple types of cancer, and the overexpression of ANLN was frequently observed in multiple types of cancer, such as breast cancer and lung cancer (10-12). Deficiency of ANLN expression could inhibit the proliferation of human non-small cell lung cancer and breast cancer cells in vitro (13,14). In human lung cancer tissue, high ANLN mRNA level and nuclear ANLN protein level predicted poor survival in patients with lung cancer (15). Moreover, ANLN mRNA levels were enhanced in several types of tumors, such as pancreatic cancer (11). It is noteworthy that several studies have suggested ANLN to be potentially involved in the development of HCC (16,17). Furthermore, a previous study showed that ANLN was required for tumor growth in HBV-associated HCC (16). Another study indicated that the CDK1-PLK1/SGOL2/ANLN pathway mediated cell division in HCC cells (17). These findings suggested that ANLN had the potential to affect the progression of HCC.

The present study aimed study the role of ANLN in HCC. IHC assays and bioinformatics analysis was performed to assess the expression of ANLN human HCC tissues. The associations between ANLN expression, the prognosis and clinicopathological characteristics of patients with HCC were also analyzed. Its role in the proliferation, invasion and migration of HCC cells in vitro, and tumor growth in vivo was subsequently detected. ANLN was considered to be a promising therapeutic target for the treatment of HCC.

Materials and methods

Bioinformatic analysis

Bioinformatic analysis was conducted via Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn/detail.php?gene=ANLN/) to analyze The Cancer Genome Atlas (TCGA; https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) data with a threshold of P<0.05 and log(fold-change) >1 or <-1 for differential genes. The median of the survival rates was used as the basis for dividing patients into two groups (low and high expression) for Kaplan-Meier survival analysis. Log-rank test was used for the survival analysis.

Antibodies, plasmids and primers

Rabbit anti-anillin antibody [for immunohistochemistry (IHC) assays: 1:200; for immunoblot assays, 1:1,000; cat. no. ab211872; Abcam), mouse anti-β-actin (1:10,000; cat. no. ab8226; Abcam]. The quantitative PCR primers were as follows: ANLN forward, 5'-CATCTCTGCCCCCTCTGC TGA-3'; ANLN reverse, 5'-GGATGACCTTGCCCACAG CCT-3'; ANLN forward, 5'-CGACCACTTTGTCAAGCTCA-3'; and ANLN reverse, 5'-GGTTGAGCACAGGGTACTTTATT-3'.

A total of 4 pre-designed short hairpin (sh)RNA constructs targeting ANLN were inserted into pAV-U6-GFP (cat. no. SH807607; Addgene, Inc.). shRNA plasmid of ANLN with the highest silencing efficiency was selected for subsequent experiments following the detection of silencing efficiency (data not shown). ANLN shRNA were listed as follows: shRNA#1: 5'-AAGGTCTATGACTCATGCTAAGC-3'; shRNA#2: 5'-AAG TGGAGCATCTGCTAGGATCA-3'; shRNA#3: 5'-AAAGCA AACAACTAGAAACCAAA-3'; shRNA#4: 5'-AATCTGGTGA TAGCCTTGGTTCT-3'.

Clinical samples, immunohistochemistry (IHC) and clinical pathological analysis

Human HCC tissues and the corresponding normal tissues were collected in the Secondary Hospital of Tianjin Medical University. This study was approved by the Ethics Committee of the Secondary Hospital of Tianjin Medical University. All clinical specimens were collected from the Secondary Hospital of Tianjin Medical University between April 2016 and December 2018. The exclusion criteria were as follows: All patients with distal metastasis or chemotherapy. All patients were treated with surgery only. All patients were enrolled with informed consent. The patients were followed up according to the items listed in Table I and then summarized for clinicopathological analysis.

Table I

Association of ANLN and clinicopathological characteristics in 66 patients with hepatocellular carcinoma.

Table I

Association of ANLN and clinicopathological characteristics in 66 patients with hepatocellular carcinoma.

 ANLN expression 
FeatureAll, n=66Low, n=28High, n=38χ2P-value
Age, years   3.6290.057
     <55461630  
     ≥5520128  
Sex   0.0040.951
     Male381622  
     Female281216  
Number of tumor nodes   6.4170.011
     Single261610  
     Multiple ≥2401228  
Tumor grade   1.1430.285
     Low281414  
     High381424  
Tumor size, cm   8.9630.003
     <522157  
     ≥5441331  
Lymph node metastasis   3.2260.073
     No391326  
     Yes271512  
AFP, ng/ml   0.2100.647
     <5016610  
     ≥50502228  

[i] ANLN, anillin; AFP, α-fetoprotein.

To examine the expression levels of ANLN in human HCC tissues and non-tumor tissues (which were normal), IHC assays were performed. Briefly, tumor tissues were cut into 5-µm slices and fixed with 4% paraformaldehyde (PFA) at room temperature for 30 min, and subsequently blocked with 2% BSA for 1 h at room temperature. Slides were subsequently incubated with indicated antibodies at room temperature for 2 h. After washing with PBS thrice, the sections were incubated with mice or rabbit biotinylated secondary antibodies (1:500; cat. noσ. ab201485 and ab99807, respectively; Abcam) for 1 h at room temperature, and diaminobenzidine was used as a chromogen substrate at room temperature.

The scoring method used for scoring was as follows. The positive staining proportion of positive cells was evaluated as: 0 (negative staining cells or 0-10% positive staining cells); 1 (10-60% positive staining cells) and 2 (over 60% positive staining cells). The staining intensity scored 0 (negative level staining), 1 (low staining), and 2 (high staining). Staining scored 0-2 was considered low expression, while staining index 3-4 was considered as high expression. The scoring system was used for patient tumor tissues.

The clinico-pathological characteristics of patients with HCC were recorded and analyzed. Patient age, sex, tumor grade, lymph node metastasis and α-fetoprotein (AFP) were analyzed. The tumors were recorded as single or multiple, as well as < or >5 cm (based upon the largest diameter in multiple tumors).

Cell culture and transfection

Hep3B and SNU475 cells were bought from American Type Culture Collection, and examined for mycoplasma contamination. Both cells were maintained in Dulbecco Modified Eagle Medium (DMEM, Gibco; Thermo Fisher Scientific, Inc.) containing 10% of fetal bovine serum (FBS Gibco; Thermo Fisher Scientific, Inc.) at 37˚C in a 5% CO2 incubator at 37˚C with 100 U/ml penicillin and 0.1 mg/ml streptomycin.

The aforementioned ANLN shRNA plasmids were transfected into both Hep3B and SNU475 cells using Lipofectamine® 2000 (cat. no. 11668019; Invitrogen; Thermo Fisher Scientific, Inc.). In 6-well plates, 5 µl transfection reagent and 1.5 µg shRNA plasmids were mixed in 300 µl serum-free DMEM, left to stand for 5 min and then mixed. Following incubation at room temperature for 20 min, the mix was added to serum-starved cells and incubated at 37˚C for 4 h. For the control group, the shRNA targeting sequence was nonsense and did not target intracellular RNAs. After transfection, Hep3B cells were further treated with 1 mg/ml puromycin (Sigma-Aldrich; Merck KGaA) to screen for stable ANLN knockdown cells and used for the animal assays. The subsequent experiments were performed after 24 h of transfection.

Quantitative PCR assays

Quantitative PCR assays were performed to detect the mRNA levels of ANLN in cells or tissues from different groups. Total RNA from human HCC cells were extracted by TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) reagent. Then RNA was reverse transcribed (42˚C, 60 min) into cDNA by reverse transcriptase (M1701; Promega Corporation). Quantitative PCR was conducted by using SYBR Ex Taq kit (Takara Biotechnology Co., Ltd.), the following thermocycling conditions were used for qPCR: Initial denaturation at 95˚C for 3 min; followed by 30 cycles of denaturation at 95˚C for 30 sec, annealing at 58˚C for 30 sec and extension at 72˚C for 30 sec. The 2-ΔΔCq method was used to quantify the results (18). The expression levels of ANLN were normalized to GAPDH level.

Immunoblotting

Total protein from cells and tissues were extracted by lysis buffer (9800; Cell Signaling Technology, Inc.). Subsequently, protein determination was performed using the BCA method, and the protein samples (20 µg loaded per lane) were separated via 10% SDS-PAGE. Protein was transferred onto the polyvinylidene fluoride (PVDF) membranes. After blocking with 5% BSA in TBST for 2 h at room temperature, the membranes were incubated with indicated primary antibody of ANLN at room temperature for 2 h. Following washing, the PVDF membranes were conjugated with mice or rabbit secondary antibodies (1:5,000; cat. no. ab201485 and ab99807; respectively; Abcam) for 1 h at room temperature. Signals were detected using an ECL kit (Novex™ ECL Chemiluminescent Substrate Reagent kit; Thermo Fisher Scientific, Inc.). ImageJ 9.0 software (National Institutes of Health) was used for densitometry analysis.

Colony-formation assays

Colony-formation assays were performed to detect the effects of ANLN on HCC cell proliferation. Hep3B or SNU475 cells (~1,000) were seeded into a 6-well culture plate for 24 h. After culturing for 2 weeks, the cells were fixed with 4% PFA for 20 min at room temperature, stained with 0.2% crystal violet buffer for 30 min at room temperature, and washed with PBS. Images of the colonies were captured and counted manually. A light microscope (IX71; Zeiss AG) was used for imaging at a magnification of x20.

Cell Counting Kit-8 (CCK-8) assay

CCK-8 assays were performed to detect the effects of ANLN on HCC cell proliferation. A total of ~1,000 HCC cells transfected with control or ANLN shRNA plasmids were seeded into 96-well plates and maintained for 48 h. HCC cells were treated with CCK-8 solutions (Dojindo Molecular Technologies, Inc.) for 1.5 h at 37˚C. Absorbance of each well was measured by a microplate reader at 450 nm.

Wound healing assays

The effects of ANLN on HCC cell migration was detected through wound healing assays. Both Hep3B and SNU-475 cells, transfected with control or ANLN shRNA plasmids, were maintained for 48 h and a mechanical lesion was made using a 20-µl-pipette tip. Subsequently, cancer cells were washed with PBS, and the serum-free culture medium was added. Images were captured were taken at 0 and 24 h, respectively, and the relative extent of wound healing was the percentage of the area migrated divided by the total wound area. A light microscope (IX71; Zeiss AG) was used for imaging at a magnification of x50.

Transwell assays

The effects of ANLN on HCC cell invasion was detected through Transwell assays. Hep3B and SNU-475 cells transfected with the indicated shRNA were re-suspended in DMEM without FBS. Matrigel (20% in DMEM) was placed on the upper chambers of filters (8.0-µm membrane pores) and incubated at 37˚C for 30 min. Approximately 1x105 cancer cells were placed into the upper chambers and induced to migrate toward the bottom chambers containing DMEM with 10% FBS. Following incubation at 37˚C for 30 min, cells in the top chamber were removed using a cotton swab, the remaining cells were fixed in 4% PFA for 30 min at room temperature, and stained with 0.2% crystal violet buffer for 30 min at room temperature. The relative cell number was quantified manually. A light microscope (IX71; Zeiss AG) was used for imaging at a magnification of x20.

Tumor growth assays

The animal study was approved by the Secondary Hospital of Tianjin Medical University (permit no. SYXK 2019-0216), to detect the effects of ANLN on tumor growth in vivo. All animal protocols were approved by the Institutional Animal Care and Use Committee (IACUC). Female BALB/c nude mice (8-week-old; weight, ~20 g) were supplied by Beijing Vital River Laboratory Animal Technology Co., Ltd. Mice were fed with food and water ad libitum, and were kept at a Specific Pathogen-Free level at 20˚C and a humidity of 60%, alternating between light and dark for 12 h. Six mice were used in each group, and all mice were given adequate food and water and did not die normally. Mice were sacrificed with their necks broken before tumor tissue was removed, and their heartbeat was checked to validate their death. Adequate humanitarian care was given.

Hep3B cells were infected with control or ANLN shRNA lentivirus to stably knockdown ANLN. Approximately 5x105 ANLN depletion or control cells were subcutaneously implanted into nude mice to induce tumor. After 14 days, the tumor began to form, and its volume was measured every 3 days. Tumor volume was calculated as follows: Tumor volume (mm3) = tumor length (mm) x tumor width (mm)2/2. After 29 days, all tumors were removed and images were captured. Subsequently, the expression levels of ANLN were measured through IHC assays. The expression of ANLN was compared between control and ANLN depletion tumors using ImageJ software 9.0 (National Institutes of Health) according the staining intensity. The staining intensity of the control group was normalized.

Statistical analysis

GraphPad 6.0 (GraphPad Software, Inc.) was used for data analysis in this study. All data are representative of at least three independent experiments and are displayed as mean ± SEM. Paired-t-test was performed to analyze the mRNA levels of ANLN in tumor and normal tissues. Additionally, the analysis between clinical overall survival and disease-free survival and ANLN expression were performed through χ2 analysis. Student's t-test was used for statistical comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

ANLN is associated with poor prognosis in patients with HCC

To analyze the expression of ANLN in human HCC and the corresponding normal liver tissues, bioinformatic analysis was first performed. ANLN mRNA level was found to be significantly enhanced in tumor tissues compared with the normal tissues (Fig. 1A; P<0.05), according to the GEPIA database. Importantly, the median ANLN level was used as the standard to divide all patients into high group (n=180) and low group (n=181). The patients with high ANLN mRNA level had poor overall survival rate as displayed in Fig. 1B. Similarly, patients with high expression of ANLN tend to have lower disease-free survival. These data suggest that ANLN is highly expressed in HCC tissues and associated with the poor prognosis of patients with HCC.

ANLN is upregulated in human HCC tissues

To further identify the expression levels of ANLN in 66 patients with HCC, qPCR and IHC assays were performed. The mRNA levels of ANLN in 66 tumor tissues and corresponding normal tissues were analyzed, which showed high ANLN mRNA levels in tumor tissues (Fig. 2A). The expression of ANLN level was higher in patients with HCC compared with the adjacent normal tissues (Fig. 2B and C). Thus, high expression level of ANLN was demonstrated in human HCC tissues.

Furthermore, the patients were also divided into two groups according to the staining level. The associations between ANLN levels and clinicopathological characteristics of 66 patients with HCC were analyzed. As shown in Table I, the patient's age, sex, number of tumor nodes, tumor grade, tumor size, lymph node metastasis and AFP level were detected. According to the results, there was no significant clinical association between ANLN expression and clinical features: Age (P=0.057), sex (P=0.951), tumor grade (P=0.285), lymph node metastasis (P=0.073), and AFP (P=0.647) (Table I). However, high ANLN expression level was significantly associated with number of tumor nodes (P=0.011) and tumor size (P=0.003).

ANLN is downregulated in Hep3B and SNU-475 cells by the transfection of ANLN shRNA plasmids

Given the associations between ANLN expression levels, clinical features, and the prognosis of patients with HCC, the function of ANLN in HCC cells was explored. Hep3B and SNU-475 cells were used as HCC cell model and were transfected with ANLN shRNA plasmids to specifically decrease ANLN expression in vitro. The expression levels of ANLN was ablated, as examined by qPCR assays (Fig. 3A). Similarly, through immunoblot assays, decreased ANLN protein level was detected following transfection with ANLN shRNA plasmids (Fig. 3B). Thus, the efficiency of ANLN shRNA plasmid transfection was confirmed.

ANLN depletion decreases the proliferation, migration and invasion of HCC cells

Subsequently, to detect the effects of ANLN on HCC cell proliferation, control or ANLN-depleted HCC cells were subjected to colony-formation assays. A significant decrease in colony number following ANLN depletion was observed in both Hep3B and SNU-475 cells (Fig. 4A). Furthermore, the proliferation capacity of control and ANLN-depleted HCC cells was monitored by CCK-8 assays. As a result, ANLN-depleted Hep3B and SNU-475 cells exhibited a markedly suppressed cell proliferation, consistent with the previous results (Fig. 4B). Furthermore, wound healing and Transwell assays were conducted to evaluate the effects of ANLN on the migration and invasion of HCC cells. ANLN ablation notably inhibited the closure of the wound in both Hep3B and SNU-475 cells (Fig. 4C). In addition, ANLN was high involved in the invasion of HCC cells. Relatively lower number of cells were observed in ANLN-depleted groups in both types of HCC cells (Fig. 4D). Therefore, the data suggest that ANLN affects the proliferation, invasion and migration of HCC cells in vitro.

ANLN promotes tumor formation in vivo

To further identify the effect of ANLN on HCC progression in vivo, the xenograft model of HCC in BALB/c nude mice was established using Hep3B-ANLN-shRNA or Hep3B-Con-shRNA cells. Hep3B cells stably transfected with control or ANLN shRNA plasmids were injected into BALB/c nude mice subcutaneously. After 14 days, the tumor began to form, and its volume was measured every 3 days. After 29 days, all tumors were isolated from mice and images were captured. In consistence with the in vitro assays, smaller tumor volume was observed in mice receiving Hep3B-ANLN-shRNA cells. The representative images of tumor in each group are displayed in Fig. 5A. The expression level of ANLN in each group was further examined to confirm the depletion of ANLN in Hep3B-ANLN-shRNA group through immunoblotting (Fig. 5B). Taken together, the results show that ANLN depletion led to the inhibition of tumor growth in vivo.

Discussion

HCC is one of the most common malignant tumors in China, and the mortality rate is second only to that of gastric cancer and esophageal cancer (19). Approximately 110,000 patients die of HCC every year in China, accounting for 45% of the death toll in the world (20). The therapeutic strategies for HCC mainly include chemotherapy, interventional therapy, surgery, liver transplantation and other comprehensive treatments (21). However, due to its high heterogeneity and high metastasis, it is still difficult to achieve good clinical effects (22). Cancer is prone to bone metastasis, and the targeted therapy with liposomes has a good effect (23,24). Targeted therapy for cancer has also been widely studied recently. The targeted therapy drugs, such as sorafenib and sunitinib, have good clinical effects (25). The present study found notably high expression of ANLN in human HCC tissues. The expression levels of ANLN were associated with the prognosis and clinical features of patients with HCC. Thus, ANLN could serve as a potential therapeutic target for HCC treatment.

Bioinformation analysis revealed high expression of ANLN in human HCC tissues. ANLN expression was associated with the prognosis of patients with HCC. IHC assays demonstrated abnormally high expression of ANLN in 66 HCC tissues. The abnormal expression of ANLN suggests a critical role for ANLN in cancer progression, which has been observed in multiple types of cancer, such as lung cancer and breast cancer. These studies, together with the present findings strongly suggest that ANLN could affect cancer progression (26). ANLN expression was associated with the clinical pathological features of patients with HCC. Similarly, the association between ANLN expression and clinical feature of patients has been widely revealed (26,27).

Previous studies indicated the wide involvement of ANLN on the progression and metastasis of multiple of cancer types (26,27). High expression of ANLN was also associated with colorectal cancer prognosis (26). In addition, ANLN could act as a prognostic factor for cancer survival (28). Similarly, transcriptome sequencing identified ANLN as a potential prognostic factor in bladder urothelial carcinoma (29). In breast cancer, ANLN was abnormally highly expressed and was associated with the prognosis of patients receiving anthracycline-based chemotherapy (30). Another study also indicated ANLN was a breast cancer prognostic factor independent of Ki-67(27).

Performing colony-formation assays, CCK-8, wound closure and Transwell assays demonstrated that ANLN affected the proliferation, migration and invasion of HCC cells in vitro. These results indicated the involvement of ANLN in HCC progression. Similarly, ANLN affected the proliferation and cell cycle progression of breast cancer and anaplastic thyroid carcinoma (27,31). ANLN expression was associated with metastasis in lung adenocarcinoma, and the depletion of ANLN also suppressed the migration of breast cancer cells in vitro (14,28). The present in vivo data demonstrated that ANLN ablation suppressed tumor growth of HCC cells in mice, consistent with the in vitro data. However, the precise molecular mechanism needs further study.

Previous studies also demonstrated ANLN affected cancer progression through different signaling pathways. ANLN promoted the progression of gastric cancer via Wnt/β-catenin pathway (32), and acted a vital role in lung carcinogenesis through the PI3K/AKT pathway (13,32). The effects of ANLN on HCC progression have been demonstrated in other studies (16,17). ANLN is required for tumor growth and is regulated through miR-15a/miR-16-1 in HCC (16). Another study showed that CDK1-PLK1/SGOL2/ANLN pathway, mediating abnormal cell division in cell cycle, might be a critical process in HCC (17). These findings are partially consistent with the present findings and suggest ANLN as a potential therapeutic target for HCC. The signaling pathways that mediated the effects of ANLN on HCC progression is worthy of further study. It is speculated that the Wnt/β-catenin and PI3K/AKT have the potential to be involved in the regulation of ANLN on HCC progression, which was reported in gastric cancer and lung cancer (13,32). Both Wnt/β-catenin and PI3K/AKT pathway could affect the proliferation, migration and apoptosis of HCC cells. Therefore, the pathways through which ANLN affects HCC progression should be investigated next.

In conclusion, through bioinformatic analysis and IHC assays, high expression of ANLN was demonstrated in human HCC tissues. ANLN expression was associated with the prognosis and clinical pathological features of patients with HCC. ANLN promoted the proliferation, migration and invasion of HCC cells in vitro, and contributed to tumor growth of HCC cells in mice. Thus, ANLN could serve as a novel therapeutic target for the treatment of HCC.

Acknowledgements

Not applicable.

Funding

Not applicable.

Availability of data and materials

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

Authors' contributions

HJ, ZG and FY carried out the experiment of molecular biology and drafted the manuscript. HG and LB participated in the design of the study and performed the statistical analysis. HJ, ZG, FY, HG and BL conceived the study, participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All procedures of both the human and animal studies (approval no. SYXK 2019-0216) performed in the current study were approved by the Ethics Committee of School of Medicine Xuchang University. Written informed consent was obtained from all patients or their families.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Nault JC, Sutter O, Nahon P, Ganne-Carrié N and Séror O: Percutaneous treatment of hepatocellular carcinoma: State of the art and innovations. J Hepatol. 68:783–797. 2018.PubMed/NCBI View Article : Google Scholar

2 

Menke K, Schwermer M, Falke K, Felenda J, Beckmann C, Stintzing F, Voigt A, Schramm A and Zuzak TJ: Taraxacum officinale extract induces antitumorigenic effects in ovarian carcinoma cell lines. Eur J Gynaecol Oncol. 40:106–112. 2019.

3 

Labeur TA, Achterbergh R, Takkenberg B, Van Delden O, Mathôt R and Klümpen HJ: Sorafenib for patients with hepatocellular carcinoma and Child-Pugh B liver cirrhosis: Lessons learned from a terminated study. Oncologist. 24:1–6. 2019.PubMed/NCBI View Article : Google Scholar

4 

Ray EM and Sanoff HK: Optimal therapy for patients with hepatocellular carcinoma and resistance or intolerance to sorafenib: Challenges and solutions. J Hepatocell Carcinoma. 4:131–138. 2017.PubMed/NCBI View Article : Google Scholar

5 

Hickson GR and O'Farrell PH: Anillin: A pivotal organizer of the cytokinetic machinery. Biochem Soc Trans. 36:439–441. 2008.PubMed/NCBI View Article : Google Scholar

6 

Piekny AJ and Maddox AS: The myriad roles of Anillin during cytokinesis. Semin Cell Dev Biol. 21:881–891. 2010.PubMed/NCBI View Article : Google Scholar

7 

D'Avino PP, Archambault V, Przewloka MR, Zhang W, Laue ED and Glover DM: Isolation of protein complexes involved in mitosis and cytokinesis from Drosophila cultured cells. Methods Mol Biol. 545:99–112. 2009.PubMed/NCBI View Article : Google Scholar

8 

Oegema K, Savoian MS, Mitchison TJ and Field CM: Functional analysis of a human homologue of the Drosophila actin binding protein anillin suggests a role in cytokinesis. J Cell Biol. 150:539–552. 2000.PubMed/NCBI View Article : Google Scholar

9 

Hickson GR and O'Farrell PH: Rho-dependent control of anillin behavior during cytokinesis. J Cell Biol. 180:285–294. 2008.PubMed/NCBI View Article : Google Scholar

10 

Hall PA, Todd CB, Hyland PL, McDade SS, Grabsch H, Dattani M, Hillan KJ and Russell SE: The septin-binding protein anillin is overexpressed in diverse human tumors. Clin Cancer Res. 11:6780–6786. 2005.PubMed/NCBI View Article : Google Scholar

11 

Olakowski M, Tyszkiewicz T, Jarzab M, Król R, Oczko-Wojciechowska M, Kowalska M, Kowal M, Gala GM, Kajor M, Lange D, et al: NBL1 and anillin (ANLN) genes over-expression in pancreatic carcinoma. Folia Histochem Cytobiol. 47:249–255. 2009.PubMed/NCBI View Article : Google Scholar

12 

Shimizu S, Seki N, Sugimoto T, Horiguchi S, Tanzawa H, Hanazawa T and Okamoto Y: Identification of molecular targets in head and neck squamous cell carcinomas based on genome-wide gene expression profiling. Oncol Rep. 18:1489–1497. 2007.PubMed/NCBI

13 

Suzuki C, Daigo Y, Ishikawa N, Kato T, Hayama S, Ito T, Tsuchiya E and Nakamura Y: ANLN plays a critical role in human lung carcinogenesis through the activation of RHOA and by involvement in the phosphoinositide 3-kinase/AKT pathway. Cancer Res. 65:11314–11325. 2005.PubMed/NCBI View Article : Google Scholar

14 

Zhou W, Wang Z, Shen N, Pi W, Jiang W, Huang J, Hu Y, Li X and Sun L: Knockdown of ANLN by lentivirus inhibits cell growth and migration in human breast cancer. Mol Cell Biochem. 398:11–19. 2015.PubMed/NCBI View Article : Google Scholar

15 

Skrzypski M, Jassem E, Taron M, Sanchez JJ, Mendez P, Rzyman W, Gulida G, Raz D, Jablons D, Provencio M, et al: Three-gene expression signature predicts survival in early-stage squamous cell carcinoma of the lung. Clin Cancer Res. 14:4794–4799. 2008.PubMed/NCBI View Article : Google Scholar

16 

Lian YF, Huang YL, Wang JL, Deng MH, Xia TL, Zeng MS, Chen MS, Wang HB and Huang YH: Anillin is required for tumor growth and regulated by miR-15a/miR-16-1 in HBV-related hepatocellular carcinoma. Aging (Albany NY). 10:1884–1901. 2018.PubMed/NCBI View Article : Google Scholar

17 

Li L, Huang K, Zhao H, Chen B, Ye Q and Yue J: CDK1-PLK1/SGOL2/ANLN pathway mediating abnormal cell division in cell cycle may be a critical process in hepatocellular carcinoma. Cell Cycle. 19:1236–1252. 2020.PubMed/NCBI View Article : Google Scholar

18 

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

19 

Abdelnabi M, Almaghraby A, Saleh Y and Abd Elsamad S: Hepatocellular carcinoma with a direct right atrial extension in an HCV patient previously treated with direct-acting antiviral therapy: A case report. Egypt Heart J. 71(5)2019.PubMed/NCBI View Article : Google Scholar

20 

Salem R, Gabr A, Riaz A, Mora R, Ali R, Abecassis M, Hickey R, Kulik L, Ganger D, Flamm S, et al: Institutional decision to adopt Y90 as primary treatment for hepatocellular carcinoma informed by a 1,000-patient 15-year experience. Hepatology. 68:1429–1440. 2018.PubMed/NCBI View Article : Google Scholar

21 

Lünse S, Heidecke CD and Partecke LI: Current topics and perspectives in surgical management of hepatocellular carcinoma. In: Hepatocellular Carcinoma. Tirnitz-Parker JE (eds). Codon Publications, Brisbane, AU, pp111-126, 2019.

22 

Colombo M: Treatment of hepatocellular carcinoma. Antiviral Res. 52:209–215. 2001.PubMed/NCBI View Article : Google Scholar

23 

Yang Y, Zhao Z, Xie C and Zhao Y: Dual-targeting liposome modified by glutamic hexapeptide and folic acid for bone metastatic breast cancer. Chem Phys Lipids. 228(104882)2020.PubMed/NCBI View Article : Google Scholar

24 

Zhao Z, Zhao Y, Xie C, Chen C, Lin D, Wang S, Lin D, Cui X, Guo Z and Zhou J: Dual-active targeting liposomes drug delivery system for bone metastatic breast cancer: Synthesis and biological evaluation. Chem Phys Lipids. 223(104785)2019.PubMed/NCBI View Article : Google Scholar

25 

Obiorah IE, Chahine J, Park BU, Ko K, deGuzman J and Kallakury B: Well differentiated arginase-1 negative hepatocellular carcinoma. Transl Gastroenterol Hepatol. 4(66)2019.PubMed/NCBI View Article : Google Scholar

26 

Wang G, Shen W, Cui L, Chen W, Hu X and Fu J: Overexpression of Anillin (ANLN) is correlated with colorectal cancer progression and poor prognosis. Cancer Biomark. 16:459–465. 2016.PubMed/NCBI View Article : Google Scholar

27 

Magnusson K, Gremel G, Rydén L, Pontén V, Uhlén M, Dimberg A, Jirström K and Pontén F: ANLN is a prognostic biomarker independent of Ki-67 and essential for cell cycle progression in primary breast cancer. BMC Cancer. 16(904)2016.PubMed/NCBI View Article : Google Scholar

28 

Long X, Zhou W, Wang Y and Liu S: Prognostic significance of ANLN in lung adenocarcinoma. Oncol Lett. 16:1835–1840. 2018.PubMed/NCBI View Article : Google Scholar

29 

Zeng S, Yu X, Ma C, Song R, Zhang Z, Zi X, Chen X, Wang Y, Yu Y, Zhao J, et al: Transcriptome sequencing identifies ANLN as a promising prognostic biomarker in bladder urothelial carcinoma. Sci Rep. 7(3151)2017.PubMed/NCBI View Article : Google Scholar

30 

Wang Z, Chen J, Zhong MZ, Huang J, Hu YP, Feng DY, Zhou ZJ, Luo X, Liu ZQ, Jiang WZ, et al: Overexpression of ANLN contributed to poor prognosis of anthracycline-based chemotherapy in breast cancer patients. Cancer Chemother Pharmacol. 79:535–543. 2017.PubMed/NCBI View Article : Google Scholar

31 

Weinberger P, Ponny SR, Xu H, Bai S, Smallridge R, Copland J and Sharma A: Cell cycle M-phase genes are highly upregulated in anaplastic thyroid carcinoma. Thyroid. 27:236–252. 2017.PubMed/NCBI View Article : Google Scholar

32 

Pandi NS, Manimuthu M, Harunipriya P, Murugesan M, Asha GV and Rajendran S: In silico analysis of expression pattern of a Wnt/β-catenin responsive gene ANLN in gastric cancer. Gene. 545:23–29. 2014.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jia H, Gao Z, Yu F, Guo H and Li B: Actin-binding protein anillin promotes the progression of hepatocellular carcinoma <em>in vitro</em> and in mice Retraction in /10.3892/etm.2023.12120. Exp Ther Med 21: 454, 2021
APA
Jia, H., Gao, Z., Yu, F., Guo, H., & Li, B. (2021). Actin-binding protein anillin promotes the progression of hepatocellular carcinoma <em>in vitro</em> and in mice Retraction in /10.3892/etm.2023.12120. Experimental and Therapeutic Medicine, 21, 454. https://doi.org/10.3892/etm.2021.9885
MLA
Jia, H., Gao, Z., Yu, F., Guo, H., Li, B."Actin-binding protein anillin promotes the progression of hepatocellular carcinoma <em>in vitro</em> and in mice Retraction in /10.3892/etm.2023.12120". Experimental and Therapeutic Medicine 21.5 (2021): 454.
Chicago
Jia, H., Gao, Z., Yu, F., Guo, H., Li, B."Actin-binding protein anillin promotes the progression of hepatocellular carcinoma <em>in vitro</em> and in mice Retraction in /10.3892/etm.2023.12120". Experimental and Therapeutic Medicine 21, no. 5 (2021): 454. https://doi.org/10.3892/etm.2021.9885