Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

TACC3 Promotes Gastric Carcinogenesis by Promoting Epithelial-mesenchymal Transition Through the ERK/Akt/cyclin D1 Signaling Pathway

MD RASHEDUNNABI AKANDA, JEE SOO PARK, MYUNG-GIUN NOH, GEUN-HYOUNG HA, YOUNG SOOK PARK, JAE-HYUK LEE, KYUNG-SUB MOON, CHUNG KWON KIM and KYUNG-HWA LEE
Anticancer Research July 2021, 41 (7) 3349-3361; DOI: https://doi.org/10.21873/anticanres.15123
MD RASHEDUNNABI AKANDA
1Department of Pathology, Chonnam National University Research Institute of Medical Science, Chonnam National University Medical School and Hwasun Hospital, Jeollanam-do, Republic of Korea;
2Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, Bangladesh;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JEE SOO PARK
3Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea;
4Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MYUNG-GIUN NOH
5Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
GEUN-HYOUNG HA
6Medical Innovation Technology Inc. (MEDINNO Inc.), Ace High-End Tower Classic 26, Seoul, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YOUNG SOOK PARK
7Department of Physical & Rehabilitation Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JAE-HYUK LEE
1Department of Pathology, Chonnam National University Research Institute of Medical Science, Chonnam National University Medical School and Hwasun Hospital, Jeollanam-do, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KYUNG-SUB MOON
8Department of Neurosurgery, Brain Tumor Clinic & Gamma Knife Center, Chonnam National University Hwasun Hospital and Medical School, Jeollanam-do, Republic of Korea
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
CHUNG KWON KIM
3Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea;
4Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea;
6Medical Innovation Technology Inc. (MEDINNO Inc.), Ace High-End Tower Classic 26, Seoul, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: kimck0405@gmail.com mdkaylee@jnu.ac.kr
KYUNG-HWA LEE
1Department of Pathology, Chonnam National University Research Institute of Medical Science, Chonnam National University Medical School and Hwasun Hospital, Jeollanam-do, Republic of Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: kimck0405@gmail.com mdkaylee@jnu.ac.kr
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: The present study investigated the oncogenic functions of TACC3 in the progression of gastric cancer (GC). Materials and Methods: We analysed TACC3 in relation to cell growth, invasion capability, expression of epithelial-mesenchymal transition (EMT)-related markers, and ERK/Akt/cyclin D1 signaling factors. The correlation between the immunohistochemically confirmed expression of TACC3 and clinical factors was also analyzed. Results: The increased proliferation and invasion of TACC3-over-expressing GC cells was accompanied by altered regulation of EMT-associated markers and activation of ERK/Akt/cyclin D1 signaling. Immunohistochemical analysis of TACC3 in human GC tissues revealed that its expression is correlated with aggressive characteristics and poor prognosis of intestinal-type GC. Conclusion: TACC3 contributes to gastric tumorigenesis by promoting EMT via the ERK/Akt/cyclin D1 signaling pathway. The correlation between TACC3 expression and multiple clinicopathological variables implies that its effective therapeutic targeting in GC will depend on the tumor subtype.

Key Words:
  • TACC3
  • gastric cancer
  • therapeutic target
  • EMT
  • invasion

Gastric cancer (GC) is the third leading cause of cancer-associated mortality worldwide, but the development of effective treatment is complicated by its high level of genetic and cellular heterogeneity, including a heterogeneous tumor microenvironment (1, 2). Despite recent advances in diagnostic, surgical, and pharmacological interventions, GC tends to progress and thus, often has a dismal prognosis with a high rate of mortality (3). Surgical resection in advanced GC is typically followed by radio-chemotherapy, which can prolong overall survival. However, the 5-year survival in advanced GC patients treated with modern chemotherapy protocols is 3.1% (4). Furthermore, the poor prognosis of patients with therapeutically resistant GC remains a major challenge in clinical oncology. Novel strategies for the identification of effective prognostic biomarkers of therapeutic resistance are needed to overcome the poor prognosis associated with GC.

Targeted cancer therapy is a novel, emerging research field that is anticipated to improve prognosis of many types of cancer. Among the several targets, the epithelial-mesenchymal transition (EMT) has been investigated as one of the critical regulators of chemo-resistance of GC (5), tumor progression, and metastatic spread of diverse cancers (6, 7). The EMT involves the dynamic switching or transdifferentiation from round-shaped, polarized epithelial cells into spindle-shaped, motile mesenchymal cells, whose invasiveness and migratory capacity contribute to cancer progression and distant metastasis (8-10). The distinctive feature of the EMT is the repression of the epithelial cell marker E-cadherin (11), accompanied by the expression of several transcription molecules and the mesenchymal cell markers snail, slug, twist and ZEB (8, 12). Regarding signaling molecules, triggers of EMT include extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/Akt, cyclin D1, and β-catenin, all of which have also been involved in cancer progression and metastasis (13-15).

The transforming acidic coiled-coil (TACC) family consists of centrosomal- and microtubule-associated proteins that participate in the regulation of cell division (16). TACC3 is located in 4p16.3; aberrant location can increase likelihood of development of several cancers and neurodegenerative diseases (17). TACC3 is a highly conserved, microtubule-related protein with a C-terminal coiled-coil domain, critically involved in cancer development (18). By contributing to genomic instability and tumorigenesis through the regulation of transcriptional activities, aberrant TACC3 expression can interfere with mitosis (19) and has been demonstrated to be associated with a poor clinical outcome in several human cancers (20-23). Knockdown of TACC3 suppressed the growth, spread, and tumorigenic properties of renal cell carcinoma cells (24). Genetic analysis of patient samples has indicated that TACC3 over-expression is related with chemoresistance in ovarian cancer and aggressiveness in breast carcinoma (25, 26). Elevated expression of TACC3 is also a poor prognostic indicator in non-small cell lung cancer (21). In cervical carcinoma, TACC3 has been shown to trigger the ERK and PI3K/Akt signaling pathways to promote the EGF-mediated EMT (27, 28).

The mechanism underlying TACC3-mediated tumor progression and metastasis in GC is unclear, but this knowledge would have important clinical implications. Thus, the purpose of this investigation was to elucidate the molecular mechanisms of TACC3 expression in correlation with carcinogenesis in GC.

Materials and Methods

Cell culture. AGS, MKN45, SNU-719 and SNU-638 cells were obtained from the American Type Culture Collection (Manassas, VA, USA) and Korean Cell Line Bank (Seoul, Republic of Korea). AGS, MKN45, SNU-719 and SNU-638 cells were maintained in RPMI-1640 medium (HyClone, Logan, UT, USA) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin and cultured in a humidified incubator at 37°C with 5% CO2.

Antibodies, reagents, and plasmid constructs. Antibodies against TACC3 and actin were purchased from Santa Cruz Biotechnology (Dallas, TX, USA); anti-flag antibody was from Sigma-Aldrich (St. Louis, MO, USA); antibodies against E-cadherin, slug, snail, β-catenin, vimentin, ERK1/2, p-ERK1/2 (T202/Y204), Akt, anti-p-Akt (S473), cyclin D1, and actin were acquired from Cell Signaling Technology (Danvers, MA, USA). Invasion assay was performed using cell invasion assay kits (Chemicon, #ECM550, Sigma-Aldrich). 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) was bought from Sigma-Aldrich.

Western blot analysis. Transfected cells were washed with PBS and lysed in nuclear extraction buffer [20 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) (pH 7.6), 20% glycerol, 250 mM NaCl, 1.5 mM MgCl2, 0.1% Triton X-100, 1 mM phenylmethylsulfonyl fluoride, 1 mM 1,4-dithiothreitol, and protease inhibitor cocktail (Roche, Basel, Switzerland)]. Proteins (30 μg) were separated by 8-12% polyacrylamide gel electrophoresis containing SDS and incubated with primary antibodies for 16 h at 4°C. Bands were quantified using an electrochemiluminescence system (Millipore, Burlington, MA, USA) and an LAS-4000 image analyzer (Fujifilm, Tokyo, Japan). Actin served as a loading control.

Cell proliferation assay. Exponentially growing GC cells were harvested and then spread in 96-well microplates (200 μl medium, 2×104 cells/ml). After the designated time elapsed, the cell suspension was dissolved with DMSO after being incubated with 2 mg MTT/ml for 3 h. The optical density was spectrophotometrically measured at 490 nm.

Cell invasion assay. The invasion assay was carried out with an invasion assay kit (Chemicon, Temecula, CA, USA), according to the manufacturer’s instructions. A cell suspension in FBS-free medium was added to the cell-culture inserts and then placed into the lower chamber containing complete medium. After passing through the extracellular matrix layer during 24 h of incubation, cells attached to the polycarbonate membrane were stained and counted.

Transfection. AGS and SNU-638 cells were transiently transfected with 5 μg Flag-TACC3 (over-expression construct) or with a small hairpin RNA (shRNA) targeting human TACC3 by electroporation using the Neon transfection system (Invitrogen, CA, USA), with one 30-msec pulse at 1300 V following the manufacturer’s recommendation. The cells were maintained in complete medium with 10% FBS for 24-48 h and harvested for western blotting, invasion, and MTT assays.

Human tissue samples and clinical data. The patient cohort included in this study had stage II/III GC and had participated in a phase III open-label multi-center randomized controlled trial (the CLASSIC trial) conducted between 2006 and 2009, in which D2 gastrectomy alone was compared with adjuvant chemotherapy after D2 gastrectomy (29, 30). The median follow-up period of the cohort was 67 months (range=1-92 months). Survival was calculated as disease-free survival (DFS) and overall survival (OS). Data regarding age, sex, stage, microsatellite instability status, Epstein-Barr virus (EBV) infection, HER2 amplification, and histological features were collected from the records of the CLASSIC trial. Gastric cancer stage was assessed following the sixth version of the American Joint Committee on Cancer (AJCC) criteria. This study was approved by the Institutional Review Board of Chonnam National University Hwasun Hospital (CNUHH-2020-149).

Immunohistochemistry (IHC). Tissue microarrays consisting of two 3-mm diameter cores representing each tumor were prepared from FFPE blocks and used for immunohistochemistry (30). Tissue sections (4 μm) were subjected to IHC using the Bond-max autostainer (Leica Microsystems, Buffalo Grove, IL, USA). The sections were pretreated with ER1 solution (for epitope retrieval using citrate buffer at pH 6.0) for 15 min and then incubated with anti-TACC3 antibody (Santa Cruz, catalogue no. sc-376883) at a 1:100 dilution. Negative controls were prepared as described above but omitting the primary antibodies. Cytoplasmic immunoreactivity was assessed using a four-tier grade: 0, null staining; 1+, weak reactivity; 2+, moderate reactivity; and 3+, strong reactivity. The labeled area as a proportion of the entire tumor area was determined as a percentage. The IHC scores for TACC3 immunopositivity were quantified by multiplication of the staining grade with the labeled area percentage as previously described (31). TACC3-positive immune cells were counted in five different densely labeled foci at 400× magnification and the counts were then averaged. IHC slides were assessed by two experienced pathologists (JHL and KHL) blinded to the clinical details; in the event of disagreement between the observers, immunohistochemical staining was re-evaluated. The two pathologists re-assessed the cases jointly and came to an agreement for inconclusive samples.

Statistical analysis. Statistical analyses were carried out using Graph Pad Prism version 6 (Graph Pad, La Jolla, CA, USA) and SPSS version 23.0 software for Windows (SPSS, Chicago, IL, USA). Student’s t-test or the Mann-Whitney test was used to compare differences between two groups. Chi-squared and Fisher’s exact tests were used to evaluate differences between clinicopathological variables and TACC3 expression. The effect of single variables on DFS or OS was determined using univariate (Kaplan–Meier method with log-rank tests) and multivariate (Cox’s proportional hazards model) analyses. Survival was calculated in R (version 4.0.2), with an optimal cut-off point analysis using the surv_cutpoint function from the R-package Survminer (version 0.4.8). A p-value of <0.05 was considered to indicate statistical significance, and a p-value between 0.05 and 0.01 marginal significance.

Results

Aberrant expression of TACC3 protein in intestinal-type gastric cancer. TACC3 expression in human GC tissues was further investigated in an IHC study using tissue microarrays using 629 gastric tumors classified according to histological type using the Lauren classification (32). TACC3 staining was observed in normal gastric epithelial cells (Figure 1A, left panel) and the four categories of the intensity of TACC3 immunostaining are shown in Figure 1A (right panel). Among the 303 intestinal-type GCs, high-level TACC3 expression was detected in 195 (64.4%), and low-level expression in the remaining 108 (35.6%). TACC3 expression correlated significantly with the tumor grade (low histologic grade vs. high histologic grade; p=0.024). Of the 326 diffuse-type GCs, high-level TACC3 expression was determined in 98 (30.1%) and low-level expression in the remaining 228 (69.9%). TACC3 was variably expressed in human gastric cancer tissues (Figure 1A) with the highest frequency in the intestinal-type (Figure 1B) among diffuse-type gastric cancers. We investigated TACC3 gene expression profiles in cancers by searching available data from the Oncomine microarray database. We found that TACC3 was significantly over-expressed in a set of 26 gastric intestinal-type adenocarcinomas, compared to that in 31 gastric mucosa (p-value=7.52×10–10; fold change=5.531) (Figure 1C, left panel). In addition, TACC3 was significantly over-expressed in a set of 65 gastric intestinal-type adenocarcinomas, compared to that in 26 gastric mucosa (p-value=3.05×10–9; fold change=1.670) (Figure 1C, right panel). TACC3 expression was observed in the intestinal-type gastric cell lines with a relative value of expression exceeding 1.5 compared with diffuse-type gastric cell lines (MKN45) (Figure 1D). These data corroborated our findings, indicating that TACC3 levels were significantly over-expressed in gastric intestinal-type adenocarcinomas (Figure 1B and C).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

TACC3 was over-expressed in gastric cancer. (A and B) Representative IHC images for TACC3 in normal human gastric tissues (left) and gastric cancers (right) (original magnification, ×400) (A). TACC3 expression rate was analyzed according to the histological subtype of human gastric cancer including Intestinal-type and diffuse-type histology (B). (C and D) Gene expression data in Oncomine was analyzed. The thick bars in the boxes are average expression levels and the boxes represent 95% of the samples. The error bars are above or below the boxes and the range of expression levels is enclosed by two dots. (D) Four different gastric cancer cells were analyzed for TACC3 expression using immunoblotting (left). TACC3 expression level was quantified through scanning densitometry with actin as an internal control (right).

Correlation between IHC expression of TACC3 and clinicopathologic variables of the gastric cancer cohort. TACC3 expression in human GC tissues was further investigated in an IHC study using tissue microarrays prepared from 629 gastric tumors classified according to histological type using the Lauren classification (32). TACC3 expression was mainly observed in the cytoplasm of both tumor cells and immune cells (Figure 1A). Among the 303 intestinal-type GCs, high-level TACC3 expression was detected in 195 (64%), and low-level expression in the remaining 108 (36%) (Table I). In addition, the expression of TACC3 correlated significantly with the histologic grade of the tumor (low grade vs. high grade; p=0.024), the depth of invasion (T1-T2 vs. T3-4; p=0.026), and perineural invasion by the tumor cells (p=0.009), with a marginal association with lymph node (LN) metastasis (N0 or N1 vs. N2 or higher; p=0.052) and tumor location (upper body vs. lower body; p=0.099) (Table I). The correlation between TACC3-positive immune cells in intestinal-type GC and EBV infection was also statistically significant (p=0.001). Of the 326 diffuse-type GCs, high-level TACC3 expression was determined in 98 (30%) and low-level expression in the remaining 228 (70%) (Table I). TACC3 expression in diffuse-type GC correlated significantly with lymphovascular invasion (p<0.001), microsatellite instability (p=0.004), and EBV infection (p<0.001) (Table II). The correlation between the number of TACC3-positive immune cells in diffuse-type GC and EBV infection was also significant (p<0.001) (Table II).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Correlation between TACC3 expression in intestinal-type gastric cancer (n=303) and clinicopathological variables.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Correlation between TACC3 expression in diffuse-type gastric cancer (n=326) and clinicopathological variables.

TACC3 over-expression increases the proliferation and migration of intestinal-type GC cells. To explore the functional role of TACC3 in intestinal-type GC, genetically modulated cell lines over-expressing TACC3 were established. SNU-638 GC cell lines were transfected with a TACC3-over-expression vector (Flag-TACC3) or the empty vector (Flag). The efficacy of TACC3 over-expression was examined by western blot analysis (Figure 2A). In SNU-638 cells over-expressing TACC3, proliferation was significantly upregulated (Figure 2B). TACC3 over-expression also expanded the invasive capacities of the cells, as revealed in a transwell cell invasion assay (Figure 2C and D).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

TACC3 over-expression enhances the growth and invasiveness of gastric cancer cells. (A) SNU-638 cells were transfected with Flag or Flag-tagged TACC3 plasmids. Forty-eight hours after transfection the cells were harvested and lysates were prepared and analyzed by immunoblotting using the indicated antibodies. (B) MTT assay was performed to estimate proliferation of SNU-638 cells over-expressing TACC3. (C and D) SNU-638 cells over-expressing Flag-TACC3 were pretreated with 10 μg/ml mitomycin C for 1 h at 37°C, washed twice with PBS and then were subjected to invasion assays. The results are expressed as the mean±standard deviation (SD) of at least three independent experiments (*p<0.05).

TACC3-knockdown decreases proliferation and migration by intestinal-type GC cells. Previous studies have demonstrated a role of TACC3 in proliferation and invasion of cervical cancer and osteosarcoma cells (27, 28). To explore the functional role of TACC3 in GC, genetically modulated TACC3-knockdown cells were established. AGS cell lines were transfected with a TACC3-knockdown vector (shTACC3) or the empty vector (shLuc). TACC3 expression was silenced with shRNA, and interference efficacy was validated by western blot analysis (Figure 3A). In AGS cells with TACC3-knockdown, proliferation was significantly down-regulated compare to control cells (shLuc) (Figure 3B). Moreover, TACC3-knockdown significantly reduced transwell invasion (Figure 3C and D). Together, these results indicated that depletion of TACC3 dramatically reduced the proliferation and invasion of gastric cancer cells.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

TACC3-knockdown decreases the growth and invasiveness of gastric cancer cells. AGS cells were transfected with shLuc (control) or TACC3-targeted shRNA (shTACC3), and subjected to immunoblotting for TACC3 and actin. (B) MTT assay was performed to estimate proliferation of AGS cells with TACC3-knockdown. (C and D) AGS cells with TACC3-knockdown (shTACC3) were pretreated with 10 μg/ml mitomycin C for 1 h at 37°C, washed twice with PBS and then were subjected to invasion assays. The results are expressed as the mean±standard deviation (SD) of at least three independent experiments (*p<0.05).

The epithelial-mesenchymal transition is regulated by TACC3 expression via the ERK/Akt/cyclin D1 signaling pathway. We have observed that TACC3 promotes epithelial-mesenchymal transition (EMT) through the activation of PI3K/Akt and ERK signaling pathways in HeLa cells (28). The results described above identified a role of TACC3 in increasing the motility of GC cells, thus implicating this protein in the progression of GC. Because cancer cell motility is closely linked to the EMT, we investigated the role of TACC3 in this process. Thus, the expression of EMT-related markers (E-cadherin, β-catenin, snail, slug, and vimentin) in SNU-638 cells over-expressing TACC3 was examined by western blot analysis. Over-expression of TACC3 induced upregulation of the mesenchymal markers β-catenin, snail, slug, and vimentin and downregulation of E-cadherin (Figure 4A). Moreover, western blot analysis showed an increase in the phosphorylation of Akt and ERK and up-regulation of cyclin D1 in cells over-expressing TACC3 (Figure 4B). By contrast, knockdown of TACC3 in AGS cells reduced expression of β-catenin, snail, slug, and vimentin, but enhanced E-cadherin expression (Figure 4C). In addition, in TACC3-knockdown AGS cells, however, the phosphorylation of Akt and ERK as well as the expression of cyclin D1 were decreased (Figure 4D). Together, these results indicated that TACC3 promotes epithelial-mesenchymal transition (EMT) through the activation of PI3K/Akt and ERK signaling pathways in GC cells.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Regulation of epithelial-mesenchymal transition (EMT) factors and altered expression of ERK/Akt/cyclin D1 signaling by the modulation of TACC3 expression in GC cells. (A and B) SNU-638 cells were transfected with Flag (control), Flag-tagged TACC3 plasmids. Forty-eight hours after transfection, the cells were harvested and lysates were prepared and analyzed by immunoblotting using the indicated antibodies. (C and D) AGS cells were transfected with shLuc (control) or TACC3-targeted shRNA (shTACC3) plasmids. Forty-eight hours after transfection, cells were harvested and lysates were prepared and analyzed by immunoblotting using the indicated antibodies.

TACC3 gene expression is upregulated and affects the prognosis of patients with intestinal-type gastric cancer. The role of TACC3 in GC was further assessed by examining the level of TACC3 expression in a GC patient dataset from the Cancer Genome Atlas (TCGA) database published in the Cbioportal (33, 34). Of the 440 identified cases, TACC3 gene expression levels were reported in 412. Of those, data on both histologic type and survival were available for 129 cases. The level of TACC3 gene expression was significantly higher in patients with intestinal-type GC than in patients with diffuse-type GC (p=0.013) (Figure 5A), a finding consistent with our IHC results, which showed that TACC3 expression was higher in intestinal-type GC than in diffuse-type GC (p<0.001) (Figure 5B). A Kaplan–Meier survival analysis in which the median value of TACC3 gene expression was divided by the cut-off showed that in patients with intestinal-type GC, the expression level of TACC3 was a significant determinant of disease-specific survival (DSS) (p=0.017) but not in OS (p=0.212) (Figure 6). In the group consisting of all histological types of GC and in the group with diffuse-type GC, expression levels of TACC3 were not significantly related with either DSS (p=0.392 and p=0.94, respectively) or OS (p=0.243 and p=0.904, respectively) (Figure 6). Moreover, we plotted the Kaplan–Meier survival curves for TACC3 the web-based curator. TACC3 gene expression affected the prognosis of patients with intestinal-type gastric cancer, but not diffuse-type GC regarding OS (Figure 7). Together, TACC3 gene expression is upregulated and affects the prognosis of patients with intestinal-type gastric cancer.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

TACC3 expression level in gastric cancer (GC) patients according to tumor histologic type. (A) Based on the TCGA data, levels of TACC3 gene expression were significantly higher in patients with intestinal-type GC than in patients with diffuse-type GC. (B) The IHC analysis of TACC3 expression in our patient cohort showed significant differences between intestinal-type GC and diffuse-type GC.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Analyses of disease-specific survival (DSS) and overall survival (OS) in patients with gastric cancer (GC) of all histologic-types, including intestinal-type and diffuse-type GC, using the TCGA data. (A) All histologic-type GC was not significantly related to a longer DSS. (B, C) Patients with intestinal-type GC whose tumor exhibited low TACC3 expression had a significantly longer DSS, whereas no significant benefit was determined for patients with diffuse-type GC. (D) All histologic-type GC was not significantly related to longer OS. (E, F) Patients with intestinal-type GC and low TACC3 expression by the tumor tended to have a longer DSS but the difference compared to those with high TACC3 expression was not significant. This difference in survival rates became even less significant in the group with diffuse-type GC.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

Analysis of overall survival (OS) in patients with gastric cancer (GC) of all histologic-types using the Kaplan–Meier Plotter database. (A and B) All histologic-type GC and Intestinal-type GC were significantly related to a longer OS. (C and D) Patients with diffuse-type GC and mixed-type GC had no significantly longer OS. This difference in survival rates became even less significant in the group with diffuse-type GC.

Discussion

TACC3 is presumably required during the process of cell growth and differentiation (35) but its function also seems to depend on the specific type of cell or organ (27). Oncogenic deregulation of TACC3 has been related to the progression of various human cancers (28) and a TACC3 inhibitor has been tested as a potent anticancer drug (36). In this study, therefore, we investigated the functional impact of TACC3 on GC to determine its clinicopathological relevance. Our results show that over-expression of TACC3 increases the motility and proliferation of GC cells and promotes the EMT, through the ERK/Akt/cyclin D1 signaling pathways, implying a role for this protein in GC progression. We also found an association of enhanced TACC3 expression with decreased DFS and OS, especially in GC patients with the intestinal type (37, 38).

We have reported that TACC3 activates the ERK and PI3K/Akt signaling pathways leading EMT process in cervical cancer (27). Among TACC family proteins, TACC3 is vital in the cellular processes of growth, differentiation, and gene regulation (19). Increased cancer cell proliferation, differentiation, invasion, and metastasis are key elements in carcinogenesis (39), with EMT playing a crucial role in the latter two events (40). A previous study showed a higher invasive capacity of TACC3-over-expressing cells through the support of EMT (41). A role for the snail and slug proteins in EMT during embryogenesis and also in tumor progression has been reported (42). β-catenin, another key cell regulator, has been shown to trigger numerous genes, including cyclin D1, involved in cancer initiation and progression (43, 44). ERK activation is required for EMT processes such as adherent junction loss and acquisition of mesenchymal features by tumor cells (45). ERK signaling increases snail and slug expression in lung cancer (46) while upregulation of the two proteins by β-catenin and Akt/ERK signals in cancer cells leads to downregulation of E-cadherin (47, 48). We also found elevated levels of phosphorylated Akt/ERK and activation of the cyclin D1 signaling pathways in TACC3-over-expressing AGS and SNU-638 GC cells. In contrary, decreased expression of EMT markers was found in TACC3-knockdown cells. These are in accordance with the result of a previous study (28). In summary, our experimental results demonstrate that TACC3 promotes EMT and upregulates the Akt/ERK and cyclin D1 signaling pathways in GC cells.

In patients with GC, higher levels of TACC3 expression by the tumor are associated with a worse prognosis regardless of the histologic type of GC (49). However, in both our cohort and the publicly open dataset TCGA, TACC3 expression was correlated with survival benefit only in patients with intestinal-type GC. Since TACC3 is enhanced in the early stage of differentiation in various cell types and highly expressed in cells undergoing rapid growth and differentiation, the degree of GC differentiation (according to the Lauren classification) may affect survival. In addition to the differential expression of TACC3 in tumor cells vs. the surrounding normal mucosa, TACC3 expression was detected in immune cells within the tumor. Strong labeling of TACC3 in an immune cell population that included lymphocytes, polymorphonuclear leukocytes, and monocytes has been reported (38). The presence of TACC3-positive immune cells may reflect the presence of tumor-infiltrating lymphocytes or a functional role for TACC3 in the immune cell population, but this remains to be determined in further studies. Clinically, high TACC3-positive immune cell counts have been related to a prolonged DFS in patients with intestinal-type and diffuse-type GCs, and in the former, it was shown to be an independent predictor of DFS. Whether the role of TACC3 in molecular signaling pathways differs according to the Lauren type of GC and within the immune cell population also requires further research.

High levels of TACC3 protein have been reported in liver (22), non-small-cell lung (21), colon (50), and cervical cancer (27). The upregulation of TACC3 gene expression during the advance of breast cancer from in situ carcinoma to invasive type implies a role for TACC3 in tumor advancement and in invasive cancer malignancy phenotypes (26). The diverse manifestations of TACC3 expression point to its various roles according to different types of primary tumor while ultimately contributing to carcinogenesis and metastasis, by acting on mitosis and oncogenic signaling and promoting genomic instability (19).

The results of our study imply that TACC3 participates in oncogenic functions in GC cells, including EMT and ERK/Akt/cyclin D1 signaling pathways. Higher expression of TACC3 may be linked to a poor prognosis in patients with intestinal-type—but not diffuse-type—GC. Consequently, TACC3 may be a therapeutic target in GC, but efficacy may depend on the histologic subtype of the tumor.

Acknowledgements

This study was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIP) (2016R1A5A2945889 & 2018 R1D1A1B07050274 for CKK, 2020R1A5A2031185 & 2019R1A2B5B01070598 for LKH), and the Chonnam National University Hwasun Hospital Biomedical Research Institute (HCRI19031 for KSM & KHL).

Footnotes

  • ↵* These Authors contributed equally to this work.

  • Authors’ Contributions

    M.R.A. investigation, writing the original draft; J.S.P. methodology, validation; M.G.N. software, visualization, data curation. G.H.H. formal analysis, investigation. Y.S.P. resources, project administration; J.H.L. supervision, project administration; K.S.M. conceptualization, writing - review & editing; C.K.K. funding acquisition, writing - review & editing; K.H.L. supervision, funding acquisition; writing - review & editing.

  • Conflicts of Interest

    All Authors have no conflicts of interest to disclose in relation to this study.

  • Received May 27, 2021.
  • Revision received June 14, 2021.
  • Accepted June 15, 2021.
  • Copyright © 2021 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

References

  1. ↵
    1. Torre LA,
    2. Bray F,
    3. Siegel RL,
    4. Ferlay J,
    5. Lortet-Tieulent J and
    6. Jemal A
    : Global cancer statistics, 2012. CA Cancer J Clin 65(2): 87-108, 2015. PMID: 25651787. DOI: 10.3322/caac.21262
    OpenUrlCrossRefPubMed
  2. ↵
    1. Wadhwa R,
    2. Song S,
    3. Lee JS,
    4. Yao Y,
    5. Wei Q and
    6. Ajani JA
    : Gastric cancer-molecular and clinical dimensions. Nat Rev Clin Oncol 10(11): 643-655, 2013. PMID: 24061039. DOI: 10.1038/nrclinonc.2013.170
    OpenUrlCrossRefPubMed
  3. ↵
    1. Aoyama T and
    2. Yoshikawa T
    : Adjuvant therapy for locally advanced gastric cancer. Surg Today 47(11): 1295-1302, 2017. PMID: 28251375. DOI: 10.1007/s00595-017-1493-y
    OpenUrlCrossRefPubMed
  4. ↵
    1. Brenner H,
    2. Rothenbacher D and
    3. Arndt V
    : Epidemiology of stomach cancer. Methods Mol Biol 472: 467-477, 2009. PMID: 19107449. DOI: 10.1007/978-1-60327-492-0_23
    OpenUrlCrossRefPubMed
  5. ↵
    1. Voon DC,
    2. Huang RY,
    3. Jackson RA and
    4. Thiery JP
    : The EMT spectrum and therapeutic opportunities. Mol Oncol 11(7): 878-891, 2017. PMID: 28544151. DOI: 10.1002/1878-0261.12082
    OpenUrlCrossRefPubMed
  6. ↵
    1. Huang L,
    2. Wu RL and
    3. Xu AM
    : Epithelial-mesenchymal transition in gastric cancer. Am J Transl Res 7(11): 2141-2158, 2015. PMID: 26807164.
    OpenUrlPubMed
  7. ↵
    1. Brabletz T,
    2. Kalluri R,
    3. Nieto MA and
    4. Weinberg RA
    : EMT in cancer. Nat Rev Cancer 18(2): 128-134, 2018. PMID: 29326430. DOI: 10.1038/nrc.2017.118
    OpenUrlCrossRefPubMed
  8. ↵
    1. Lamouille S,
    2. Xu J and
    3. Derynck R
    : Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15(3): 178-196, 2014. PMID: 24556840. DOI: 10.1038/nrm3758
    OpenUrlCrossRefPubMed
    1. Tanos B and
    2. Rodriguez-Boulan E
    : The epithelial polarity program: machineries involved and their hijacking by cancer. Oncogene 27(55): 6939-6957, 2008. PMID: 19029936. DOI: 10.1038/onc.2008.345
    OpenUrlCrossRefPubMed
  9. ↵
    1. Huber MA,
    2. Kraut N and
    3. Beug H
    : Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol 17(5): 548-558, 2005. PMID: 16098727. DOI: 10.1016/j.ceb.2005.08.001
    OpenUrlCrossRefPubMed
  10. ↵
    1. Kang Y and
    2. Massagué J
    : Epithelial-mesenchymal transitions: twist in development and metastasis. Cell 118(3): 277-279, 2004. PMID: 15294153. DOI: 10.1016/j.cell.2004.07.011
    OpenUrlCrossRefPubMed
  11. ↵
    1. Rosivatz E,
    2. Becker I,
    3. Specht K,
    4. Fricke E,
    5. Luber B,
    6. Busch R,
    7. Höfler H and
    8. Becker KF
    : Differential expression of the epithelial-mesenchymal transition regulators snail, SIP1, and twist in gastric cancer. Am J Pathol 161(5): 1881-1891, 2002. PMID: 12414534. DOI: 10.1016/S0002-9440(10)64464-1
    OpenUrlCrossRefPubMed
  12. ↵
    1. Shin S,
    2. Buel GR,
    3. Nagiec MJ,
    4. Han MJ,
    5. Roux PP,
    6. Blenis J and
    7. Yoon SO
    : ERK2 regulates epithelial-to-mesenchymal plasticity through DOCK10-dependent Rac1/FoxO1 activation. Proc Natl Acad Sci USA 116(8): 2967-2976, 2019. PMID: 30728292. DOI: 10.1073/pnas.1811923116
    OpenUrlAbstract/FREE Full Text
    1. Xu W,
    2. Yang Z and
    3. Lu N
    : A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adh Migr 9(4): 317-324, 2015. PMID: 26241004. DOI: 10.1080/19336918.2015.1016686
    OpenUrlCrossRefPubMed
  13. ↵
    1. Jiang YG,
    2. Luo Y,
    3. He DL,
    4. Li X,
    5. Zhang LL,
    6. Peng T,
    7. Li MC and
    8. Lin YH
    : Role of Wnt/beta-catenin signaling pathway in epithelial-mesenchymal transition of human prostate cancer induced by hypoxia-inducible factor-1alpha. Int J Urol 14(11): 1034-1039, 2007. PMID: 17956532. DOI: 10.1111/j.1442-2042.2007.01866.x
    OpenUrlCrossRefPubMed
  14. ↵
    1. Thakur HC,
    2. Singh M,
    3. Nagel-Steger L,
    4. Prumbaum D,
    5. Fansa EK,
    6. Gremer L,
    7. Ezzahoini H,
    8. Abts A,
    9. Schmitt L,
    10. Raunser S,
    11. Ahmadian MR and
    12. Piekorz RP
    : Role of centrosomal adaptor proteins of the TACC family in the regulation of microtubule dynamics during mitotic cell division. Biol Chem 394(11): 1411-1423, 2013. PMID: 23787465. DOI: 10.1515/hsz-2013-0184
    OpenUrlCrossRefPubMed
  15. ↵
    1. Kiemeney LA,
    2. Sulem P,
    3. Besenbacher S,
    4. Vermeulen SH,
    5. Sigurdsson A,
    6. Thorleifsson G,
    7. Gudbjartsson DF,
    8. Stacey SN,
    9. Gudmundsson J,
    10. Zanon C,
    11. Kostic J,
    12. Masson G,
    13. Bjarnason H,
    14. Palsson ST,
    15. Skarphedinsson OB,
    16. Gudjonsson SA,
    17. Witjes JA,
    18. Grotenhuis AJ,
    19. Verhaegh GW,
    20. Bishop DT,
    21. Sak SC,
    22. Choudhury A,
    23. Elliott F,
    24. Barrett JH,
    25. Hurst CD,
    26. de Verdier PJ,
    27. Ryk C,
    28. Rudnai P,
    29. Gurzau E,
    30. Koppova K,
    31. Vineis P,
    32. Polidoro S,
    33. Guarrera S,
    34. Sacerdote C,
    35. Campagna M,
    36. Placidi D,
    37. Arici C,
    38. Zeegers MP,
    39. Kellen E,
    40. Gutierrez BS,
    41. Sanz-Velez JI,
    42. Sanchez-Zalabardo M,
    43. Valdivia G,
    44. Garcia-Prats MD,
    45. Hengstler JG,
    46. Blaszkewicz M,
    47. Dietrich H,
    48. Ophoff RA,
    49. van den Berg LH,
    50. Alexiusdottir K,
    51. Kristjansson K,
    52. Geirsson G,
    53. Nikulasson S,
    54. Petursdottir V,
    55. Kong A,
    56. Thorgeirsson T,
    57. Mungan NA,
    58. Lindblom A,
    59. van Es MA,
    60. Porru S,
    61. Buntinx F,
    62. Golka K,
    63. Mayordomo JI,
    64. Kumar R,
    65. Matullo G,
    66. Steineck G,
    67. Kiltie AE,
    68. Aben KK,
    69. Jonsson E,
    70. Thorsteinsdottir U,
    71. Knowles MA,
    72. Rafnar T and
    73. Stefansson K
    : A sequence variant at 4p16.3 confers susceptibility to urinary bladder cancer. Nat Genet 42(5): 415-419, 2010. PMID: 20348956. DOI: 10.1038/ng.558
    OpenUrlCrossRefPubMed
  16. ↵
    1. Mortuza GB,
    2. Cavazza T,
    3. Garcia-Mayoral MF,
    4. Hermida D,
    5. Peset I,
    6. Pedrero JG,
    7. Merino N,
    8. Blanco FJ,
    9. Lyngsø J,
    10. Bruix M,
    11. Pedersen JS,
    12. Vernos I and
    13. Montoya G
    : XTACC3-XMAP215 association reveals an asymmetric interaction promoting microtubule elongation. Nat Commun 5: 5072, 2014. PMID: 25262927. DOI: 10.1038/ncomms6072
    OpenUrlCrossRefPubMed
  17. ↵
    1. Ha GH,
    2. Kim JL and
    3. Breuer EK
    : Transforming acidic coiled-coil proteins (TACCs) in human cancer. Cancer Lett 336(1): 24-33, 2013. PMID: 23624299. DOI: 10.1016/j.canlet.2013.04.022
    OpenUrlCrossRefPubMed
  18. ↵
    1. Matsuda K,
    2. Miyoshi H,
    3. Hiraoka K,
    4. Hamada T,
    5. Nakashima K,
    6. Shiba N and
    7. Ohshima K
    : Elevated expression of transforming acidic coiled-coil containing protein 3 (TACC3) is associated with a poor prognosis in osteosarcoma. Clin Orthop Relat Res 476(9): 1848-1855, 2018. PMID: 30024460. DOI: 10.1097/CORR.0000000000000379
    OpenUrlCrossRefPubMed
  19. ↵
    1. Jung CK,
    2. Jung JH,
    3. Park GS,
    4. Lee A,
    5. Kang CS and
    6. Lee KY
    : Expression of transforming acidic coiled-coil containing protein 3 is a novel independent prognostic marker in non-small cell lung cancer. Pathol Int 56(9): 503-509, 2006. PMID: 16930330. DOI: 10.1111/j.1440-1827.2006.01998.x
    OpenUrlCrossRefPubMed
  20. ↵
    1. Zhou DS,
    2. Wang HB,
    3. Zhou ZG,
    4. Zhang YJ,
    5. Zhong Q,
    6. Xu L,
    7. Huang YH,
    8. Yeung SC,
    9. Chen MS and
    10. Zeng MS
    : TACC3 promotes stemness and is a potential therapeutic target in hepatocellular carcinoma. Oncotarget 6(27): 24163-24177, 2015. PMID: 26219398. DOI: 10.18632/oncotarget.4643
    OpenUrlCrossRefPubMed
  21. ↵
    1. Lin ZR,
    2. Wang MY,
    3. He SY,
    4. Cai ZM and
    5. Huang WR
    : TACC3 transcriptionally upregulates E2F1 to promote cell growth and confer sensitivity to cisplatin in bladder cancer. Cell Death Dis 9(2): 72, 2018. PMID: 29358577. DOI: 10.1038/s41419-017-0112-6
    OpenUrlCrossRefPubMed
  22. ↵
    1. Guo F and
    2. Liu Y
    : Knockdown of TACC3 inhibits the proliferation and invasion of human renal cell carcinoma cells. Oncol Res 26(2): 183-189, 2018. PMID: 28109075. DOI: 10.3727/096504017X14837020772250
    OpenUrlCrossRefPubMed
  23. ↵
    1. L’Espérance S,
    2. Popa I,
    3. Bachvarova M,
    4. Plante M,
    5. Patten N,
    6. Wu L,
    7. Têtu B and
    8. Bachvarov D
    : Gene expression profiling of paired ovarian tumors obtained prior to and following adjuvant chemotherapy: molecular signatures of chemoresistant tumors. Int J Oncol 29(1): 5-24, 2006. PMID: 16773180.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Ma XJ,
    2. Salunga R,
    3. Tuggle JT,
    4. Gaudet J,
    5. Enright E,
    6. McQuary P,
    7. Payette T,
    8. Pistone M,
    9. Stecker K,
    10. Zhang BM,
    11. Zhou YX,
    12. Varnholt H,
    13. Smith B,
    14. Gadd M,
    15. Chatfield E,
    16. Kessler J,
    17. Baer TM,
    18. Erlander MG and
    19. Sgroi DC
    : Gene expression profiles of human breast cancer progression. Proc Natl Acad Sci U S A 100(10): 5974-5979, 2003. PMID: 12714683. DOI: 10.1073/pnas.0931261100
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Ha GH,
    2. Kim JL and
    3. Breuer EK
    : TACC3 is essential for EGF-mediated EMT in cervical cancer. PLoS One 8(8): e70353, 2013. PMID: 23936413. DOI: 10.1371/journal.pone.0070353
    OpenUrlCrossRefPubMed
  26. ↵
    1. Ha GH,
    2. Park JS and
    3. Breuer EK
    : TACC3 promotes epithelial-mesenchymal transition (EMT) through the activation of PI3K/Akt and ERK signaling pathways. Cancer Lett 332(1): 63-73, 2013. PMID: 23348690. DOI: 10.1016/j.canlet.2013.01.013
    OpenUrlCrossRefPubMed
  27. ↵
    1. Bang YJ,
    2. Kim YW,
    3. Yang HK,
    4. Chung HC,
    5. Park YK,
    6. Lee KH,
    7. Lee KW,
    8. Kim YH,
    9. Noh SI,
    10. Cho JY,
    11. Mok YJ,
    12. Kim YH,
    13. Ji J,
    14. Yeh TS,
    15. Button P,
    16. Sirzén F,
    17. Noh SH and CLASSIC trial investigators
    : Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet 379(9813): 315-321, 2012. PMID: 22226517. DOI: 10.1016/S0140-6736(11)61873-4
    OpenUrlCrossRefPubMed
  28. ↵
    1. Choi YY,
    2. Kim H,
    3. Shin SJ,
    4. Kim HY,
    5. Lee J,
    6. Yang HK,
    7. Kim WH,
    8. Kim YW,
    9. Kook MC,
    10. Park YK,
    11. Kim HH,
    12. Lee HS,
    13. Lee KH,
    14. Gu MJ,
    15. Choi SH,
    16. Hong S,
    17. Kim JW,
    18. Hyung WJ,
    19. Noh SH and
    20. Cheong JH
    : Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: Post hoc analysis of the CLASSIC randomized controlled study. Ann Surg 270(2): 309-316, 2019. PMID: 29727332. DOI: 10.1097/SLA.0000000000002803
    OpenUrlCrossRefPubMed
  29. ↵
    1. Kho DH,
    2. Bae JA,
    3. Lee JH,
    4. Cho HJ,
    5. Cho SH,
    6. Lee JH,
    7. Seo YW,
    8. Ahn KY,
    9. Chung IJ and
    10. Kim KK
    : KITENIN recruits Dishevelled/PKC delta to form a functional complex and controls the migration and invasiveness of colorectal cancer cells. Gut 58(4): 509-519, 2009. PMID: 18653728. DOI: 10.1136/gut.2008.150938
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Lokuhetty D,
    2. White VA,
    3. Watanabe R and
    4. Cree IA
    1. Carneiro F,
    2. Fukayama M,
    3. Grabsch HI and
    4. Yasui W
    : Gastric adenocarcinoma. In: Digestive system tumours who classification of tumours. 5th ed. Lokuhetty D, White VA, Watanabe R and Cree IA (eds.). IARC Press, Lyon, pp. 85-95, 2019.
  31. ↵
    1. Cerami E,
    2. Gao J,
    3. Dogrusoz U,
    4. Gross BE,
    5. Sumer SO,
    6. Aksoy BA,
    7. Jacobsen A,
    8. Byrne CJ,
    9. Heuer ML,
    10. Larsson E,
    11. Antipin Y,
    12. Reva B,
    13. Goldberg AP,
    14. Sander C and
    15. Schultz N
    : The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2(5): 401-404, 2012. PMID: 22588877. DOI: 10.1158/2159-8290.CD-12-0095
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Gao J,
    2. Aksoy BA,
    3. Dogrusoz U,
    4. Dresdner G,
    5. Gross B,
    6. Sumer SO,
    7. Sun Y,
    8. Jacobsen A,
    9. Sinha R,
    10. Larsson E,
    11. Cerami E,
    12. Sander C and
    13. Schultz N
    : Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6(269): pl1, 2013. PMID: 23550210. DOI: 10.1126/scisignal.2004088
    OpenUrlAbstract/FREE Full Text
  33. ↵
    1. Gangisetty O,
    2. Lauffart B,
    3. Sondarva GV,
    4. Chelsea DM and
    5. Still IH
    : The transforming acidic coiled coil proteins interact with nuclear histone acetyltransferases. Oncogene 23(14): 2559-2563, 2004. PMID: 14767476. DOI: 10.1038/sj.onc.1207424
    OpenUrlCrossRefPubMed
  34. ↵
    1. Akbulut O,
    2. Lengerli D,
    3. Saatci O,
    4. Duman E,
    5. Seker UOS,
    6. Isik A,
    7. Akyol A,
    8. Caliskan B,
    9. Banoglu E and
    10. Sahin O
    : A highly potent TACC3 inhibitor as a novel anticancer drug candidate. Mol Cancer Ther 19(6): 1243-1254, 2020. PMID: 32217742. DOI: 10.1158/1535-7163.MCT-19-0957
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Sadek CM,
    2. Jalaguier S,
    3. Feeney EP,
    4. Aitola M,
    5. Damdimopoulos AE,
    6. Pelto-Huikko M and
    7. Gustafsson JA
    : Isolation and characterization of AINT: a novel ARNT interacting protein expressed during murine embryonic development. Mech Dev 97(1-2): 13-26, 2000. PMID: 11025203. DOI: 10.1016/s0925-4773(00)00415-9
    OpenUrlCrossRefPubMed
  36. ↵
    1. Sadek CM,
    2. Pelto-Huikko M,
    3. Tujague M,
    4. Steffensen KR,
    5. Wennerholm M and
    6. Gustafsson JA
    : TACC3 expression is tightly regulated during early differentiation. Gene Expr Patterns 3(2): 203-211, 2003. PMID: 12711550. DOI: 10.1016/s1567-133x(02)00066-2
    OpenUrlCrossRefPubMed
  37. ↵
    1. Gupta SC,
    2. Kim JH,
    3. Prasad S and
    4. Aggarwal BB
    : Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev 29(3): 405-434, 2010. PMID: 20737283. DOI: 10.1007/s10555-010-9235-2
    OpenUrlCrossRefPubMed
  38. ↵
    1. Chen HT,
    2. Liu H,
    3. Mao MJ,
    4. Tan Y,
    5. Mo XQ,
    6. Meng XJ,
    7. Cao MT,
    8. Zhong CY,
    9. Liu Y,
    10. Shan H and
    11. Jiang GM
    : Crosstalk between autophagy and epithelial-mesenchymal transition and its application in cancer therapy. Mol Cancer 18(1): 101, 2019. PMID: 31126310. DOI: 10.1186/s12943-019-1030-2
    OpenUrlCrossRefPubMed
  39. ↵
    1. Son H and
    2. Moon A
    : Epithelial-mesenchymal transition and cell invasion. Toxicol Res 26(4): 245-252, 2010. PMID: 24278531. DOI: 10.5487/TR.2010.26.4.245
    OpenUrlCrossRefPubMed
  40. ↵
    1. Nieto MA
    : The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3(3): 155-166, 2002. PMID: 11994736. DOI: 10.1038/nrm757
    OpenUrlCrossRefPubMed
  41. ↵
    1. Lecarpentier Y,
    2. Schussler O,
    3. Hébert JL and
    4. Vallée A
    : Multiple targets of the canonical WNT/β-catenin signaling in cancers. Front Oncol 9: 1248, 2019. PMID: 31803621. DOI: 10.3389/fonc.2019.01248
    OpenUrlCrossRefPubMed
  42. ↵
    1. Diehl JA
    : Cycling to cancer with cyclin D1. Cancer Biol Ther 1(3): 226-231, 2002. PMID: 12432268. DOI: 10.4161/cbt.72
    OpenUrlCrossRefPubMed
  43. ↵
    1. Santamaria PG and
    2. Nebreda AR
    : Deconstructing ERK signaling in tumorigenesis. Mol Cell 38(1): 3-5, 2010. PMID: 20385084. DOI: 10.1016/j.molcel.2010.03.012
    OpenUrlCrossRefPubMed
  44. ↵
    1. Shih JY and
    2. Yang PC
    : The EMT regulator slug and lung carcinogenesis. Carcinogenesis 32(9): 1299-1304, 2011. PMID: 21665887. DOI: 10.1093/carcin/bgr110
    OpenUrlCrossRefPubMed
  45. ↵
    1. Hong KO,
    2. Kim JH,
    3. Hong JS,
    4. Yoon HJ,
    5. Lee JI,
    6. Hong SP and
    7. Hong SD
    : Inhibition of Akt activity induces the mesenchymal-to-epithelial reverting transition with restoring E-cadherin expression in KB and KOSCC-25B oral squamous cell carcinoma cells. J Exp Clin Cancer Res 28: 28, 2009. PMID: 19243631. DOI: 10.1186/1756-9966-28-28
    OpenUrlCrossRefPubMed
  46. ↵
    1. Conacci-Sorrell M,
    2. Simcha I,
    3. Ben-Yedidia T,
    4. Blechman J,
    5. Savagner P and
    6. Ben-Ze’ev A
    : Autoregulation of E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug, and MAPK. J Cell Biol 163(4): 847-857, 2003. PMID: 14623871. DOI: 10.1083/jcb.200308162
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Yun M,
    2. Rong J,
    3. Lin ZR,
    4. He YL,
    5. Zhang JX,
    6. Peng ZW,
    7. Tang LQ,
    8. Zeng MS,
    9. Zhong Q and
    10. Ye S
    : High expression of transforming acidic coiled coil-containing protein 3 strongly correlates with aggressive characteristics and poor prognosis of gastric cancer. Oncol Rep 34(3): 1397-1405, 2015. PMID: 26133271. DOI: 10.3892/or.2015.4093
    OpenUrlCrossRefPubMed
  48. ↵
    1. Du Y,
    2. Liu L,
    3. Wang C,
    4. Kuang B,
    5. Yan S,
    6. Zhou A,
    7. Wen C,
    8. Chen J,
    9. Wu Y,
    10. Yang X,
    11. Feng G,
    12. Liu B,
    13. Iwamoto A,
    14. Zeng M,
    15. Wang J,
    16. Zhang X and
    17. Liu H
    : TACC3 promotes colorectal cancer tumourigenesis and correlates with poor prognosis. Oncotarget 7(27): 41885-41897, 2016. PMID: 27248823. DOI: 10.18632/oncotarget.9628
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 41 (7)
Anticancer Research
Vol. 41, Issue 7
July 2021
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
TACC3 Promotes Gastric Carcinogenesis by Promoting Epithelial-mesenchymal Transition Through the ERK/Akt/cyclin D1 Signaling Pathway
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
2 + 3 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
TACC3 Promotes Gastric Carcinogenesis by Promoting Epithelial-mesenchymal Transition Through the ERK/Akt/cyclin D1 Signaling Pathway
MD RASHEDUNNABI AKANDA, JEE SOO PARK, MYUNG-GIUN NOH, GEUN-HYOUNG HA, YOUNG SOOK PARK, JAE-HYUK LEE, KYUNG-SUB MOON, CHUNG KWON KIM, KYUNG-HWA LEE
Anticancer Research Jul 2021, 41 (7) 3349-3361; DOI: 10.21873/anticanres.15123

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
TACC3 Promotes Gastric Carcinogenesis by Promoting Epithelial-mesenchymal Transition Through the ERK/Akt/cyclin D1 Signaling Pathway
MD RASHEDUNNABI AKANDA, JEE SOO PARK, MYUNG-GIUN NOH, GEUN-HYOUNG HA, YOUNG SOOK PARK, JAE-HYUK LEE, KYUNG-SUB MOON, CHUNG KWON KIM, KYUNG-HWA LEE
Anticancer Research Jul 2021, 41 (7) 3349-3361; DOI: 10.21873/anticanres.15123
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Role of Platelet Interactions in Promoting Melanoma Malignancy With Insights into Proliferation, Cyclin D1 Expression, and Migration
  • Recombinant Methioninase and Cisplatinum Act Synergistically to Inhibit Lewis Lung Carcinoma Cells But Not Normal Fibroblasts
  • Light Bladder Net: Non-invasive Bladder Cancer Prediction by Weighted Deep Learning Approaches and Graphical Data Transformation
Show more Experimental Studies

Similar Articles

Keywords

  • TACC3
  • gastric cancer
  • therapeutic target
  • EMT
  • invasion
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire