Open Access

Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma

Retraction in: /10.3892/ijo.2023.5603

  • Authors:
    • Qingping Guo
    • Jiale Wang
    • Zeyu Cao
    • Yongchang Tang
    • Chao Feng
    • Feizhou Huang
  • View Affiliations

  • Published online on: June 5, 2018     https://doi.org/10.3892/ijo.2018.4431
  • Pages: 592-602
  • Copyright: © Guo 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

Despite advances in surgery and chemotherapy, the prognosis of patients with hepatocellular carcinoma (HCC) remains poor. In the present study, the role of S100A1 in the progression of HCC was investigated. Immunohistochemical staining was used to measure the expression of S100A1 in HCC tissues. S100A1 was knocked down by siRNA. A battery of experiments was used to evaluate the biology functions of S100A1. It was found that S100A1 was upregulated in HCC tissues, and its upregulation was associated with a large tumor size, low differentiation and shorter survival time. The biological experiments demonstrated that S100A1 functions as an oncogene in HCC. It was also found that S100A1 knockdown enhanced the inhibitory effects of cisplatin on HCC cells. The results showed that the downregulation of S100A1 induced the phosphorylation of yes‑associated protein (YAP), and treatment with CHX demonstrated that the downregulation of S100A1 accelerated YAP protein degradation. The downregulation of S100A1 did not alter the expression of mammalian sterile 20‑like kinase (MST)1/2 or phosphorylated MST1/2, but upregulated the phosphorylation of large tumor suppressor kinase 1 (LATS1). It was further confirmed that S100A1 interacted with LATS1. LATS1 depletion significantly reduced the effects of S100A1 on cell growth rate and apoptosis, and there was a positive correlation between phosphorylated LATS1 and S100A1 in clinical samples, indicating that LATS1 was responsible for the S100A1-induced changes in cancer cell growth and Hippo signaling. In conclusion, the results of the present study indicated that S100A1 functions as an oncogene and may be a biomarker for the prognosis of patients with HCC. S100A1 exerted its oncogenic function by interacting with LATS1 and activating YAP. S100A1 may serve as a target for novel therapies in HCC.

Introduction

Hepatocellular carcinoma (HCC) has become the leading cause of cancer-associated mortality worldwide, particularly in China (1). Despite advances in surgery and chemotherapy, the prognosis of patients with HCC remains poor (2). There is substantial interest in obtaining an improved understanding of the potential molecular targets associated with HCC.

The Hippo signaling pathway is a conserved kinase cascade in mammals and controls tissue homeostasis. Yes-associated protein (YAP) is the downstream effector of this pathway (3). Activation of the Hippo signaling pathway is a common phenomenon in the progression of HCC (4). YAP is expressed at high levels in HCC specimens and serves as an independent prognostic marker for disease-free survival and overall survival rates in patients with HCC (5). Inhibition of the activity of YAP may offer a novel therapeutic approach for patients with HCC.

S100A1 is an important member of the S100 family of Ca(2+)-binding proteins (6). Upregulation of the expression of S100A4 has been associated with the progression of various tumors (7,8). The expression of S100A1 increased with increasing Silverberg grade in serous tumors and is associated with decreased relapse-free survival (9). S100A1 also enhances the ovarian cancer cell proliferation and migration (10). However, the mechanisms underlying the role of S100A1 in HCC remain to be fully elucidated.

In the present study, it was demonstrated that S100A1 protein was upregulated in human HCC, and was associated with tumor size, differentiation and tumor-node-metastasis (TNM) stage in patients with HCC. A high expression of S100A1 was found to be an independent prognostic factor for poor outcome in patients with HCC. Its biological effects and the potential molecular mechanism were also investigated in HCC cell lines.

Materials and methods

Study population

A total of 104 HCC tissues and matched adjacent tissues were collected from The Third Xiang-Ya Hospital of Central South University (Changsha, China) between October 2008 and October 2015. The information of the patients was obtained from their medical records. The present study was approved by the Ethics Committee of the Third Xiangya Hospital of Central South University. Writ ten inform consent was obtained from all participants involved in the study.

Cell culture

The HL7702 human normal hepatocyte cell line and the MHCC97-H, HCCLM3, Hep3B, SMMC-7721 and Huh7 human HCC cell lines were obtained from the Chinese Academy of Medical Sciences (Beijing, China). The cells were cultured with Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and maintained in a humidified atmosphere of 5% CO2 in air at 37°C.

Antibodies

The following antibodies were used in the present study: GAPDH (cat. no. sc-293335) from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA); S100A1 (cat. no. #5066), P-glycoprotein (p-gp) (cat. no. 13342), multidrug-resistant (MDR) (cat. no. # 13978S), multidrug resistance-associated protein (MRP) (cat. no. 14685), mammalian sterile 20-like kinase (MST)1/2 (cat. no. 14946), phosphorylated (p-)MST1/2 (cat. no. #3681), large tumor suppressor kinase 1 (LATS1) (cat. no. 3477), p-LATS1 (cat. no. 9157), p-YAP (cat. no. 13008) and YAP (cat. no. 15028) from Cell Signaling Technology, Inc. (Danvers, MA, USA); and Lamin B1 (cat. no. ab133741), B-cell lymphoma 2 (Bcl-2) (cat. no. ab32124), Bcl-2-associated X protein (Bax) (cat. no. ab32503) and Bcl-2 antagonist/killer (Bak) (cat. no. ab32371) from Abcam (Cambridge, UK).

Cell treatment

The lentiviruses containing S100A1 small interfering (si)RNA (siRNA sequences: CCGGGTTGAGCAGACAGCCACATTGCTCGAGCAATGTGGCTGTCTGCTCAAC) or LATS1 siRNA (siRNA sequences: CCGGTAGTCAATTCTTGGTACTTAACTCGAGTTAAGTACCAAGAATTGACTA) and empty control were purchased from Genepharma Company (Beijing, China). The Hep3B and SMMC-7721 cells were transfected with these lentiviruses using Lip3000 (Thermo Fisher Scientific, Inc.) according to the manufacturer’s protocol.

To investigate the effect of S100A1 on drug sensitivity, the cells were treated with cisplatin (DDP, 1 μg/ml) for 48 h or the indicated time points (0, 24, 48 and 72 h) at 37°C. To assess the half-life of S100A1, the cells were treated with protein synthesis inhibitor cycloheximide (CHX; 10 μg/ml) for 0, 2, 4 or 6 h at 37°C.

Immunohistochemical staining

The paraffin-embedded tissue sections were serially cut at 4-μm. Slides were regularly deparaffinized and dehydrated, and retrieval was performed with citric acid buffer (pH 6.0) in a microwave oven. Following cooling, the sections were blocked in normal goat serum (cat. no. AR0009, Boster, Wuhan, China), and incubated with primary anti-S100A1 (1:200), or anti-p-LATS1 (1:100) antibody overnight at 4°C. The sections were then washed with PBS and incubated with secondary antibody (cat. no. BA1050 and BA1054, dilution, 1:3,000; Boster) for 2 h at 37°C. The sections were then washed with PBS and stained using a DAB detection kit (Maxim Biological Technology, Xiamen, China). Finally, the sections were counterstained with hematoxylin. Images were captured using a microscope (Eclipse Ni-E/Ni-U, Nikon Corp., Tokyo, Japan). The integral optical density (IOD) of positive signaling was obtained using ImageJ software (version 1.8.0_112, National Institutes of Health, Bethesda, MD, USA). The tumor samples with an IOD higher than the mean were regarded to have a high expression of S100A1.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA was extracted from cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The expression levels of S100A1, Bcl-2, Bax, Bak, p-gp, MDR and MRP was measured using a SYBR-Green qPCR assay (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturer’s protocol. The expression of β-actin was used as an endogenous control. The primer sequences are listed in Table I. The qPCR procedure [SYBR Premix (2X) 10 μl, PCR forward primer (10 μM), 0.8 μl, PCR reverse primer (10 μM), 0.8 μl, ROX reference dye (50X), 0.4 μl, cDNA, 2 μl, sterile purified water 6 μl, total volume of 20 μl] was performed under the following conditions: 95.0°C for 3 min, and 39 cycles of 95.0°C for 10 sec and 60°C for 30 sec. Data were processed using the 2-ΔΔCq method (11).

Table I

Primer sequences used in reverse transcription-quantitative polymerase chain reaction analysis.

Table I

Primer sequences used in reverse transcription-quantitative polymerase chain reaction analysis.

GeneSense (5′-3′)Antisense (5′-3′)
S100A1 GAGTATGTGGTGCTTGTGGC CTTGGACCGCTACTCTTGCG
Bcl-2 GGGAGGATTGTGGCCTTCTT ACTTGTGGCCCAGATAGGCA
Bax GTCTTTTTCCGAGTGGCAGC GGAGACAGGGACATCAGTCG
Bak GATCCCGGCAGGCTGATCC GTTCCTGCTGATGGCGGTAA
p-gp CTGGGCTTCATCACCAACAAC TCGGGGTTGATGCCGTATTC
MDR CCTGTGAAGAGTAGAACATGAAGA CGAATGAGCTCAGGCTTCCT
MRP CTAGATGACCCCCTGTCTGC ACTGCCATCATGGATCAGACTT
β-actin TTGTTACAGGAAGTCCCTTGCC ATGCTATCACCTCCCCTGTGTG

[i] Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X protein; Bak, Bcl-2 antagonist/killer; p-gp, p-glycoprotein; MDR, multidrug resistant; MRP, multidrug resistance-associated protein.

Cell viability assay

The cells in the indicated treatment groups were plated in 96-well plates at a density of 3,000 cells in 100 μl medium per well 24 h prior to the experiment. The cell viability was assessed using a CKK-8 assay (Beyotime Institute of Biotechnology, Haimen, China) at the indicated time points according to the manufacturer’s protocol.

Flow cytometry for analysis of apoptosis

Staining was performed using an Annexin V-FITC kit (KeyGen BioTech Co., Ltd., Nanjing, China) following the manufacturer’s protocol. Briefly, 2×105 Hep3B and SMMC-7721 cells were harvested by centrifugation at 1,000 g for 5 min at room temperature and resuspended in 100 μl binding buffer. The cells were incubated with 5 μl Annexin V-FITC for 15 min in the dark at 37°C and then incubated with 10 μl PI with gentle shaking for 10 min. Flow cytometric analysis (BD Biosciences, Franklin Lakes, NJ, USA) was employed for detecting apoptotic events.

Western blot analysis and immunoprecipitation

Total protein was extracted from cells using cold RIPA buffer, and the nuclear protein was extracted using an EpiQuik nuclear extraction kit (EpiGentek, Farmingdale, NY, USA) following the manufacturer’s protocol. The protein concentration was determined using the BCA Protein assay kit (cat. no. AR0197, Boster). A total of 60 μg protein was separated by 10% SDS-PAGE, which was then transferred onto a PVDF membrane (Thermo Fisher Scientific, Inc.). Subsequently, the membrane was incubated in PBS with 5% non-fat dried milk (Mengniu Dairy, Hohhot, China) for 3 h at 4°C. The membrane was then incubated with primary antibodies (GAPDH, dilution, 1:3,000; Lamin B1, dilution, 1:2,000; S100A1, dilution, 1:1,000; p-gp, dilution, 1:1,000; MDR, dilution, 1:1,000; MRP, dilution, 1:1,000; MST1/2, dilution, 1:1,000; p-MST1/2, dilution, 1:1,000; LATS1, dilution, 1:1,000; p-LATS1, dilution, 1:1,000; p-YAP, dilution, 1:1,000; YAP, dilution, 1:1,000; Bcl-2, dilution, 1:1,000; Bax, dilution, 1:1,000; Bak, dilution, 1:1,000) overnight at 4°C, followed by incubation with appropriate secondary antibody [goat anti-rabbit IgG (HRP), cat. no. ab6721, dilution, 1:3,000; or goat anti-mouse IgG (HRP), cat. no. ab205719, dilution, 1:3,000; both from Abcam] for 1 h at 37°C. The immune complexes were detected using an ECL Western Blotting kit (EMD Millipore, Billerica, MA, USA). The relative protein expression was analyzed using Image-Pro plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA), and GAPDH was used as the internal reference.

For immunoprecipitation, magnetic beads (Bio-Rad SureBeads; Bio-Rad Laboratories, Inc., Hercules, CA, USA) were incubated with antibodies (S100A1, 10 μg/ml; LATS1, 10 μg/ml) for 3 h at 4°C. The bead-antibody complex was then incubated with target protein (S100A1, 10 μg/ml; LATS1, 10 μg/ml) overnight at 4°C. The beads were magnetized using the SureBeads magnetic rack and supernatant was discarded. The elution buffer was then used for western blot analysis.

Luciferase reporter assay

For detecting the change of Hippo signaling and YAP transcription activity, a luciferase assay was performed according to the manufacturer’s protocol (Dual-Luciferase Assay kit, Promega Corporation, Madison, WI, USA). The Hep3B and SMMC-7721 cells were plated in 96-well clusters, and then cotransfected with the luciferase reporter plasmid with 100 ng pGL3-basic vector (NC) or pGL3-S100A1 siRNA. At 48 h post-transfection, luciferase activity was detected using a dual-luciferase reporter assay system and normalized to Renilla activity.

Statistical analysis

In the present study, all experiments were repeated at least three times, and data are expressed as the mean ± standard error of the mean. The SPSS 18.0 software package (SPSS, Inc., Chicago, IL, USA) was used to perform statistical analysis. Differences between two groups were compared using an independent-samples t-test. Differences among three or more groups were compared using one-way analysis of variance with the Bonferroni post hoc test. The clinical association between the expression of S100A1 and clinicopathological variables in the patients with lung cancer was evaluated using χ2 test. Kaplan-Meier analysis was used to analyze the overall survival rate of patients with HCC. Pearson’s correlation was used to analyze the correlation between p-LAST1 and S100A1 in HCC tissues. P<0.05 was considered to indicate a statistically significant difference.

Results

High expression of S100A1 is a predictor of poor prognosis in HCC

To investigate the role of S100A1 in HCC, the present study first evaluated the expression of S100A1 in HCC tissues. It was found that S100A1 was expressed in the cytoplasm and was significantly increased in HCC tissues compared with adjacent control tissues (Fig. 1A). In addition, the expression of S100A1 was higher in HCC tissues than in the matched adjacent tissues (Fig. 1B). In line with the results in the tissues, the expression of S100A1 was significantly increased in HCC cells compared with normal hepatocytes at the mRNA and protein levels (Fig. 1C and D), and the highest expression of S100A1 was found in Hep3B and SMMC-7721 cells.

The patients were divided into two groups according to the mean IOD of S100A1 and it was found that the expression of S100A1 was associated with differentiation (P=0.0027), tumor size (P=0.0065), number of tumor nodes (P=0.0165), lymph node metastasis (P=0.0246), distant metastasis (P=0.0028) and TNM stage (P=0.0018), but was not associated with age (P=0.843), sex (P=0.626) or cirrhosis (P=0.671) (Table II). In addition, the present study investigated the factors that predicate the prognosis of patients with HCC by univariate and multivariate analyses. The univariate analysis indicated that the level of S100A1 (P=0.02), in addition to differentiation (P=0.03), tumor size (P=0.01), lymph node metastasis (P=0.01), distant metastasis (P=0.02) and TNM stage (P=0.02) were significantly associated with patients prognosis (Table III). Multivariate analysis revealed that the level of S100A1 (P=0.01), differentiation (P=0.02), the tumor size (P=0.03), lymph node metastasis (P=0.02), distant metastasis (P=0.02) and TNM stage (P=0.02) were independent factors for predicating the prognosis of patients with HCC (Table IV). Furthermore, it was found that the patients with low S100A1 had higher overall survival rates, compared with patients with high S100A1 (Fig. 1E).

Table II

Clinical association between the expression of S100A1 and clinicopathological variables in patients with hepatocellular carcinoma.

Table II

Clinical association between the expression of S100A1 and clinicopathological variables in patients with hepatocellular carcinoma.

VariableS100A1
Low expression
(n=46)
High expression
(n=58)
χ2 test P-value
Age (years)
 <5018240.843
 ≥502834
Sex
 Male38450.626
 Female813
Differentiation
 High1880.0027
 Moderate1618
 Low1232
Tumor size
 <5 cm29210.0065
 ≥5 cm1737
Tumor nodes
 Single25180.0165
 Multiple2140
Lymph node metastasis
 N0–126200.0246
 N2–32038
Distant metastasis
 No31220.0028
 Yes1536
TNM stage
 I–II30200.0018
 III–IV1638
Cirrhosis
 Yes35420.671
 No1116

[i] TNM, tumor-node-metastasis.

Table III

Univariate analysis of prognostic factors of hepatocellular carcinoma.

Table III

Univariate analysis of prognostic factors of hepatocellular carcinoma.

VariableHazard ratioP-value
Age (≥50/<50 years)1.160.65
Sex (male/female)1.080.92
Tumor size (≥5 cm/<5 cm)2.340.01
Differentiation (high-moderate/low)3.180.03
Lymph node metastasis (N0–1/N2–3)2.460.01
Distant metastasis (yes/no)3.840.02
TNM stage (III–IV/I–II)2.560.02
S100A1 expression (high/low)3.070.02

[i] TNM, tumor-node-metastasis.

Table IV

Multivariate analysis of independent prognostic factors of hepatic carcinoma.

Table IV

Multivariate analysis of independent prognostic factors of hepatic carcinoma.

VariableHazard ratioP-value
Tumor size2.240.03
Differentiation (high-moderate/low)3.130.02
Lymph node metastasis2.170.03
Distant metastasis3.890.02
TNM stage2.320.02
S100A1 expression3.350.01

[i] TNM, tumor-node-metastasis.

Knockdown of S100A1 by siRNA inhibits HCC cell growth and sensitizes HCC cells to DDP

The expression of S100A1 was knocked down by siRNA in Hep3B and SMMC7721 cells (Fig. 2A and B), and it was found that the knockdown of S100A1 significantly inhibited cell viability (Fig. 2C), and induced apoptosis (Fig. 2D) in the Hep3B and SMMC7721 cells. In addition, the mRNA levels of apoptosis markers Bcl-2, Bax and Bak were measured. The knockdown of S100A1 significantly reduced the expression of Bcl-2 and increased the expression of Bax and Bak (Fig. 2E). These results revealed that S100A1 functions as an oncogene in liver cancer.

The present study further investigated whether knockdown of the expression of S100A1 sensitizes HCC cells to DDP. It was observed that the knockdown of S100A1 significantly reduced cell viability, compared with negative control, in the Hep3B and SMMC7721 cells treated with DDP (Fig. 3A). It was also found that the knockdown of S100A1 enhanced the inhibitory effects of DDP on the promotion of cell apoptosis, indicated by the significant increase in the apoptotic rate compared with the negative control (Fig. 3B). In addition, the knockdown of S100A1 decreased the expression of drug resistance-related genes, including p-gp, MRP and MDR (Fig. 3C–E). Therefore, the knockdown of S100A1 exhibited a synergistic inhibitory effect with DDP on HCC cell growth.

S100A1 regulates Hippo signaling through its interaction with LATS1

The present study also investigated the mechanism by which the downregulation of S100A1 suppresses HCC cell growth. Hippo signaling is important in the development of HCC. The present study found that the downregulation of S100A1 significantly increased the phosphorylation of LATS1 and YAP, and decreased total YAP protein, but did not alter the expression of total MST1/2 or p-MST1/2 in the Hep3B and SMMC7721 cells, compared with the control group (Fig. 4A). YAP stabilization is controlled by its nuclear/ cytoplasmic distribution and the phosphorylation of YAP. Western blot analysis was performed using nuclear/cytoplasmic fractionation, which revealed that nuclear YAP was downregulated following S100A1 siRNA treatment, whereas YAP cytoplasm localization was upregulated (Fig. 4B). To further demonstrate whether S100A1 stabilizes YAP protein, the Hep3B and SMMC7721 cells were treated with the protein synthesis inhibitor CHX. Following CHX treatment for 6 h, the knockdown of S100A1 by siRNA significantly downregulated total YAP protein, compared with that in the control group (Fig. 4C), indicating that the half-life of endogenous YAP decreased following siRNA treatment. As YAP acts downstream of LATS1 in Hippo signaling, the present study examined whether S100A1 regulates YAP through LAST1, as the downregulation of S100A1 significantly increased the phosphorylation of LATS1. Co-immunoprecipitation was performed to examine whether there is any interaction between S100A1 and LATS1. The results showed that S100A1 and LATS1 were co-immunoprecipitated in the Hep3B cells overexpressing S100A1 (Fig. 4D). A luciferase reporter assay was then performed to measure the activity of TEA domain (TEAD), which indicates the transcriptional activity of YAP. S100A1 knockdown inhibited the transcription of TEAD (Fig. 4E). Therefore, it was demonstrated that S100A1 stabilized the YAP protein, leading to inhibition of Hippo signaling.

Downregulation of S100A1 suppresses HCC cell growth through LATS1/YAP signaling

Due to the regulatory effect between S100A1 and LATS1, the present study further examined the biological function between S100A1 and LATS1. The Hep3B and SMMC7721 cells were transfected with S100A1 siRNA or LATS1 siRNA alone, or with both together. It was found that the downregulation of LATS1 largely attenuated the S100A1 siRNA-mediated induction of cell apoptosis (Fig. 5A), and inhibition of cell survival (Fig. 5B). In addition, compared with the control group, the knockdown of S100A1 inhibited the expression of pro-survival gene Bcl-2 and increased the expression of pro-apoptotic genes Bak and Bax (Fig. 5C). However, these inhibitory effects were reversed by LATS1 siRNA (Fig. 5C). The results indicated that S100A1 inhibited HCC cell proliferation via LATS1/YAP signaling.

To further validate the association between S100A1 and Hippo signaling in HCC tissues. The present study examined the association between the protein expression of S100A1 and p-LATS in HCC tissues using immunohistochemistry. S100A1 staining was weak positive in normal liver tissues, whereas p-LATS1 showed strong positive cytoplasmic staining. S100A1 showed marked staining in cancer tissues, whereas p-LATS1 showed weak staining in cancer tissues (Fig. 6A). The correlation analysis showed that S100A1 was negatively correlated with p-LATS1 (Fig. 6B).

Discussion

In the present study, it was found that S100A1 was upregulated in HCC tissues, and its upregulation was associated with large tumor size, low differentiation, and shorter survival rates. The biological experiments demonstrated that S100A1 functions as an oncogene in HCC. It was also found that S100A1 knockdown enhanced the inhibitory effects of DDP in HCC cells.

The S100 protein family consists of 25 relatively small calcium-binding proteins. These proteins have been reported to be involved in tumorigenesis by interacting with enzymes, cytoskeletal proteins, receptors, transcription factors, and nucleic acids (12,13). The interaction between these members is also critical in tumor development. S100A1 interacts with S100A4, and modulates the metastasis-inducing capability of S100A4 (14,15). They have been identified as potential biomarkers for various tumor types, including lung cancer and ovarian cancer (16). Under physiological conditions, the expression of S100A1 is low in kidney, lungs, ovaries and liver (1719). However, S100A1 is expressed at high levels under pathological conditions, including tumorigenesis. Sviatoha et al found that the expression of S100A1 was low in benign melanocytic tumors and increased in malignant melanomas (20). The expression of S100A1 was also increased with increasing Silverberg grade in serous tumors, and its upregulation predicted a decreased relapse-free survival rate in the endometrioid subtype of ovarian and endometrial cancer (9). S100A1 was found to be differentially expressed in clear cell renal cell carcinoma and papillary renal cell carcinoma, and the differential expression may be a potentially useful marker to differentiate the chromophobe renal cell carcinoma from renal oncocytoma (7,8). S100A1 can also be used to differentiate other tumors (2125).

S100A1 not only functions as a biomarker, but also an oncogene. Tian et al found that the overexpression of S100A1 enhanced ovarian cancer cell proliferation and migration (10). In the present study, it was also shown that S100A1 knockdown inhibited HCC cell growth and induced apoptosis. It was also found that the knockdown of S100A1 exhibited a synergistic inhibitory effect with DDP on HCC cell growth. The modulation of calcium signaling has been demonstrated to alter the sensitivity of chemotherapeutic agents to apoptotic signals (26). S100A4 is overexpressed in several methotrexate-resistant cells. The overexpression of S100A4 decreases the sensitivity of HT29 colon cancer human cells to methotrexate, whereas its knockdown causes chemosensitization towards methotrexate (27). Considering the interaction of S100A1 with S100A4 and their regulatory association (13,14,28,29), it is likely that S100A1 sensitizes HCC cells to DDP through S100A4.

The present study also investigated the mechanism by which S100A1 promotes HCC cell growth. It was demonstrated that S100A1 inactivated the Hippo signaling pathway. YAP, the downstream effector of Hippo signaling, is frequently overexpressed in various types of human cancer (30). The present study found that the depletion of S100A1 reduced total and nuclear YAP protein, and decreased TEAD luciferase reporter activity, serving as a marker of YAP downstream function (31). The functions of YAP were determined by its phosphorylation status, with p-YAP remaining in the cytoplasm for degradation, and dephosphorylated YAP translocating into the nucleus and binding to TEAD proteins to promote proliferation by activating downstream targets (32). The results of the present study showed that the downregulation of S100A1 induced YAP phosphorylation, and CHX treatment demonstrated that the downregulation of S100A1 accelerated YAP protein degradation. MST1/2 and LATS1 are the upstream regulators of YAP (33). The findings of the present study showed that the downregulation of S100A1 did not alter the expression of MST1/2 or p-MST1/2, but upregulated the phosphorylation of LATS1. It was further confirmed that S100A1 interacted with LATS1. LATS1 depletion significantly reduced the effects of S100A1 on cell growth rate and apoptosis, and there was a positive correlation between p-LATS1 and S100A1 in clinical samples, supporting the hypothesis that LATS1 is responsible for S100A1-induced changes in cancer cell growth and Hippo signaling. A previous study demonstrated that S100A7 repressed the expression of YAP and inhibited the expression of ΔNp63, which directly binds to the region of the YAP promoter and induces its expression, inhibiting the Hippo pathway and enhancing YAP activity. S100A7 also enhances the resistance of squamous cell carcinoma cancer cells by inhibiting the expression and activity of YAP (34). Taken together, the findings of the present study demonstrate that S100A1 regulated the aggressiveness of HCC mainly through the YAP and Hippo signaling pathways. Therefore S100A1 is a novel upstream regulator of the Hippo signaling pathway.

In conclusion, the present study supports the hypothesis that S100A1 functions as an oncogene and may be a biomarker for the prognosis of patients with HCC. S100A1 exerted its oncogenic function by interacting with LATS1 and activating YAP. S100A1 may serve as a target for novel therapies in HCC.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors’ contributions

QG and FH designed the study. QG, JW and ZC analyzed and interpreted the patient data. YT, CF and QG performed cell biological experiments. JW and ZC performed qPCR analysis, western blot analysis, luciferase reporter assay and IHC staining. All authors contributed to the writing of the manuscript. All authors have read and approved the final manuscript.

Ethical approval and consent to participate

The present study was approved by the Ethics Committee of the Third Xiangya Hospital of Central South University. Writ ten inform consent was obtained from all participants involved in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Costentin CE, Ferrone CR, Arellano RS, Ganguli S, Hong TS and Zhu AX: Hepatocellular carcinoma with macrovascular invasion: Defining the optimal treatment strategy. Liver Cancer. 6:360–374. 2017. View Article : Google Scholar

2 

Liu S, Miao R, Zhai M, Pang Q, Deng Y, Liu S, Qu K, Liu C and Zhang J: Effects and related mechanisms of serotonin on malignant biological behavior of hepatocellular carcinoma via regulation of Yap. Oncotarget. 8:47412–47424. 2017.

3 

Kim W, Khan SK, Liu Y, Xu R, Park O, He Y, Cha B, Gao B and Yang Y: Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma. Gut gutjnl-2017-314061. 2017.

4 

Luo X, Liu Y, Feng W, Lei L, Du Y, Wu J and Wang S: NUP37, a positive regulator of YAP/TEAD signaling, promotes the progression of hepatocellular carcinoma. Oncotarget. 8:98004–98013. 2017. View Article : Google Scholar

5 

Valero V III, Pawlik TM and Anders RA: Emerging role of Hpo signaling and YAP in hepatocellular carcinoma. J Hepatocell Carcinoma. 2:69–78. 2015.

6 

Wright NT, Cannon BR, Zimmer DB and Weber DJ: S100A1: Structure, function, and therapeutic potential. Curr Chem Biol. 3:138–145. 2009.

7 

Rocca PC, Brunelli M, Gobbo S, Eccher A, Bragantini E, Mina MM, Ficarra V, Zattoni F, Zamò A and Pea M: Diagnostic utility of S100A1 expression in renal cell neoplasms: An immu-nohistochemical and quantitative RT-PCR study. Mod Pathol. 20:722–728. 2007. View Article : Google Scholar

8 

Li G, Barthelemy A, Feng G, Gentil-Perret A, Peoc’h M, Genin C and Tostain J: S100A1: A powerful marker to differentiate chromophobe renal cell carcinoma from renal oncocytoma. Histopathology. 50:642–647. 2007. View Article : Google Scholar

9 

DeRycke MS, Andersen JD, Harrington KM, Pambuccian SE, Kalloger SE, Boylan KL, Argenta PA and Skubitz AP: S100A1 expression in ovarian and endometrial endometrioid carcinomas is a prognostic indicator of relapse-free survival. Am J Clin Pathol. 132:846–856. 2009. View Article : Google Scholar

10 

Tian T, Li X, Hua Z, Ma J, Liu Z, Chen H and Cui Z: S100A1 promotes cell proliferation and migration and is associated with lymph node metastasis in ovarian cancer. Discov Med. 23:235–245. 2017.

11 

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

12 

Wang T, Huo X, Chong Z, Khan H, Liu R and Wang T: A review of S100 protein family in lung cancer. Clin Chim Acta. 476:54–59. 2018. View Article : Google Scholar

13 

Tong L, Lan W, Lim RR and Chaurasia SS: S100A proteins as molecular targets in the ocular surface inflammatory diseases. Ocul Surf. 12:23–31. 2014. View Article : Google Scholar

14 

Wang G, Rudland PS, White MR and Barraclough R: Interaction in vivo and in vitro of the metastasis-inducing S100 protein, S100A4 (9Ka) with S100A1. J Biol Chem. 275:11141–11146. 2000. View Article : Google Scholar

15 

Wang G, Zhang S, Fernig DG, Martin-Fernandez M, Rudland PS and Barraclough R: Mutually antagonistic actions of S100A4 and S100A1 on normal and metastatic phenotypes. Oncogene. 24:1445–1454. 2005. View Article : Google Scholar

16 

Donato R, Sorci G and Giambanco I: S100A6 protein: Functional roles. Cell Mol Life Sci. 74:2749–2760. 2017. View Article : Google Scholar

17 

Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A and Edlund K: Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 13:397–406. 2014. View Article : Google Scholar

18 

Teratani T, Watanabe T, Kuwahara F, Kumagai H, Kobayashi S, Aoki U, Ishikawa A, Arai K and Nozawa R: Induced transcriptional expression of calcium-binding protein S100A1 and S100A10 genes in human renal cell carcinoma. Cancer Lett. 175:71–77. 2002. View Article : Google Scholar

19 

Li G, Gentil-Perret A, Lambert C, Genin C and Tostain J: S100A1 and KIT gene expressions in common subtypes of renal tumours. Eur J Surg Oncol. 31:299–303. 2005. View Article : Google Scholar

20 

Sviatoha V, Tani E, Kleina R, Sperga M and Skoog L: Immunohistochemical analysis of the S100A1, S100B, CD44 and Bcl-2 antigens and the rate of cell proliferation assessed by Ki-67 antibody in benign and malignant melanocytic tumours. Melanoma Res. 20:118–125. 2010. View Article : Google Scholar

21 

Cossu-Rocca P, Contini M, Brunelli M, Festa A, Pili F, Gobbo S, Eccher A, Mura A, Massarelli G and Martignoni G: S-100A1 is a reliable marker in distinguishing nephrogenic adenoma from prostatic adenocarcinoma. Am J Surg Pathol. 33:1031–1036. 2009. View Article : Google Scholar

22 

Carvalho JC, Wasco MJ, Kunju LP, Thomas DG and Shah RB: Cluster analysis of immunohistochemical profiles delineates CK7, vimentin, S100A1 and C-kit (CD117) as an optimal panel in the differential diagnosis of renal oncocytoma from its mimics. Histopathology. 58:169–179. 2011. View Article : Google Scholar

23 

Kuroda N, Kanomata N, Yamaguchi T, Imamura Y, Ohe C, Sakaida N, Hes O, Michal M, Shuin T and Lee GH: Immunohistochemical application of S100A1 in renal onco-cytoma, oncocytic papillary renal cell carcinoma, and two variants of chromophobe renal cell carcinoma. Med Mol Morphol. 44:111–115. 2011. View Article : Google Scholar

24 

Yordanov A, Ivanov I, Popovska S, Dineva T, Dimitrov T and Ivanova Z: Evaluation of the expression of S100A1 protein in serous, mucinous and endometroid ovarian carcinoma. Akush Ginekol (Sofiia). 52:22–26. 2013.In Bulgarian.

25 

Conner JR, Hirsch MS and Jo VY: HNF1β and S100A1 are useful biomarkers for distinguishing renal oncocytoma and chro-mophobe renal cell carcinoma in FNA and core needle biopsies. Cancer Cytopathol. 123:298–305. 2015. View Article : Google Scholar

26 

Hatoum D, Yagoub D, Ahadi A, Nassif NT and McGowan EM: Annexin/S100A protein family regulation through 14ARF-53 activation: A role in cell survival and predicting treatment outcomes in breast cancer. PLoS One. 12:e01699252017. View Article : Google Scholar

27 

Mencía N, Selga E, Rico I, de Almagro MC, Villalobos X, Ramirez S, Adan J, Hernández JL, Noé V and Ciudad CJ: Overexpression of S100A4 in human cancer cell lines resistant to methotrexate. BMC Cancer. 10:2502010. View Article : Google Scholar

28 

Tsuchiya M, Yamaguchi F, Shimamoto S, Fujimoto T, Tokumitsu H, Tokuda M and Kobayashi R: Oxidized S100A4 inhibits the activation of protein phosphatase 5 through S100A1 in MKN-45 gastric carcinoma cells. Int J Mol Med. 34:1713–1719. 2014. View Article : Google Scholar

29 

Tarabykina S, Kriajevska M, Scott DJ, Hill TJ, Lafitte D, Derrick PJ, Dodson GG, Lukanidin E and Bronstein I: Heterocomplex formation between metastasis-related protein S100A4 (Mts1) and S100A1 as revealed by the yeast two-hybrid system. FEBS Lett. 475:187–191. 2000. View Article : Google Scholar

30 

Watt KI, Harvey KF and Gregorevic P: Regulation of tissue growth by the mammalian Hippo signaling pathway. Front Physiol. 8:9422017. View Article : Google Scholar

31 

Lin KC, Park HW and Guan KL: Regulation of the Hippo pathway transcription factor TEAD. Trends Biochem Sci. 42:862–872. 2017. View Article : Google Scholar

32 

Hong AW, Meng Z and Guan KL: The Hippo pathway in intestinal regeneration and disease. Nat Rev Gastroenterol Hepatol. 13:324–337. 2016. View Article : Google Scholar

33 

Yimlamai D, Fowl BH and Camargo FD: Emerging evidence on the role of the Hippo/YAP pathway in liver physiology and cancer. J Hepatol. 63:1491–1501. 2015. View Article : Google Scholar

34 

Li Y, Kong F, Shao Q, Wang R, Hu E, Liu J, Jin C, He D and Xiao X: YAP expression and activity are suppressed by S100A7 via 65/NFκB-mediated repression of ΔNp63. Mol Cancer Res. 15:1752–1763. 2017. View Article : Google Scholar

Related Articles

Journal Cover

August-2018
Volume 53 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Q, Wang J, Cao Z, Tang Y, Feng C and Huang F: Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma Retraction in /10.3892/ijo.2023.5603. Int J Oncol 53: 592-602, 2018
APA
Guo, Q., Wang, J., Cao, Z., Tang, Y., Feng, C., & Huang, F. (2018). Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma Retraction in /10.3892/ijo.2023.5603. International Journal of Oncology, 53, 592-602. https://doi.org/10.3892/ijo.2018.4431
MLA
Guo, Q., Wang, J., Cao, Z., Tang, Y., Feng, C., Huang, F."Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma Retraction in /10.3892/ijo.2023.5603". International Journal of Oncology 53.2 (2018): 592-602.
Chicago
Guo, Q., Wang, J., Cao, Z., Tang, Y., Feng, C., Huang, F."Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma Retraction in /10.3892/ijo.2023.5603". International Journal of Oncology 53, no. 2 (2018): 592-602. https://doi.org/10.3892/ijo.2018.4431