Open Access

Rsf‑1 regulates malignant melanoma cell viability and chemoresistance via NF‑κB/Bcl‑2 signaling

  • Authors:
    • Jiani He
    • Lin Fu
    • Qingchang Li
  • View Affiliations

  • Published online on: August 23, 2019     https://doi.org/10.3892/mmr.2019.10610
  • Pages: 3487-3498
  • Copyright: © He 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

Remodeling and spacing factor 1 (Rsf‑1) has been reported as overexpressed in numerous cancers; however, its expression, biological functions and mechanisms in malignant melanoma remain unknown. In the present study, the expression of Rsf‑1 was investigated in 50 cases of malignant melanoma samples using immunohistochemistry. The results revealed that Rsf‑1 expression was elevated in 38% of specimens. MTT, colony formation, Transwell and flow cytometry assays were performed to investigate the functions of Rsf‑1. Knockdown of Rsf‑1 in the MV3 and A375 melanoma cell lines decreased the viability, invasion and cell cycle transition of cells. Conversely, overexpression of Rsf‑1 in M14 cells with low endogenous Rsf‑1 expression induced opposing effects. Further analysis revealed that Rsf‑1 knockdown decreased matrix metalloproteinase‑2, cyclin E and phosphorylated‑IκB expression. Additionally, Rsf‑1 depletion reduced cisplatin resistance and significantly increased the cisplatin‑associated apoptotic rate, whereas Rsf‑1 overexpression exhibited opposing effects. Rsf‑1 also maintained the mitochondrial membrane potential following cisplatin treatment. Analysis of apoptosis‑associated proteins revealed that Rsf‑1 positively regulated B‑cell lymphoma 2 (Bcl‑2), cellular inhibitor of apoptosis 1 (cIAP1) and cIAP2, and downregulated Bcl‑2‑associated X protein expression. Nuclear factor κ‑light‑chain‑enhancer of activated B‑cells (NF‑κB) inhibition reversed the effects of Rsf‑1 on Bcl‑2. In conclusion, Rsf‑1 was overexpressed in malignant melanoma and may contribute to the malignant behaviors of melanoma cells, possibly via the regulation of NF‑κB signaling. Therefore, Rsf‑1 may be a potential therapeutic target in the treatment of malignant melanoma.

Introduction

Malignant melanoma arises from melanocytes, which are responsible for pigment production (13). The incidence of melanoma has increased at an alarming rate and patients with advanced malignancies exhibit poor prognoses, with an average survival time of 3–11 months (48). Melanoma can be removed via surgical resection in patients with early diagnosis; however, melanoma has high metastatic potential and treatment options for metastatic melanoma are limited (912). Therefore, novel targets against melanoma are urgently required for the identification of effective therapies.

Remodeling and spacing factor 1 (Rsf-1), also known as hepatitis B X-antigen associated protein, is a subunit of RSF (13,14). Rsf-1 protein is located in the nucleus and binds to human sucrose nonfermenting protein 2 homolog (hSNF2H), forming a chromatin remodeling complex (15). The Rsf-1/hSNF2H complex regulates adenosine 5′-triphosphate-dependent chromatin remodeling and alters the chromatin structure of nucleosomes (15,16), which are required for biological processes, including activation or repression of transcription, DNA replication and cell cycle progression (17,18).

Rsf-1 overexpression has been reported in a number of solid tumors, including breast cancer, ovarian cancer and oral squamous cell carcinoma (1925); increased Rsf-1 expression was associated with poor prognosis in bladder cancer (15) and nasopharyngeal cancer (26). Additionally, ectopic expression of Rsf-1 promoted cell and tumor growth in a mouse xenograft model (27). Furthermore, Rsf-1 was associated with paclitaxel resistance in ovarian cancer (28); however, there are no reports concerning the expression profile of Rsf-1 in malignant melanoma. The aim of the present study was to determine the status of Rsf-1 in malignant melanoma tissues, and the effects of Rsf-1 on the biological behavior of melanoma cell lines.

Materials and methods

Patients and specimens

The present study was approved by the Ethics Committee of China Medical University (Shenyang, China). Informed consent was obtained from all patients. Melanoma and adjacent normal specimens were obtained from 50 patients diagnosed with malignant melanoma who underwent resection at The First Affiliated Hospital of China Medical University (Shenyang, China) between November 2009 and March 2012. Patients did not receive chemotherapy or radiation therapy prior to surgical resection. Histological classification was performed according to the American Joint Committee on Cancer (29). There were 20 female and 30 male patients, with an age range of 25–82 years (mean, 53.2±8.67 years).

Immunohistochemical staining

Tumor samples were obtained from The First Affiliated Hospital of China Medical University. The samples were fixed in 37% formaldehyde at room temperature for 18 h and embedded in paraffin. Immunostaining was performed using the Elivision Plus method (Fuzhou Maixin Biotech. Co., Ltd., Fuzhou, China). Sections of 4-µm thickness were deparaffinized in xylene and rehydrated with a graded alcohol series (100, 95, 80 and 70%). Sections were permeabilized with Triton X-100 and then boiled in citrate buffer. Sections were blocked with goat serum (Fuzhou Maixin Biotech Co., Ltd.) at room temperature for 20 min. Hydrogen peroxide (0.3%) was used to block peroxidase activity. Sections were incubated with rabbit anti-Rsf-1 polyclonal antibody (1:1,000; HPA046129, Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) at 4°C overnight. Sections were then incubated for 2 h at 37°C with a biotinylated anti-rabbit horseradish peroxidase (HRP) polymer (KIT-9902, Fuzhou Maixin Biotech. Co., Ltd.). Sections were developed with 3,3′-diaminobenzidine plus from Fuzhou Maixin Biotech. Co., Ltd. Sections were counterstained with hematoxylin at room temperature for 2 min.

All tumor slides were analyzed by two independent investigators randomly under a light microscope (magnification, ×400; BX53; Olympus Corporation, Tokyo, Japan). Immunostaining of Rsf-1 was scored using a semi-quantitative scale by evaluating the intensity and percentage of tumor cells. Nuclear immunostaining was considered positive. The intensity of Rsf-1 staining was scored as 0 (no signal), 1 (moderate) or 2 (strong). Percentage scores were assigned as 1 (1–25%), 2 (26–50%), 3 (51–75%) or 4 (76–100%) (30). The scores of each tumor sample were multiplied to provide a final score of 0–8; tumor samples that scored 4–8 were considered to demonstrate Rsf-1 overexpression.

Cell culture and reagents

M14 cells with low Rsf-1 expression, and MV3 and A375 cells with high Rsf-1 expression were purchased from the American Type Culture Collection (Manassas, VA, USA). M14 and A375 cells were cultivated in Dulbecco's Modified Eagle's medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.). MV3 cells were cultured in RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS. Cells were cultured under conditions of 37°C and 5% CO2, and seeded at a density of 1×106 cells/ml. Then, cells were treated with cisplatin (final concentration, 5 µM) following attachment of cells to plates at 37°C for 24 h. Additionally, M14 cells were treated with NF-κB inhibitor (Bay11-7082; cat. no. S2913, Selleck Chemicals, Houston, TX, USA) at a concentration of 10 µM for 12 h at 37°C.

Small interfering RNA (siRNA) and plasmid transfection

Oligonucleotide pools of siRNA targeting Rsf-1 and non-targeting siRNA (control siRNA) were purchased from GE Healthcare Dharmacon, Inc. (Lafayette, CO, USA), and MV3 and A375 cells were transfected with 50 nM siRNA using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. The targeting sequences were as follows: Rsf-1 siRNA, 5′-GGAAAGACAUCUCUACUAU-3′; and control siRNA, 5′-GCGCGATAGCGCGAATATA-3′. pCMV6-Rsf-1 and control empty plasmids were purchased from OriGene Technologies, Inc. (Rockville, MD, USA), and M14 cells were transfected with 1 µg plasmid using Lipofectamine 3000 according to the manufacturer's protocols. Subsequent experiments were performed 48–72 h following transfection.

Western blotting

Total protein from cells was extracted using Pierce™ Universal Nuclease for Cell Lysis (Pierce; Thermo Fisher Scientific, Inc.) and quantified by the Bradford method. A total of 40 µg protein was separated by 8–12% SDS-PAGE. Samples were transferred to polyvinylidene difluoride membranes (EMD Millipore), blocked at room temperature for 1 h in 3% bovine serum albumin (BioSharp Co., Hefei, China), and incubated overnight at 4°C with antibodies against: Rsf-1 (1:2,000; cat. no. HPA046129, Sigma-Aldrich; Merck KGaA), cyclin E (1:700; cat. no. 4129, Cell Signaling Technology, Inc., Danvers, MA, USA), matrix metalloproteinase-2 (MMP2; 1:1,000; cat. no. 4022, Cell Signaling Technology, Inc.), IκB (1:1,000; cat. no. 9242, Cell Signaling Technology, Inc.), phosphorylated (p)-IκB (1:1,000; cat. no. 9246, Cell Signaling Technology, Inc.), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB; 1:1,000; cat. no. 4764, Cell Signaling Technology, Inc.), B-cell lymphoma (Bcl-2; 1:1,000; cat. no. 15071, Cell Signaling Technology, Inc.), Bcl-2-associated X protein (Bax; 1:1,000; cat. no. 2774, Cell Signaling Technology, Inc.), cellular inhibitor of apoptosis protein 1 (cIAP1; 1:1,000; cat. no. 7065, Cell Signaling Technology, Inc.), cIAP2 (1:1,000; cat. no. 3130, Cell Signaling Technology, Inc.) and β-actin (1:2,000; cat. no. 4970, Cell Signaling Technology, Inc.). Following incubation with HRP-conjugated anti-mouse/rabbit IgG (1:1,000; cat nos. 7076/7074, Cell Signaling Technology, Inc.) at 37°C for 2 h, proteins were visualized using an enhanced chemiluminescence kit (Pierce; Thermo Fisher Scientific, Inc.) and detected using a DNR Bio-Imaging System (DNR Bio-Imaging Systems, Ltd., Neve Yamin, Israel). Relative protein levels were quantified using ImageJ 1.8.0 software (National Institutes of Health, Bethesda, MD, USA).

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

Total RNA was extracted from MV3, M14 and A375 cells using TRIzol® reagent (Thermo Fisher Scientific, Inc.). Total RNA (500 ng) was then reverse-transcribed using PrimeScript RT Master Mix (10X; Takara Biotechnology Co., Ltd., Dalian, China) at 85°C for 2 min and 37°C for 30 min. qPCR was conducted using the Reverse Transcription System kit (Promega Corporation, Madison, WI, USA) according to the manufacturer's protocols. An ABI 7500 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used for gene amplification, under the conditions of: 95°C for 2 min, and 40 cycles of 95°C for 2 sec and annealing/extension at 60°C for 30 sec. A dissociation step was performed at 60–95°C for 6 sec to generate a melting curve. β-actin was used as the reference gene, and relative levels of gene expression were represented as: ΔCq=Cq gene-Cq reference. The fold change in gene expression was calculated using the 2−ΔΔCq method (31). The experiment was performed in triplicate. The primers were as follows: Rsf-1, forward 5′-GATACTATGCGTCTCCAGCCAA-3′, reverse, 5′-CAACTCGTTTCGATTTCTGACAA-3′; and β-actin, forward 5′-CCAACCGCGAGAAGATGACC-3′ and reverse, 5′-GATAGCACAGCCTGGATAGCAAC-3′.

MTT assay

A total of 5,000 cells were plated in 96-well plates and cultured overnight, followed by the addition of 20 µl of 5 mg/ml MTT solution to each well; cells were incubated for 4 h at 37°C. The supernatant was removed from each well, and dimethyl sulfoxide (150 µl) was added to dissolve the formazan crystals. The absorbance was detected at 490 nm using a microplate reader (Infinite F50; Tecan Group, Ltd., Mannedorf, Switzerland).

Colony formation assay

For colony formation, cells were seeded into three 6-cm cell culture dishes (~800 cells/dish) 48 h following transfection. Cells were incubated for 14 days at 37°C. Plates were washed with PBS and then stained with Giemsa at room temperature for 10 min. The number of colonies with >50 cells was manually counted under a light microscope (magnification, ×200; BX53).

Transwell invasion assay

A Transwell invasion assay was performed using a 24-well Transwell chamber with a pore size of 8 µm (Costar; Corning Inc., Corning, NY, USA), and the inserts were coated with 20 µl Matrigel (1:3; BD Biosciences, San Jose, CA, USA). After 48 h following transfection, cells were trypsinized (0.25% trypsin) at 37°C for 30 sec and then transferred to the upper Matrigel-coated chamber in 100 µl serum-free medium (1×105 cells/ml). Medium (DMEM for M14 and A375 cells, RPMI-1640 for MV3 cells) supplemented with 10% FBS was added to the lower chamber as the chemoattractant. Cells were incubated for 18 h at 37°C. Non-invading cells on the upper membrane surface were then removed with a cotton tip, and the cells that passed via the filter were fixed in 4% paraformaldehyde at room temperature for 20 min. Cells were stained with hematoxylin at room temperature for 5 min. Cells were observed under a light microscope (magnification, ×200; BX53). The experiments were performed in triplicate.

Flow cytometry for cell cycle and apoptosis analyses

Cells in 6-well plates were collected using tryptase 48 h following transfection. Cells were washed twice with PBS, followed by resuspension in 250 µl binding buffer (BD Pharmingen; BD Biosciences). Cells were fixed in 1% paraformaldehyde at 4°C overnight and then stained with 5 mg/ml propidium iodide (PI) alone or together with Annexin V/fluorescein isothiocyanate (BD Pharmingen; BD Biosciences) at room temperature for 15 min for cell cycle or apoptosis analysis, respectively. Incubation was performed in the dark for 15 min. Flow cytometry was performed using flow cytometer and analyzed using NovoExpress 1.2.5 software (ACEA Biosciences, Inc.; Agilent Technologies, Inc., Santa Clara, CA, USA). The apoptotic rate was calculated by adding the percentage of early apoptotic (Annexin V-positive, PI-negative) and late apoptotic cells (Annexin V-positive, PI-positive).

Detection of the mitochondrial membrane potential (MMP)

The MMP was detected via the JC-1 staining method. Briefly, cells (300 cells/µl) were harvested, washed with PBS and incubated with 5 µM JC-1 (Cell Signaling Technology, Inc.) at 37°C for 30 min in an incubator. Cells were then washed and analyzed using a flow cytometer. Data were analyzed using NovoExpress 1.2.5 software.

Statistical analysis

SPSS version 16 for Windows (SPSS, Inc., Chicago, IL, USA) was used for all statistical analyses. A χ2 test was used to examine potential associations between Rsf-1 expression and the clinicopathological features of patients with melanoma. A Student's t-test was used to compare differences between the control and treatment groups. Data were presented as the mean ± standard deviation of at least three experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of Rsf-1 in human malignant melanoma

Rsf-1 expression in 50 cases of malignant melanoma was determined by immunohistochemistry (Fig. 1). Normal skin tissue exhibited weak or negative staining (Fig. 1A). In total, 19/50 (38%) cases of skin melanoma demonstrated high Rsf-1 immunoreactivity (Rsf-1 overexpression, or an immunostaining score of ≥4), which was localized to the nuclear compartment of tumor cells (Fig. 1B-D). The association between Rsf-1 expression and the clinicopathological characteristics of patients with melanoma was analyzed (Table I). The frequency of Rsf-1 overexpression was increased in melanomas of advanced tumor, node and metastasis (TNM) stages (III+IV vs. II, P=0.0494). The results revealed that no significant association was observed between Rsf-1 expression and patient age (P=0.5655) and gender (P=0.122), or T stage (P=0.8842).

Table I.

Distribution of Rsf-1 status in melanoma according to the clinicopathological characteristics of patients.

Table I.

Distribution of Rsf-1 status in melanoma according to the clinicopathological characteristics of patients.

Clinicopathological characteristicsNumber of patientsRsf-1 low expressionRsf-1 high expressionχ2P-value
Age (years) 0.33020.5655
  <60342212
  ≥601697
Sex 2.39110.1220
  Female20155
  Male301614
TNM stage 3.86060.0494
  II432914
  III+IV725
T stage 0.02120.8842
  T1-31064
  T4402515

[i] Rsf-1, remodeling and spacing factor 1; TNM, tumor, node, and metastasis.

Rsf-1 promotes malignant melanoma cell viability and invasion

Rsf-1 expression levels in malignant melanoma cell lines (MV3, M14 and A375) were investigated using western blotting and RT-qPCR. It was revealed that the Rsf-1 expression levels were low in M14 cells, and high in MV3 and A375 cell lines (Fig. 2A). To investigate the biological roles of Rsf-1 in malignant melanoma, Rsf-1 siRNA-mediated interference was performed in the MV3 and A375 melanoma cell lines, whilst Rsf-1-encoding plasmid transfection was performed in the M14 cell line. As presented in Fig. 2B, Rsf-1 siRNA significantly downregulated Rsf-1 protein and mRNA expression, whereas the Rsf-1 plasmid significantly upregulated Rsf-1 expression. An MTT assay was performed to investigate cell viability, which demonstrated that Rsf-1 depletion in MV3 and A375 cells notably decreased cell viability compared with the controls, whereas Rsf-1 overexpression in M14 cells markedly promoted cell viability (Fig. 3A). A colony formation assay also revealed that Rsf-1 depletion significantly decreased colony number in the MV3 and A375 cell lines, while Rsf-1 overexpression significantly increased the colony number in the M14 cell line compared with the control (Fig. 3B). To determine the effects of Rsf-1 on cell invasion, a Transwell invasion assay was performed. The results revealed that the number of invasive cells was significantly reduced following Rsf-1 depletion in the MV3 (control siRNA vs. Rsf-1 siRNA, 243±20 vs. 52±12 cells; P<0.05; Fig. 3C) and A375 cell lines (control siRNA vs. Rsf-1 siRNA, 214±15 vs. 90±8 cells; P<0.05) compared with the control. Conversely, Rsf-1 overexpression significantly increased the invasive ability of M14 cells compared with the control (empty plasmid vs. Rsf-1 plasmid, 100±7 vs. 221±15 cells; P<0.05).

Rsf-1 regulates cell cycle progression and associated protein expression

Cell cycle analysis was performed in melanoma cell lines. Rsf-1 depletion in MV3 and A375 cells significantly increased the percentage of cells in G1 phase and decreased that in S phase compared with the control (Fig. 4A). Rsf-1 overexpression in M14 cells had opposing effects; the percentage of cells in G1 phase was significantly reduced, while the percentage of cells in S phase increased compared with the control, suggesting that Rsf-1 depletion suppresses G1/S cell cycle transition (Fig. 4A). To analyze the potential molecular mechanisms underlying the effects of Rsf-1 on the cell cycle, the expression of associated proteins was examined by western blotting. As presented in Fig. 4B, the expression levels of MMP2, cyclin E and p-IκB were decreased in Rsf-1-depleted MV3 and A375 cells compared with control cells. Conversely, Rsf-1 overexpression upregulated MMP2, cyclin E and p-IκB expression in M14 cells.

Rsf-1 enhances cisplatin resistance and MMP

To investigate the role of Rsf-1 in the chemoresistance of malignant melanoma cells, Rsf-1 depleted and control cells were treated with cisplatin (5 µM). The results of the MTT assay revealed that Rsf-1 siRNA significantly decreased cell survival rate following 3 days of cisplatin treatment in MV3 and A375 cells compared with the control. Rsf-1 overexpression significantly increased cell viability in M14 cells treated with cisplatin (Fig. 5A). Furthermore, apoptosis analysis revealed that the cell apoptotic rate was significantly increased following Rsf-1 depletion in MV3 and A375 cells treated with cisplatin, and reduced in Rsf-1-overexpressing M14 cells treated with cisplatin compared with the control (Fig. 5B). Collectively, the results demonstrated that Rsf-1 expression promotes cisplatin resistance in melanoma cells.

As resistance to chemotherapeutic drugs is closely associated with mitochondrial function, whether Rsf-1 affected the MMP was investigated. JC-1 staining was used to monitor alterations in MMP following cisplatin treatment. JC-1 staining exhibits red fluorescence under normal conditions; however, green fluorescence is observed when the MMP is depolarized following cisplatin treatment. As presented in Fig. 5C, in M14 cells treated with cisplatin, Rsf-1 overexpression notably decreased the percentage of cells exhibiting green fluorescence, suggesting that Rsf-1 promoted mitochondrial membrane polarization. Conversely, Rsf-1 depletion led to notable depolarization of the MMP in MV3 and A375 cells treated with cisplatin.

Rsf-1 regulates Bcl-2 expression via NF-κB signaling

Furthermore, Rsf-1-induced alterations in apoptosis-associated protein expression were investigated via western blot analysis (Fig. 6A). Rsf-1 depletion downregulated Bcl-2, cIAP1 and cIAP2 expression levels, and upregulated Bax expression in MV3 and A375 cell lines compared with the control; opposing effects were observed in Rsf-1-overexpressing M14 cells. As Rsf-1 positively regulates p-IκB, and Bcl-2 expression was reported as associated with NF-κB signaling (32), whether Rsf-1 regulated the activity of Bcl-2 via its effects on NF-κB was investigated. To validate this, M14 cells were treated with NF-κB inhibitor (10 µM). As presented in Fig. 6B, NF-κB inhibition significantly downregulated p-IκB and NF-κB p65 protein levels in control and Rsf-1 plasmid-transfected M14 cells. Furthermore, treatment with the NF-κB inhibitor eliminated the effects of Rsf-1 upregulation on Bcl-2 expression.

Discussion

Previous studies have reported that Rsf-1 overexpression occurs in numerous cancers, including ovarian cancer, breast cancer, nasopharyngeal carcinoma, non-small cell lung cancer, gastric adenocarcinoma and colon cancer (2023,25,26,28,3335); however, its involvement in melanoma has not been investigated. In the present study, the expression of Rsf-1 was analyzed in 50 malignant melanoma specimens via immunohistochemistry. Overexpression of Rsf-1 was detected in 19 cases, which was positively associated with advanced TNM stage. As surgical therapy is not the preferred treatment for patients with advanced melanomas, particularly stage IV melanoma, the number of stage III and IV melanoma specimens analyzed was markedly lower than that for stage II melanoma. It was observed that the incidence of Rsf-1 overexpression was notably higher in melanomas with advanced TNM stage (III and IV vs. II). Accordingly, overexpression of Rsf-1 was reported to be associated with advanced TNM stage, nodal metastasis and poorly differentiated tumor cells in other cancers (20,22,23,33,34). Thus, Rsf-1 tends to be overexpressed in advanced stage melanomas, suggesting its association with the malignant progression of melanoma cells. To the best of our knowledge, the present study is the first to demonstrate the clinical significance of Rsf-1 in melanoma.

Rsf-1 has been reported to regulate cell behaviors including proliferation, invasion and cell cycle progression (16,26,3639). A previous study reported that Rsf-1 depletion significantly decreased the proliferation rate and colony formation ability in colon cancer cell lines HT29 and HCT116 (23). Rsf-1 depletion also inhibited proliferation in lung cancer cells (21). The present findings support these previous reports, demonstrating that Rsf-1 depletion decreased cell viability and colony number, while its overexpression promoted viability. For cell invasion, it was observed that the number of invading cells decreased significantly following Rsf-1 depletion, but markedly increased following overexpression of Rsf-1, which was consistent with previous reports demonstrating that Rsf-1 depletion inhibited invasiveness in the prostate cancer cell line DU145 (20) and lung cancer cells (30).

The regulatory effect of Rsf-1 on cell viability suggested that Rsf-1 may serve an important role in cell cycle progression. The present study revealed that Rsf-1 depletion enhanced the percentage of G1 phase cells and downregulated that of S phase, demonstrating that Rsf-1 can facilitate G1/S transition. Western blotting revealed that Rsf-1 depletion decreased the levels of MMP2, cyclin E and p-IκB, consistent with previous reports of MMP2 downregulation following Rsf-1 depletion in lung cancer cells (30). MMP2 is a member of the matrix metalloproteinase family, the members of which are involved in various pathological and physiological processes, including cancer cell growth, invasion and metastasis, suggesting that Rsf-1 regulates melanoma invasion via MMP2 (40). However, the possibility that other effects regulated by Rsf-1 may also be responsible for its effects on invasion cannot be excluded.

Transfection of cell lines exhibiting high Rsf-1 expression with siRNA targeted against Rsf-1 also increased the rate of apoptosis, which may also contribute to the decreased invasive ability of cells following Rsf-1 depletion. Cyclin E serves an essential role in fundamental biological processes, including cell cycle control and DNA replication (4143). Sheu et al (13) revealed that cyclin E1 interacts with the first 441 amino acids of Rsf-1, and that their interaction promotes G1-S transition. Additionally, Rsf-1 depletion downregulated cyclin E in hepatocellular carcinoma (25). These reports further support the findings of the present study.

Furthermore, the present study proposed that Rsf-1 positively regulated the chemoresistance of melanoma cells, which has not been previously reported, to the best of our knowledge. In cells treated with cisplatin, MTT and Annexin V/PI analysis were performed to examine the effects of Rsf-1. The cell survival rate decreased, while the apoptotic rate increased significantly following Rsf-1 depletion. The role of Rsf-1 in chemoresistance has been indicated in various cancers including ovarian cancer (28), lung cancer (44) and glioma (36); however, the association between Rsf-1 and mitochondrial regulation has not yet been reported. Mitochondrial function serves an important role in the development of chemoresistance. Depolarization of the MMP induces apoptosis via the mitochondria-dependent pathway (45). It was demonstrated that Rsf-1 depletion depolarized the MMP, with opposing effects observed following Rsf-1 overexpression in M14 cells. To the best of our knowledge, the present study is the first to report of the association between the role of Rsf-1 in chemoresistance and the regulation of mitochondrial function.

It was revealed that expression of the pro-apoptotic protein Bax increased, while the levels of anti-apoptotic proteins, including cIAP1, cIAP2 and Bcl-2 decreased significantly following Rsf-1 depletion, as reported in previous studies (4648); Rsf-1 overexpression induced opposing effects. cIAP1 and cIAP2 are members of the IAP family, which regulate apoptosis and chemoresistance (49).

The NF-κB signaling pathway is induced via activation of IκB, and is involved in numerous biological processes, including cell growth, tumorigenesis and apoptosis (50). Bcl-2 is a downstream effector of NF-κB, and serves as an important anti-apoptotic mediator in melanoma (51,52). The present study proposed that Rsf-1 could positively regulate the NF-κB pathway via upregulation of p-IκB. NF-κB signaling was considered particularly noteworthy for two reasons. A previous study using Ingenuity Pathways Analysis Systems revealed that various molecular hubs including NF-kB, extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) were identified in an Rsf-1-regulated gene network (28). In addition, analysis of numerous other signaling pathways was conducted, including p-ERK and p-Akt (data not shown); however, significant alterations were not observed in the expression profile of these proteins (data not shown). Notable alterations in p-IκB expression were observed. Thus, the NF-κB pathway was selected for further study, and its importance was confirmed via the use of an NF-κB inhibitor. Rsf-1 overexpression failed to induce Bcl-2 upregulation in cells treated by NF-κB inhibitor, supporting the association between Rsf-1 and Bcl-2 in melanoma cells.

There are two novel points to highlight based upon the findings of the present study. The clinical significance of Rsf-1, which has not been previously reported in melanoma, was demonstrated in this study. Additionally, the role of Rsf-1 in chemosensitivity was associated with mitochondrial function. In conclusion, the present study demonstrated that Rsf-1 is overexpressed in malignant melanoma, and may contribute to the proliferation, invasion and cell cycle progression of malignant cells by modulating the expression of MMP2, cyclin E and NF-κB. Furthermore, Rsf-1 may regulate chemoresistance and MMP in melanoma cells, with concomitant alterations in cIAP1, cIAP2, Bax and Bcl-2 protein expression. Thus, Rsf-1 may serve as a potential therapeutic target in the treatment of malignant melanoma.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

JH and LF performed the experiments, evaluated the data, drafted the manuscript and prepared the figures. QL made significant contributions towards the design of the study, evaluated the data and drafted the manuscript. All authors reviewed the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of China Medical University (Shenyang, China). Informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Merrill SJ, Subramanian M and Godar DE: Worldwide cutaneous malignant melanoma incidences analyzed by sex, age, and skin type over time (1955–2007): Is HPV infection of androgenic hair follicular melanocytes a risk factor for developing melanoma exclusively in people of European-ancestry? Dermatoendocrinol. 8:e12153912016. View Article : Google Scholar : PubMed/NCBI

2 

Li H, Pedersen L, Nørgaard M, Ulrichsen SP, Thygesen SK and Nelson JJ: The occurrence of non-melanoma malignant skin lesions and non-cutaneous squamous-cell carcinoma among metastatic melanoma patients: An observational cohort study in Denmark. BMC Cancer. 16:2952016. View Article : Google Scholar : PubMed/NCBI

3 

Nahar VK, Allison Ford M, Brodell RT, Boyas JF, Jacks SK, Biviji-Sharma R, Haskins MA and Bass MA: Skin cancer prevention practices among malignant melanoma survivors: A systematic review. J Cancer Res Clin Oncol. 142:1273–1283. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Peterson M, Albertini MR and Remington P: Remington, incidence, survival, and mortality of malignant cutaneous melanoma in wisconsin, 1995–2011. WMJ. 114:196–201. 2015.PubMed/NCBI

5 

Johnson-Obaseki SE, Labajian V, Corsten MJ and McDonald JT: Incidence of cutaneous malignant melanoma by socioeconomic status in Canada: 1992–2006. J Otolaryngol Head Neck Surg. 44:532015. View Article : Google Scholar : PubMed/NCBI

6 

Dzambova M, Sečníková Z, Jiráková A, Jůzlová K, Viklický O, Hošková L, Göpfertovà D and Hercogová J: Malignant melanoma in organ transplant recipients: Incidence, outcomes, and management strategies: A review of literature. Dermatol Ther. 29:64–68. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Brewer JD, Shanafelt TD, Call TG, Cerhan JR, Roenigk RK, Weaver AL and Otley CC: Increased incidence of malignant melanoma and other rare cutaneous cancers in the setting of chronic lymphocytic leukemia. Int J Dermatol. 54:e287–e293. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Chang HY, Feng HL, Wang L, Chou P and Wang PF: The Incidence, prevalence, and survival of malignant melanoma in Taiwan. Value Health. 17:A7402014. View Article : Google Scholar : PubMed/NCBI

9 

Nowak-Sadzikowska J, Walasek T, Jakubowicz J, Blecharz P and Reinfuss M: Current treatment options of brain metastases and outcomes in patients with malignant melanoma. Rep Pract Oncol Radiother. 21:271–277. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Schmid-Wendtner M and Wendtner CM: Treatment of metastatic malignant melanoma. Dtsch Med Wochenschr. 141:10002016.(In German). PubMed/NCBI

11 

Kozovska Z, Gabrisova V and Kucerova L: Malignant melanoma: Diagnosis, treatment and cancer stem cells. Neoplasma. 63:510–517. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Harries M, Malvehy J, Lebbe C, Heron L, Amelio J, Szabo Z and Schadendorf D: Treatment patterns of advanced malignant melanoma (stage III–IV)-A review of current standards in Europe. Eur J Cancer. 60:179–189. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Sheu JJ, Choi JH, Guan B, Tsai FJ, Hua CH, Lai MT, Wang TL and Shih IeM: Rsf-1, a chromatin remodelling protein, interacts with cyclin E1 and promotes tumour development. J Pathol. 229:559–568. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Hanai K, Furuhashi H, Yamamoto T, Akasaka K and Hirose S: RSF governs silent chromatin formation via histone H2Av replacement. PLoS Genet. 4:e10000112008. View Article : Google Scholar : PubMed/NCBI

15 

Liang PI, Wu LC, Sheu JJ, Wu TF, Shen KH, Wang YH, Wu WR, Shiue YL, Huang HY, Hsu HP, et al: Rsf-1/HBXAP overexpression is independent of gene amplification and is associated with poor outcome in patients with urinary bladder urothelial carcinoma. J Clin Pathol. 65:802–807. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Min S, Jo S, Lee HS, Chae S, Lee JS, Ji JH and Cho H: ATM-dependent chromatin remodeler Rsf-1 facilitates DNA damage checkpoints and homologous recombination repair. Cell Cycle. 13:666–677. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Goldfarb DM, Gukova LA, Chernin LS, Avdienko ID, Mnatsakanian GG, Kushner IC, Kuznetsova VN and Strachova TS: Rsf mutants of Escherichia coli HfrC defective in the production of the factor stimulating recombination in conjugation. Mol Gen Genet. 129:295–310. 1974. View Article : Google Scholar : PubMed/NCBI

18 

Iwasa H, Kuroyanagi H, Maimaiti S, Ikeda M, Nakagawa K and Hata Y: Characterization of RSF-1, the Caenorhabditis elegans homolog of the Ras-association domain family protein 1. Exp Cell Res. 319:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Fang FM, Li CF, Huang HY, Lai MT, Chen CM, Chiu IW, Wang TL, Tsai FJ, Shih IeM and Sheu JJ: Overexpression of a chromatin remodeling factor, RSF-1/HBXAP, correlates with aggressive oral squamous cell carcinoma. Am J Pathol. 178:2407–2415. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Li H, Zhang Y, Zhang Y, Bai X, Peng Y and He P: Rsf-1 overexpression in human prostate cancer, implication as a prognostic marker. Tumour Biol. 35:5771–5776. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Li Q, Dong Q and Wang E: Rsf-1 is overexpressed in non-small cell lung cancers and regulates cyclinD1 expression and ERK activity. Biochem Biophys Res Commun. 420:6–10. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Lin CY, Tian YF, Wu LC, Chen LT, Lin LC, Hsing CH, Lee SW, Sheu MJ, Lee HH, Wang YH, et al: Rsf-1 expression in rectal cancer: With special emphasis on the independent prognostic value after neoadjuvant chemoradiation. J Clin Pathol. 65:687–692. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Liu S, Dong Q and Wang E: Rsf-1 overexpression correlates with poor prognosis and cell proliferation in colon cancer. Tumour Biol. 33:1485–1491. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Maeda D, Chen X, Guan B, Nakagawa S, Yano T, Taketani Y, Fukayama M, Wang TL and Shih IeM: Rsf-1 (HBXAP) expression is associated with advanced stage and lymph node metastasis in ovarian clear cell carcinoma. Int J Gynecol Pathol. 30:30–35. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Xie C, Fu L, Xie L, Liu N and Li Q: Rsf-1 overexpression serves as a prognostic marker in human hepatocellular carcinoma. Tumour Biol. 35:7595–7601. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Tai HC, Huang HY, Lee SW, Lin CY, Sheu MJ, Chang SL, Wu LC, Shiue YL, Wu WR, Lin CM and Li CF: Associations of Rsf-1 overexpression with poor therapeutic response and worse survival in patients with nasopharyngeal carcinoma. J Clin Pathol. 65:248–253. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Sheu JJ, Choi JH, Yildiz I, Tsai FJ, Shaul Y, Wang TL and Shih IeM: The roles of human sucrose nonfermenting protein 2 homologue in the tumor-promoting functions of Rsf-1. Cancer Res. 68:4050–4057. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Choi JH, Sheu JJ, Guan B, Jinawath N, Markowski P, Wang TL and Shih IeM: Functional analysis of 11q13.5 amplicon identifies Rsf-1 (HBXAP) as a gene involved in paclitaxel resistance in ovarian cancer. Cancer Res. 69:1407–1415. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Trinidad CM, Torres-Cabala CA, Curry JL, Prieto VG and Aung PP: Update on eighth edition American Joint Committee on Cancer classification for cutaneous melanoma and overview of potential pitfalls in histological examination of staging parameters. J Clin Pathol. 72:265–270. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Zhang X, Fu L, Xue D, Zhang X, Hao F, Xie L, He J, Gai J, Liu Y, Xu H, et al: Overexpression of Rsf-1 correlates with poor survival and promotes invasion in non-small cell lung cancer. Virchows Arch. 470:553–560. 2017. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Jang JH and Surh YJ: Bcl-2 attenuation of oxidative cell death is associated with up-regulation of gamma-glutamylcysteine ligase via constitutive NF-kappaB activation. J Biol Chem. 279:38779–38786. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Sheu JJ, Guan B, Choi JH, Lin A, Lee CH, Hsiao YT, Wang TL, Tsai FJ and Shih IeM: Rsf-1, a chromatin remodeling protein, induces DNA damage and promotes genomic instability. J Biol Chem. 285:38260–38269. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Mao TL, Hsu CY, Yen MJ, Gilks B, Sheu JJ, Gabrielson E, Vang R, Cope L, Kurman RJ, Wang TL and Shih IeM: Expression of Rsf-1, a chromatin-remodeling gene, in ovarian and breast carcinoma. Hum Pathol. 37:1169–1175. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Davidson B, Trope' CG, Wang TL and Shih IeM: Expression of the chromatin remodeling factor Rsf-1 is upregulated in ovarian carcinoma effusions and predicts poor survival. Gynecol Oncol. 103:814–819. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Zhao XC, An P, Wu XY, Zhang LM, Long B, Tian Y, Chi XY and Tong DY: Overexpression of hSNF2H in glioma promotes cell proliferation, invasion, and chemoresistance through its interaction with Rsf-1. Tumour Biol. 37:7203–7212. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Ren J, Chen QC, Jin F, Wu HZ, He M, Zhao L, Yu ZJ, Yao WF, Mi XY, Wang EH and Wei MJ: Overexpression of Rsf-1 correlates with pathological type, p53 status and survival in primary breast cancer. Int J Clin Exp Pathol. 7:5595–5608. 2014.PubMed/NCBI

38 

Chae S, Ji JH, Kwon SH, Lee HS, Lim JM, Kang D, Lee CW and Cho H: HBxAPalpha/Rsf-1-mediated HBx-hBubR1 interactions regulate the mitotic spindle checkpoint and chromosome instability. Carcinogenesis. 34:1680–1688. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Hu BS, Yu HF, Zhao G and Zha TZ: High RSF-1 expression correlates with poor prognosis in patients with gastric adenocarcinoma. Int J Clin Exp Pathol. 5:668–673. 2012.PubMed/NCBI

40 

Rotte A, Martinka M and Li G: MMP2 expression is a prognostic marker for primary melanoma patients. Cell Oncol (Dordr). 35:207–216. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Santala S, Talvensaari-Mattila A, Soini Y and Santala M: Cyclin E expression correlates with Cancer-specific survival in endometrial endometrioid adenocarcinoma. Anticancer Res. 35:3393–3397. 2015.PubMed/NCBI

42 

Alsina M, Landolfi S, Aura C, Caci K, Jimenez J, Prudkin L, Castro S, Moreno D, Navalpotro B, Tabernero J and Scaltriti M: Cyclin E amplification/overexpression is associated with poor prognosis in gastric cancer. Ann Oncol. 26:438–439. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Deng W, Zhou Y, Tiwari AF, Su H, Yang J, Zhu D, Lau VM, Hau PM, Yip YL, Cheung AL, et al: p21/Cyclin E pathway modulates anticlastogenic function of Bmi-1 in cancer cells. Int J Cancer. 136:1361–1370. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Li HC, Chen YF, Feng W, Cai H, Mei Y, Jiang YM, Chen T, Xu K and Feng DX: Loss of the Opa interacting protein 5 inhibits breast cancer proliferation through miR-139-5p/NOTCH1 pathway. Gene. 603:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Chen X, Wong JY, Wong P and Radany EH: Low-dose valproic acid enhances radiosensitivity of prostate cancer through acetylated p53-dependent modulation of mitochondrial membrane potential and apoptosis. Mol Cancer Res. 9:448–461. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Matsuyama S, Palmer J, Bates A, Poventud-Fuentes I, Wong K, Ngo J and Matsuyama M: Bax-induced apoptosis shortens the life span of DNA repair defect Ku70-knockout mice by inducing emphysema. Exp Biol Med (Maywood). 241:1265–1271. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Gill C, Dowling C, O'Neill AJ and Watson RW: Effects of cIAP-1, cIAP-2 and XIAP triple knockdown on prostate cancer cell susceptibility to apoptosis, cell survival and proliferation. Mol Cancer. 8:392009. View Article : Google Scholar : PubMed/NCBI

48 

Vassina EM, Yousefi S, Simon D, Zwicky C, Conus S and Simon HU: cIAP-2 and survivin contribute to cytokine-mediated delayed eosinophil apoptosis. Eur J Immunol. 36:1975–1984. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Gyrd-Hansen M and Meier P: IAPs: From caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nat Rev Cancer. 10:561–574. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Hussain AR, Ahmed SO, Ahmed M, Khan OS, Al Abdulmohsen S, Platanias LC, Al-Kuraya KS and Uddin S: Cross-talk between NFkB and the PI3-kinase/AKT pathway can be targeted in primary effusion lymphoma (PEL) cell lines for efficient apoptosis. PLoS One. 7:e399452012. View Article : Google Scholar : PubMed/NCBI

51 

Benimetskaya L, Ayyanar K, Kornblum N, Castanotto D, Rossi J, Wu S, Lai J, Brown BD, Popova N, Miller P, et al: Bcl-2 protein in 518A2 melanoma cells in vivo and in vitro. Clin Cancer Res. 12:4940–4948. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Leiter U, Schmid RM, Kaskel P, Peter RU and Krähn G: Antiapoptotic bcl-2 and bcl-xL in advanced malignant melanoma. Arch Dermatol Res. 292:225–232. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He J, Fu L and Li Q: Rsf‑1 regulates malignant melanoma cell viability and chemoresistance via NF‑κB/Bcl‑2 signaling. Mol Med Rep 20: 3487-3498, 2019
APA
He, J., Fu, L., & Li, Q. (2019). Rsf‑1 regulates malignant melanoma cell viability and chemoresistance via NF‑κB/Bcl‑2 signaling. Molecular Medicine Reports, 20, 3487-3498. https://doi.org/10.3892/mmr.2019.10610
MLA
He, J., Fu, L., Li, Q."Rsf‑1 regulates malignant melanoma cell viability and chemoresistance via NF‑κB/Bcl‑2 signaling". Molecular Medicine Reports 20.4 (2019): 3487-3498.
Chicago
He, J., Fu, L., Li, Q."Rsf‑1 regulates malignant melanoma cell viability and chemoresistance via NF‑κB/Bcl‑2 signaling". Molecular Medicine Reports 20, no. 4 (2019): 3487-3498. https://doi.org/10.3892/mmr.2019.10610