Open Access

Downregulated RRS1 inhibits invasion and metastasis of BT549 through RPL11‑c‑Myc‑SNAIL axis

  • Authors:
    • Runze Wang
    • Cuixiu Peng
    • Junying Song
    • Yanan Hua
    • Qinglan Wu
    • Lin Deng
    • Yi Cao
    • Jinyu Zhang
    • Li Zhang
    • Li Wu
    • Lin Hou
  • View Affiliations

  • Published online on: February 17, 2022     https://doi.org/10.3892/ijo.2022.5323
  • Article Number: 33
  • Copyright: © Wang 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

Regulator of ribosome synthesis 1 (RRS1) is a key factor in ribosome biosynthesis and other cellular functions. High level of RRS1 in breast cancer cell lines is associated with increased cell proliferation, invasion and migration. RRS1 controls the assembly of the 60s subunit and maturation of 25S rRNA during ribosome biosynthesis. In this study, lentiviral transfection of sh‑RNA was used to knock down the level of RRS1, to detect the effect of RRS1 on cell function and to explore the specific mechanism of RRS1 affecting cell invasion and metastasis by COIP and dual‑luciferase reporter gene assays. The present study found that RRS1 knockdown reduced the accumulation of ribosome protein L11 (RPL11) in the nucleolus, which then migrated to the nucleoplasm and bound to c‑Myc. This inhibited trans‑activation of SNAIL by c‑Myc and eventually decreased the invasion and metastasis capacity of the human breast cancer cell line BT549. Taken together, RRS1 regulates invasion and metastasis of human breast cancer cells through the RPL11‑c‑Myc‑SNAIL axis. The findings are of great significance for exploring the mechanism of breast cancer invasion and metastasis and the corresponding regulatory factors.

Introduction

Breast cancer is a highly prevalent malignancy among women and remains fatal in spite of the decrease in mortality rate in recent years (1). Clinically, three types of biomarkers are used to classify breast cancer: Estrogen receptor α (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER-2) (2,3). Among them, Luminal A subtype highly expresses ER and PR, lacks HER-2 expression and has the characteristics of low proliferation rate and good prognosis (4). Luminal B subtype is similar to Luminal A subtype, but lacks PR expression. It demonstrates higher proliferation rate, worse prognostic effect and tolerance to hormone therapy (5). The HER-2 subtype is characterized by HER-2 overexpression, which can be further divided into HER-2 positive and HER-2 negative according to the expression of ER. It is more resistant to chemotherapeutics and the prognosis is worse than that of the Luminal subtypes. Triple negative breast cancer (TNBC) is characterized by absence of HER-2, PR and ER expression. It has strong chemotherapy resistance and metastasis. The prognosis of patients with triple-negative breast cancer is extremely poor and relapse common (6,7). Distant organ metastasis, especially to the brain, bones, lungs and liver, is a characteristic feature of breast cancer (8,9). Currently, metastatic breast cancer is treated by surgery, radiotherapy and adjuvant chemotherapy. Surgical resection has inherent disadvantages and is associated with high recurrence rates (10,11), whereas radiotherapy and chemotherapy have a number of adverse effects, such as cardiac damage caused by combined chemoradiotherapy (12,13). Therefore, it is critical to identify novel therapeutic targets for metastatic triple negative breast cancer (TNBC).

The ribosomal regulatory protein RRS1 was discovered in Saccharomyces cerevisiae by Tsuno et al in 1999 (14). The human RRS1 gene is located on chromosome 8q13.1 and contains only one exon (15). RRS1 regulates ribosome biosynthesis by recruiting 5S ribonucleoprotein (RNP) to form the pre-60S ribosomal subunit with ribosomal production factor 2 (Rpf2) and promoting the maturation of 25S rRNA (16-18). RRS1 also mediates the export of pre-60S ribosomal subunit from the nucleolus to the cytoplasm. Thus, depletion of RRS1 results in the accumulation of the pre-60 subunit in the nucleoplasm, eventually stalling ribosome biosynthesis (18). Studies show that RRS1 also regulates chromosome rearrangement during mitosis (19) and telomere aggregation (20) and serves an important role in delayed cell aging (21) and in the development of Huntington's disease (22). In addition, RRS1 is abnormally expressed in various cancers (23-30). Through binding of RPL11, which inhibits the interaction between RPL11 and murine doubleminute 2 (MDM2), it promotes the proliferation of breast cancer cells and reduces p53 levels (26). Knocking down RRS1 in breast cancer cells significantly reduced their proliferation rates by inducing cell cycle arrest (26).

RPL11 is a component of 5S ribonucleoprotein particles (5S RNP) and is translocated to the nucleus along with RPL5 by the nuclear import protein Syo1, where they bind to 5S rRNA (31). RPL11 regulates MDM2 (32), which prevents p53 accumulation during cellular stress by inducing protein ubiquitination and degradation (33-37). Furthermore, RPL11 suppresses c-Myc activity through a negative feedback pathway (38), which inhibits the transcription of downstream genes regulating cell growth, proliferation, metabolism, apoptosis, differentiation and ribosomal biosynthesis (39), whereas c-Myc can transcriptionally activate RPL11 (40). In addition, miR-150 and miR-383 suppresses 5S rRNA in esophageal squamous cell carcinoma cells, which strengthening RPL11-c-Myc interaction and reducing c-Myc-induced proliferation (41). C-myc regulates the epithelial mesenchymal transition (EMT) of various tumor cells (42-47), which allows the cells to detach from the primary tumors, extravasate and metastasize to distant organs (48).

The present study found that RRS1 regulated the invasion and metastasis of breast cancer cells through the RPL11-c-Myc-SNAIL axis. RRS1 knockdown disrupted ribosomal biosynthesis and prevented the export of RPL11 from the nucleus. The accumulation of RPL11 in the nucleoplasm inhibited c-Myc-dependent transcription of EMT-related genes. The findings revealed a new role of RRS1 in regulating breast cancer cell invasion and metastasis and demonstrate its potential as a therapeutic target.

Materials and methods

Cell line authentication

The breast cancer cell lines BT549, MDA-MB-231, MDA-MB-468, HMEC and MCF-7 was purchased from Procell Life Science & Technology Co., Ltd. in March, 2021. The cell line was verified in March, 2021 and the follow-up experiments were performed in the same month. Briefly, DNA was extracted from 1×106 cells using Chelex100 and 20 STR sites and sex loci were identified using the 21 CELL ID System (Procell Life Science & Technology Co., Ltd.), including D19S433, D5S818, D21S11, D18S51, D6S1043, D3S1358, D13S317, D7S820, D16S539, CSF1PO, PentaD, vWA, D8S1179, TPOX, PentaE, TH01, D12S391, D2S1338, D1656 and Amelogenin1. The PCR products were analyzed with the ABI3130x1 genetic analyzer and Gene Mapper IDX software (Applied Biosystems; Thermo Fisher Scientific, Inc.) and compared with the ATCC, DSMZ, JCRB and Cellosaurus databases. BT549, MDA-MB-231, MDA-MB-468 are Triple negative breast cancer breast cancer. MCF-7 is Luminal A breast cancer. 293T cells were from the collection of our research group.

Cell culture and transfection

All the cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% FBS (Shanghai ExCell Biology, Inc.) and 1% penicillin and streptomycin solution at 37°C. On reaching 20-30% confluency, the cells were transduced with the GV493-GFP lentivirus RNAi expression system (Shanghai GeneChem Co., Ltd.) expressing RPL11-shRNA or scrambled shRNA with Hitrans A&P (Shanghai GeneChem Co., Ltd.). The multiplicity of infection (MOI) was 10. The virus was transfected with serum-free medium for 10 h at 37°C. The medium was changed and replaced with complete medium and subsequent experiments were carried out after culturing for 48 h at 37°C. The siRNAs and scrambled sequence for siRNAs were synthesized by Sangon Biotech Co., Ltd. The target sequences for RPL11 were as follows: Forward 5′-GGU GCU GGA GUA UGA GUU ATT-3′ and reverse 5′-UAA CUC AUA CUC CCG CAC CTT-3′. The cell was transfected with siRNA using RNAFit (Hanbio Biotechnology Co., Ltd.) at 37°C according to the manufacturer's protocol.

Western blotting

Total protein was extracted from cells using RIPA buffer (Shandong Sparkjade Scientific Instruments Co., Ltd.) and quantified using a BCA kit (Beijing Solarbio Science & Technology Co., Ltd.). Then 30 µg of protein per sample was resolved by 10% SDS-PAGE and blotted onto PVDF membranes. After blocking with 5% BSA (Beijing Solarbio Science & Technology Co., Ltd., A8020) for 2 h at room temperature, the membranes were incubated overnight with primary antibodies against RRS1 (1:1,000; Abcam; cat. no. ab188161), c-Myc (1:1,000; Abcam; cat. no. ab32072), NPM1 (1:1,000; Proteintech; cat. no. 60096-1-Ig), Lamin B1 (1:1,000; Proteintech; cat. no. 12987-1-AP), N-cadherin (1:1,000; Proteintech; cat. no. 22018-1-AP), E-cadherin (1:1,000; Proteintech; cat. no. 20874-1-AP), c-Myc (1:1,000; Proteintech; cat. no. 67447-1-Ig), RPL11 (1:1,000; Proteintech; cat. no. 16277-1-AP), Vimentin (1:1,000; Proteintech; cat. no. 10366-1-AP), RPL23 (1:1,000; ABclona; cat. no. A4292) and Snail (1:1,000; ABclona; cat. no. A11794) at 4°C, followed by the secondary antibodies at room temperature (1:3,000, Bioss; cat. nos. bs-40296G-HRP and bs-40295G-HRP) for 1 h. The positive bands were visualized using enhanced chemiluminescence (ECL) kit and the bands were measured using ImageJ (v1.53, National Institutes of Health).

Cell proliferation assay

BT549 cells were seeded into 96-well plates at the density of 2,000 cells/well. After 1, 2, 3, 4, 5 days of incubation at 37°C, 10 µl of CCK8 solution (Beijing Solarbio Science & Technology Co., Ltd.) solution was added to each well and the cells were incubated for 2 h at 37°C. The absorbance at 450 nm was measured using a microplate reader.

Reverse transcription-quantitative (RT-q) PCR analysis

Total RNA was extracted by adding 1 ml of TRIzol® Reagent (Thermo Fisher Scientific, Inc.) per 2×106 cells and using M-MLV Reverse Transcriptase (HiScript® III All-in-one RT SuperMix kit, Vazyme Biotech Co., Ltd.) reverse transcription 1 µg. qPCR was performed using the ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech Co., Ltd.) on ABI QuantStudio 3 (Applied Biosystems; Thermo Fisher Scientific, Inc.). The PCR amplification conditions were: Pre-denaturing at 95°C for 30 sec, followed by 45 cycles of denaturing at 95°C for 30 sec, annealing at 60°C for 30 sec and the melting curve was the system default. The relative gene expression levels were normalized to GAPDH. The primer sequences were as follows: RRS1 Forward: 5′-CCC TAC CGG ACA CCA GAG TAA-3′, Reverse: 5′-CCG AAA AGG GGT TGA AAC TTC C-3′; GAPDH Forward: 5′-AGA AGG CTG GGG CTC ATT TG-3′, Reverse: 5′-AGG GGC CAT CCA CAG TCT TC-3′. The above experiments followed the manufacturer′s protocol. The relative expression levels were calculated using the 2−∆∆Cq method (49).

Transwell and invasion assay

Transwell units were used to measure the migration and invasion capacity of BT549 cells in vitro. BT549 cells were seeded into the upper compartment of Transwell inserts (Corning, Inc.) at the density of 10,000 cells/well in serum-free media. For the invasion assay, the inserts were coated with Matrigel (35 µl per Transwell unit; diluted 1:8 with serum-free DMEM) at 4°C and incubated at 37°C for 2 h to gel the Matrigel. The lower compartments were filled with complete media. After 48 h of culture, the cells remaining on the upper surface of the membranes were removed with a cotton swab and those that migrated/invaded through the membranes were fixed with 4% paraformaldehyde for 10 min at room temperature and then stained with 0.1% hexamethylpararosaniline for 10 min at room temperature. Then three fields of view were randomly selected under an inverted microscope, images captured at ×100 and ×200 magnification, with counting and data analyzed from the ×100 image. Migration assay is used to detect the ability of cells to metastasize and invasion assay is used to detect the ability of cells to lyse the cell matrix.

Scratch test

The cells were seeded into a 6-well plate at the density of 2×105 cells/well and cultured until 90% confluent. Three lines were scratched on the monolayers with a 200 µl sterile pipette tip and were cultivated in serum-free medium. The scratched area was measured using ImageJ (v1.53, National Institutes of Health) at 0, 12, 24 and 48 h and the migration rate was calculated as (scratch area at 0 h-scratch area at 12, 24, 48 h)/scratch area at 0 h ×100%.

Co-immunoprecipitation (Co-IP)

Co-IP was performed with anti-c-Myc (1:1,000; Proteintech; cat. no. 67447-1-Ig) and anti-RPL11 (1:1,000; Proteintech; cat. no. 16277-1-AP) antibodies using the Classic Magnetic Protrin A/G IP/Co-IP kit (Epizyme, Inc.) according to the manufacturer's protocol. The kit contained Protein A/G magnetic beads, lysis/wash buffer, SDS-PAGE protein loading buffer (5X), elution buffer, neutralization buffer. Lysis/wash buffer and PMSF (1:100, Solarbio, P0100) were added at a ratio of 30 µl per 1.0×105 cells, mixed well and incubated on ice for 30 min (mixing several times during this period); collected by centrifugation (4°C; 12,000 × g; 10 min) with supernatant placed on ice for later use. Then 500 µl of the prepared sample was added to a 1.5 ml EP tube, followed by 4 µg antibody and incubated on a flip mixer (4°C overnight) to form antigen-antibody complexes. Magnetic bead suspension (25 µl) was placed into a 1.5 ml EP tube and 500 µl of lysis/wash buffer added, the magnetic beads were resuspended by gently pipetting and then let stand on a magnetic stand for 1 min. When the magnetic beads were adsorbed to the sidewall of the EP tube, the supernatant was aspirated and this step repeated twice. The antigen-antibody complex was added to the pretreated magnetic beads and incubated on an inversion mixer (4°C overnight). Then it was stood on the magnetic stand for 1 min, until the magnetic beads were adsorbed on the side wall of the EP tube. The supernatant was aspirated and discarded and what remained in the centrifuge tube was the antigen-antibody-magnetic bead complex. Lysis/rinse buffer (500 µl) was added to the antigen-antibody-magnetic bead complex, the magnetic beads resuspended by gently pipetting and agitation and then allowed to stand on the magnetic stand for 1 min until the magnetic beads were adsorbed to the side-wall of the centrifuge tube. The supernatant was aspirated and discarded and this step repeated twice. An appropriate amount of 5XSDS-PAGE loading buffer was added to the antigen-antibody-magnetic bead complex, mixed well and heated at 100°C for 10 min. Following cooling, the EP tube was placed on a magnetic stand for 1 min. After the magnetic beads were adsorbed on the side wall of the EP tube, the supernatant was collected and detected by SDS-PAGE. The immuno-precipitates were detected by western blotting as described above.

Luciferase activity assay

The dual-luciferase reporter gene detected the relationship between the transcription factor c-Myc and the Snail promoter and the 3′untranslated regions (UTRs) 2,000 bp upstream of the Snail transcription start site. The RPL11 and c-Myc overexpression plasmids and the luciferase SNAIL-pro reporter plasmid and Renilla luciferase plasmid were designed and constructed by Sangon Biotech Co., Ltd. LipoFiter 3.0 (Hanbio Biotechnology Co., Ltd.) was used for transfection according to the manufacturer's protocol. The BT549 cells were co-transfected with these constructs and lysed 48 h after transfection. Luciferase activity was measured using a luciferase detection kits (TransGen Biotech Co., Ltd.; cat. no. FR201-01) on a SpectraMax i3x Microplate Reader (Molecular Devices, LLC). The method of normalization was firefly luciferase activity comparison with Renilla luciferase activity.

Immunofluorescence

BT549 cells were seeded onto a round coverslip (Biosharp; cat. no. BS-14-RC) at the density of 2,000 cells/well. After 24 h, the cells were fixed with 4% paraformaldehyde for 10 min, permeabilized for with 2% Triton X-100 10 min, blocked in 10% goat serum (Wuhan Boster Biological Technology, Ltd.; cat. no. AR0009) for 30 min (the above steps were carried out at room temperature) and then incubated overnight with anti-RRS1 (1:200, Abcam; cat. no. ab188161), anti-RPL11 (1:200, Proteintech; cat. no. 16277-1-AP), anti-c-Myc (1:50, Thermo, MA5-12080), anti-NPM (1:200, Proteintech; cat. no. 60096-1-Ig), anti-Lamin B1 (1:200, Proteintech 12987-1-AP) and anti-Fibrillarin/U3 RNP (1:200, ABclona; cat. no. A0850) at 4°C. Subsequently, the cells were incubated with the appropriate fluorescent secondary antibody (1:200, ABclona; cat. no. AS039 and AS011) for 1 h at room temperature. Nuclei were stained using DAPI (Beijing Solarbio Science & Technology Co., Ltd.; cat. no. C0065) for 10 min at room temperature according to the manufacturer's protocol. The stained cells were viewed with a laser-scanning confocal microscope (Leica Stellaris 5; Leica Microsystems GmbH), 3 fields of view were randomly selected, observed and images captured at ×200 and ×630 magnification.

Cellular fractionation

BT549 cells were harvested and 6×106 cells were resuspended in 500 µl hypotonic buffer. After 10 min incubation on ice, they were centrifuged at 500 × g for 10 min at 4°C, and the supernatant containing the cytoplasmic fraction aspirated. The precipitate was resuspended in 300 µl S1 buffer, layered on 300 µl S2 buffer and centrifuged at 4°C, 1,500 × g for 5 min. The precipitated nuclear fraction was resuspended in 300 µl S2 buffer, sonicated for 30 sec with 60 sec intervals for 5 cycles at 4°C, 25 kHz, 60 W, layered on 300 µl S3 buffer and centrifuged at 4°C, 3,000 × g for 10 min. The nucleoplasmic fraction in the supernatant was separated and the precipitated nucleolar fraction was resuspended in RIPA buffer. Protease inhibitor cocktail was added throughout.

Reagent composition: Hypotonic buffer: 10 mM HEPES (pH 7.9), 10 mM KCl, 1.5 mM MgCl2, 0.5 mM DTT S1 buffer: 0.25 M sucrose, 10 mM MgCl2 S2 buffer: 0.35 M sucrose, 0.5 mM MgCl2 S3 buffer: 0.88 M sucrose, 0.05 mM MgCl2

Statistical analyses

The data was analyzed using the SPSS v13.0 program (SPSS, Inc.). All data was expressed as mean ± standard deviation (SD) and statistical significance was determined through one-way ANOVA analysis with Bonferroni post hoc test. P<0.05 was considered to indicate a statistically significant difference. Each experiment was repeated three times.

Results

RRS1 is upregulated in breast cancer cell lines

Tumor cells have a significantly higher rate of ribosome biosynthesis compared to normal cells in order to sustain rapid proliferation, which coincides with increased levels of RRS1. Consistent with this, we found that the expression of RRS1 mRNA and protein were significantly higher in four breast cancer cell lines (MDA-MB-231, BT549, MDA-MB-468 and MCF-7) compared to normal mammary epithelial cells (HMEC; Fig. 1A) and the highest levels were detected in the BT549 cells (Fig. 1B and C). Fibrillarin/U3 RNP as a nucleolar marker can accurately locate the nucleolus, immunofluorescence assay using in BT549 cells indicated that RRS1 was mainly located in the nucleus and nucleolus, followed by the cytoplasm (Fig. 1D). The sub-cellular localization of RRS1 was consistent with its role in ribosome biosynthesis.

RRS1 knockdown inhibited the proliferation, invasion and metastasis of BT549 cells

To determine the biological relevance of RRS1 in breast cancer progression, the present study knocked down RRS1 in BT-549 cells using an sh-RNA construct (Fig. 2A) and the knockdown efficiency was validated by the reduced protein and mRNA levels of RRS1 (Fig. 2B-D). RRS1 knockdown significantly decreased cell proliferation rates (Fig. 2E), as well as the migration and invasion abilities in vitro (Fig. 2F-K). Similarly, in the triple-negative breast cancer cell line MDA-MB-231, knocking down RRS1 also inhibited its invasion and metastasis ability (Fig. S1). Taken together, RRS1 is a key factor promoting the malignant phenotype of breast cancer cells.

RRS1 knockdown disrupted the ribosome assembly by altering the cellular localization of RPL11

RRS1 recruits RPL11 to the nucleolus during the synthesis of large ribosomal subunits, which are then exported to the cytoplasm for further maturation. Cytoplasmic and nuclear proteins were analyzed to determine whether RRS1 affected the cellular localization of RPL11 and it was found that knocking down RRS1 did not alter the levels of RPL11 in the cytoplasmic or nuclear fractions (Fig. 3A and B). By contrast, RRS1 knockdown significantly decreased RPL11 levels in the nucleolus and increased that it in nucleoplasm (Fig. 3C). Rea1 disengages the Rsa4 and Rpf2/RRS1 complex from Pre-60S, which converts 5S RNP to the correct configuration and exports it out of the nucleus. Deletion of RRS1 can inhibit the interaction between Rea1 and Rsa4 (50). Consistent with this, the present study found that the knockdown of RRS1 significantly decreased the interaction of RPL23 (a marker protein of Pre-60S) with RPL11 and RPL5 (Fig. 3D). Taken together, RRS1 disrupted ribosomal assembly by preventing the incorporation of RPL11 into the Pre-60S subunit, which in turn led to its dissociation into the nucleoplasm. The dissociation of RPL11 from the ribosomal complex allows it to bind to c-Myc and inhibit its transactivation (41). There is also evidence that RRS1 knockout inhibits the nuclear export of RPL11 and increases its accumulation in the nucleus (30). Consistent with these previous observations, the present study found that RRS1 knockdown significantly enhanced the interaction between RPL11 and c-Myc (Fig. 3E and F). Furthermore, the absence of RRS1 also increased the c-Myc accumulation in the nucleus, with a concomitant decrease in its cytoplasmic levels (Fig. 3A and B). These findings suggested that knocking down RRS1 enhances the interaction between RPL11 and c-Myc.

RPL11 inhibits c-Myc-driven SNAIL transcription

A previous study showed that c-Myc could promote the EMT, invasion and metastasis of tumor cells by activating the mesenchymal transcription factor SNAIL (51). As RPL11 inhibits c-Myc transactivation (38), the present study next examined whether RPL11 also affected c-Myc-dependent SNAIL transcription using the luciferase reporter assay. As shown in Fig. 4, while overexpression of c-Myc increased luciferase activity in 293T cells, the expression of RPL11 inhibited the function of c-Myc and reduced the increase in luciferase activity (Fig. 4). This indicated that RPL11 reduces SNAIL transcription by inhibiting c-Myc activity and may therefore block the EMT process.

RPL11 inhibits EMT during RRS1 knockdown

EMT is a dynamic process in which serves an important role in tumor invasion and metastasis. Consistent with our hypothesis, RRS1 knockdown increased the expression of the epithelial marker E-cadherin (Fig. 5) and Cytokeratin (Fig. S2) and reduced that of the mesenchymal markers such as SNAIL, N-cadherin and vimentin in BT549 cell (Fig. 5). Simultaneous knockdown of RRS1 and RPL11 restored the levels of the mesenchymal markers and downregulated E-cadherin (Fig. 5). Similar results were obtained in the MDA-MB-231 cell line (Fig. S3). Taken together, RRS1 may promotes EMT of breast cancer cells through the RPL11-c-Myc axis, which in turn promotes tumor invasion and metastasis.

Discussion

Breast cancer has a high mortality rate due to its metastatic nature (52). The clinical outcomes of surgery, radiotherapy, chemotherapy, endocrine therapy and targeted therapy are sub-optimal and the survival rate of breast cancer patients remains poor (53). The aim of the present study was to determine the role and underlying mechanism of RRS1 in the invasion and metastasis of breast cancer cells.

Increased ribosome biosynthesis is a prerequisite for sustaining the high proliferation rates of tumor cells (54,55). RRS1 is a regulator of ribosome biosynthesis and shows high expression levels in multiple tumors (14,24-28,30). Likewise, the present study also detected significantly higher levels of RRS1 in the breast cancer cell lines compared to normal breast epithelial cells. Knocking down RRS1 inhibited the invasion, migration and proliferation of BT549 cells, indicating that RRS1 functions as an oncogene in breast cancer. From a previous study, miRNA-148a inhibits proliferation, migration and invasion by downregulating the expression of RRS1 and promotes the apoptosis of cervical cancer cells (23). RRS1 forms a complex with Rpf2, which recruits 5S RNP onto the Pre-60S subunit. RPL5 also interacts with Rsa4, which integrates the 5S RNP onto Pre-60S (56). Subsequently, Rea1, an AAA+ (ATPases associated with various cellular activities) family member, interacts with the ubiquitin-like (UBL) domain of Rsa4 and releases Rsa4 in an ATP-dependent manner, which is followed by the release of the Rpf2/RRS1 complex (50,57,58). This rotates the conformation of 5S RNP, which fixes it on Pre-60S and is necessary for Pre-60S export. RRS1 ensures correct binding of the Rsa4 UBL domain with Rea1 (50). In the absence of RRS1 therefore, Pre-60S maturation is blocked and it is released from the nucleolus to the nucleoplasm and cannot be exported to the cytoplasm (18,50). The present study found that while RRS1 knockdown did not affect the total content of RPL11, it shifted the localization of RPL11 from the nucleolus to the nucleoplasm. Consistent with this, RPL23 also accumulated in the nucleus after RRS1 knockdown. In addition, deletion of RRS1 is known to inhibit the incorporation of RPL11 and RPL5 into Pre-60S, thereby affecting Pre-60S assembly (18). Taken together, RRS1 depletion prevents the incorporation of 5S RNP into Pre-60S, which hinders the nuclear export of pre ribosomes and inhibits ribosome biosynthesis.

C-Myc is a pro-survival protein (59), which is often overexpressed in tumor cells (60,61). RPL11 can regulate the expression of c-Myc at both mRNA and protein levels (38,62). Dai et al (38) found that RPL11 could bind to the Myc box II (MB II) of c-Myc at c-Myc target gene promoter and inhibit the histone H4 acetylation at c-Myc target nucleolin gene promoter and recruitment of c-Myc coactivator TRRAP, finally reducing the transcription of the c-Myc downstream target gene. In addition, RPL11 can bind to 3′-UTR at c-Myc mRNA, recruit microRNA-induced silencing complex (miRISC) assembled by mir-24 and combine with Ago2 promote the interaction of miRISC and 3′-UTR, leading to c-Myc mRNA degradation (63). RPL11 and RPL5 can also suppress c-Myc expression in a synergistic manner (62). The present study found that RRS1 knockdown increased the interaction between RPL11 and c-Myc and inhibited the transactivation of SNAIL by c-Myc. Similarly, knocking down RRS1 reduced the levels of SNAIL, N-cadherin and VIM proteins and increased that of E-cadherin. Knockdown of RPL11 reversed the inhibitory effect of RRS1 deletion. Thus, RRS1 regulated the function of c-Myc and EMT of breast cancer cells through RPL11.

To summarize, the present study showed that RRS1 may promote the EMT and metastasis of breast cancer cells by regulating ribosome assembly and biosynthesis. Depletion of RRS1 prevented nuclear export of the Pre-60s subunit and increased the accumulation of RPL11 in the nucleoplasm. This enhanced the interaction between RPL11 and c-Myc and decreased the transcription of the c-Myc target SNAIL, eventually inhibiting EMT (Fig. 6). Therefore, the RPL11-c-Myc-SNAIL axis may as a potential therapeutic target in breast cancer. However, it remains to expound the effect of RRS1 on the RPL11-c-Myc-SNAIL axis and its role in breast cancer.

Supplementary Data

Availability of data and materials

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

Authors' contributions

LH provided the study conception and design. RW analyzed the data, edited the figures and wrote the manuscript. CP and JS contributed to data curation. RW, YH, QW, LD, YC, JZ, LZ, LW performed the experiments. LH reviewed the manuscript. LH and RW confirm the authenticity of all the raw data. All authors contributed to the article and all authors reviewed and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81472542) and Chinese Medicine Projects in Shandong Province (grant no. 2021Z036).

References

1 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Roulot A, Héquet D, Guinebretière JM, Vincent-Salomon A, Lerebours F, Dubot C and Rouzier R: Tumoral heterogeneity of breast cancer. Ann Biol Clin (Paris). 74:653–660. 2016.

3 

Yeo SK and Guan JL: Breast cancer: Multiple Subtypes within a Tumor? Trends Cancer. 3:753–760. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Gao JJ and Swain SM: Luminal A breast cancer and molecular assays: A review. Oncologist. 23:556–565. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Ades F, Zardavas D, Bozovic-Spasojevic I, Pugliano L, Fumagalli D, de Azambuja E, Viale G, Sotiriou C and Piccart M: Luminal B breast cancer: Molecular characterization, clinical management, and future perspectives. J Clin Oncol. 32:2794–2803. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Prat A, Pineda E, Adamo B, Galván P, Fernández A, Gaba L, Díez M, Viladot M, Arance A and Muñoz M: Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast. 24(Suppl 2): S26–S35. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Yin L, Duan JJ, Bian XW and Yu SC: Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 22:612020. View Article : Google Scholar : PubMed/NCBI

8 

Hao S, Ha L, Cheng G, Wan Y, Xia Y, Sosnoski DM, Mastro AM and Zheng SY: A Spontaneous 3D Bone-On-a-Chip for bone metastasis study of breast cancer cells. Small. 14:e17027872018. View Article : Google Scholar : PubMed/NCBI

9 

Watase C, Shiino S, Shimoi T, Noguchi E, Kaneda T, Yamamoto Y, Yonemori K, Takayama S and Suto A: Breast cancer brain metastasis-overview of disease state, treatment options and future perspectives. Cancers (Basel). 13:10782021. View Article : Google Scholar

10 

Maughan KL, Lutterbie MA and Ham PS: Treatment of breast cancer. Am Fam Physician. 81:1339–1346. 2010.PubMed/NCBI

11 

Elsayed M, Alhussini M, Basha A and Awad AT: Analysis of loco-regional and distant recurrences in breast cancer after conservative surgery. World J Surg Oncol. 14:1442016. View Article : Google Scholar : PubMed/NCBI

12 

Salata C, deAlmeida CE, Ferreira-Machado SC, Barroso RC, Nogueira LP, Mantuano A, Pickler A, Mota CL and de Andrade CBV: Preliminary pre-clinical studies on the side effects of breast cancer treatment. Int J Radiat Biol. 97:877–887. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Taylor CW and Kirby AM: Cardiac Side-effects from breast cancer radiotherapy. Clin Oncol (R Coll Radiol). 27:621–629. 2015. View Article : Google Scholar

14 

Tsuno A, Miyoshi K, Tsujii R, Miyakawa T and Mizuta K: RRS1, a conserved essential gene, encodes a novel regulatory protein required for ribosome biogenesis in Saccharomyces cerevisiae. Mol Cell Biol. 20:2066–2074. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Hua Y, Song J, Peng C, Wang R, Ma Z, Zhang J, Zhang Z, Li N and Hou L: Advances in the relationship between regulator of ribosome Synthesis 1 (RRS1) and diseases. Front Cell Dev Biol. 9:6209252021. View Article : Google Scholar : PubMed/NCBI

16 

Granneman S and Baserga SJ: Ribosome biogenesis: Of knobs and RNA processing. Exp Cell Res. 296:43–50. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Baßler J and Hurt E: Eukaryotic ribosome assembly. Annu Rev Biochem. 88:281–306. 2019. View Article : Google Scholar

18 

Zhang J, Harnpicharnchai P, Jakovljevic J, Tang L, Guo Y, Oeffinger M, Rout MP, Hiley SL, Hughes T and Woolford JL Jr: Assembly factors Rpf2 and Rrs1 recruit 5S rRNA and ribosomal proteins rpL5 and rpL11 into nascent ribosomes. Genes Dev. 21:2580–2592. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Gambe AE, Matsunaga S, Takata H, Ono-Maniwa R, Baba A, Uchiyama S and Fukui K: A nucleolar protein RRS1 contributes to chromosome congression. FEBS Lett. 583:1951–1956. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Horigome C, Okada T, Shimazu K, Gasser SM and Mizuta K: Ribosome biogenesis factors bind a nuclear envelope SUN domain protein to cluster yeast telomeres. EMBO J. 30:3799–3811. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Miyoshi K, Tsujii R, Yoshida H, Maki Y, Wada A, Matsui Y, Toh-E A and Mizuta K: Normal assembly of 60S ribosomal subunits is required for the signaling in response to a secretory defect in Saccharomyces cerevisiae. J Biol Chem. 277:18334–18339. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Carnemolla A, Fossale E, Agostoni E, Michelazzi S, Calligaris R, De Maso L, Del Sal G, MacDonald ME and Persichetti F: Rrs1 is involved in endoplasmic reticulum stress response in Huntington disease. J Biol Chem. 284:18167–18173. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Sun B, Zhao L, Liu Z, Xu Z, Tian Y and Hao C: Up-regulation of miRNA-148a inhibits proliferation, invasion, and migration while promoting apoptosis of cervical cancer cells by down-regulating RRS1. Biosci Rep. 39:BSR201818152019. View Article : Google Scholar : PubMed/NCBI

24 

Wang J, Li Z, Zuo C, Xie Q, Li H, Jia J, Zhen Z, Qi R, Li Z, Liu D and Sun B: Knockdown of RRS1 by lentiviral-mediated RNAi promotes apoptosis and suppresses proliferation of human hepatocellular carcinoma cells. Oncol Rep. 38:2166–2172. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Hua YN, Song JL, Ma ZL, Wu L, Zhang Z, Zhang L, Li N, Cong SB and Hou L: Effect of RRS1 gene knockdown on BT549 cell line proliferation and apoptosis in breast cancer. Neoplasma. 66:28–32. 2019. View Article : Google Scholar

26 

Song J, Ma Z, Hua Y, Xu J, Li N, Ju C and Hou L: Functional role of RRS1 in breast cancer cell proliferation. J Cell Mol Med. 22:6304–6313. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Chen F, Jin Y, Feng L, Zhang J, Tai J, Shi J, Yu Y, Lu J, Wang S, Li X, et al: RRS1 gene expression involved in the progression of papillary thyroid carcinoma. Cancer Cell Int. 18:202018. View Article : Google Scholar : PubMed/NCBI

28 

XL Wu, Yang ZW, He L, Dong PD, Hou MX, Meng XK, Zhao HP, Wang ZY, Wang F, Baoluri, et al: RRS1 silencing suppresses colorectal cancer cell proliferation and tumorigenesis by inhibiting G2/M progression and angiogenesis. Oncotarget. 8:82968–82980. 2017. View Article : Google Scholar

29 

Ma Y, Yan F, Wei W, Deng J, Li L, Liu L and Sun J: MicroRNA-598 inhibits the growth and maintenance of gastric cancer stem-like cells by down-regulating RRS1. Cell Cycle. 18:2757–2769. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Cao P, Yang A, Li P, Xia X, Han Y, Zhou G, Wang R, Yang F, Li Y, Zhang Y, et al: Genomic gain of RRS1 promotes hepatocellular carcinoma through reducing the RPL11-MDM2-p53 signaling. Sci Adv. 7:eabf43042021. View Article : Google Scholar :

31 

Calviño FR, Kharde S, Ori A, Hendricks A, Wild K, Kressler D, Bange G, Hurt E, Beck M and Sinning I: Symportin 1 chaperones 5S RNP assembly during ribosome biogenesis by occupying an essential rRNA-binding site. Nat Commun. 6:65102015. View Article : Google Scholar : PubMed/NCBI

32 

Zhang Y, Wolf GW, Bhat K, Jin A, Allio T, Burkhart WA and Xiong Y: Ribosomal protein L11 negatively regulates oncoprotein MDM2 and mediates a p53-dependent ribosomal-stress checkpoint pathway. Mol Cell Biol. 23:8902–8912. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Haupt Y, Maya R, Kazaz A and Oren M: Mdm2 promotes the rapid degradation of p53. Nature. 387:296–299. 1997. View Article : Google Scholar : PubMed/NCBI

34 

Momand J, Zambetti GP, Olson DC, George D and Levine AJ: The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 69:1237–1245. 1992. View Article : Google Scholar : PubMed/NCBI

35 

Bursać S, Brdovčak MC, Pfannkuchen M, Orsolić I, Golomb L, Zhu Y, Katz C, Daftuar L, Grabušić K, Vukelić I, et al: Mutual protection of ribosomal proteins L5 and L11 from degradation is essential for p53 activation upon ribosomal biogenesis stress. Proc Natl Acad Sci USA. 109:20467–20472. 2012. View Article : Google Scholar

36 

Horn HF and Vousden KH: Cooperation between the ribosomal proteins L5 and L11 in the p53 pathway. Oncogene. 27:5774–5784. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Wang HT, Chen TY, Weng CW, Yang CH and Tang MS: Acrolein preferentially damages nucleolus eliciting ribosomal stress and apoptosis in human cancer cells. Oncotarget. 7:80450–80464. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Dai MS, Arnold H, Sun XX, Sears R and Lu H: Inhibition of c-Myc activity by ribosomal protein L11. EMBO J. 26:3332–3345. 2007. View Article : Google Scholar : PubMed/NCBI

39 

van Riggelen J, Yetil A and Felsher DW: MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer. 10:301–309. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Dai MS, Sears R and Lu H: Feedback regulation of c-Myc by ribosomal protein L11. Cell Cycle. 6:2735–2741. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Wang X, Ren Y, Wang Z, Xiong X, Han S, Pan W, Chen H, Zhou L, Zhou C, Yuan Q and Yang M: Down-regulation of 5S rRNA by miR-150 and miR-383 enhances c-Myc-rpL11 interaction and inhibits proliferation of esophageal squamous carcinoma cells. FEBS Lett. 589:3989–3997. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Gao X, Liu X, Lu Y, Wang Y, Cao W, Liu X, Hu H and Wang H: PIM1 is responsible for IL-6-induced breast cancer cell EMT and stemness via c-myc activation. Breast Cancer. 26:663–671. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Yang J, Wu SP, Wang WJ, Jin ZR, Miao XB, Wu Y, Gou DM, Liu QZ and Yao KT: A novel miR-200c/c-myc negative regulatory feedback loop is essential to the EMT process, CSC biology and drug sensitivity in nasopharyngeal cancer. Exp Cell Res. 391:1118172020. View Article : Google Scholar : PubMed/NCBI

44 

Tao L, Shu-Ling W, Jing-Bo H, Ying Z, Rong H, Xiang-Qun L, Wen-Jie C and Lin-Fu Z: MiR-451a attenuates doxorubicin resistance in lung cancer via suppressing epithelialmesenchymal transition (EMT) through targeting c-Myc. Biomed Pharmacother. 125:1099622020. View Article : Google Scholar : PubMed/NCBI

45 

Liu N, Wang Z, Liu D and Xie P: HOXC13-AS-miR-122-5p-S ATB1-C-Myc feedback loop promotes migration, invasion and EMT process in glioma. Onco Targets Ther. 12:7165–7173. 2019. View Article : Google Scholar :

46 

Lin X, Sun R, Zhao X, Zhu D, Zhao X, Gu Q, Dong X, Zhang D, Zhang Y, Li Y and Sun B: C-myc overexpression drives melanoma metastasis by promoting vasculogenic mimicry via c-myc/snail/Bax signaling. J Mol Med (Berl). 95:53–67. 2017. View Article : Google Scholar

47 

Wang K, Zheng J, Yu J, Wu Y, Guo J, Xu Z and Sun X: Knockdown of MMP-1 inhibits the progression of colorectal cancer by suppressing the PI3K/Akt/c-myc signaling pathway and EMT. Oncol Rep. 43:1103–1112. 2020.PubMed/NCBI

48 

Nieto MA, Huang RY, Jackson RA and Thiery JP: EMT: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI

49 

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

50 

Madru C, Lebaron S, Blaud M, Delbos L, Pipoli J, Pasmant E, Réty S and Leulliot N: Chaperoning 5S RNA assembly. Genes Dev. 29:1432–1446. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Smith AP, Verrecchia A, Fagà G, Doni M, Perna D, Martinato F, Guccione E and Amati B: A positive role for Myc in TGFbeta-induced Snail transcription and epithelial-to-mesenchymal transition. Oncogene. 28:422–430. 2009. View Article : Google Scholar

52 

Azamjah N, Soltan-Zadeh Y and Zayeri F: Global trend of breast cancer mortality rate: A 25-year study. Asian Pac J Cancer Prev. 20:2015–2020. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Henson KE, McGale P, Darby SC, Parkin M, Wang Y and Taylor CW: Cardiac mortality after radiotherapy, chemotherapy and endocrine therapy for breast cancer: Cohort study of 2 million women from 57 cancer registries in 22 countries. Int J Cancer. 147:1437–1449. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Penzo M, Montanaro L, Treré D and Derenzini M: The ribosome biogenesis-cancer connection. Cells. 8:552019. View Article : Google Scholar

55 

Nait Slimane S, Marcel V, Fenouil T, Catez F, Saurin JC, Bouvet P, Diaz JJ and Mertani HC: Ribosome biogenesis alterations in colorectal cancer. Cells. 9:23612020. View Article : Google Scholar :

56 

Baßler J, Paternoga H, Holdermann I, Thoms M, Granneman S, Barrio-Garcia C, Nyarko A, Lee W, Stier G, Clark SA, et al: A network of assembly factors is involved in remodeling rRNA elements during preribosome maturation. J Cell Biol. 207:481–498. 2014. View Article : Google Scholar

57 

Ulbrich C, Diepholz M, Bassler J, Kressler D, Pertschy B, Galani K, Böttcher B and Hurt E: Mechanochemical removal of ribosome biogenesis factors from nascent 60S ribosomal subunits. Cell. 138:911–922. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Micic J, Li Y, Wu S, Wilson D, Tutuncuoglu B, Gao N and Woolford JL Jr: Coupling of 5S RNP rotation with maturation of functional centers during large ribosomal subunit assembly. Nat Commun. 11:37512020. View Article : Google Scholar : PubMed/NCBI

59 

Davis AC, Wims M, Spotts GD, Hann SR and Bradley A: A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev. 7:671–682. 1993. View Article : Google Scholar : PubMed/NCBI

60 

Destefanis F, Manara V and Bellosta P: Myc as a regulator of ribosome biogenesis and cell competition: A link to cancer. Int J Mol Sci. 21:40372020. View Article : Google Scholar :

61 

Wu H, Yang TY, Li Y, Ye WL, Liu F, He XS, Wang JR, Gan WJ, Li XM, Zhang S, et al: Tumor necrosis factor receptor-associated Factor 6 promotes hepatocarcinogenesis by interacting with histone deacetylase 3 to enhance c-Myc Gene expression and protein stability. Hepatology. 71:148–163. 2020. View Article : Google Scholar

62 

Liao JM, Zhou X, Gatignol A and Lu H: Ribosomal proteins L5 and L11 co-operatively inactivate c-Myc via RNA-induced silencing complex. Oncogene. 33:4916–4923. 2014. View Article : Google Scholar

63 

Challagundla KB, Sun XX, Zhang X, DeVine T, Zhang Q, Sears RC and Dai MS: Ribosomal protein L11 recruits miR-24/miRISC to repress c-Myc expression in response to ribosomal stress. Mol Cell Biol. 31:4007–4021. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2022
Volume 60 Issue 3

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
Wang R, Peng C, Song J, Hua Y, Wu Q, Deng L, Cao Y, Zhang J, Zhang L, Wu L, Wu L, et al: Downregulated RRS1 inhibits invasion and metastasis of BT549 through RPL11‑c‑Myc‑SNAIL axis. Int J Oncol 60: 33, 2022
APA
Wang, R., Peng, C., Song, J., Hua, Y., Wu, Q., Deng, L. ... Hou, L. (2022). Downregulated RRS1 inhibits invasion and metastasis of BT549 through RPL11‑c‑Myc‑SNAIL axis. International Journal of Oncology, 60, 33. https://doi.org/10.3892/ijo.2022.5323
MLA
Wang, R., Peng, C., Song, J., Hua, Y., Wu, Q., Deng, L., Cao, Y., Zhang, J., Zhang, L., Wu, L., Hou, L."Downregulated RRS1 inhibits invasion and metastasis of BT549 through RPL11‑c‑Myc‑SNAIL axis". International Journal of Oncology 60.3 (2022): 33.
Chicago
Wang, R., Peng, C., Song, J., Hua, Y., Wu, Q., Deng, L., Cao, Y., Zhang, J., Zhang, L., Wu, L., Hou, L."Downregulated RRS1 inhibits invasion and metastasis of BT549 through RPL11‑c‑Myc‑SNAIL axis". International Journal of Oncology 60, no. 3 (2022): 33. https://doi.org/10.3892/ijo.2022.5323