Abstract
Aim: To analyze the biological role of the long non-coding RNA LINK-A. Materials and Methods: An 850-bp segment from the second exon of LINK-A was removed using the CRISPR/Cas9 system in OVCA433 ovarian serous carcinoma cells. Spheroid formation, migration, invasion, proliferation, matrix metalloproteinase (MMP) activity and expression of cell-signaling proteins were assessed in vitro. Results: OVCA433 cells with LINK-A deletion were more invasive (p=0.0008) but had reduced migration and MMP9 secretion compared to controls (p=0.003 and p=0.005, respectively). LINK-A deletion did not affect proliferation but induced phosphorylation of extracellular signal-regulated kinase (10-fold; p=0.005). LINK-A knock out additionally reduced spheroid formation. Conclusion: Added to our previous data from analysis of clinical specimens, LINK-A is likely to be a tumor suppressor.
Ovarian cancer (OC) is the most aggressive gynecological cancer. OC was predicted to be diagnosed in 22,530 women and lead to 13,980 fatalities in the U.S. in 2019 (1). Globally, OC was diagnosed in 295,414 women and led to 184,799 deaths in 2018, making it the eighth most common and most lethal cancer in women (2). Late diagnosis and chemotherapy resistance are two major factors contributing to the fact that prognosis remains dismal for the majority of patients (3).
Recent years have brought growing interest in non-coding RNA. Long non-coding RNAs (lncRNAs) are longer than 200 nucleotides and are involved in many different cellular processes and pathways. In cancer, lncRNAs have been found to be involved in post-transcriptional modifications and decoy of mRNA, proliferation and immortality, maintenance of genomic stability, invasion and metastasis, and drug resistance (4). LINK-A (LINC01139; also known as LOC339535 and NR_015407) is a ~1.5-kb intergenic noncoding RNA located on chromosome 1 at the 238,480,372-238,486,05 site on the antisense strand. It consists of two exons, of which the second contains functional sites consisting of a phosphatidylcholine-binding site, breast tumor kinase (BRK)-binding sites #1 and #2, a phosphatidylinositol-3,4,5-trisphosphate-binding loop and a leucine-rich repeat kinase 2 binding site. This lncRNA was first described in triple-negative breast cancer, where it was shown to phosphorylate and stabilize hypoxia-inducible factor 1α (HIF1α), and thereby promote tumorigenesis (5). In a subsequent study by the same group, LINK-A was shown to hyperactivate protein kinase B (AKT) and mediate resistance to AKT inhibitors (6). The ability of LINK-A to activate HIF1α was also subsequently demonstrated in OC (7). In OC, LINK-A was further found to activate the transforming growth factor-β pathway and to promote migration and invasion (8).
High-grade serous carcinoma (HGSC) is the most common histotype of OC, and is responsible for the majority of deaths from this disease. In a recent study of clinical HGSC specimens, we compared LINK-A expression at different anatomic sites affected by HGSC. LINK-A was overexpressed in solid metastases from the abdominal cavity compared to HGSC effusions (malignant ascites and pleural effusions), the ovarian tumors and extracellular material (exosomes) derived from HGSC effusion supernatants, suggesting upregulation during tumor progression. Conversely, higher LINK-A levels in post-chemotherapy HGSC effusions were significantly related to longer progression-free and overall survival, the latter finding being retained as independent prognosticator in multivariate Cox analysis, suggesting that LINK-A may have a tumor suppressor role (9). Given this apparent discrepancy, we wished to study the biological role of this molecule in OC. In the present study, we used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology to knock out LINK-A and study the effect of this change in vitro in OC cells.
Materials and Methods
Cell culture. OC cell lines included in this study were OVCAR-3 (HGSC), OVCAR-8 (HGSC), OVCA433 (serous adenocarcinoma), OC-238 (serous cystadenocarcinoma), OVCA 429 (cystadenocarcinoma), DOV13 (adenocarcinoma) and ES-2 (clear-cell carcinoma). OVCA433, OVCA429 and DOV13 cells were a kind gift from Professor Robert Bast, MD Anderson Cancer Center, Houston TX, USA. The remaining cell lines were purchased from the American Tissue Culture Collection (Manassas VA, USA).
Cell lines were cultured at 37°C with 5% CO2 in the following media: OVCAR-3, OC-238 and ES-2: Dulbecco’s modified Eagle’s medium-high glucose (Sigma-Aldrich, St. Louis, MO, USA) with 10% fetal calf serum (FCS; Sigma-Aldrich); OVCA433, OVCA429 and DOV13: MEM-EAGLA Earle’s Salts Base (MEM; Biological Industries, Kibbutz Beit-Haemek, Israel) with 10% FCS; OVCAR-8: RPMI medium 1640 (GIBCO, Thermo Fisher Scientific, Waltham, MA, USA) with 5% FCS.
All media were supplemented with 1% MEM-Eagle non-essential amino acid solution, 1% L-glutamine solution, 1% MEM vitamin solution, 1% sodium pyruvate solution, 1% penicillin-streptomycin-amphotericin B Solution (Biological Industries).
CRISPR/Cas9 knock out. LINK-A active sites are found on the second exon, and we therefore decided to delete it. To achieve this goal, we designed two guides that cut out ~870 bases of the second exon. The guides were designed with the help of Massachusetts Institute of Technology (https://zlab.bio/guide-design-resources) and University of California, Santa Cruz (http://genome.ucsc.edu/index.html) web sites. Each guide was cloned into a PX330 plasmid (pX330-U6-Chimeric_BB-CBh-hSpCas9; a kind gift from Dr. Yosef Buganim, The Hebrew University of Jerusalem, Israel).
PX330 plasmid transfection into OVCA433 cells was performed using Avalanche-Everyday (EZ Biosystems, College Park, MD, USA) with 1.25 μg of each guide plasmid and 0.5 μg of puromycin-resistant plasmid (Fuw-original-puro-2A-EGFP was a kind gift from Dr. Yosef Buganim) co-transfected for 24 hours, followed by puromycin selection for another 48 hours. The transfected cells were then seeded as single cells in a 96-well plate.
Spheroid formation. Control and knock out OVCA433 cells (n=400,000) were seeded on a 6-well plate dish and agitated at 20 rpm for 24 hours, then photographed. The medium was collected for RNA isolation.
Scratch assay/migration assay. OVCA433 cells (n=100-200,000) were seeded on a 12-well plate dish and cultured to confluence. A scratch was made with a 1 ml pipette tip and images were taken at 0, 6 and 24 hours. Results were analyzed using the ImageJ program (NIH, Bethesda, MD, USA).
Cell-proliferation assay. OVCA433 cells (n=100,000) were seeded on a 12-well plate. At 0 and 24 hours, the cells were treated with 2 mg/ml of 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Calbiochem, San Diego, CA, USA) for 20 minutes at 37°C in an incubator with 5% CO2. Cells were subsequently lysed using 150 μl dimethyl sulfoxide (Sigma-Aldrich) and absorption at 570 nm was then measured by a Cytation 3 instrument (BioTek Instruments, Inc., Winooski, VT, USA).
Matrigel invasion assay and motility assay. Nuclepore polycarbonate filters (13 mm, 8 μm pore size, polyvinylpyrrolidone-free; Whatman International Ltd, Maidstone, UK) were coated with extract of basement membrane components (Matrigel, 25 μg per filter) for the chemo-invasion assay or with 5 μg collagen IV for the motility assay, and placed in Boyden chambers. OVCA433 cells (n=200,000) were resuspended in serum-free medium and placed in the upper compartment of the Boyden chambers. As chemoattractant, fibroblast-conditioned medium (obtained from NIH-3T3 cells) was used. After 5 or 24 hours of incubation at 37°C in an incubator with 5% CO2, the lower surface of each filter was stained in DiffQuik (Medion Diagnostics International Inc., Miami, FL, USA) and the number of cells that invaded was counted in five random fields.
Matrix metalloproteinase (MMP) activity assay (zymography). The assay was performed on medium collected from Boyden chambers at the end of the Matrigel invasion experiment. Medium from OVCA433 control and knock out cells was loaded onto 10% sodium-dodecylsulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) gels with 1 mg/ml gelatin (Sigma-Aldrich), as previously described (10).
RNA extraction and cDNA generation. RNA was extracted from OVCA433 cells using Bio-Tri reagent (Bio-Lab Ltd, Jerusalem, Israel) according to the manufacturer’s protocol. RNA concentration was measured by a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific).
Prior to cDNA formation, 1 μg of extracted RNA was subjected to DNA degradation by PerfeCTa DNase I (Quanta Biosciences, Gaithersburg, MD, USA). cDNA was created by qScript cDNA synthesis kit (Quanta Biosciences).
Quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR) and RT-PCR. qRT-PRC was performed using the Fast SYBRTM Green Master Mix (Applied Biosystems by Thermo Fisher Scientific, Foster City, CA, USA) with specific primers in CFX Connect Real-Time system (Bio-Rad Laboratories, Hercules, CA). RT-PCR was carried out using Hy-Taq Ready Mix (2X) (Hylabs, Rehovot, Israel) with specific primers in PCR TOUCH T960 (Hangzhou Jingle Scientific Instrument Co., Ltd, Hangzhou, China). PCR products were loaded onto 1.5% agarose gel with 100-bp DNA ladder (GeneDireX Inc., Taoyuan, Taiwan, ROC). Gene expression was normalized to that for ribosomal protein lateral stalk subunit P0 (RPLP0).
Primer sequences were as follows: LINK-A: Forward: AACCAGTCAC CCAACCAGAG, reverse: CACAGGCCAGATGGAGTTTT; RPLP0: forward: CCAACTACTTCCTTAAGATCATCCAACTA, reverse: ACATGCGGATCTGCTGCA.
Western blot. Total protein isolation was performed using RIPA (1% NP-40, 20 mM Tris-HCl (pH 7.5), 137 mM NaCl, 0.5 mM EDTA, 10% glycerol, 1% protease inhibitor cocktail (Millipore, Burlington, MA, USA), 1 mM sodium orthovanadate and 0.1% sodium dodecyl sulfate (SDS). Twenty-five micrograms of protein was loaded onto 10% SDS-PAGE gel. The proteins were then transferred to Immobilon polyvinylidene difluoride membrane (Millipore). Subsequently, the membrane was blocked by 5% Difco skim milk (BD Biosciences, San Jose, CA, USA) for 1 hour, and then incubated for 16 hours at 4°C with one of the following rabbit monoclonal antibodies (all from Cell Signaling Biotechnology, Danvers, MA, USA): glyceraldehyde 3-phosphate dehydrogenase , clone 14C10; p44/42 mitogen-activated protein kinase (MAPK) (ERK1/2), clone 137F5; p-p44/42 MAPK (ERK1/2) (Thr202/Tyr204), clone 197G2.
Statistical analysis. Student’s t-test was applied.
Results
LINK-A expression in OC cell lines. LINK-A expression was analyzed in seven OC cell lines, namely OVCAR-3, OVCAR-8, OC-238, OVCA429, OVCA433, DOV13 and ES2, and in two forms of culture: 2D monolayer and 3D spheroids. LINK-A was expressed in the OVCA433, DOV13 and ES2 cell lines. In OVCA433 cells, expression was low in 2D monolayer and high in spheroids (p<0.0001; Figure 1).
LINK-A CRISPR/Cas9 knock out. We performed knock out of the second exon of LINK-A using the CRISPR/Ca9 system in OVCA433 cells. Figure 2 shows the change in LINK-A secondary structure following knock out.
LINK-A knock out affects invasion and migration. The invasiveness of knock out cells was strongly up-regulated following the loss of LINK-A to 452% of the control level (p=0.0008; Figure 3A). In contrast, in wound-healing assay, knock out cells closed the gap less efficiently than did control cells, with closure at 54% vs. 70%, respectively, after 24 hours (p=0.003; Figure 3B).
LINK-A knock out reduces MMP secretion. We examined MMP secretion in two states: cells cultured in serum-free medium for 24 hours and cells in the invasion assay, studied at 5 hours. No MMPs were found in the medium from the nontransfected cells, whereas the medium from the invasion assay, obtained from both the OVCA433 nontransfected and knock out cells, had MMP2 and MMP9 activity. A significant difference was found only for MMP9, which was reduced to 70% in knock out cell cultures (p=0.005; Figure 3C).
LINK-A does not affect cell proliferation. To find whether LINK-A is involved in cell proliferation we performed an MTT assay for 24 hours. No significant difference was found between the numbers of control and knock out cells (p>0.05; Figure 3D).
Suppression of LINK-A leads to a change in spheroid structure and size. As LINK-A was highly expressed in spheroids, we assessed the effect of knock out on their formation. Spheroids from OVCA433 knock-out cells were much smaller and less spherical compared to those formed by nontransfected control cells (Figure 4).
LINK-A suppresses ERK1/2 phosphorylation. In OVCA433 knock-out cells, levels of ERK1/2 expression were not significantly altered but the phosphorylation was elevated about 2.5-fold (p=0.0015; Figure 5).
Discussion
Research focusing on lncRNAs is a relatively new field in molecular biology but in recent years an increasing number of studies have documented the important role of these molecules in the evolution and progression of cancer. In OC, lncRNAs have been shown to be involved in invasion, proliferation, cell-cycle regulation, apoptosis, and drug resistance (11-13). Moreover, the association between lncRNA expression and patient survival has been reported by us and others (9, 14).
In the current study, we focused on the lncRNA LINK-A. In addition to its role in breast carcinoma and OC discussed above, LINK-A was recently reported to have a biological and clinical role in other cancer types. LINK-A promotes migration, invasion and proliferation in pancreatic adenocarcinoma, apparently through up-regulation of Rho-associated protein kinase 1 (ROCK1) (15). It was similarly reported to promote migration and invasion in non-small cell lung carcinoma. In this tumor type, plasma LINK-A levels were higher in patients with metastatic disease compared to those without and healthy controls (16).
Similar results have been reported in non-epithelial cancer. Plasma LINK-A levels were higher in patients with glioma and mantle-cell lymphoma compared to healthy controls, and this molecule prevented apoptosis through up-regulation of survivin in vitro (17, 18). Promotion of migration and invasion through increased HIF1α level in vitro along with higher plasma levels compared to controls were also seen in osteosarcoma (19).
Using the CRISPR/Cas9 system, we deleted the majority of the second exon and thereby eliminated LINK-A functional sites and disrupted its secondary structure. We found that LINK-A knock out suppressed migration and MMP secretion, although it increased invasion, with no effect on proliferation. Importantly, we observed that OVCA433 knock out cells did not form spheroids. Together with the high level of LINK-A initially found in spheroids, we hypothesize that this lncRNA is critical for their formation. Additionally, LINK-A knock out increased ERK1/2 phosphorylation. A previous study showed that LINK-A can activate AKT (3) but in our knock-out cells, we found no significant change in the level of AKT or its phosphorylation (data not shown).
In summary, the present study showed that LINK-A is involved in critical cellular processes in serous OC. While LINK-A knock out has both tumor-promoting and tumor-suppressing effects in an experimental model of OC, a combination of the present findings with our earlier analysis of clinical specimens may favor its tumor-suppressor role in this type of cancer. This does not concur with the above-discussed studies of different malignant tumors nor with the recent report of Hu and co-workers, in which LINK-A was reported to negatively regulate cancer cell antigen presentation and intrinsic tumor suppression (20). This discrepancy may suggest that LINK-A has different roles in different cancer types. In clinical OC, it may promote invasion and metastasis in early-stage disease but mediate undefined tumor-suppressive activities later in the clinical course, when tumor cells are metastatic. Analysis of the role of this molecule in early-stage disease, including in primary fallopian tube lesions in HGSC, may shed light on this issue.
Footnotes
Authors’ Contributions
NFL: Performed the experiments and wrote the article. BD: Participated in designing the study and co-wrote the article. RR: Designed the study, supervised the experiments and critically read the article.
Conflicts of Interest
The Authors declare no conflicts of interest in regard to this study.
- Received October 2, 2020.
- Revision received October 13, 2020.
- Accepted October 14, 2020.
- Copyright © 2020 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.