Purpose:

The development of resistance to platinum-based chemotherapy remains the unsurmountable obstacle in cancer treatment and consequently leads to tumor relapse. This study aims to investigate the mechanism by which loss of RBMS3 induced chemoresistance in epithelial ovarian cancer (EOC).

Experimental Design:

FISH and IHC were used to determine deletion frequency and expression of RBMS3 in 15 clinical EOC tissues and 150 clinicopathologically characterized EOC specimens. The effects of RBMS3 deletion and CBP/β-catenin antagonist PRI-724 in chemoresistance were examined by clone formation and Annexin V assays in vitro, and by intraperitoneal tumor model in vivo. The mechanism by which RBMS3 loss sustained activation of miR-126-5p/β-catenin/CBP signaling and the effects of RBMS3 and miR-126-5p competitively regulating DKK3, AXIN1, BACH1, and NFAT5 was explored using CLIP-seq, RIP, electrophoretic mobility shift, and immunoblotting and immunofluorescence assays.

Results:

Loss of RBMS3 in EOC was correlated with the overall and relapse-free survival. Genetic ablation of RBMS3 significantly enhanced, whereas restoration of RBMS3 reduced, the chemoresistance ability of EOC cells both in vitro and in vivo. RBMS3 inhibited β-catenin/CBP signaling through directly associating with and stabilizing multiple negative regulators, including DKK3, AXIN1, BACH1, and NFAT5, via competitively preventing the miR-126-5p–mediated repression of these transcripts. Importantly, cotherapy of CBP/β-catenin antagonist PRI-724 induced sensitization of RBMS3-deleted EOC to platinum therapy.

Conclusions:

Our results demonstrate that genetic ablation of RBMS3 contributes to chemoresistance and PRI-724 may serve as a potential tailored treatment for patients with RBMS3-deleted EOC.

Translational Relevance

The chemoresistance of platinum-based therapy is a serious limitation for lasting effective treatment of epithelial ovarian cancer, while the underlying mechanisms remain unclear. Herein, we reported that loss of RBMS3, an RNA-binding protein, augmented the resistance of epithelial ovarian cancer to platinum therapy and promoted recurrence of epithelial ovarian cancer. Mechanistically, we demonstrated that genetic ablation of RBMS3 sustained activation of β-catenin/CBP signaling via promoting miR-126-5p–mediated repression of multiple negative regulators of Wnt/β-catenin signaling. Cotherapy of PRI-724, a specific CBP/β-catenin antagonist, was sufficient to induce RBMS3-deleted epithelial ovarian cancer, but not RBMS3-nondeleted ovarian cancer, sensitization to platinum therapy, and thus inhibit tumor growth in vivo and in vitro. Our findings represent combined platinum and PRI-724 as a potential tailored treatment for RBMS3-deleted EOC and uncover a novel mechanism underlying hyperactivation miR-126-5p/Wnt/β-catenin/CBP signaling–mediated platinum therapy resistance in human ovarian cancer.

Among the various therapeutic modalities currently used for epithelial ovarian cancer (EOC), cytoreductive surgery combined with platinum-based chemotherapy is still acknowledged to be of the most effective treatments (1, 2). Despite the fact that more than 80% of patients with EOC exhibit encouraging initial clinical responses to chemotherapy with platinum-based drugs, the responses are often temporary. Approximately 25% of patients with EOC display multiple recurrences within 6 months after platinum therapy and more than 75% patients suffer relapse and progression years later, leading to a 5-year survival rate of only about 30% (3–5). Thus, the development of resistance to platinum-based chemotherapy is a serious limitation of long-lasting, effective treatments for patients with EOC.

Diverse range of intrinsic (de novo) mechanisms underlying platinum resistance, such as escape from platinum-induced apoptosis, increased platinum efflux, and enhanced DNA adduct repair, have been identified and usually coexist within the same tumor (6, 7). Meanwhile, intracellular signaling pathways, activated by heritable genetic/epigenetic alterations and the tumor microenvironment, also play critical roles in chemoresistance (8, 9). For instance, constitutively active Wnt/β-catenin signaling frequently occurs in various cancer types, confers resistance to oxaliplatin and gemcitabine through inhibition of apoptosis via upregulation of the antiapoptotic proteins Survivin and MMP-7, and contributes to cancer therapy failure through the repair of damaged DNA by inducing the DNA repair proteins MRE11 and LIG4 (10–12). Moreover, the drug transporters ABCB1 and ABCG2 can be directly upregulated by Wnt/β-catenin signaling, resulting in the extrusion of drugs out of cancer cells and multidrug resistance (13, 14). These results indicate that pharmacologic targeting of the Wnt/β-catenin signaling cascade may increase the clinical efficacy of chemotherapy (10, 15).

The acetyltransferase CREB-binding protein (CBP) is essential for Wnt/β-catenin signaling–mediated chemoresistance via diverse mechanisms (16–18). Thus, a specific small-molecule inhibitor of the CBP/β-catenin interaction, ICG-001, was screened and tested, and appears to have distinctive antitumor effects in multiple cancer types via inactivation of the Wnt/CBP/β-catenin signaling (19–24). In addition, the potential therapeutic value of PRI-724, which was derived from ICG-001 and is a second-generation CBP/β-catenin antagonist, is currently being tested in multiple clinical trials for the treatment of cancers, such as advanced colorectal and pancreatic cancers and myeloid malignancies (ClinicalTrials.gov ID: NCT02413853, NCT01764477, and NCT01606579). Hence, identifying key regulator(s) of Wnt/β-catenin/CBP signaling may reveal how PRI-724 can be successfully utilized for cancer treatment.

Genetic aberrations, such as chromosomal deletions, were recently reported to be associated with the cancer therapy response and may serve as potent predictive markers. For instance, loss of chromosome 8p contributes to taxane resistance and may server as a biomarker for microtubule inhibitor–based chemotherapy in breast cancer, and loss of PTEN predicts trastuzumab (Herceptin) resistance (25, 26). Anaplastic oligodendroglioma lacking 1p and 19q alleles are more responsive to radiochemotherapy (27). Heterozygous chromosomal deletions on the 3p arm are one of the most frequent allelic imbalances and a common early event in the formation of multiple types of cancers (28, 29). Herein, we found that loss of RNA-binding protein RBMS3, located on chromosome 3p, significantly enhanced the resistance of EOC to cisplatin (CDDP) via activation of Wnt/β-catenin/CBP signaling. Cotherapy of CDDP with PRI-724 exhibited remarkable therapeutic efficacy in preclinical models of EOC containing an RBMS3 deletion. These findings substantiate the importance of RBMS3 deletion in chemoresistance and may represent a new therapeutic strategy for EOC treatment.

Ethics statement

Signed informed consent was obtained from all patients, and the investigation has been performed in accordance with the ethical standards according to the Declaration of Helsinki, national and international guidelines, and that the studies were approved by an authors' institutional review board.

EOC patients, cancer tissue samples, and cells

All of the patients received standardized platinum-based chemotherapy. Platinum resistance or sensitivity was defined as relapse or progression within 6 months or after 6 months from the last platinum-based chemotherapy, respectively. Informed consent was obtained from all patients. Approvals from Sun Yat-sen University Cancer Center Institutional Research Ethics Committee were obtained for this study. A total of 150 paraffin-embedded, archived EOC specimens and 15 freshly collected EOC tissues were histopathologically and clinically diagnosed at Sun Yat-sen University Cancer Center (Guangdong, China) between 2005 and 2016. The clinical information of the samples is shown in Supplementary Tables S1 and S2. Detailed description provided in Supplementary Materials and Methods. Patient-derived EOC cells were prepared from fresh EOC tissues as described previously (30). EOC cell lines HEY were purchased from Bioleaf and SKOV3 were purchased from ECACC, which were cultured in completed DMEM (Gibco). All the cell lines been tested for Mycoplasma contamination and authenticated by short tandem repeat (STR) fingerprinting at Medicine Lab of Forensic Medicine Department of Sun Yat-Sen University (Guangzhou, China).

Xenografted tumor models, IHC, and H&E staining

In the subcutaneous patient-derived xenografts (PDX) tumor model, freshly isolated clinical EOC patient tissues were subdivided into 2–3 mm3 pieces, coated with Matrigel (BD Biosciences) and media in a 1:1 ratio, and embedded within the subcutaneous space underneath the skin of female NOD/Shi-scid/IL-2Rγnull (NOG) mouse (6–8 weeks old; CREA Japan Inc.). Tumor growth was monitored by measuring the tumor diameters, and the tumor volume was calculated using the equation (L × W2)/2. When the tumor became palpable, mice were intraperitoneally treated with vehicle (control) or CDDP (5 mg/kg) three times per week (as per cycle) for up to 6 weeks. At the end of treatment, the mice were sacrificed and the tumors were excised and weighed, and confirmed by histology. In the intraperitoneal tumor model, the indicated luciferase-expressing cells (1 × 106) were injected intraperitoneally into female NOD/SCID or NOG mice. When the luminescence signal reached 2 × 107 p/sec/cm2/sr, mice were intraperitoneally treated with vehicle (control), CDDP (5 mg/kg), or a combination of CDDP (5 mg/kg) and ICG-001 (5 mg/kg) three times per week (as per cycle) for up to 6 weeks. Mice were sacrificed when moribund as determined by an observer blinded to the treatment, and tumors were excised and paraffin-embedded. The indicated CDDP-resistant cells were isolated from tumors and reinjected intraperitoneally into a new cohort of mice to examine relapse within 4 months. Serial 4.0-μm sections were cut and subjected to IHC and hematoxylin and eosin (H&E) staining. After deparaffinization, sections were H&E-stained with Mayer hematoxylin solution, or IHC stained using antibodies of RBMS3 (1:100; Novus Biologicals, LLC), Ki-67 (Dako), or stained with TUNEL (In Situ Cell Death Detection Kit, TMR red, Roche Applied Science), and counterstained with Phalloidin (Alexa Fluor 488, Invitrogen) and DAPI (Sigma-Aldrich) according to manufacturer's protocols. The images were captured using the AxioVision Rel.4.6 computerized image analysis system (Carl Zeiss). All of the animal procedures were approved by the Sun Yat-sen University Animal Care Committee.

HITS-CLIP assay

In brief, HEY cells expressing FLAG-RBMS3 were rinsed, resuspended in ice-cold 1 × PBS, and irradiated at 400 mJ/cm2 and 200 mJ/cm2 subsequently using a Stratalinker 2400. Cells were then scraped in ice-cold PBS and RNA-IP performed as described previously (31) using FLAG-agarose beads (Sigma-Aldrich). Furthermore, RBMS3-associated RNAs were immunoprecipitated and subjected to adapter ligation and RT-PCR to generate libraries for high-throughput sequencing, done on an Illumina Hi-Seq. Sequencing reads were preprocessed and adapter sequences were removed, and trimmed reads were mapped to the human genome (hg19). Identical alignments were collapsed in each sample to remove PCR replicates. Strand-specific read coverage was then calculated using the alignments from the sample.

Statistical analysis

Statistical tests for data analysis included Fisher exact test, log-rank test, χ2 test and Student two-tailed t test. Bivariate correlations between study variables were calculated by Spearman rank correlation coefficients. Survival curves were plotted by the Kaplan–Meier method and compared by the log-rank test. The significance of various variables for survival was analyzed by univariate and multivariate Cox regression analyses. Statistical analyses were performed using the SPSS 11.0 statistical software package. Data represent mean ± SD. P values of 0.05 or less were considered statistically significant.

Deletion of chromosome 3p is associated with poor survival in multiple cancers

To better define the aberrant structure within chromosome 3 in tumors, we examined the patterns of chromosome 3 across 10,823 tumors spanning 33 human cancer types (TCGA dataset) using high-resolution single nucleotide polymorphism (SNP)-based copy number analysis. Chr3p was markedly deleted in 13 types of cancer, but deletion of Chr3q arm was only observed in pheochromocytoma and paraganglioma (false discovery rate (FDR)-corrected q value < 0.25, Fig. 1A). Copy number alterations of the 3p were significantly correlated with poorer overall and relapse-free survival (P = 0.016 and P = 0.045, respectively) in patients with these 13 cancer types (Supplementary Fig. S1A; Fig. 1B), indicating that deletion of 3p is extensively involved in cancer progression. Furthermore, statistical analysis showed that three deleted regions in 3p, namely Chr3p14.3-21.33, 3p22, and 3p23-24.1, were strongly correlated with shorter relapse-free survival (all P < 0.001), but only the 3p23-24.1 deletion was recognized as an independent prognostic factor for relapse-free survival in patients with cancer (P = 0.035; Fig. 1C and D). These pan-cancer data suggest that loss of 3p23-24.1 may contribute to poor patient outcome and cancer recurrence.

Figure 1.

Loss of RBMS3 correlates with chemoresistance in pan-cancer. A, GISTIC2.0 analysis across 10,823 SNP6.0 Affymetrix copy number arrays spanning 33 human cancer types (The Cancer Genome Atlas, TCGA) showed significant deletion of the Chr3p arm in 13 cancer types (FDR-corrected q value < 0.25). B, Loss of the Chr3p arm correlated with relapse-free survival in the TCGA Pan-Cancer cohort (P = 0.045; n = 4528; 13 cancer types). C, Loss of Chr3p14.3-21.33, Chr3p22, or Chr3p23-24.1 was associated with shorter relapse-free survival in TCGA Pan-Cancer cohort (P < 0.001; n = 4,528; 13 cancer types). D, Multivariate analysis showed Chr3p23-24.1 deletion as an independent prognostic factor for relapse-free survival in patients with cancer (P = 0.035; n = 4528; 13 cancer types). E, MTT cell viability analysis of the response of 368 genes to CDDP treatment in HEY and SKOV3 EOC cells. F, EOCs analyzed by FISH for RBMS3 deletion and IHC for RBMS3 protein (left), correlation between RBMS3 deletion and protein expression (middle) and correlation between RBMS3 deletion and chemoresistance (right). G, Fold change of RBMS3 mRNA expression and corresponding RBMS3 protein expression and deletion in 15 freshly collected clinical EOC tissues. Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. H and I, Deletion (left) and downregulation (right) of RBMS3 correlated with relapse-free survival in EOC tissues (P = 0.008; P < 0.001; n = 150).

Figure 1.

Loss of RBMS3 correlates with chemoresistance in pan-cancer. A, GISTIC2.0 analysis across 10,823 SNP6.0 Affymetrix copy number arrays spanning 33 human cancer types (The Cancer Genome Atlas, TCGA) showed significant deletion of the Chr3p arm in 13 cancer types (FDR-corrected q value < 0.25). B, Loss of the Chr3p arm correlated with relapse-free survival in the TCGA Pan-Cancer cohort (P = 0.045; n = 4528; 13 cancer types). C, Loss of Chr3p14.3-21.33, Chr3p22, or Chr3p23-24.1 was associated with shorter relapse-free survival in TCGA Pan-Cancer cohort (P < 0.001; n = 4,528; 13 cancer types). D, Multivariate analysis showed Chr3p23-24.1 deletion as an independent prognostic factor for relapse-free survival in patients with cancer (P = 0.035; n = 4528; 13 cancer types). E, MTT cell viability analysis of the response of 368 genes to CDDP treatment in HEY and SKOV3 EOC cells. F, EOCs analyzed by FISH for RBMS3 deletion and IHC for RBMS3 protein (left), correlation between RBMS3 deletion and protein expression (middle) and correlation between RBMS3 deletion and chemoresistance (right). G, Fold change of RBMS3 mRNA expression and corresponding RBMS3 protein expression and deletion in 15 freshly collected clinical EOC tissues. Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. H and I, Deletion (left) and downregulation (right) of RBMS3 correlated with relapse-free survival in EOC tissues (P = 0.008; P < 0.001; n = 150).

Close modal

Loss of RBMS3 correlates with CDDP resistance in EOC

Cancer recurrence is mainly attributed to resistance to chemotherapy (7). Because EOC is the most common recurrent tumor disease, we used the HEY and SKOV3 EOC cell line to determine whether candidate genes in the Chr3p arm are involved in chemoresistance. The MTT cell viability assay showed that among 368 genes in three deleted regions of the Chr3p arm, silencing of RBMS3, one of the genes located in Chr3p23-24.1, had a significant inhibitory effect on CDDP-induced apoptosis both in HEY and SKOV3 EOC cells (Fig. 1E), suggesting that loss of RBMS3 may contribute to resistance of EOC to platinum chemotherapy. Consistent with this hypothesis, the low expression of RBMS3 mRNA was associated with shorter progression-free survival of patients with platinum-treated EOC as well as with gene signature–related CDDP resistance (Supplementary Fig. S1B–S1D).

Furthermore, we found that RBMS3 protein levels were clearly lower in RBMS3-deleted EOC tissues than in RBMS3-nondeleted tissues, as assessed by IHC and FISH methodologies (Fig. 1F). Furthermore, of the 15 freshly collected clinical EOC tissues, 5 RBMS3-deleted EOC tissues had lower RBMS3 mRNA and protein expression than RBMS3-nondeleted EOC (n = 10; Fig. 1G), suggesting that RBMS3 gene copy alterations represent one mechanism of protein deregulation. Importantly, RBMS3 deletion or low RBMS3 expression was significantly correlated with chemoresistance and shorter overall/relapse-free survival in patients with EOC treated with platinum-based therapy (Fig. 1H and I; Supplementary Fig. S1E and S1F; Supplementary Tables S1 and S2).

In addition, statistical analysis revealed that RBMS3 deletion was significantly associated with poorer overall/relapse-free survival in patients with cancer and with signature-related chemoresistance in 13 cancer types (Supplementary Fig. S1G–S1I). These data suggest a widespread correlation between RBMS3 deletion and chemoresistance in human cancers.

RBMS3 deletion promotes chemoresistance in EOC in a PDX model

To examine the biological role of RBMS3 deletion in chemoresistance, a patient-derived xenograft (PDX) model was employed using the abovementioned 15 collected EOC tissues, of which 5 RBMS3+/+ and 3 RBMS3+/− tissues were successfully engrafted in female NOD/Shi-scid/IL-2Rγnull (NOG) mice (Fig. 2A). Tumor-bearing mice were further treated with vehicle or CDDP, and both groups of xenograft tumors in vehicle-treated mice showed similar tumor growth kinetics (Fig. 2A), which suggest that the loss of RBMS3 did not affect tumor growth. However, compared with vehicle-treated tumors, CDDP chemotherapy dramatically reduced the growth rate of RBMS3+/+ tumors, but RBMS3+/− tumors displayed higher CDDP-resistant capability as indicated by their faster growth rate (Fig. 2A).

Figure 2.

Dysregulation of RBMS3 is involved in chemoresistance in EOC in vivo. A, A PDX model was established in NOG mice by inoculating them with the 15 freshly collected clinical EOC tissues and relative volume changes in xenograft tumor upon CDDP (5 mg/kg body weight) or vehicle treatment (n = 6/group). Five RBMS3+/+- and 3 RBMS3+/−-EOC tissues successfully engrafted in NOG mouse. The tumor volume of each group was recorded. B, Primary OV-2 and OV-11 cells isolated from ovarian serous carcinoma tissues (OV#2 and OV#11) analyzed by FISH for RBMS3 deletion (left) and immunoblot for RBMS3 protein (right). C, Representative images of CDDP-treated intraperitoneal tumor-bearing NOG mice at indicated time. D, IHC staining of RBMS3 and Ki67, TUNNEL staining, and H&E analysis (left) and quantification of RBMS3 expression, apoptotic rate, and proliferation index in the indicated xenograft tumors. E, Relative changes in bioluminescence signal of intraperitoneal tumors in NOG mice upon CDDP chemotherapy. F, Kaplan–Meier survival of CDDP-treated intraperitoneal tumor-bearing NOG mice. G, Representative images of CDDP-treated intraperitoneal tumor-bearing NOD/SCID mice at the indicated time. H, Relative change in bioluminescence signal of intraperitoneal tumors in CDDP-treated NOD/SCID mice. I, Kaplan–Meier survival of CDDP-treated intraperitoneal tumor-bearing mice. J, Kaplan–Meier relapse-free survival of mice intraperitoneally injected with different dose of the indicated cells isolated form tumors in Fig. 2G and Supplementary Fig. S2E.

Figure 2.

Dysregulation of RBMS3 is involved in chemoresistance in EOC in vivo. A, A PDX model was established in NOG mice by inoculating them with the 15 freshly collected clinical EOC tissues and relative volume changes in xenograft tumor upon CDDP (5 mg/kg body weight) or vehicle treatment (n = 6/group). Five RBMS3+/+- and 3 RBMS3+/−-EOC tissues successfully engrafted in NOG mouse. The tumor volume of each group was recorded. B, Primary OV-2 and OV-11 cells isolated from ovarian serous carcinoma tissues (OV#2 and OV#11) analyzed by FISH for RBMS3 deletion (left) and immunoblot for RBMS3 protein (right). C, Representative images of CDDP-treated intraperitoneal tumor-bearing NOG mice at indicated time. D, IHC staining of RBMS3 and Ki67, TUNNEL staining, and H&E analysis (left) and quantification of RBMS3 expression, apoptotic rate, and proliferation index in the indicated xenograft tumors. E, Relative changes in bioluminescence signal of intraperitoneal tumors in NOG mice upon CDDP chemotherapy. F, Kaplan–Meier survival of CDDP-treated intraperitoneal tumor-bearing NOG mice. G, Representative images of CDDP-treated intraperitoneal tumor-bearing NOD/SCID mice at the indicated time. H, Relative change in bioluminescence signal of intraperitoneal tumors in CDDP-treated NOD/SCID mice. I, Kaplan–Meier survival of CDDP-treated intraperitoneal tumor-bearing mice. J, Kaplan–Meier relapse-free survival of mice intraperitoneally injected with different dose of the indicated cells isolated form tumors in Fig. 2G and Supplementary Fig. S2E.

Close modal

Serous ovarian carcinoma represents the largest proportion (up to 70%) of EOC cases. To further clarify the effects of RBMS3 deletion in EOC on CDDP resistance, two primary serous ovarian carcinoma cells OV-2 and OV-11, isolated from EOC tissues OV-#2 (RBMS3+/+) and OV-#11 (RBMS3+/−), were engineered to stably overexpress the luciferase reporter and were then intraperitoneally injected into NOG mice (Fig. 2B and C). When the bioluminescence signal of intraperitoneal tumors reached 2 × 107 p/sec/cm2/sr, NOG mice were treated with CDDP (Fig. 2C). After 6 weeks/cycles of CDDP therapy, the tumors formed by OV-11 cells remained higher growth rate, as shown by stronger Ki67 signals and less terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells compared with OV-2/tumors (Fig. 2C–E), indicating that RBMS3-deleted EOC cells possessed higher chemoresistant capability. In contrast, silencing RBMS3 via RBMS3 siRNA incorporated into dioleoylphosphatidylcholine (DOPC) nanoliposomes dramatically abrogated the inhibitory effect of CDDP on OV-2/tumors growth, which exhibited a decreased apoptotic index and increased proliferation, resulting in shorter survival of tumor-bearing mice (Fig. 2C–F).

To further support the contribution of RBMS3 deletion to CDDP resistance, serous ovarian cancer cell line HEY, the major subtype of EOC (32), was utilized to establish HEY/RBMS3 gRNA#1 and gRNA#2 EOC cells via CRISPR/Cas9 genome editing technology (Supplementary Fig. S2A). Concordant with the PDX results, both HEY/RBMS3 gRNA#1 and gRNA#2 tumors treated with CDDP exhibited a stronger bioluminescence signal, a lower percentage of TUNEL-positive apoptotic cells, and a higher proliferative index compare to control tumors, and the mice bearing HEY/RBMS3 gRNA#1 and gRNA#2 tumors survived significantly shorter than the CRISPR-Control mice (Fig. 2G–I; Supplementary Fig. S2B). Strikingly, as few as 100 HEY/RBMS3 gRNA#1 EOC cells, which were isolated from CDDP-treated mice and reinjected into secondary mouse recipients, were able to develop tumors in NOG mice around 3 months, while no tumor was formed by HEY cells after 4 months (Fig. 2J; Supplementary Fig. S2C), demonstrating the vital role of RBMS3 deletion in EOC recurrence.

Restoration of RBMS3 promotes chemosensitivity of EOC cells

Next, the effect of RBMS3 restoration on chemosensitivity was assessed in OV-11 and HEY cells with ectopic expression of RBMS3 cDNA (Supplementary Fig. S2D). As expected, upregulation of RBMS3 significantly enhanced the antitumor efficacy of CDDP on growth of OV-11/tumors and HEY/tumors, as indicated by the increased CDDP-induced apoptotic rate and decreased Ki67 signals (Supplementary Fig. S2E and S2F), which demonstrated that RBMS3 restores the sensitivity of EOC cells to CDDP in vivo.

Ablation of RBMS3 confers resistance of EOC cells to cisplatin in vitro

In concordance with the in vivo results, RBMS3-deleted or silenced cells did not show obvious alterations of apoptotic rate or colony-forming ability compared with control cells under vehicle treatment. However, deletion or silencing of RBMS3 dramatically reduced CDDP-induced apoptotic death and promoted colony formation in EOC cells (Supplementary Fig. S3A–S3C). Moreover, we found that the IC50 value of CDDP was significantly increased in RBMS3-deleted or RBMS3-silenced cells (Supplementary Fig. S3D). These results indicate that loss of RBMS3 confers resistance of EOC cells to CDDP in vitro.

Loss of RBMS3 promotes CDDP efflux and DNA repair in EOC cells

There is ample evidence demonstrating that reduced drug accumulation is a major mechanism underlying CDDP resistance (6, 7). Interestingly, we found that the CDDP concentration was much lower in RBMS3-ablated/tumors, and that the content of intracellular and genomic DNA-bound CDDP was reduced in RBMS3–deleted/silenced cells (Supplementary Fig. S3E and F), suggesting that loss of RBMS3 promotes CDDP efflux. This was further confirmed by an efflux kinetic assay, which demonstrated that deletion/silencing of RBMS3 significantly increased the efflux velocity of CDDP (Supplementary Fig. S3G).

Formation and persistence of DNA adducts of CDDP are vital in inducing apoptosis (33). Concomitantly, we found that the number of CDDP-induced γH2AX foci was drastically decreased in RBMS3–deleted/silenced cells (Supplementary Fig. S3H). Furthermore, a comet assay showed that deletion or silencing of RBMS3 dramatically abrogated CDDP-induced DNA damage (Supplementary Fig. S3I). These results suggest that loss of RBMS3 may also promote DNA damage repair.

Loss of RBMS3 activates Wnt/β-catenin signaling

Consistent with our results that the loss of RBMS3 contributes to chemoresistance, apoptosis, drug efflux, and DNA repair, Gene ontology (GO) enrichment analysis in public datasets has shown that genes with GO biological process terms “Response to drug,” “Apoptosis,” “Transporter activity,” and “DNA repair” were enriched in RBMS3-downregulated cells (enrichment cut-off P < 0.05; Fig. 3A). Similar enrichments were also observed in our analysis results obtained from mRNA microarray and high-throughput sequencing crosslink immunoprecipitation (HITS-CLIP; Fig. 3B). Strikingly, we found that genes with GO terms, “Wnt signaling pathway,” “Negative regulation of canonical Wnt signaling pathway,” and “Regulation of Wnt signaling pathway” were also enriched (Fig. 3A and B). Meanwhile, GSEA analysis showed that RBMS3 levels were inversely associated with related gene signatures of Wnt signaling in EOC (Fig. 3C). These results suggest that RBMS3 may be involved in modulating Wnt signaling.

Figure 3.

Loss of RBMS3 activates Wnt/β-catenin signaling. A, GO enrichment analysis of RBMS3-regulated transcripts from public datasets (NCBI/GEO/GSE 4122/27651). B, GO enrichment analysis of RBMS3-regulated transcripts identified in microarray profiling analysis or HITS-CLIP analysis. C, GSEA analysis showing that RBMS3 expression was inversely correlated with related gene signatures of Wnt/β-catenin signaling in a published EOC dataset (TCGA, n = 419; NCBI/GEO/GSE27651, n = 41). D, Relative TOPflash or FOPflash luciferase reporter activity analyzed in the indicated cells. Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. E, EMSA analysis of DNA-binding activity of β-catenin in the indicated cells. F, The pseudocolor represents the relative mRNA expression of chemoresistance-related genes regulated by Wnt/β-catenin signaling in the indicated cells. G, Immunoblot analysis of expression levels of indicated protein in the indicated cells. α-Tubulin served as the loading control. H, Subcellular localization of β-catenin in the indicated cells, as analyzed by an immunofluorescence staining assay. I, IHC analysis (left) and correlation (right) of expression of RBMS3 and nuclear β-catenin in EOC tissues (n = 150).

Figure 3.

Loss of RBMS3 activates Wnt/β-catenin signaling. A, GO enrichment analysis of RBMS3-regulated transcripts from public datasets (NCBI/GEO/GSE 4122/27651). B, GO enrichment analysis of RBMS3-regulated transcripts identified in microarray profiling analysis or HITS-CLIP analysis. C, GSEA analysis showing that RBMS3 expression was inversely correlated with related gene signatures of Wnt/β-catenin signaling in a published EOC dataset (TCGA, n = 419; NCBI/GEO/GSE27651, n = 41). D, Relative TOPflash or FOPflash luciferase reporter activity analyzed in the indicated cells. Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. E, EMSA analysis of DNA-binding activity of β-catenin in the indicated cells. F, The pseudocolor represents the relative mRNA expression of chemoresistance-related genes regulated by Wnt/β-catenin signaling in the indicated cells. G, Immunoblot analysis of expression levels of indicated protein in the indicated cells. α-Tubulin served as the loading control. H, Subcellular localization of β-catenin in the indicated cells, as analyzed by an immunofluorescence staining assay. I, IHC analysis (left) and correlation (right) of expression of RBMS3 and nuclear β-catenin in EOC tissues (n = 150).

Close modal

This hypothesis was further confirmed by several lines of evidence, which showed that deletion or silencing of RBMS3 increased, but overexpression of RBMS3 decreased, the transcriptional activity and DNA-binding activity of β-catenin, as well as the expression of multiple Wnt/β-catenin downstream genes and nuclear β-catenin levels (Fig. 3D–H). The inverse correlation of RBMS3 expression and activation of Wnt/β-catenin signaling, as indicated by nuclear β-catenin levels, was further verified in a cohort of clinical EOC tissues (Fig. 3I). Consistently, the reverse correlation of RBMS3 expression and activation of Wnt/β-catenin signaling was also found in multiple types of cancer via GSEA analysis (Supplementary Fig. S4A). Moreover, the duration of Wnt/β-catenin signaling, as indicated by the mRNA expression of Survivin, was dramatically prolonged in RBMS3-deleted cells but was much shortened in RBMS3-transduced cells (Supplementary Fig. S4B), suggesting that loss of RBMS3 sustains Wnt/β-catenin signaling in EOC cells. Importantly, inactivation of Wnt/β-catenin signaling in RBMS3-ablated cells by silencing β-catenin or TCF4 drastically decreased the levels of multiple Wnt/β-catenin signaling–regulated chemoresistance-related genes, such as ABCB1, Survivin, and MRE11, and cisplatin IC50 and CDDP content, but increased CDDP-induced apoptotic rates (Supplementary Fig. S4C–S4F), demonstrating that Wnt/β-catenin signaling is required for RBMS3 deletion–mediated chemoresistance.

RBMS3 stabilizes multiple negative regulators in Wnt/β-catenin cascade

Next, we identified the molecular basis for the regulation of RBMS3 in Wnt/β-catenin signaling. Our HITS-CLIP data indicated that RBMS3 may associate with 22 genes involved in the negative regulation of Wnt/β-catenin signaling (Fig. 4A; Supplementary Fig. S5A). Furthermore, RNA immunoprecipitation (RIP) assay revealed that RBMS3 most significantly interacted with the mRNA of DKK3, AXIN1, BACH1, and NFAT5 in both OV-2 and HEY EOC cells (Fig. 4B). A concomitant decrease in DKK3, AXIN1, BACH1, and NFAT5, at both the mRNA and protein levels, was also observed in RBMS3-deleted/silenced cells, which were increased in RBMS3-transduced cells (Fig. 4C and D; Supplementary Fig. S5B). Importantly, loss of RBMS3 reduced, while overexpression of RBMS3 increased the mRNA half-life of these transcripts (Fig. 4E and F). Taken together, these results suggested that RBMS3 mediated the stabilization of these transcripts at the posttranscriptional level through association with their 3′UTR.

Figure 4.

RBMS3 stabilizes multiple negative regulators in Wnt/β-catenin cascade. A, List of 22 RBMS3-associated negative regulators in Wnt/β-catenin cascade analyzed by HITS-CLIP dataset (left) and represented DKK3, AXIN1, BACH1 and NFAT5 (right). B. RIP analysis of association of RBMS3 with identified 22 transcripts in the indicated cells. C, Relative expression of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells as quantified by qRT-PCR analysis. D, Immunoblot analysis of protein expression of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells. E, RIP analysis of association of RBMS3 with target mRNAs (AXIN1, BACH1, and NFAT5) in indicated cells. F, Relative mRNA levels of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells treated with actinomycin D (10 μg/mL). G, IHC analysis (top) and correlation (bottom) of expression of RBMS3 and DKK3, AXIN1, BACH1, and NFAT5 in clinical EOC tissues (n = 150). H, Kaplan–Meier survival analysis of association of DKK3, AXIN1, BACH1, and NFAT5 protein expression with relapse-free survival in EOC (P < 0.05). I, Relative TOP/FOPflash luciferase reporter activity analyzed in the indicated cells. J, Quantification of apoptotic index in the indicated cells, as analyzed by Annexin V assay. K, IC50 value of CDDP in the indicated cells analyzed by MTT cell viability assay. Each bar represents the mean ± SD of three independent experiments (*, P <0.05).

Figure 4.

RBMS3 stabilizes multiple negative regulators in Wnt/β-catenin cascade. A, List of 22 RBMS3-associated negative regulators in Wnt/β-catenin cascade analyzed by HITS-CLIP dataset (left) and represented DKK3, AXIN1, BACH1 and NFAT5 (right). B. RIP analysis of association of RBMS3 with identified 22 transcripts in the indicated cells. C, Relative expression of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells as quantified by qRT-PCR analysis. D, Immunoblot analysis of protein expression of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells. E, RIP analysis of association of RBMS3 with target mRNAs (AXIN1, BACH1, and NFAT5) in indicated cells. F, Relative mRNA levels of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells treated with actinomycin D (10 μg/mL). G, IHC analysis (top) and correlation (bottom) of expression of RBMS3 and DKK3, AXIN1, BACH1, and NFAT5 in clinical EOC tissues (n = 150). H, Kaplan–Meier survival analysis of association of DKK3, AXIN1, BACH1, and NFAT5 protein expression with relapse-free survival in EOC (P < 0.05). I, Relative TOP/FOPflash luciferase reporter activity analyzed in the indicated cells. J, Quantification of apoptotic index in the indicated cells, as analyzed by Annexin V assay. K, IC50 value of CDDP in the indicated cells analyzed by MTT cell viability assay. Each bar represents the mean ± SD of three independent experiments (*, P <0.05).

Close modal

To complement our assessment of the effects of RBMS3 deletion on chemoresistance, the clinical relevance of these RBMS3-associated regulators was examined in EOC tissues. Integrating Kaplan–Meier plot and IHC analyses revealed that low levels of the mRNA or protein of these regulators coincided with failure of platinum therapy in EOCs (Supplementary Fig. S5C; Fig. 4G and H), which further supported the notion that RBMS3 deletion results in chemoresistance and recurrence of EOC. Moreover, individual force expressing DKK3, AXIN1, BACH1, and NFAT5 in RBMS3+/− significantly inhibited TOP/FOP flash reporter activity and reduced the CDDP IC50 value, but increased CDDP-induced apoptosis (Supplementary Fig. S5D; Fig. 4I–K), demonstrating that these regulators are functional effectors of RBMS3 loss–induced activation of Wnt/β-catenin signaling and chemoresistance.

miR-126-5p reverses RBMS3-mediated inhibition of Wnt/β-catenin pathway

Interestingly, we found that the RNA-binding site of RBMS3, AAUAAU, was complementary to the “seed” sequences of miR-126-5p (UUAUUA), suggesting that miR-126-5p may be involved in regulation of DKK3, AXIN1, BACH1, and NFAT5 mRNAs (Fig. 5A and B). As expected, miR-126-5p could associate with 3′UTR of DKK3, AXIN1, BACH1, and NFAT5 mRNA, resulting in a consistent reduction in luciferase activity of target mRNA 3′UTRs (DKK3, AXIN1, BACH1, and NFAT5; Supplementary Fig. S6A and S6B). However, mutation of the miR-126-5p “seed” sequence abrogated the suppressive ability of miR-126-5p (Supplementary Fig. S6B). Meanwhile, the expression of these negative regulators, at both the mRNA and protein levels, was decreased in miR-126-5p–transduced cells, but increased in miR-126-5p–silenced cells (Supplementary Fig. S6C; Fig. 5C).

Figure 5.

RBMS3 prevents miR-126-5p–mediated repression of negative regulators in Wnt/β-catenin pathway. A, The RNA-binding site of RBMS3, AAUAAU, was complementary to the “seed” sequences of miR-126-5p (UUAUUA). B, Predicted miR-126-5p target sequences in the 3′UTRs of DKK3, AXIN1, BACH1, and NFAT5, and mutant containing three altered nucleotides in the miR-126-5p seed sequence (miR-126-5p-mu). The RBMS3-binding site has been predicted by RBPmap Website and labeled with asterisks. C and D, Western blotting analysis of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells. α-Tubulin served as the loading control. E, RIP analysis of association of RBMS3 (left) and miR-126-5p (right) with DKK3, AXIN1, BACH1, and NFAT5 in indicated cells. F, Relative mRNA levels of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells treated with Actinomycin D (10 μg/mL). Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. G, EMSA analysis of DNA-binding activity of β-catenin in the indicated cells. OCT-1 served as the loading control. H, The pseudocolor represents the relative mRNA expression regulated by Wnt/β-catenin signaling in the indicated cells.

Figure 5.

RBMS3 prevents miR-126-5p–mediated repression of negative regulators in Wnt/β-catenin pathway. A, The RNA-binding site of RBMS3, AAUAAU, was complementary to the “seed” sequences of miR-126-5p (UUAUUA). B, Predicted miR-126-5p target sequences in the 3′UTRs of DKK3, AXIN1, BACH1, and NFAT5, and mutant containing three altered nucleotides in the miR-126-5p seed sequence (miR-126-5p-mu). The RBMS3-binding site has been predicted by RBPmap Website and labeled with asterisks. C and D, Western blotting analysis of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells. α-Tubulin served as the loading control. E, RIP analysis of association of RBMS3 (left) and miR-126-5p (right) with DKK3, AXIN1, BACH1, and NFAT5 in indicated cells. F, Relative mRNA levels of DKK3, AXIN1, BACH1, and NFAT5 in the indicated cells treated with Actinomycin D (10 μg/mL). Each bar represents the mean ± SD of three independent experiments. *, P < 0.05. G, EMSA analysis of DNA-binding activity of β-catenin in the indicated cells. OCT-1 served as the loading control. H, The pseudocolor represents the relative mRNA expression regulated by Wnt/β-catenin signaling in the indicated cells.

Close modal

We then further investigate the competitive effect between RBMS3 and miR-126-5p upon target mRNAs. As shown in Fig. 5D, the upregulated effect of RBMS3 on expression of DKK3, AXIN1, BACH1, and NFAT5 was dramatically decreased upon miR-126-5p overexpression, while blocking miR-126-5p enhanced the expression of these transcripts in RBMS3+/− cells. Furthermore, RIP assays indicated that enforced expression of miR-126-5p significantly reduced the binding activity of RBMS3 with these target mRNAs, and upregulation of RBMS3 tremendously impaired the association of miR-126-5p with mRNA of DKK3, AXIN1, BACH1, and NFAT5 (Fig. 5E), indicating that RBMS3 and miR-126-5p exerted competitive regulation in these mRNAs. Accordingly, miR-126-5p overexpression suppressed the RBMS3-mediated stabilization of target mRNAs, resulting in Wnt/β-Catenin signaling activation, while downregulation of miR-126-5p significantly increased these negative regulators and inhibited Wnt pathway (Fig. 5F–H; Supplementary Fig. S6D and S6E). These observations suggest that miR-126-5p abrogates RBMS3-mediated stabilization of the mRNAs of DKK3, AXIN1, BACH1, and NFAT5, which activates Wnt/β-catenin pathway in ovarian cancer.

Blocking Wnt/β-catenin/CBP signaling confers chemosensitivity to RBMS3-deleted cells

Acetyltransferase CBP is reported to play crucial roles in Wnt/β-catenin signaling–mediated chemoresistance (16–18). Multiple RBMS3-associated regulators, such as BACH1, KLF4, NFAT5, and WT1, modulate Wnt/CBP/β-catenin signaling via blocking the interaction of β-catenin and CBP (34–36), which raises the possibility that blocking CBP/β-catenin signaling confers chemosensitivity to RBMS3-deleted cells. To test this hypothesis, we first examined the effects of RBMS3 on the association of β-catenin with CBP. As shown in Supplementary Fig. S7A, the level of β-catenin–associated CBP, through pulling down the same amount of nuclear β-catenin, was greatly increased in RBMS3 knockdown cells, but was decreased in RBMS3-transduced cells compared with control cells, suggesting that RBMS3 modulated the binding efficiency of β-catenin and CBP. Furthermore, we found that deletion or silencing of RBMS3 significantly increased, but overexpressing RBMS3 decreased levels of β-catenin, CBP, and acylated-histone 3 in promoter of ABCB1, Survivin, and MRE11 (Supplementary Fig. S7B).

As expected, silencing of CBP resulted in significant decreased β-catenin transactivity and expression of ABCB1, Survivin, and MRE11 in RBMS3-deleted cells compared with control cells (Supplementary Fig. S7C–S7E), indicating that CBP is essential for RBMS3 loss–induced chemoresistance. Similar to the effect of CBP silencing in RBMS3-deleted cells, treatment with ICG-001, a specific inhibitor of CBP/β-catenin signaling via blockage of β-catenin/CBP interaction, in RBMS3-deleted cells also overtly reduced expression of ABCB1, BIRC5, and MRE11 compared with RBMS3-nondeleted cells (Supplementary Fig. S7F). Importantly, ICG-001 treatment of RBMS3-deleted/silenced cells significantly enhanced the inhibitory effects of CDDP on clonogenic growth and CDDP-induced apoptosis compared with RBMS3-nondeleted cells (Supplementary Fig. S7G and S7H). The superiority of the combination effects was further confirmed by the drug combination index, which indicated a synergistic effect of ICG-001 and CDDP in RBMS3-deleted cells, but not in RBMS3-nondeleted cells (Supplementary Fig. S7I). Collectively, these results demonstrate that ICG-001 chemosensitizes RBMS3-deleted EOC to CDDP.

Combined PRI-724 sensitizes RBMS3-deleted EOC to CDDP therapy

To assess whether the in vitro findings could be recapitulated in vivo, the therapeutic efficacy of CDDP combined with PRI-724, a second-generation ICG-001 that has been tested in clinical trials in multiple cancers, was examined in a mouse model. As shown in Fig. 6A–F and Supplementary Fig. S8A and S8B, the curative effects of CDDP were significantly augmented by PRI-724 cotherapy in RBMS3-ablated/tumors, in a striking reduction in tumor volume, higher apoptotic index, and longer survival of tumor-bearing mice. In contrast, PRI-724 treatment only moderately increased the antitumor effects of CDDP on RBMS3+/+/tumors (Fig. 6D–F). From these experiments, we concluded that the combination of PRI-724 and CDDP produced synergistic cytotoxicity in RBMS3-deleted EOCs. To validate our findings in patients with cancer, the PDX model in Fig. 2A was employed, and showed that cotherapy of PRI-724 with CDDP considerably impaired the tumor growth of RBMS3+/− xenografts, whereas no additional antitumor effects of PRI-724 were observed in CDDP-treated RBMS3+/+-implanted tumors (Fig. 6G). Therefore, our results demonstrate that RBMS3 loss enhances platinum chemoresistance via activating Wnt/β-Catenin/CBP pathway, suggesting cotherapy of PRI-724 and CDDP might be a valuable therapeutic strategy for RBMS3-deleted EOC (Fig. 6H).

Figure 6.

Combined PRI-724 sensitizes RBMS3-deleted EOC to CDDP therapy. A, Representative images of intraperitoneal tumor-bearing NOD/SCID mice (n = 6/group) treated with vehicle, or CDDP (5mg/kg), or ICG-001 (5 mg/kg) or ICG001 (5 mg/kg) plus CDDP (5 mg/kg or 1 mg/kg) for 6 weeks compared with pretreated tumors (week 0). B, Relative changes of bioluminescence signal of indicated reagent-treated intraperitoneal tumors formed by HEY/Control or HEY/RBMS3 gRNA#1 cell. C, Kaplan–Meier survival of HEY/RBMS3 gRNA#1 mice treated with indicated reagent(s), n = 6/group. D, Representative images of tumor-bearing mice (n = 6/group) treated with indicated reagent(s). E, Relative change of bioluminescence signal of intraperitoneal tumors treated for 6 weeks compared with pretreated tumors (week 0) formed by the indicated cells. F, Kaplan–Meier survival of OV-2/mice and OV-11/mice treated with indicated reagent(s). G, PDX model showing that relative volume change of PDX tumors (n = 6) treated with indicated reagent(s) for 6 weeks compared with pretreated tumors (week 0) formed by the indicated EOC tissues (mean ± SD, n = 3; *, P < 0.05). H, Hypothetical model illustrating that RBMS3 loss promotes chemoresistance in EOC via constitutively activating Wnt/β-Catenin/CBP pathway.

Figure 6.

Combined PRI-724 sensitizes RBMS3-deleted EOC to CDDP therapy. A, Representative images of intraperitoneal tumor-bearing NOD/SCID mice (n = 6/group) treated with vehicle, or CDDP (5mg/kg), or ICG-001 (5 mg/kg) or ICG001 (5 mg/kg) plus CDDP (5 mg/kg or 1 mg/kg) for 6 weeks compared with pretreated tumors (week 0). B, Relative changes of bioluminescence signal of indicated reagent-treated intraperitoneal tumors formed by HEY/Control or HEY/RBMS3 gRNA#1 cell. C, Kaplan–Meier survival of HEY/RBMS3 gRNA#1 mice treated with indicated reagent(s), n = 6/group. D, Representative images of tumor-bearing mice (n = 6/group) treated with indicated reagent(s). E, Relative change of bioluminescence signal of intraperitoneal tumors treated for 6 weeks compared with pretreated tumors (week 0) formed by the indicated cells. F, Kaplan–Meier survival of OV-2/mice and OV-11/mice treated with indicated reagent(s). G, PDX model showing that relative volume change of PDX tumors (n = 6) treated with indicated reagent(s) for 6 weeks compared with pretreated tumors (week 0) formed by the indicated EOC tissues (mean ± SD, n = 3; *, P < 0.05). H, Hypothetical model illustrating that RBMS3 loss promotes chemoresistance in EOC via constitutively activating Wnt/β-Catenin/CBP pathway.

Close modal

Platinum-based chemotherapy, the cornerstone of modern treatment for EOC, can induce considerable clinical remission. However, most patients with advanced EOC invariably experience relapse and eventually die from this disease (1–4). In this scenario, understanding the molecular basis of platinum resistance may aid in the stratification of patients with EOC who are most likely to benefit from platinum-based chemotherapy, as well as the delineation of new targets for the pharmacologic intervention of patients with poor outcomes. Herein, we demonstrated that loss of RBMS3 significantly enhanced the resistance of EOC to cisplatin through activation of Wnt/β-catenin/CBP signaling, and that combined CDDP with PRI-724 exhibited significant therapeutic efficacy in preclinical models of RBMS3-deleted EOC. These results provided mechanistic and clinical insights into platinum-based resistance and suggested that the combination of platinum-based therapy with PRI-724 may serve as a potential tailored treatment for patients with RBMS3-deleted EOC. Considering that the action mechanisms of carboplatin and cisplatin are similar, loss of RBMS3 may also confer the resistance of EOC to carboplatin therapy. Meanwhile, because the gold-standard chemotherapeutic regimen for EOC is carboplatin combined with paclitaxel, the resistant effect of RBMS3 deletion on paclitaxel–carboplatin chemotherapy in EOC is investigated currently in our laboratory.

Aberrant activation of Wnt/β-catenin signaling has been observed in multiple tumors and contributed to tumor cell properties characteristic of the malignant phenotype, which raises the possibility that targeting the members of this signaling cascade may be an attractive therapeutic approach for the treatment of cancer (10, 15). Preclinical studies have underscored the potential value of inhibiting Wnt/β-catenin activation in multiple tumor types such as colorectal cancer, advanced myeloid malignancies, and prostate cancer (NCT01931046). Under this circumstance, rational combination with Wnt/β-catenin signaling inhibitors may improve the curative effects and reduce the side effects of platinum-based therapies. Recently, it has been reported that CBP plays essential roles in Wnt/β-catenin pathway–mediated chemoresistance via its association with β-catenin (16–24). Herein, we demonstrated that the inactivation of Wnt/CBP/β-catenin signaling via PRI-724 significantly enhanced the therapeutic efficacy of platinum therapy in RBMS3-deleted EOC, which further supports the notion that Wnt/β-catenin signaling is a druggable target for preventing chemoresistance and provides an attractive clinical strategy for treating RBMS3-deleted EOC. Because the potential therapeutic value of PRI-724 is currently tested in clinical trials for the treatment of multiple cancer types, such as advanced colorectal and pancreatic cancers, a clinical trial that incorporates PRI-724 in patients with ovarian cancer is also worthy to design.

Aberrant RNA metabolism induced by RNA-binding proteins has emerged as an important determinant of oncogenesis and chemotherapy resistance via the global rewiring of pathways. For instance, the RNA-binding protein hnRNPA0 was recognized as the “successor” to p53 for checkpoint control, which drives chemoresistance of p53-mutant tumor cells via p27/Gadd45a mRNAs (37). The RNA-binding protein LARP1, acts as a central posttranscriptional regulator of survival and promotes chemotherapy resistance by differentially regulating the stability of a selection of mRNAs (38). These studies suggest that dysregulation of RNA-binding proteins contribute to chemotherapy failure. RBMS3, a newly identified RNA-binding protein with two RNA-binding motifs, plays roles in craniofacial and pancreas development and liver fibrosis by modulating the half-life of mRNA via binding to 3′UTR (39–41). Although its downregulation has been found in multiple cancers (42–45), RBMS3 reduction–mediated cancer progression was found to be through the transcriptional repression c-Myc (45). In contrast, the RBMS3 protein is predominantly restricted to the cytoplasm, suggesting that its major role is in RNA metabolism rather than transcription. In this study, we demonstrated that RBMS3 directly associated and stabilized the mRNA of multiple negative regulators in Wnt/β-catenin cascade via competitively preventing the repression ability of miR-126-5p, and that the loss of RBMS3 provided refuge protection for EOC cells from platinum therapy via activation of Wnt/β-catenin/CBP signaling. These results present reveal a novel RNA metabolic function of RBMS3 and may shed light on the breadth and extent of aberrant RNA metabolism in the context of chemoresistance.

It was recently reported that approximately 25% of the genome is affected by arm-level chromosomal deletions in cancers (46), and analysis of TCGA data in pan-cancer revealed that heterozygous chromosomal deletions on the 3p arm, among copy number alterations, is most associated with the poor survival of cancer patient (29). We found that the loss of Chr3p23-24.1 in 13 cancer types was significantly correlated with shorter relapse-free survival, and as such, may serve as an independent prognostic factor. We further demonstrated that loss of RBMS3, one of the genes in Chr3p23-24.1, correlates with the poor response and resistance of platinum therapy in EOC. Therefore, future studies are needed to determine the molecular mechanisms underlying chemoresistance induced by loss Chr3p23-24.1. The results of these studies will not only contribute to our understanding of 3p23-24.1-contanining genes to cancer progression and responses to platinum-based therapy but will also lead to the characterization of specific tumor patients' groups who may benefit from personalized therapies.

No potential conflicts of interest were disclosed.

Conception and design: L. Song, J. Li

Development of methodology: G. Wu, L. Cao, J. Zhu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.):

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Wu, L. Cao, J. Zhu, Z. Tan, M. Tang, Z. Li, Y. Hu, R. Yu, S. Zhang

Writing, review, and/or revision of the manuscript: J. Li

Study supervision: L. Song, J. Li

This work was supported by Natural Science Foundation of China [no. 81830082 (to J. Li), 91740119 (to J. Li), 91529301 (to J. Li), 81621004 (to J. Li), 91740118 (to L. Song), 81773106 (to L. Song), and 81530082 (to L. Song)]; Guangzhou Science and Technology Plan Projects [201803010098 (to J. Li)]; Guandong Natural Science Foundation [2018B030311009 (to J. Li), 2016A030308002 (to L. Song)]; The Fundamental Research Funds for the Central Universities [no. 17ykjc02 (to J. Li)].

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jayson
GC
,
Kohn
EC
,
Kitchener
HC
,
Ledermann
JA
. 
Ovarian Cancer
.
Lancet
2014
;
384
:
1376
88
.
2.
Schneble
EJ
,
Graham
LJ
,
Shupe
MP
,
Flynt
FL
,
Banks
KP
,
Kirkpatrick
AD
, et al
Current approaches and challenges in early detection of breast cancer recurrence
.
J Cancer
2014
;
5
:
281
90
.
3.
McGuire
WP
. 
Maintenance therapy for ovarian cancer: of Helsinki and Hippocrates
.
J Clin Oncol
2009
;
27
:
4633
4
.
4.
Vaughan
S
,
Coward
JI
,
Bast
RC
 Jr
,
Berchuck
A
,
Berek
JS
,
Brenton
JD
, et al
Rethinking ovarian cancer: recommendations for improving outcomes
.
Nat Rev Cancer
2011
;
11
:
719
25
.
5.
Pinato
DJ
,
Graham
J
,
Gabra
H
,
Sharma
R
. 
Evolving concepts in the management of drug resistant ovarian cancer: dose dense chemotherapy and the reversal of clinical platinum resistance
.
Cancer Treat Rev
2013
;
39
:
153
60
.
6.
Kelland
L
. 
The resurgence of platinum-based cancer chemotherapy
.
Nat Rev Cancer
2007
;
7
:
573
84
.
7.
Holohan
C
,
Van Schaeybroeck
S
,
Longley
DB
,
Johnston
PG
. 
Cancer drug resistance: an evolving paradigm
.
Nat Rev Cancer
2013
;
13
:
714
26
.
8.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
9.
Meads
MB
,
Gatenby
RA
,
Dalton
WS
. 
Environment-mediated drug resistance: a major contributor to minimal residual disease
.
Nat Rev Cancer
2009
;
9
:
665
74
.
10.
Barker
N
,
Clevers
H
. 
Mining the Wnt pathway for cancer therapeutics
.
Nat Rev Drug Discov
2006
;
5
:
997
1014
.
11.
Guo
Q
,
Chen
Y
,
Zhang
B
,
Kang
M
,
Xie
Q
,
Wu
Y
. 
Potentiation of the effect of gemcitabine by emodin in pancreatic cancer is associated with survivin inhibition
.
Biochem Pharmacol
2009
;
77
:
1674
83
.
12.
Almendro
V
,
Ametller
E
,
Garcia-Recio
S
,
Collazo
O
,
Casas
I
,
Auge
JM
, et al
The role of MMP7 and its cross-talk with the FAS/FASL system during the acquisition of chemoresistance to oxaliplatin
.
PLoS One
2009
;
4
:
e4728
.
13.
Jun
S
,
Jung
YS
,
Suh
HN
,
Wang
W
,
Kim
MJ
,
Oh
YS
, et al
LIG4 mediates Wnt signalling-induced radioresistance
.
Nat Commun
2016
;
7
:
10994
.
14.
Yamada
T
,
Takaoka
AS
,
Naishiro
Y
,
Hayashi
R
,
Maruyama
K
,
Maesawa
C
, et al
Transactivation of the multidrug resistance 1 gene by T-cell factor 4/beta-catenin complex in early colorectal carcinogenesis
.
Cancer Res
2000
;
60
:
4761
6
.
15.
Anastas
JN
,
Moon
RT
. 
WNT signalling pathways as therapeutic targets in cancer
.
Nat Rev Cancer
2013
;
13
:
11
26
.
16.
Ma
H
,
Nguyen
C
,
Lee
KS
,
Kahn
M
. 
Differential roles for the coactivators CBP and p300 on TCF/beta-catenin-mediated survivin gene expression
.
Oncogene
2005
;
24
:
3619
31
.
17.
He
K
,
Xu
T
,
Xu
Y
,
Ring
A
,
Kahn
M
,
Goldkorn
A
. 
Cancer cells acquire a drug resistant, highly tumorigenic, cancer stem-like phenotype through modulation of the PI3K/Akt/beta-catenin/CBP pathway
.
Int J Cancer
2014
;
134
:
43
54
.
18.
Xia
Z
,
Guo
M
,
Liu
H
,
Jiang
L
,
Li
Q
,
Peng
J
, et al
CBP-dependent Wnt/beta-catenin signaling is crucial in regulation of MDR1 transcription
.
Curr Cancer Drug Targets
2015
;
15
:
519
32
.
19.
Emami
KH
,
Nguyen
C
,
Ma
H
,
Kim
DH
,
Jeong
KW
,
Eguchi
M
, et al
A small molecule inhibitor of beta-catenin/CREB-binding protein transcription [corrected]
.
Proc Natl Acad Sci U S A
2004
;
101
:
12682
7
.
20.
Gang
EJ
,
Hsieh
YT
,
Pham
J
,
Zhao
Y
,
Nguyen
C
,
Huantes
S
, et al
Small-molecule inhibition of CBP/catenin interactions eliminates drug-resistant clones in acute lymphoblastic leukemia
.
Oncogene
2014
;
33
:
2169
78
.
21.
Wend
P
,
Fang
L
,
Zhu
Q
,
Schipper
JH
,
Loddenkemper
C
,
Kosel
F
, et al
Wnt/beta-catenin signalling induces MLL to create epigenetic changes in salivary gland tumours
.
EMBO J
2013
;
32
:
1977
89
.
22.
Arensman
MD
,
Telesca
D
,
Lay
AR
,
Kershaw
KM
,
Wu
N
,
Donahue
TR
, et al
The CREB-binding protein inhibitor ICG-001 suppresses pancreatic cancer growth
.
Mol Cancer Ther
2014
;
13
:
2303
14
.
23.
Nagaraj
AB
,
Joseph
P
,
Kovalenko
O
,
Singh
S
,
Armstrong
A
,
Redline
R
, et al
Critical role of Wnt/beta-catenin signaling in driving epithelial ovarian cancer platinum resistance
.
Oncotarget
2015
;
6
:
23720
34
.
24.
Lin
HH
,
Feng
WC
,
Lu
LC
,
Shao
YY
,
Hsu
CH
,
Cheng
AL
. 
Inhibition of the Wnt/beta-catenin signaling pathway improves the anti-tumor effects of sorafenib against hepatocellular carcinoma
.
Cancer Lett
2016
;
381
:
58
66
.
25.
Cai
Y
,
Crowther
J
,
Pastor
T
,
Abbasi Asbagh
L
,
Baietti
MF
,
De Troyer
M
, et al
Loss of chromosome 8p governs tumor progression and drug response by altering lipid metabolism
.
Cancer Cell
2016
;
29
:
751
66
.
26.
Nagata
Y
,
Lan
KH
,
Zhou
X
,
Tan
M
,
Esteva
FJ
,
Sahin
AA
, et al
PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients
.
Cancer Cell
2004
;
6
:
117
27
.
27.
Intergroup Radiation Therapy Oncology Group
,
Cairncross
G
,
Berkey
B
,
Shaw
E
,
Jenkins
R
,
Scheithauer
B
, et al
Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402
.
J Clin Oncol
2006
;
24
:
2707
14
.
28.
Zabarovsky
ER
,
Lerman
MI
,
Minna
JD
. 
Tumor suppressor genes on chromosome 3p involved in the pathogenesis of lung and other cancers
.
Oncogene
2002
;
21
:
6915
35
.
29.
Gross
AM
,
Orosco
RK
,
Shen
JP
,
Egloff
AM
,
Carter
H
,
Hofree
M
, et al
Multi-tiered genomic analysis of head and neck cancer ties TP53 mutation to 3p loss
.
Nat Genet
2014
;
46
:
939
43
.
30.
Shepherd
TG
,
Theriault
BL
,
Campbell
EJ
,
Nachtigal
MW
. 
Primary culture of ovarian surface epithelial cells and ascites-derived ovarian cancer cells from patients
.
Nat Protoc
2006
;
1
:
2643
9
.
31.
Keene
JD
,
Komisarow
JM
,
Friedersdorf
MB
. 
RIP-Chip: the isolation and identification of mRNAs, microRNAs and protein components of ribonucleoprotein complexes from cell extracts
.
Nat Protoc
2006
;
1
:
302
7
.
32.
Domcke
S
,
Sinha
R
,
Levine
DA
,
Sander
C
,
Schultz
N
. 
Evaluating cell lines as tumour models by comparison of genomic profiles
.
Nat Commun
2013
;
4
:
2126
.
33.
Siddik
ZH
. 
Cisplatin: mode of cytotoxic action and molecular basis of resistance
.
Oncogene
2003
;
22
:
7265
79
.
34.
Jiang
L
,
Yin
M
,
Wei
X
,
Liu
J
,
Wang
X
,
Niu
C
, et al
Bach1 represses Wnt/beta-catenin signaling and angiogenesis
.
Circ Res
2015
;
117
:
364
75
.
35.
Evans
PM
,
Chen
X
,
Zhang
W
,
Liu
C
. 
KLF4 interacts with beta-catenin/TCF4 and blocks p300/CBP recruitment by beta-catenin
.
Mol Cell Biol
2010
;
30
:
372
81
.
36.
Wang
Q
,
Zhou
Y
,
Rychahou
P
,
Liu
C
,
Weiss
HL
,
Evers
BM
. 
NFAT5 represses canonical Wnt signaling via inhibition of beta-catenin acetylation and participates in regulating intestinal cell differentiation
.
Cell Death Dis
2013
;
4
:
e671
.
37.
Cannell
IG
,
Merrick
KA
,
Morandell
S
,
Zhu
CQ
,
Braun
CJ
,
Grant
RA
, et al
A pleiotropic RNA-binding protein controls distinct cell cycle checkpoints to drive resistance of p53-defective tumors to chemotherapy
.
Cancer Cell
2015
;
28
:
623
37
.
38.
Hopkins
TG
,
Mura
M
,
Al-Ashtal
HA
,
Lahr
RM
,
Abd-Latip
N
,
Sweeney
K
, et al
The RNA-binding protein LARP1 is a post-transcriptional regulator of survival and tumorigenesis in ovarian cancer
.
Nucleic Acids Res
2016
;
44
:
1227
46
.
39.
Jayasena
CS
,
Bronner
ME
. 
Rbms3 functions in craniofacial development by posttranscriptionally modulating TGF-beta signaling
.
J Cell Biol
2012
;
199
:
453
66
.
40.
Lu
CK
,
Lai
YC
,
Chen
HR
,
Chiang
MK
. 
Rbms3, an RNA-binding protein, mediates the expression of Ptf1a by binding to its 3′UTR during mouse pancreas development
.
DNA Cell Biol
2012
;
31
:
1245
51
.
41.
Fritz
D
,
Stefanovic
B
. 
RNA-binding protein RBMS3 is expressed in activated hepatic stellate cells and liver fibrosis and increases expression of transcription factor Prx1
.
J Mol Biol
2007
;
371
:
585
95
.
42.
Zhang
T
,
Wu
Y
,
Fang
Z
,
Yan
Q
,
Zhang
S
,
Sun
R
, et al
Low expression of RBMS3 and SFRP1 are associated with poor prognosis in patients with gastric cancer
.
Am J Cancer Res
2016
;
6
:
2679
89
.
43.
Liang
YN
,
Liu
Y
,
Meng
Q
,
Li
X
,
Wang
F
,
Yao
G
, et al
RBMS3 is a tumor suppressor gene that acts as a favorable prognostic marker in lung squamous cell carcinoma
.
Med Oncol
2015
;
32
:
459
.
44.
Chen
J
,
Kwong
DL
,
Zhu
CL
,
Chen
LL
,
Dong
SS
,
Zhang
LY
, et al
RBMS3 at 3p24 inhibits nasopharyngeal carcinoma development via inhibiting cell proliferation, angiogenesis, and inducing apoptosis
.
PLoS One
2012
;
7
:
e44636
.
45.
Li
Y
,
Chen
L
,
Nie
CJ
,
Zeng
TT
,
Liu
H
,
Mao
X
, et al
Downregulation of RBMS3 is associated with poor prognosis in esophageal squamous cell carcinoma
.
Cancer Res
2011
;
71
:
6106
15
.
46.
Beroukhim
R
,
Mermel
CH
,
Porter
D
,
Wei
G
,
Raychaudhuri
S
,
Donovan
J
, et al
The landscape of somatic copy-number alteration across human cancers
.
Nature
2010
;
463
:
899
905
.