Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Cis-acting circ-CTNNB1 promotes β- signaling and cancer progression via

DDX3-mediated transactivation of YY1

Feng Yang 1, #, Erhu Fang 1, #, Hong Mei 1, Yajun Chen 2, Huanhuan Li 1, Dan Li 1, Huajie Song 1, Jianqun Wang 1, Mei Hong 1, Wenjing Xiao 2, Xiaojing Wang 3, Kai Huang 3, Liduan Zheng 2, 3, *, Qiangsong Tong 1, 3, *

1 Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China 2 Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China 3 Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, P. R. China

Running title: circ-CTNNB1 promotes cancer progression

Key words: circular RNA; DEAD-box polypeptide 3; Yin Yang 1; beta-catenin; cancer progression

*Corresponding author:

Qiangsong Tong, Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China. Tel: +86-27-85726005; Fax: +86-27-85726821; Email: [email protected]

Liduan Zheng, Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China. Tel: +86-27-85726129; Fax: +86-27-85726821; Email: [email protected]

# Yang F. and Fang E. contributed equally to this work.

Conflict of Interest Statement: The authors disclose no potential conflicts of interest.

Word count: 4997; Total number of figures and tables: 7

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

Circular RNAs (circRNA), a subclass of noncoding RNA characterized by covalently closed continuous loops, play emerging roles in tumorigenesis and aggressiveness. However, the functions and underlying mechanisms of circRNA in regulating Wnt/β-catenin signaling and cancer progression remain elusive. Here we screen cis-acting circRNA generated by β-catenin

(CTNNB1)/transcription factor 7 like 2 and identify one intronic circRNA derived from

CTNNB1 (circ-CTNNB1) as a novel driver of cancer progression. Circ-CTNNB1 was predominantly expressed in the nucleus, upregulated in cancer tissues and cell lines, and associated with unfavorable outcome in cancer patients. Circ-CTNNB1 promoted β-catenin activation, growth, invasion, and metastasis in cancer cells. Circ-CTNNB1 bound DEAD-box polypeptide 3 (DDX3) to facilitate its physical interaction with transcription factor Yin Yang 1 (YY1), resulting in transactivation of YY1 and transcriptional alteration of downstream genes associated with β-catenin activation and cancer progression. Pre-clinically, administration of lentivirus-mediated short hairpin

RNA targeting circ-CTNNB1 or a cell-penetrating inhibitory peptide blocking the circ-CTNNB1-DDX3 interaction inhibited downstream expression, tumorigenesis, and aggressiveness in cancer cells. Taken together, these results demonstrate cis-acting circ-CTNNB1 as a mediator of β-catenin signaling and cancer progression through DDX3-mediated transactivation of

YY1.

2

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Significance

Findings reveal the oncogenic functions of a cis-acting circular RNA in β-catenin activation and cancer progression, with potential value as a therapeutic target for human cancers.

3

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

The Wnt/β-catenin signaling pathway plays essential roles in biological processes, including embryogenesis, tissue homeostasis, and proliferation or differentiation of mammalian cells (1).

Within the cytoplasm, β-catenin is constitutively phosphorylated at (Ser)/ (Thr) residues by glycogen synthase kinase 3 beta (GSK-3β), and phosphorylated β-catenin is degraded by the ubiquitin/proteasome pathway (2). Upon Wnt signals (such as WNT3), the phosphorylation of

β-catenin is reduced, resulting in stabilization, cytoplasmic accumulation, and subsequent nuclear translocation, whereas it interacts with T-cell factor (TCF)/lymphoid enhancer-binding factor to modulate target (2). As characterized by nuclear β-catenin accumulation, activation of Wnt/β-catenin signaling is documented in many types of cancer, and contributes to cancer progression (3,4). However, the mechanisms underlying β-catenin activation in cancers still remain to be determined.

Circular RNAs (circRNA), a group of transcripts characterized by covalently closed continuous loops, are generated from exons or introns of parent genes (5). Exonic circRNAs are predominantly localized within the cytoplasm (6), and some of them (such as ciRS-7 and circSry) regulate the expression of other genes in trans by serving as sponges for microRNA (miRNA) (7,8).

Beside the trans-regulatory mode, circRNAs are able to regulate gene expression in a cis-acting manner. For example, circRNA derived from the second exon of muscleblind (circMbl) regulates parent gene expression by competing with linear splicing (9). Exonic circRNA generated from poly(A) binding protein nuclear 1 (circPABPN1) represses the translation of PABPN1 via preventing the enrichment of human antigen R on mRNA (10). In addition, ci-ankrd52, originating from the intron of repeat domain 52 (ANKRD52), binds RNA polymerase II to regulate the

4

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

transcription of ANKRD52 (11). A special class of exon–intron circRNAs are abundant in the nucleus, lack of miRNA binding sites, and involved in transcriptional regulation of parent gene expression

(12). Dysregulated circRNAs have been identified in almost all types of cancers, such as hepatocellular carcinoma, prostate cancer, and colorectal cancer (13). However, the roles of cis-acting circRNA in β-catenin activation and cancer progression still remain elusive.

In this study, through screening cis-regulatory circRNAs derived from β-catenin (CTNNB1) and transcription factor 7 like 2 (TCF7L2) genes, essential components of β-catenin/TCF transcriptional complexes (14), we identified one intronic circRNA derived from CTNNB1

(circ-CTNNB1) as a driver of cancer progression. We demonstrate that circ-CTNNB1 is upregulated in cancer tissues, and facilitates the β-catenin activation, tumorigenesis, and aggressiveness.

Mechanistically, circ-CTNNB1 binds DEAD-box polypeptide 3 (DDX3) to facilitate its physical interaction with Yin Yang 1 (YY1), which results in transactivation of YY1 and subsequent transcriptional alteration of downstream genes associated with β-catenin activation, indicating the essential roles of circ-CTNNB1/DDX3/YY1 axis in cancer progression.

Materials and Methods

Cell culture

Human embryonic kidney (HEK293T) cells and cell lines of gastric cancer (MKN-45 and

AGS), cervical cancer (HeLa), prostate cancer (PC-3), breast cancer (MCF-7), colon cancer

(HCT-116 and LoVo), liver cancer (HepG2), and neuroblastoma [BE(2)-C and SH-SY5Y] were obtained from American Type Culture Collection (Rockville, MD) and Japanese Collection of

Research Bioresources Cell Bank (Osaka, Japan), authenticated by short tandem repeat profiling, and used within 6 months after resuscitation of frozen aliquots. Mycoplasma contamination was

5

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

regularly examined using Lookout Mycoplasma PCR Detection Kit (Sigma, St. Louis, MO). Cells were cultured with RPMI1640 containing 10% fetal bovine serum (Life Technologies, Inc.,

Gaithersburg, MD), incubated in serum-free RPMI1640 for 4 hrs, and treated with XAV939

(Selleckchem, Houston, TX), WNT3a (R&D Systems, Inc., Minneapolis, MN), or lithium chloride

(LiCl, Sigma).

RT-PCR and real-time quantitative RT-PCR

Nuclear, cytoplasmic, and total RNAs were extracted using RNA Subcellular Isolation Kit

(Active Motif, Carlsbad, CA) or RNeasy Mini Kit (Qiagen Inc., Redwood City, CA). For circRNA detection, RNase R (3 U/mg, Epicenter, Madison, WI) digestion was undertaken at 37°C for 15 min.

Reverse transcription and real-time PCR were performed using Transcriptor First Strand cDNA

Synthesis Kit (Roche, Indianapolis, IN), SYBR Green PCR Master Mix (Applied Biosystems, Foster

City, CA), and primer sets (Supplementary Table S1). The results of transcript levels were analyzed by 2-△△Ct method.

Northern blot

The junction or non-junction probe for circ-CTNNB1 was synthesized and labeled by digoxigenin (DIG, Supplementary Table S2). Northern blot was performed as previously described

(7,8). Blots were washed stringently, detected by anti-DIG antibody, and recorded on X-ray films with chemiluminescence substrate CSPD (Roche).

Western blot

Tissue or cellular protein was extracted with 1× cell lysis buffer (Promega, Madison, WI).

Western blot was performed as previously described (15,16), with antibodies specific for β-catenin

(ab32572), c-myc (ab32072), cyclin D1 (ab134175), DDX3 (ab37160), RNA binding motif protein

6

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14 (RBM14, ab70636), RNA binding motif protein 39 (RBM39, ab12326), YY1 (ab109228), Wnt family member 1 (WNT1, ab15251), WNT3 (ab172612), axin 2 (AXIN2, ab109307), frizzled class receptor 10 (FZD10, ab83044), bone morphogenetic protein 4 (BMP4, ab39973), FLAG (ab1162),

Myc (ab9106), β- (ab8226, Abcam Inc., Cambridge, MA), phosphorylated β-catenin

(Ser33/Ser37, sc-57535), glutathione S-transferase (GST, sc-33614), histone H3 (sc-24516), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH, sc-47724, Santa Cruz Biotechnology, Santa

Cruz, CA).

Over-expression or knockdown of genes

Linear circ-CTNNB1 (378 bp) was obtained from gastric cancer tissues by PCR

(Supplementary Table S2) and inserted into pLCDH-ciR (Geenseed Biotech Co., Guangzhou, China).

Human DDX3 cDNA (1989 bp) and its Myc-tagged truncations were provided by Dr. Christof

Niehrs, while YY1 cDNA (1245 bp) was a gift from Dr. Makoto Miyagishi. The amplified truncations of DDX3 and YY1 (Supplementary Table S2) were subcloned into pGEX-6P-1 or pCMV-3Tag-1A (Addgene, Cambridge, MA). Mutation of DDX3 was prepared with GeneTailorTM

Site-Directed Mutagenesis System (Invitrogen, Carlsbad, CA) and PCR primers (Supplementary

Table S2). Oligonucleotides encoding short hairpin RNA (shRNA) against circRNAs, DDX3, YY1,

ETS-related gene (ERG), or specificity protein 1 (SP1) (Supplementary Table S3) were inserted into

GV298 (Genechem Co., Ltd, Shanghai, China). After selection for neomycin or puromycin

(Invitrogen) resistance, stable cell lines were obtained.

Lentiviral vector construction

Lentiviral vectors were co-transfected with packaging plasmids psPAX2 and pMD2G into

HEK293T cells. Infectious lentiviruses were harvested at 36 and 60 hrs after transfection, and

7

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

filtered through 0.45 μm PVDF filters. Recombinant lentiviruses were concentrated 100-fold by ultracentrifugation (2 hrs at 120,000 g).

RNA sequencing (RNA-seq) assay

Total RNA was extracted using TRIzol® reagent (Life Technologies, Inc.). Transcriptome sequencing on an Illumina HiSeq X Ten platform was carried out at Novogene Bioinformatics

Technology Co., Ltd. (Beijing, China). Sequencing results were deposited in Gene Expression

Omnibus database (accession code GSE120496).

Dual-luciferase reporter assay

The TOP-FLASH and FOP-FLASH reporters were obtained from Millipore (Temecula, CA).

Human YY1 luciferase reporter was established by inserting complementary oligonucleotides containing four canonical YY1 binding sites (Supplementary Table S2) into pGL3-Basic (Promega).

Dual-luciferase assay was performed as previously described (15,16). Relative β-catenin activation was determined by the TOP-FLASH/FOP-FLASH ratio.

Biotin-labeled RNA pull-down and mass spectrometry analysis

The 5'-monophosphorylated linear or random probe of circ-CTNNB1 was in vitro transcribed using Biotin RNA Labeling Mix (Roche), reaction mixture containing excessive guanosine monophosphate (7.5 mM) and guanosine triphosphate (1.5 mM), and T7 RNA polymerase (17,18).

After incubation with guide oligonucleotide targeting circular junction (Supplementary Table S2), the 5'-monophosphorylated linear probe was circularized using T4 RNA ligase I, treated with RNase

R, and purified with RNeasy Mini Kit (Qiagen Inc.). The lysates of 2×107 cancer cells were incubated with 3 μg of biotin-labeled probe, and treated with 35 μl of Streptavidin C1 magnetic beads (Invitrogen) for 1 hr. The retrieved protein was detected by western blot or mass spectrometry

8

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

analysis (Wuhan Institute of Biotechnology, Wuhan, China) (15).

RNA fluorescence in situ hybridization (RNA-FISH)

Cells were seeded on coverslips, fixed with 4% paraformaldehyde, and incubated with 50% formamide, 2× SSC, E. coli transfer RNA (0.25 mg/ml), salmon sperm DNA (0.25 mg/ml, Life

Technologies, Inc.), bovine serum albumin (2.5 mg/ml, Roche), and DIG-labeled antisense or sense probe for circ-CTNNB1 junction (Supplementary Table S2). The probes for GAPDH and U1 were generated by in vitro transcription of PCR products (Supplementary Table S1) using DIG Labeling

Kit (MyLab Corporation, Beijing, China). Hybridization was undertaken using Fluorescent In Situ

Hybridization Kit (RiboBio, Guangzhou, China), with or without RNase R (3 U/mg) treatment, while nuclei were counterstained using 4',6-diamidino-2-phenylindole (DAPI).

Fluorescence immunocytochemical staining

Cancer cells were grown on coverslips, and incubated with antibodies specific for β-catenin

(ab32572; Abcam Inc.; 1:100 dilution) or DDX3 (ab37160; Abcam Inc.; 1:100 dilution). Then, coverslips were treated with Alexa Fluor 594 or 498 goat anti-rabbit IgG (1:1000 dilution) and DAPI

(300 nmol/L) staining.

Co-immunoprecipitation (co-IP) assay

Co-IP was undertaken as previously described (15), with antibodies specific for DDX3

(sc-365768, Santa Cruz Biotechnology), YY1 (ab109228), FLAG (ab1162), or Myc (ab9106, Abcam

Inc.).

Bimolecular fluorescence complementation (BiFC) assay

Human DDX3 cDNA (1989 bp) and YY1 cDNA (1245 bp) were subcloned into BiFC vectors pBiFC-VN173 and pBiFC-VC155 (Addgene), respectively (Supplementary Table S2). The

9

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

recombinant constructs were co-transfected into cancer cells for 24 hrs. The fluorescence emission was observed under a confocal microscope, with excitation and emission wavelengths of 488 and

500 nm, respectively.

Rescue of target gene expression

To rescue the circ-CTNNB1 knockdown-altered levels of target genes, the expression vector of

DDX3 or YY1 was transfected into stable cell lines. To restore the altered expression of target genes induced by circ-CTNNB1, shRNAs against DDX3 or YY1 (Supplementary Table S3) were transfected into cancer cells using Genesilencer Transfection Reagent (Genlantis, San Diego, CA).

Chromatin immunoprecipitation (ChIP)

ChIP assay was undertaken using EZ-ChIP kit (Upstate Biotechnology, Temacula, CA) (15,16), with antibodies specific for YY1 (ab38422), enhancer of zeste homologue 2 (EZH2, ab228697), histone deacetylase 1 (HDAC1, ab7028), or p300 (ab14984, Abcam Inc.) and primer sets

(Supplementary Table S1).

Cross-linking RNA immunoprecipitation (RIP) assay

Cells were cross-linked by ultraviolet light at 254 nm (200 J/cm2) (15,16). RIP assay was performed using Magna RIPTM RNA-Binding Protein Immunoprecipitation Kit (Millipore), using antibodies specific for DDX3 (sc-365768, Santa Cruz Biotechnology) or FLAG (ab1162, Abcam

Inc.). Co-precipitated RNAs were detected by RT-PCR or real-time quantitative RT-PCR (qRT-PCR) with primers (Supplementary Table S1).

In vitro binding assay

A series of DDX3 truncates were amplified with primer sets (Supplementary Table S2), subcloned into pGEX-6P-1 (Addgene), and transformed into E. coli to produce GST-tagged DDX3

10

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

truncation (15,16). The DDX3-circRNA complexes were pulled down using GST beads

(Sigma). Protein was detected by SDS-PAGE and western blot, while circRNA was measured by

RT-PCR with divergent primers (Supplementary Table S1).

RNA electrophoretic mobility shift assay (EMSA)

Biotin-labeled circ-CTNNB1 probe was prepared as described above. RNA EMSA using nuclear extracts or recombinant DDX3 protein was performed using LightShift Chemiluminescent

RNA EMSA Kit (Thermo Fisher Scientific, Inc., Grand Island, NY).

Design and synthesis of inhibitory peptides

The inhibitory peptides blocking the circ-CTNNB1-DDX3 interaction were designed, and chemically synthesized by linking with biotin-labeled cell-penetrating peptide (YGRKKRRQRRR) from Tat protein transduction domain at the N-terminus and conjugating with fluorescein isothiocyanate (FITC) at the C-terminus (ChinaPeptides Co. Ltd, Shanghai, China), with purity larger than 95%.

Biotin-labeled peptide pull-down assay

Cellular total RNA was isolated using RNeasy Mini Kit (Qiagen Inc.), and incubated with biotin-labeled peptide and streptavidin-agarose at 4°C. The beads were extensively washed, and circRNA pulled down was measured by RT-PCR or real-time qRT-PCR.

In vitro cell viability, growth, and invasion assays

The 2-(4,5-dimethyltriazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT, Sigma) colorimetric

(15), soft agar (15,16), and matrigel invasion (15,16) assays were undertaken to measure the in vitro viability, growth, and invasiveness of cancer cells.

In vivo growth, metastasis, and therapeutic assays

11

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

All animal experiments were carried out in accordance with NIH Guidelines for the Care and

Use of Laboratory Animals, and approved by the Animal Care Committee of Tongji Medical

College (approval number: Y20080290). The in vivo tumor growth (1×106 cancer cells per mouse, n=5 per group) and experimental metastasis (0.4×106 cancer cells per mouse, n=5 per group) studies were performed with blindly randomized four-week-old female BALB/c nude mice as previously described (15,16). For in vivo therapeutic studies, after cancer cell inoculation, mice were blindly randomized and treated by intratumoral or tail vein injection of lentivirus-mediated shRNA (1×107 plaque-forming units) or synthesized cell-penetrating peptide (ChinaPeptides, Shanghai, China) as indicated. The In Vivo Optical Imaging System (In-Vivo FX PRO, Bruker Corporation, Billerica,

MA) was applied to acquire fluorescent images of xenografts in nude mice.

Patient tissue samples

Human tissue study was approved by the Institutional Review Board of Tongji Medical

College (approval number: 2011-S085). All procedures were undertaken in accordance with guidelines set forth by Declaration of Helsinki. Written informed consent was obtained from all patients. Fresh cancer tissues were collected at surgery, validated by pathological diagnosis, and stored at -80°C.

Immunohistochemistry

Immunohistochemical staining and quantitative evaluation were performed as previously described (15,16), with antibodies specific for Ki-67 (sc-23900, Santa Cruz Biotechnology) or CD31

(ARG52748, Arigo, Hsinchu City, Taiwan).

Statistical Analysis

All data were shown as mean ± standard error of the mean (SEM). The cutoff for gene

12

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expression was defined by average values. Student’s t test, analysis of variance, and χ2 analysis were used to compare the difference in cancer cells or tissues. Fisher's exact test was used to analyze the overlapping of genes. Pearson’s correlation coefficient assay was used to analyze the expression correlation. Log-rank test was used to assess survival difference. All statistical tests were two-sided.

Results circ-CTNNB1 is a cis-acting circRNA upregulated in cancer tissues and cells

Mining of circRNA databases circBase (19) and circInteractome (20) revealed 38 potential cis-acting circRNAs derived from CTNNB1 and TCF7L2 genes, with 15 having more than 2 read scores (Fig. 1A). Further validating RT-PCR with divergent primers revealed 7 detectable circRNAs in gastric cancer AGS cells (Supplementary Fig. S1A). Three circRNAs, hsa_circ_0020050, hsa_circ_0004030, and hsa_circ_0123778 (circ-CTNNB1), were upregulated in the tissues of gastric cancer, prostate cancer, and colorectal cancer (Fig. 1B and Supplementary Fig. S1B), cancer types with obvious β-catenin activation (3,4,21). Knockdown of circ-CTNNB1, but not of hsa_circ_0020050 or hsa_circ_0004030, attenuated the β-catenin activity in gastric cancer AGS and colon cancer HCT-116 cells, with similar effects to established β-catenin inhibitor XAV939 (22) (Fig.

1C and Supplementary Fig. S1C). The existence of 378-nt circ-CTNNB1 generated from intron 10 of

CTNNB1 (Fig. 1D) was further confirmed by RT-PCR with divergent primers and Sanger sequencing

(Fig. 1E). Endogenous or ectopic expression of circ-CTNNB1 resistant to RNase R digestion was also detected by a junction-specific probe in Northern blot (Fig. 1F), which did not detect the pre-mRNA or mRNA bands of CTNNB1 (Fig. 1F). Higher circ-CTNNB1 levels were observed in cancer cell lines, than those of embryonic kidney HEK293T cells (Fig. 1G). In addition, circ-CTNNB1 was elevated in gastric cancer tissues with metastasis, than those without metastasis

13

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(P<1.0×10-4, Fig. 1H). Kaplan–Meier survival analysis of 81 gastric cancer cases indicated that high circ-CTNNB1 expression was associated with poor overall survival of patients (P<1.0×10-4, Fig. 1I).

These results indicated that circ-CTNNB1 was a cis-acting circRNA upregulated in cancer tissues and cells.

Circ-CTNNB1 promotes the growth and aggressiveness of cancer cells

To explore the roles of circ-CTNNB1 in cancer progression, cancer cell lines representing moderate expression levels were applied. The circ-CTNNB1 vector-induced circularization efficiency was approximately 80% in Northern blot (Supplementary Fig. S2A). Stable transfection of circ-CTNNB1 and three independent shRNAs against circ-CTNNB1 (sh-circ-CTNNB1) into gastric cancer cell lines MKN-45 and AGS, prostate cancer PC-3 cells, and colon cancer HCT-116 cells resulted in increased and reduced levels of circ-CTNNB1, respectively (Supplementary Fig. S2B), which were resistant to efficient RNase R digestion (Supplementary Fig. S2C). Stable over-expression or knockdown of circ-CTNNB1 facilitated and reduced the viability, anchorage-independent growth and invasiveness of cancer cells, respectively (Fig. 2A, Fig. 2B, Fig.

2C, Supplementary Fig. S2D and Fig. S2E). Consistently, stable transfection of circ-CTNNB1 or sh-circ-CTNNB1 #3 resulted in a significant increase and decrease in the growth, tumor weight,

Ki-67 proliferation index, and CD31-positive intratmoral microvessels of MKN-45 cells-formed subcutaneous xenografts, respectively (Fig. 2D and Fig. 2E). In experimental metastasis assay, athymic nude mice treated with tail vein injection of MKN-45 cells stably transfected with circ-CTNNB1 or sh-circ-CTNNB1 #3 displayed more and less lung metastatic colonies, with lower and greater survival probability, respectively (Fig. 2F and Fig. 2G). These results suggested that circ-CTNNB1 promoted the growth and aggressiveness of cancer cells in vivo and in vitro.

14

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Circ-CTNNB1 increases the expression of β-catenin in cancer cells

To elucidate the mechanisms underlying the oncogenic roles of circ-CTNNB1, we investigated its effects on the expression of its parent gene CTNNB1. Stable transfection of circ-CTNNB1 or sh-circ-CTNNB1 #3 increased and decreased the protein levels, but not the promoter activity or transcript levels, of CTNNB1 in MKN-45, AGS, PC-3, and HCT-116 cells (Fig. 3A, Fig. 3B, Fig. 3C,

Supplementary Fig. S3A and Fig. S3B). Increased cytoplasmic and nuclear accumulation of

β-catenin was observed in MKN-45 and AGS cells stably over-expressing circ-CTNNB1 (Fig. 3D,

Fig. 3E, and Supplementary Fig. S3C). In addition, stable transfection of circ-CTNNB1 or sh-circ-CTNNB1 #3 resulted in increased and decreased β-catenin activity, and abolished the effects of Wnt signaling inhibitor (XAV939) and activator (WNT3a) in cancer cells, respectively (Fig. 3F).

To determine whether circ-CTNNB1 affected the degradation of β-catenin, we observed the alteration in phosphorylated β-catenin levels in cancer cells. Ectopic expression of circ-CTNNB1 decreased the phosphorylation of β-catenin, and increased the expression of β-catenin and its downstream genes c-Myc and cyclin D1 (Fig. 3G). Meanwhile, knockdown of circ-CTNNB1 led to increased phosphorylation of β-catenin and decreased β-catenin levels (Fig. 3H), which was rescued by treatment of cancer cells with WNT3a or GSK-3β inhibitor LiCl (Fig. 3H). In addition, treatment with proteasome inhibitor (MG-132) abolished the decrease in β-catenin expression induced by knockdown of circ-CTNNB1 (Fig. 3H). These findings indicated that circ-CTNNB1 increased the expression of β-catenin in cancer cells.

Circ-CTNNB1 is predominantly localized in the nucleus and interacts with DDX3

We further investigated the localization and protein partners of circ-CTNNB1 within cancer cells. RNA-FISH and subcellular fractionation assays revealed abundant signals and enrichment of

15

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

circ-CTNNB1 in the nucleus (Fig. 4A and Fig. 4B). To identify the protein partner of circ-CTNNB1, we performed the biotin-labeled RNA pull-down followed by a proteomic analysis of

RNA-associated protein complex in cancer cells, using circRNA probe generated by ligation of in vitro transcribed linear circ-CTNNB1 (17,18) (Fig. 4C). Mass spectrometry assay revealed 69 and 77 differential proteins between circ-CTNNB1 and linear transcript pull-down groups form MKN-45 and AGS cells, respectively (Fig. 4D and Supplementary Table S4), and over-lapping with established RNA-binding proteins (RBPs, https://www.ablife.cc) indicated three RBPs consistently pulled down by biotin-labeled circ-CTNNB1 in both cell lines (Fig. 4D), including DDX3, RBM14, and RBM39. Validating biotin-labeled RNA pull-down assay further indicated that circ-CTNNB1 was able to dose-dependently interact with DDX3, but not with RBM14 or RBM39, in the lysates of

MKN-45 cells (Fig. 4E). However, no interaction was detected between DDX3 and linear or random probe (Fig. 4E). Dual RNA-FISH and immunofluorescence assay confirmed the co-localization of circ-CTNNB1 and DDX3 in MKN-45 cells, which was increased by ectopic expression of circ-CTNNB1 (Fig. 4F). RIP assay indicated the endogenous enrichment of circ-CTNNB1 in RNA co-precipitated by DDX3 antibody in MKN-45 cells, which was increased by stable over-expression of circ-CTNNB1 (Fig. 4G). Consistently, RNA EMSA showed that circ-CTNNB1 strongly interacted with endogenous DDX3 in nuclear extracts or GST-tagged recombinant DDX3 protein (Fig. 4H).

These findings indicated that circ-CTNNB1 was predominantly localized in the nucleus and interacted with DDX3.

Circ-CTNNB1 cooperates with DDX3 to facilitate the expression and activity of β-catenin

We further investigated the interplay between circ-CTNNB1 and DDX3 in regulating the expression and activity of β-catenin. Knockdown of DDX3 or treatment with an established DDX3

16

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

inhibitor (RK-33) (23) abolished the increased β-catenin expression induced by ectopic expression of circ-CTNNB1 in MKN-45 cells (Fig. 5A and Supplementary Fig. S3D). Meanwhile, ectopic expression of DDX3 prevented the decrease of β-catenin levels in MKN-45 and AGS cells with stable knockdown of circ-CTNNB1 (Supplementary Fig. S3E). In addition, RK-33 treatment and DDX3 over-expression abolished the increased and decreased β-catenin activity in MKN-45 and AGS cells stably transfected with circ-CTNNB1 or sh-circ-CTNNB1 #3, respectively (Fig. 5B). In vitro binding assay indicated that RNA binding domain [256-315 amino acids (aa)] of FLAG-tagged or GST-tagged

DDX3 protein was crucial for its interaction with circ-CTNNB1 (Fig. 5C). Mutation of amino acids

(274-279, PTRELA) within Ia domain analyzed by RNABindRPlus (24) abolished the interaction of

DDX3 with circ-CTNNB1 (Fig. 5C). Notably, ectopic expression or knockdown of DDX3 did not affect the circ-CTNNB1 levels in MKN-45 and AGS cells (Fig. 5D), while the DDX3 protein levels were not affected by over-expression or knockdown of circ-CTNNB1 (Fig. 5E). These results suggested that circ-CTNNB1 cooperated with DDX3 to facilitate the expression and activity of

β-catenin in cancer cells.

Circ-CTNNB1 facilitates DDX3-mediated transactivation of YY1 and target gene expression

To explore the downstream targets of circ-CTNNB1, we observed the circ-CTNNB1-induced differentially expressed genes by RNA-seq in AGS cells. There were 2230 genes, including 1244 upregulated and 986 downregulated ones, that showed differential expression (fold change >1.5,

P<0.05) upon circ-CTNNB1 over-expression (Fig. 6A). analysis indicated that these circ-CTNNB1 target genes were significantly associated with Wnt signaling pathway, including

WNT1, WNT3, AXIN2, FZD10, and BMP4 (Fig. 6A). Comprehensive analysis using ChIP-X program (25) and DDX3-interacting proteins derived from BioGRID database (26) revealed three

17

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

potential transcription factors regulating these target genes, including ERG (27), SP1 (28), and YY1

(29) (Fig. 6B and Supplementary Table S5). Knockdown of YY1, but not of ERG or SP1, attenuated the increased β-catenin activity induced by stable circ-CTNNB1 over-expression in MKN-45 and

AGS cells (Fig. 6C, Supplementary Fig. S3D and Fig. S4A). Co-IP and BiFC (30) assays revealed endogenous interaction between DDX3 and YY1 in cancer cells, which was increased by ectopic expression of circ-CTNNB1 (Fig. 6D and Fig. 6E). In addition, treatment with RK-33 attenuated the increased interaction between DDX3 and YY1 in MKN-45 cells stably transfected with circ-CTNNB1 (Fig. 6D). The zinc finger (ZnF) domain (228-414 aa) or recruitment of polycomb

(REPO) domain (205–226 aa), but not N-terminus (1-200 aa), of FLAG-tagged YY1 was essential for its interaction with DDX3 protein (Supplementary Fig. S4B). Meanwhile, the V domain (425-456 aa) of Myc-tagged DDX3 was essential for its interaction with YY1 (Supplementary Fig. S4B).

In dual-luciferase assay with a reporter containing four canonical YY1 binding sites, ectopic expression or knockdown of circ-CTNNB1 prevented the decrease and increase of YY1 transactivation induced by silencing or over-expression of DDX3 in cancer cells, respectively (Fig.

6F). Ectopic expression of circ-CTNNB1 facilitated the interaction of YY1 with p300, and repressed its binding to EZH2 or HDAC1 in MKN-45 cells, which was rescued by knockdown of DDX3

(Supplementary Fig. S4C). Stable over-expression of circ-CTNNB1 led to increased binding of YY1 and p300 to the promoters of target genes WNT1 and WNT3, accompanied by decreased recruitment of EZH2 or HDAC1, which were prevented by silencing of DDX3 (Supplementary Fig. S5A).

Consistently, ectopic expression of circ-CTNNB1 increased the promoter activity of WNT1 or WNT3 in MKN-45 and AGS cells, which was abolished by knockdown of DDX3 or YY1 (Fig. 6G). The expression of WNT1, WNT3, AXIN2, FZD10, and BMP4 was increased in cancer cells stably

18

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

transfected with circ-CTNNB1, which was suppressed by knockdown of DDX3 or YY1 (Fig. 6H,

Supplementary Fig. S5B and Fig. S5C). Notably, higher levels of DDX3 (P<1.0×10-4) or YY1

(P<1.0×10-4) were observed in 81 primary gastric cancer tissues, than their paired adjacent normal tissues, especially in specimens with metastasis (P=0.023 and P<1.0×10-4, Supplementary Fig. S6A).

Kaplan–Meier survival analysis of this cohort and well-defined gastric cancer cases derived from

Kaplan-Meier Plotter (http://kmplot.com/analysis) revealed that patients with higher expression of

DDX3 (P<1.0×10-4 and P=8.0×10-6), YY1 (P<1.0×10-4 and P=1.0×10-4), WNT1 (P=1.0×10-11), WNT3

(P=2.5×10-11), AXIN2 (P=3.0×10-4), FZD10 (P=7.2×10-5), or BMP4 (P=1.7×10-4) had lower overall survival probability (Supplementary Fig. S6B and Fig. S6C). These results showed that circ-CTNNB1 facilitated DDX3-mediated transactivation of YY1 and target gene expression in cancer cells.

Therapeutic effects of lentivirus-mediated circ-CTNNB1 knockdown

We further explored the therapeutic efficiencies of circ-CTNNB1 knockdown on tumor growth and survival of athymic nude mice bearing xenografts. Knockdown of circ-CTNNB1 significantly reduced the growth, tumor weight, Ki-67 proliferation index, CD31-positive intratmoral microvessels, and intratumoral expression of CTNNB1 and downstream genes of AGS cells-formed subcutaneous xenografts (Supplementary Fig. S7A, Fig. S7B, Fig. S7C, and Fig. S7D). In addition, administration of lentivirus-mediated sh-circ-CTNNB1 #3 significantly decreased the lung metastatic colonies and prolonged the survival time of nude mice treated with tail vein injection of

AGS cells (Supplementary Fig. S7E). These results suggested that lentivirus-mediated knockdown of circ-CTNNB1 suppressed the tumorigenesis and aggressiveness.

Therapeutic peptide blocking the circ-CTNNB1-DDX3 interaction

19

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Based on the importance of RNA binding domain Ia (274-279 aa) of DDX3 in interacting with circ-CTNNB1, we further designed a cell-penetrating peptide, named as DDX3 inhibitory peptide with 13 amino acids in length (DIP-13), that might potentially block the circ-CTNNB1-DDX3 interaction. Biotin-labeled peptide pull-down assay revealed the binding of DIP-13 to circ-CTNNB1

(Supplementary Fig. S8A). Administration of DIP-13 resulted in its obvious aggregation within the nucleus (Fig. 7A), abolished the circ-CTNNB1-DDX3 interaction (Fig. 7B), and prevented the transactivation of YY1 induced by stable over-expression of circ-CTNNB1 in cancer cells

(Supplementary Fig. S8B). Administration of DIP-13 suppressed the viability of MKN-45 and AGS cells in a dose- and time-dependent manner (Supplementary Fig. S8C). In soft agar and matrigel invasion assays, the growth and invasion of viable cancer cells were significantly inhibited by

DIP-13 treatment (Fig. 7C, Fig. 7D, Supplementary Fig. S8D and Fig. S8E). Intratumoral administration of DIP-13 significantly reduced the volume, tumor weight, Ki-67 proliferation index, and CD31-positive microvessels, and downstream gene expression of subcutaneous xenografts formed by injection of AGS cells (Fig. 7E, Fig. 7F, and Supplementary Fig. S8F). In experimental metastasis assay, administration of DIP-13 via the tail vein significantly reduced the lung metastatic colonies and prolonged the survival time of athymic nude mice treated with tail vein injection of

AGS cells (Fig. 7G). Collectively, these results indicated that inhibitory peptide blocking the circ-CTNNB1-DDX3 interaction suppressed the growth and aggressiveness of cancer cells.

Discussion

Recent studies show that circRNAs exert functions in multiply ways, such as binding to proteins or possessing protein-coding abilities (6,12,31). For example, circ-Foxo3 induces cell apoptosis through interacting with mouse double minute 2 homolog and increasing p53 stability (32).

20

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

In this study, we discover that cis-acting circ-CTNNB1 is upregulated in cancer tissues, and associated with unfavorable outcome of patients. The action mode of circ-CTNNB1 is different from previously reported intronic or exon–intron circRNAs (11,12). Circ-CTNNB1 does no affect the transcription of CTNNB1 in cancer cells. Instead, circ-CTNNB1 is able to activate Wnt signaling and increase the levels of β-catenin protein. Mechanistically, circ-CTNNB1 interacts with DDX3 to increase the transactivation of YY1, which regulates the expression of genes associated with

β-catenin activation and cancer progression (Fig. 7H), such as WNT1 (33), WNT3 (34), AXIN2 (35),

FZD10 (36), and BMP4 (37). Our evidence demonstrates that circ-CTNNB1 possesses oncogenic properties to drive cancer progression, representing a promising therapeutic target for cancers.

DDX3, a member of the DEAD-box RNA helicase family, plays multiple roles in RNA metabolism, such as transcription, pre-mRNA splicing, and mRNA export (38). High DDX3 expression is associated with tumor progression and poor prognosis in breast cancer and gallbladder cancer (38). In glioblastoma, DDX3 facilitates the motility of cancer cells by increasing Snail expression (39). Meanwhile, DDX3 promotes the migration and invasion of colorectal cancer cells via Snail/E-cadherin pathway (40). On the contrary, downregulation of DDX3 is associated with poor prognosis in hepatocellular carcinoma (41) and lung cancer (42). These findings indicate that DDX3 exerts oncogenic or tumor suppressive functions in a context-dependent manner. In the current study, we demonstrated that DDX3 was associated with poor outcome of gastric cancer patients. Gain- and loss-of-function studies indicated that DDX3 facilitated the -catenin activity of cancer cells, suggesting the oncogenic roles of DDX3 in cancer progression.

As a transcriptional regulator, DDX3 has been reported to regulate the promoter activity of p21 (43) and E-cadherin (44). DDX3 also acts as a co-factor for transcription factor SP1 to

21

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

synergistically activate the transcription of oncogenic KRAS (45). In addition, DDX3 promotes the stabilization and nuclear accumulation of p53 (46). Recent studies reveal that DDX3 is able to activate Wnt signaling through binding with casein kinase 1 to stimulate its kinase activity (47), or interacting with 5'-untranslated region of Rac family small GTPase 1 to facilitate its translation (48).

However, the roles of noncoding RNA in regulating the DDX3 activity in cancer progression remain to be determined. In this study, we demonstrate that DDX3 functions as a co-factor to facilitate the transactivation of YY1. As a transcription factor, YY1 is upregulated in many types of cancers, and exerts its oncogenic effects through initiating, activating, or repressing the transcription of target genes (49). Notably, we found that the electrophoresis speed of N-terminus (1-200 amino acids) of

YY1 protein was slower than that of C-terminus (201-414 amino acids), which was line with previous findings that special structure affects its electrophoresis speed (50). Our results indicate that circ-CTNNB1 binds the Ia domain of DDX3 protein to increase its interaction with YY1, resulting in transactivation of YY1. We believe that the binding of circ-CTNNB1 might result in altered DDX3 protein structure that facilitates its interaction with YY1, with the underlying mechanisms warranting further investigation. Since inhibition of circ-CTNNB1-DDX3 interaction abolishes the aggressive behavior of cancer cells, our findings indicate that the tumor promoting functions of circ-CTNNB1 are mediated, at least in part, through interacting with DDX3.

In summary, for the first time, we have demonstrated that circ-CTNNB1 is upregulated in cancer tissues, and associated with poor outcome of gastric cancer patients. Circ-CTNNB1 cooperates with DDX3 to promote the tumorigenesis and aggressiveness of cancer cells through facilitating transactivation of YY1 and transcriptional alteration of target genes associated with

β-catenin activation and cancer progression. Administration of lentivirus-mediated shRNA targeting

22

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

circ-CTNNB1 or a cell-penetrating inhibitory peptide blocking the circ-CTNNB1-DDX3 interaction suppresses the growth and aggressiveness of cancer cells. This study extends our knowledge about the regulation of Wnt/β-catenin signaling by cis-acting circRNA, and suggests that circ-CTNNB1 and DDX3 may be potential therapeutic targets for human cancers. Meanwhile, the roles of other trans-acting circRNA in β-catenin activation and cancer progression warrant further investigation.

Acknowledgements

We appreciate Drs. Christof Niehrs and Makoto Miyagishi for providing vectors. This work was supported by National Natural Science Foundation of China [81272779, 81472363, 81402301,

81672500, 81772967, 81874085 (Q. Tong), 81372667, 81572423, 81874066 (L. Zheng), 81402408,

81773094 (H. Mei), 81802925 (D. Li)].

References

1. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell

Dev Biol 2004, 20: 781-810.

2. van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, et al. The

beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells.

Cell 2002, 111: 241-250.

3. Mir R, Pradhan SJ, Patil P, Mulherkar R, Galande S. Wnt/β-catenin signaling regulated

SATB1 promotes colorectal cancer tumorigenesis and progression. Oncogene 2015, 35:

1679-1691.

4. Zhang K, Guo Y, Wang X, Zhao H, Ji Z, Cheng C, et al. WNT/β-catenin directs self-renewal

symmetric cell division of hTERThigh prostate cancer stem cells. Cancer Res 2017, 77:

2534-2547.

23

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

5. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nature Biotechnol

2014, 32: 453-461.

6. Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, et al. Circular RNA is enriched and stable in

exosomes: a promising biomarker for cancer diagnosis. Cell Res 2015, 25: 981-984.

7. Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a

large class of animal RNAs with regulatory potency. Nature 2013, 495: 333-338.

8. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, et al. Natural

RNA circles function as efficient microRNA sponges. Nature 2013, 495: 384-388.

9. Ashwal-Fluss R, Meyer M, Pamudurti Nagarjuna R, Ivanov A, Bartok O, Hanan M, et al.

circRNA biogenesis competes with pre-mRNA splicing. Mol Cell 2014, 56: 55-66.

10. Abdelmohsen K, Panda AC, Munk R, Grammatikakis I, Dudekula DB, De S, et al.

Identification of HuR target circular RNAs uncovers suppression of PABPN1 translation by

circPABPN1. RNA Biol 2017, 14: 361-369.

11. Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, et al. Circular intronic long

noncoding RNAs. Mol Cell 2013, 51: 792-806.

12. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate

transcription in the nucleus. Nat Struct Mol Biol 2015, 22: 256-264.

13. Huang G, Li S, Yang N, Zou Y, Zheng D, Xiao T. Recent progress in circular RNAs in human

cancers. Cancer Lett 2017, 404: 8-18.

14. Cadigan KM, Waterman ML. TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harb

Perspect Biol 2012, 4: a007906.

15. Li D, Wang X, Mei H, Fang E, Ye L, Song H, et al. Long noncoding RNA pancEts-1

24

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

promotes neuroblastoma progression through hnRNPK-mediated β-catenin stabilization.

Cancer Res 2018, 78: 1169-1183.

16. Zhao X, Li D, Pu J, Mei H, Yang D, Xiang X, et al. CTCF cooperates with noncoding RNA

MYCNOS to promote neuroblastoma progression through facilitating MYCN expression.

Oncogene 2016, 35: 3565-3576.

17. Petkovic S, Muller S. RNA circularization strategies in vivo and in vitro. Nucleic Acids Res

2015, 43: 2454-2465.

18. Chen C, Sarnow P. Initiation of protein synthesis by the eukaryotic translational apparatus on

circular RNAs. Science 1995, 268: 415-417.

19. Glažar P, Papavasileiou P, Rajewsky N. circBase: a database for circular RNAs. RNA 2014,

20: 1666-1670.

20. Dudekula DB, Panda AC, Grammatikakis I, De S, Abdelmohsen K, Gorospe M.

CircInteractome: A web tool for exploring circular RNAs and their interacting proteins and

microRNAs. RNA Biol 2016, 13: 34-42.

21. Tang X, Zheng D, Hu P, Zeng Z, Li M, Tucker L, et al. Glycogen synthase kinase 3 beta

inhibits microRNA-183-96-182 cluster via the β-Catenin/TCF/LEF-1 pathway in gastric

cancer cells. Nucleic Acids Res 2014, 42: 2988-2998.

22. Afifi MM, Austin LA, Mackey MA, El-Sayed MA. XAV939: From a small inhibitor to a

potent drug bioconjugate when delivered by gold nanoparticles. Bioconjug Chem 2014, 25:

207-215.

23. Xie M, Vesuna F, Tantravedi S, Bol GM, Heerma van Voss MR, Nugent K, et al. RK-33

radiosensitizes prostate cancer cells by blocking the RNA helicase DDX3. Cancer Res 2016,

25

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

76: 6340-6350.

24. Walia RR, Xue LC, Wilkins K, El-Manzalawy Y, Dobbs D, Honavar V. RNABindRPlus: a

predictor that combines machine learning and -based methods to improve

the reliability of predicted RNA-binding residues in proteins. PLoS One 2014, 9: e97725.

25. Lachmann A, Xu H, Krishnan J, Berger SI, Mazloom AR, Ma'ayan A. ChEA: transcription

factor regulation inferred from integrating genome-wide ChIP-X experiments.

Bioinformatics 2010, 26: 2438-2444.

26. Chatr-aryamontri A, Oughtred R, Boucher L, Rust J, Chang C, Kolas NK, et al. The

BioGRID interaction database: 2017 update. Nucleic Acids Res 2017, 45: D369-D379.

27. Wu L, Zhao JC, Kim J, Jin HJ, Wang CY, Yu J. ERG is a critical regulator of Wnt/LEF1

signaling in prostate cancer. Cancer Res 2013, 73: 6068-6079.

28. Kwon YJ, Baek HS, Ye DJ, Shin S, Kim D, Chun YJ. CYP1B1 enhances cell proliferation

and metastasis through induction of EMT and activation of Wnt/beta-catenin signaling via

Sp1 upregulation. PLoS One 2016, 11: e0151598.

29. Lu C, Thoeni C, Connor A, Kawabe H, Gallinger S, Rotin D. Intestinal knockout of Nedd4

enhances growth of Apc(min) tumors. Oncogene 2016, 35: 5839-5849.

30. Hu CD, Kerppola TK. Simultaneous visualization of multiple protein interactions in living

cells using multicolor fluorescence complementation analysis. Nature Biotechnol 2003, 21:

539-545.

31. Du WW, Yang W, Liu E, Yang Z, Dhaliwal P, Yang BB. Foxo3 circular RNA retards cell

cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res

2016, 44: 2846-2858.

26

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

32. Du WW, Fang L, Yang W, Wu N, Awan FM, Yang Z, et al. Induction of tumor apoptosis

through a circular RNA enhancing Foxo3 activity. Cell Death Differ 2017, 24: 357-370.

33. Zhang JG, Shi Y, Hong DF, Song M, Huang D, Wang CY, et al. MiR-148b suppresses cell

proliferation and invasion in hepatocellular carcinoma by targeting WNT1/β-catenin pathway.

Sci Rep 2015, 5: 8087.

34. Wu Y, Ginther C, Kim J, Mosher N, Chung S, Slamon D, et al. Expression of Wnt3 activates

Wnt/β-catenin pathway and promotes EMT-like phenotype in trastuzumab resistant

HER2-overexpressing breast cancer cells. Mol Cancer Res 2012, 10: 1597-1606.

35. Li Y, Jin K, van Pelt GW, van Dam H, Yu X, Mesker WE, et al. c-Myb enhances breast

cancer invasion and metastasis through the Wnt/β-catenin/axin2 pathway. Cancer Res 2016,

76: 3364-3375.

36. Gong C, Qu S, Lv XB, Liu B, Tan W, Nie Y, et al. BRMS1L suppresses breast cancer

metastasis by inducing epigenetic silence of FZD10. Nat Commun 2014, 5: 5406.

37. Kim JS, Crooks H, Dracheva T, Nishanian TG, Singh B, Jen J, et al. Oncogenic beta-catenin

is required for bone morphogenetic protein 4 expression in human cancer cells. Cancer Res

2002, 62: 2744-2748.

38. Bol GM, Xie M, Raman V. DDX3, a potential target for cancer treatment. Mol Cancer 2015,

14: 188.

39. Sun M, Song L, Zhou T, Gillespie GY, Jope RS. The role of DDX3 in regulating snail.

Biochim Biophys Acta 2011, 1813: 438-47.

40. Su CY, Lin TC, Lin YF, Chen MH, Lee CH, Wang HY, et al. DDX3 as a strongest prognosis

marker and its downregulation promotes metastasis in colorectal cancer. Oncotarget 2015, 6:

27

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

18602-18612.

41. Zhang H, Xing Z, Mani SK, Bancel B, Durantel D, Zoulim F, et al. RNA helicase DEAD box

protein 5 regulates Polycomb repressive complex 2/Hox transcript antisense intergenic RNA

function in hepatitis B virus infection and hepatocarcinogenesis. Hepatology 2016, 64:

1033-1048.

42. Wu DW, Liu WS, Wang J, Chen CY, Cheng YW, Lee H. Reduced p21(WAF1/CIP1) via

alteration of p53-DDX3 pathway is associated with poor relapse-free survival in early-stage

human papillomavirus-associated lung cancer. Clin Cancer Res 2011, 17: 1895-1905.

43. Chao CH, Chen CM, Cheng PL, Shih JW, Tsou AP, Lee YH. DDX3, a DEAD box RNA

helicase with tumor growth-suppressive property and transcriptional regulation activity of the

p21waf1/cip1 promoter, is a candidate tumor suppressor. Cancer Res 2006, 66: 6579-6588.

44. Botlagunta M, Vesuna F, Mironchik Y, Raman A, Lisok A, Winnard P, et al. Oncogenic role

of DDX3 in breast cancer biogenesis. Oncogene 2008, 27: 3912-3922.

45. Wu DW, Lin PL, Cheng YW, Huang CC, Wang L, Lee H. DDX3 enhances oncogenic

KRASinduced tumor invasion in colorectal cancer via the betacatenin/ZEB1 axis. Oncotarget

2016, 7: 22687-22699.

46. Sun M, Zhou T, Jonasch E, Jope RS. DDX3 regulates DNA damage-induced apoptosis and

p53 stabilization. Biochim Biophys Acta 2013, 1833: 1489-1497.

47. Cruciat CM, Dolde C, de Groot RE, Ohkawara B, Reinhard C, Korswagen HC, et al. RNA

helicase DDX3 is a regulatory subunit of casein kinase 1 in Wnt-beta-catenin signaling.

Science 2013, 339: 1436-1441.

48. Chen HH, Yu HI, Cho WC, Tarn WY. DDX3 modulates cell adhesion and motility and cancer

28

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cell metastasis via Rac1-mediated signaling pathway. Oncogene 2015, 34: 2790-2800.

49. Shi J, Hao A, Zhang Q, Sui G. The role of YY1 in oncogenesis and its potential as a drug

target in cancer therapies. Curr Cancer Drug Targets 2015, 15: 145-157.

50. Lorente M, Pérez C, Sánchez C, Donohoe M, Shi Y, Vidal M. Homeotic transformations of

the axial skeleton of YY1 mutant mice and genetic interaction with the polycomb group gene

Ring1/Ring1A. Mech Dev 2006, 123: 312-320.

Figure Legends

Figure 1 Identification of circ-CTNNB1 as a cis-acting circRNA upregulated in cancer tissues and cells. A, flowchart delineating the identification of circ-CTNNB1 as a cis-acting circRNA regulating β-catenin activity. B, real-time qRT-PCR assay showing the relative levels of circ-CTNNB1 (normalized to β-actin) in the peri-tumor and tumor tissues of gastric cancer (n=81), prostate cancer (n=21), and colon cancer (n=15). C, TOP/FOP flash assay indicating the β-catenin activity in AGS cells transfected with sramble shRNA (sh-Scb) or shRNA specific for circRNAs, and in those treated with solvent control (DMSO) or XAV939 (5.0 μmol/L) for 6 hrs. D, schematic illustration showing the genomic location of circ-CTNNB1 derived from its parent gene CTNNB1. E,

RT-PCR or PCR assay (left panel) indicating the detection of circ-CTNNB1 using divergent and convergent primers from cDNA or genomic DNA (gDNA) of cancer cells, with further validation by

Sanger sequencing (right panel). F, Northern blot using a junction-specific probe showing the endogenous existence of circ-CTNNB1 in HEK293T cells and cancer cell lines (left panel), and that of AGS cells transfected by circ-CTNNB1 and treated with RNase R (3 U/mg, right panel). G, real-time qRT-PCR assay revealing the relative levels of circ-CTNNB1 (normalized to β-actin) in

HEK293T and cancer cell lines (mean ± SEM, n=4). H, real-time qRT-PCR assay showing the levels

29

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

of circ-CTNNB1 (normalized to -actin) in gastric cancer tissues with differential status of metastasis.

I, Kaplan-Meier curves indicating overall survival of 81 gastric cancer patients with low or high expression of circ-CTNNB1 (cutoff value=12.14). * P<0.01 vs. DMSO, sh-Scb, or HEK293T.

Student’s t test compared gene expression levels in B, C, G and H. Log-rank test for survival comparison in I.

Figure 2 Circ-CTNNB1 promotes the growth and aggressiveness of cancer cells in vitro and in vivo. A, MTT colorimetric assay showing the viability of MKN-45, AGS, PC-3 and HCT-116 cells stably transfected with empty vector (mock), circ-CTNNB1, scramble shRNA (sh-Scb), or sh-circ-CTNNB1. B and C, representative images (left panel) and quantification (right panel) of soft agar (B) and matrigel invasion (C) assays indicating the anchorage-independent growth and invasion of MKN-45 and AGS cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1.

D, representative images (upper and middle panels), in vivo growth curve (lower left panel) and weight at the end points (lower right panel) of xenografts formed by subcutaneous injection of

MKN-45 cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1 #3 into the dorsal flanks of nude mice (n=5 for each group). E, representative images (upper panel) and quantification (lower panel) of immunohistochemical staining showing the intratumoral expression of Ki-67 and CD31 within xenografts (n=5 for each group). Scale bars: 50 μm. F and G, representative images (F), HE staining (G, upper panel), and quantification (G, lower left panel) of lung metastatic colonization and Kaplan-Meier curves (G, lower right panel) of nude mice treated with tail vein injection of MKN-45 cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1 #3 (n=5 for each group). Student’s t test and analysis of variance compared the difference in A-E and G. Log-rank test for survival comparison in G. *P<0.01 vs. mock or sh-Scb.

30

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3 Circ-CTNNB1 increases the expression of β-catenin in cancer cells. A, western blot assay showing the protein levels of β-catenin in MKN-45, AGS, PC-3, and HCT-116 cells stably transfected with empty vector (mock), circ-CTNNB1, scramble shRNA (sh-Scb), or sh-circ-CTNNB1. B and C, dual-luciferase (B) and real-time qRT-PCR (C) assays indicating the promoter activity and transcript levels of β-catenin in MKN-45 and AGS cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1 (mean ± SEM, n=4). D and E, western blot

(D) and immunofluorescence (E) assays showing the cytoplasmic and nuclear accumulation of

β-catenin in MKN-45 and AGS cells stably transfected with mock or circ-CTNNB1. Scale bar: 10

m. F, dual-luciferase assay indicating the activity of β-catenin in cancer cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1 #3, and those treated with solvent control

(DMSO), XAV-939 (5.0 μmol/L), PBS, or WNT3a (50 ng/ml). G, western blot showing the expression of phosphorylated β-catenin (Ser33/Ser37), β-catenin, c-myc, and cyclin D1 in cancer cells stably transfected with mock or circ-CTNNB1. H, western blot indicating the expression of phosphorylated β-catenin (Ser33/Ser37) and β-catenin in MKN-45 cells stably transfected with sh-Scb or sh-circ-CTNNB1 #3, and those treated with WNT3a (50 ng/ml), LiCl (30 mM), or proteasome inhibitor (MG132, 12.5 μmol/L). Student’s t test and analysis of variance compared the difference in B, C, and F. *P<0.01 vs. mock+DMSO or sh-Scb+PBS.

Figure 4 Circ-CTNNB1 is predominantly localized in the nucleus and interacts with DDX3.

A, RNA-FISH assay showing the nuclear localization of circ-CTNNB1 in MKN-45 cells incubated with a 58-bp junction antisense probe (green) and RNase R (3 U/mg), with the nuclei staining by

DAPI (blue). Sense probe with RNase R (3 U/mg) treatment was used as a negative control. The U1

31

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and GAPDH were applied as positive controls. Scale bar: 10 m. B, real-time qRT-PCR indicating the distribution of GAPDH, U1, and circ-CTNNB1 in the cytoplasmic and nuclear fractions (mean ±

SEM, n=4). C, schematic diagram showing the process to generate biotin-labeled circ-CTNNB1 probe by ligation of in vitro transcribed linear transcript (left panel), with validation by denaturing

PAGE, SYBR Green II staining, and RNase R (3 U/mg) treatment (right panel). D, Coomassie bright blue staining (upper left panel), Venn diagram (upper right panel), and mass spectrometry (MS, lower panel) assays revealing the proteins pulled down by biotin-labeled circ-CTNNB1 from the lysates of MKN-45 and AGS cells, and their over-lapping analysis with established RBP. E, western blot assay showing the proteins pulled down by biotin-labeled random (R), linear (L) or circular (C)

RNA probes from the lysates of MKN-45 cells. F, dual RNA-FISH and immunofluorescence staining assay indicating the co-localization of circ-CTNNB1 (red) and DDX3 (green) in MKN-45 cells stably transfected with empty vector (mock) or circ-CTNNB1, with nuclei staining with DAPI

(blue). Scale bar: 10 m. G, RIP (upper panel) and real-time qRT-PCR (lower panel) assays using an antibody specific for DDX3 showing the interaction between circ-CTNNB1 and DDX3 in MKN-45 cells stably transfected with mock or circ-CTNNB1. H, RNA EMSA determining the interaction between endogenous (within nuclear extracts of MKN-45 cells) or recombinant DDX3 protein and biotin-labeled RNA probe for circ-CTNNB1 (arrowheads), with or without treatment using DDX3 antibody or competition using an excess of unlabeled homologous circRNA probe. Student’s t test compared the difference in G. *P<0.01 vs. IgG. Δ P<0.01 vs. mock.

Figure 5 Circ-CTNNB1 cooperates with DDX3 to facilitate the expression and activity of

β-catenin. A, western blot assay showing the expression of β-catenin in MKN-45 cells stably transfected with empty vector (mock) or circ-CTNNB1, and those co-transfected with sh-DDX3 #2

32

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

or treated with DDX3 inhibitor (RK-33, 5.0 μmol/L). B, TOP/FOP flash assay indicating the

β-catenin activity in MKN-45 and AGS cells stably transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1 #3, and those treated with RK-33 (5.0 μmol/L) or co-transfected with DDX3. C, in vitro binding assay depicting the recovered circ-CTNNB1 levels detected by RT-PCR (upper panels) after incubation with full-length, truncation, or mutation (Mut) forms of FLAG-tagged or

GST-tagged recombinant DDX3 protein (lower left panel) validated by western blot (middle panels), with mutation of amino acids (PTRELA) within Ia domain as indicated (lower right panel). D, real-time qRT-RCR assay indicating the levels of circ-CTNNB1 in MKN-45 and AGS cells transfected with mock, DDX3, sh-Scb, or sh-DDX3. E, western blot assay showing the expression of

DDX3 in MKN-45 and AGS cells transfected with mock, circ-CTNNB1, sh-Scb, or sh-circ-CTNNB1

#3. Student’s t test for analyzing the difference in B and D. *P<0.01 vs. mock+DMSO or sh-Scb+mock.

Figure 6 Circ-CTNNB1 facilitates DDX3-mediated transactivation of YY1 and target gene expression. A, Volcano plots (left upper panel) showing the differentially expressed genes (fold change>1.5, P<0.05) in AGS cells stably transfected with empty vector (mock) or circ-CTNNB1.

Gene ontology (GO) analysis with PANTHER classification system (https://www.pantherdb.org) indicating the involved pathway of target genes (left lower panel), and heatmap showing the differentially expressed genes of Wnt/β-catenin signaling pathway (right panel). B, Venn diagram indicating the discovery of transcriptional regulators essential for altered genes using

DDX3-interacting proteins from BioGRID database and transcription factor (TF) list from ChIP-X program. C, TOP/FOP flash assay showing the β-catenin activity in cancer cells stably transfected with mock or circ-CTNNB1, and those co-transfected with sh-Scb or sh-YY1. D, co-IP and western

33

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

blot assays indicating the interaction between DDX3 and YY1 in MKN-45 cells stably transfected with mock or circ-CTNNB1, and those treated with DMSO or RK-33 (5.0 μmol/L). E, BiFC assay showing the interaction between DDX3 and YY1 in MKN-45 cells stably transfected with mock or circ-CTNNB1. F, dual-luciferase assay revealing the transactivation of YY1 in MKN-45 and AGS cells stably transfected with mock, circ-CTNNB1, or DDX3, and those co-transfected with sh-Scb, sh-DDX3, or sh-circ-CTNNB1. G, dual-luciferase assay indicating the promoter activity of WNT1 and WNT3 in MKN-45 and AGS cells stably transfected with mock or circ-CTNNB1, and those co-transfected with sh-Scb, sh-DDX3 #2, or sh-YY1 #2. H, western blot assay showing the expression of DDX3, YY1, β-catenin, WNT1, WNT3, AXIN2, FZD10, and BMP4 in MKN-45 cells stably transfected with mock or circ-CTNNB1, and those co-transfected with sh-Scb, sh-DDX3 #2, or sh-YY1 #2. Student’s t test for analyzing the difference in C, F, and G. *P<0.01 vs. mock+sh-Scb.

Figure 7 Therapeutic peptide blocking the circ-CTNNB1-DDX3 interaction. A, confocal images showing the distribution of FITC-labeled control (Ctrl) or inhibitory (DIP-13) peptide (30

μmol/L) within MKN-45 cells, with nuclei and cellular membrane stained with DAPI (blue) or Dil

(red), respectively. Scale bars: 10 m. B, western blot indicating the DDX3 protein pulled down by biotin-labeled random (R), linear (L) or circular (C) circ-CTNNB1 probe from the lysates of

MKN-45 cells treated with Ctrl peptide or DIP-13 (30 μmol/L). C and D, representative images (left panel) and quantification (right panel) of soft agar (C) and matrigel invasion (D) assays showing the anchorage-independent growth and invasion capability of viable MKN-45 cells treated with Ctrl peptide or DIP-13 (30 μmol/L) for 24 hrs. E, representative images (left panel), in vivo growth curve

(right upper panel), and tumor weight at the end points (right lower panel) of AGS cells-formed

34

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

subcutaneous xenografts in nude mice (n=5 for each group) that were treated with intratumoral injection of Ctrl peptide or DIP-13 (3 mg/kg) as indicated (left panel). F, representative images

(upper panel) and quantification (lower panel) of immunohistochemical staining indicating the expression of Ki-67 and CD31 in xenografts treated with intratumoral injection of Ctrl peptide or

DIP-13 (3 mg/kg). G, representative images (upper panel), HE staining (lower left panel) and quantification (lower middle right panel) of lung metastatic colonization, and Kaplan-Meier curves

(lower right panel) of nude mice (n=5 for each group) treated with tail vein injection of AGS cells and subsequent administration of Ctrl peptide or DIP-13 (3 mg/kg) as indicated. H, the mechanisms underlying circ-CTNNB1-promoted cancer progression: as a cis-acting circRNA, circ-CTNNB1 binds DDX3 to facilitate its interaction with YY1, resulting in transactivation of YY1 and transcriptional alteration of target genes associated with β-catenin activation and cancer progression.

An inhibitory peptide DIP-13 is able to block the circ-CTNNB1-DDX3 interaction and suppress cancer progression. Student’s t test and analysis of variance analyzed the difference in C-G.

Log-rank test for survival comparison in G. *P<0.01 vs. Ctrl.

35

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1 A B Gastric cancer Prostate cancer Colon cancer CTNNB1/TCF7L2-derived circRNA 15 10 15 in circBase (n=38) P<0.0001 P=0.0049 P=0.013

Read scores≥2 (n=15) 10 10 5 Detected in AGS cells (n=7) 5 5

Differential expression between Relative circ-CTNNB1 qRT-PCR) (by levels Relative circ-CTNNB1 qRT-PCR) (by levels Relative circ-CTNNB1 qRT-PCR) (by levels tumor and peri-tumor tissues (n=3) 0 0 0 peri-tumor tumor peri-tumor tumor peri-tumor tumor TOP/FOP flash reporter assay (n=1) (n=81) (n=81) (n=21) (n=21) (n=15) (n=15)

circ-CTNNB1 C D AGS 2.0 Chr 3 Exon89 10 11 12 13 CTNNB1 … 1.5

1.0

-catenin activity -catenin GTAT Linear AAAG  * * 0.5 * 378 nt circ-CTNNB1

(TOP/FOP flash ratio) flash (TOP/FOP 0.0

Relative b 1 SO # #1 #2 #2 #3 M V939 -Sc 0 1 1 D A B B sh 4030 4030 N N X 2005 0 0 N N 0 T T _00 -C c c G irc_0 rc-C _cir 60 a_c a_circ_0020050a #2h-cir h-ci s s s s * -h 50 sh sh-h sh-hsa_circ_00sh-hs E 40 30 * 30 * * * * MKN-45 AGS 20 * cDNA gDNA cDNA gDNA * bp 10 * * levels (by qRT-PCR) (by levels 500 Relativecirc-CTNNB1 0 250 circ-CTNNB1 T S 3 )-C 100 eLa F-7-116oVo 293 AGH PC- C L (2 SY5Y M T - EK MKN-45 HepG2BE 250 H HC SH -actin 100 H AATAAAAGGTATATAA P<0.0001 24

F 16

T Y 6 8 3 5 5 1

9 4 qRT-PCR) (by 2 - Y -1 RNase R a S o 3 ++ K N S L - T V - E K G e H C o C circ-CTNNB1 H M A H S H L P + + 0 Relative circ-CTNNB1 levels circ-CTNNB1 Relative 28 S 28 S No (n=22) Yes (n=59) Metastasis 18 S 18 S

circ-CTNNB1 I 1.0 Log-rank P<0.0001 378 nt 378 nt 0.8 Low n=41 5 S 5 S 0.6 0.4 28 S 28 S High n=40

Percent survivalPercent 0.2 18 S 18 S 0.0 0 20 40 60 80 Time (month)

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2

A B sh-circ-CTNNB1 800 2.5 Mock Mockcirc-CTNNB1 sh-Scb #2 #3 Mock circ-CTNNB1 circ-CTNNB1 sh-Scb 2.0 sh-Scb sh-circ-CTNNB1 #2 600 sh-circ-CTNNB1 #2 sh-circ-CTNNB1 #3 sh-circ-CTNNB1 #3 1.5 * * * * 400 * * * * MKN-45 * * * 1.0 *

Numbers of 200 0.5 colonies per well per colonies Relativecell viability 0.0 AGS 0 MKN-45 AGS MKN-45 AGS

2.5 C 800 Mock sh-circ-CTNNB1 Mock circ-CTNNB1 circ-CTNNB1 2.0 sh-Scb Mock circ-CTNNB1 sh-Scb #2 #3 sh-Scb sh-circ-CTNNB1 #2 600 sh-circ-CTNNB1 #2 sh-circ-CTNNB1 #3 sh-circ-CTNNB1 #3 1.5 * * * * * * 400 * * * * 1.0 MKN-45 * * 200 0.5 cells Relative invadedper field AGS Relative cell viability 0.0 0 PC-3 HCT-116 MKN-45 AGS E D Mock circ-CTNNB1 sh-Scb sh-circ-CTNNB1 #3 Mock circ-CTNNB1 sh-Scb sh-circ-CTNNB1 #3 1.0e +003 Ki-67

0 CD31

Mock 2.5 * 50 * 2.0 40 circ-CTNNB1 1.5 * 30 * 1.0 20 0.5 10 × area Ki-67)by count (by CD31) (by count Score (intensity Score

0.0 Intratumoral vessel 0 sh-Scb B1 N Mock Mock sh-Scb TN sh-Scb C

sh-circ-CTNNB1 #3 circ-CTNNB1 circ- rc-CTNNB1 #3 rc-CTNNB1 #3

sh-ci sh-ci 2.0 Mock 3 Mock circ-CTNNB1 circ-CTNNB1 G )

3 circ-CTNNB1 sh-Scb sh-circ-CTNNB1 #3 sh-Scb sh-Scb Mock 1.5 sh-circ-CTNNB1 #3 sh-circ-CTNNB1 #3 2 * * 1.0 * * 1 0.5 Tumor weight (g) weight Tumor Tumor volume (cm

0.0 0 0 1 2 3 4 week

F 3.0e +003 25 * 100 Mock P=0.0085 20 * 75 15 circ-CTNNB1 50 10 25 5 P<0.0001 Metastasis count

sh-Scb survival Percent 0 0 0 10 20 30 40 50 Mock Days sh-Scb sh-circ-CTNNB1 #3 Mock 0 circ-CTNNB1 circ-CTNNB1 sh-S cb sh-circ-CTNNB1 #3 sh-circ-CTNNB1 #3

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3 A MKN-45 BC circ-CTNNB1 + sh-circ-CTNNB1 #2 #3 2 Mock 2 Mock -catenin 92 kDa circ-CTNNB1 circ-CTNNB1 sh-Scb sh-Scb -actin 42 kDa sh-circ-CTNNB1 #2 sh-circ-CTNNB1 #2 sh-circ-CTNNB1 #3 sh-circ-CTNNB1 #3 Ratio)

AGS 1 1

circ-CTNNB1 + Renilla sh-circ-CTNNB1 #3#2 level qRT-PCR) (by

-catenin 92 kDa (Firefly/ Relative CTNNB1 mRNA -actin 42 kDa Relative luciferase activity 0 0 MKN-45 AGS MKN-45 AGS PC-3 circ-CTNNB1 + sh-circ-CTNNB1 #3#2 D MKN-45 AGS

-catenin 92 kDa cytoplasm nucleus cytoplasm nucleus circ-CTNNB1 -actin 42 kDa + + + + -catenin 92 kDa HCT-116 circ-CTNNB1 + GAPDH 37 kDa sh-circ-CTNNB1 #3#2 Histone H3 15 kDa -catenin 92 kDa

-actin 42 kDa

E G MKN-45 AGS MKN-45 AGS DAPI -catenin Merged DAPI -catenin Merged circ-CTNNB1 + + P--catenin 92 kDa

-catenin 92 kDa Mock Mock

c-Myc 57 kDa

CCND1 34 kDa

-actin 42 kDa circ-CTNNB1 circ-CTNNB1 F H sh-circ-CTNNB1 #3 + ++ WNT3a + + 6 Mock+DMSO 6 sh-Scb+PBS LiCl + + sh-circ-CTNNB1 #3+PBS circ-CTNNB1+DMSO P--catenin 92 kDa 5 Mock+XAV939 5 sh-Scb+WNT3a circ-CTNNB1+XAV939 sh-circ-CTNNB1 #3+WNT3a 4 4 -catenin 92 kDa * 3 * 3 * -actin 42 kDa -catenin activity -catenin * activity-catenin  2  2 sh-circ-CTNNB1 #3 + + 1 (TOP/FOP flash ratio) flash (TOP/FOP 1 MG-132

(TOP/FOP flash ratio) flash (TOP/FOP ++ * * * * Relative 0 Relative 0 -catenin 92 kDa MKN-45 AGS MKN-45 AGS -actin 42 kDa

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 4

A B MKN-45 AGS PC-3 HCT-116 DAPI FITC Merged 1.5 cytoplasm 1.5 cytoplasm 1.5 cytoplasm 1.5 cytoplasm nucleus nucleus nucleus nucleus

1.0 1.0 1.0 1.0 Antisense +RNase R

0.5 0.5 0.5 0.5 Sense +RNase R Relative transcript abundance Relativetranscript Relative transcript abundance Relativetranscript Relative transcript abundance Relativetranscript 0.0 abundance transcript Relative 0.0 0.0 0.0 H 1 1 H 1 U1 U1 U1 B U1 B PD NB PDH N A N G TN GA TNNB1 GAPDH T GAPD -C -C C -CTNN rc- c U1 irc i circ c c cir C RNase R ++ Transcription in vitro T4 RL1 + + T7 promoter circ-CTNNB1 Biotin labeled B B B B B B GAPDH Self-ligation Circular Circular circ-CTNNB1 probe Linear Linear probe T4 RNA ligase 1 B B

D Differential proteins in MS E Input R-probeL-probeC-probe MKN-45 AGS kDa 130 Input R-probe L-probe C-probe 100 70 55 MS analysis DDX3 40 73 kDa 35 RBM14 DDX3 RBM39 25 RBM14 75 kDa

15 DDX3 RBM39 59 kDa

MS DDX3 peptide RBP Histone H3 15 kDa

F DAPI circ-CTNNB1 DDX3 Merged

HVINFDLPSDIEEYVHR Mock

GCHLLVATPGR Circ-CTNNB1 MKN-45 G H Junction Non-Junction Input (20%)DDX3 IgG circ-CTNNB1 + + + bp Nuclear extract + + + + + + + circ-CTNNB1 120 Labeled probe + + + + + + + + + GST-DDX3 + + + Unlabeled probe Labeled probe + + + + -actin 156 + + Anti-DDX3 + Unlabeled probe + MKN-45 10 Mock Super-shift circ-CTNNB1 Shift 8 Shift Δ 6

4 Free probe 2 *Δ levels (by qRT-PCR) (by levels Relativecirc-CTNNB1 * Free probe 0 Input DDX3 IgG

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5 A B

sh-DDX3 #2 ++ 4 Mock+DMSO 4 sh-Scb+Mock circ-CTNNB1 + + circ-CTNNB1+DMSO sh-circ-CTNNB1 #3+Mock Mock+RK-33 sh-Scb+DDX3 -catenin 92 kDa 3 circ-CTNNB1+RK-33 3 sh-circ-CTNNB1 #3+DDX3 * * -actin 42 kDa * * 2 2 -catenin activity-catenin -catenin activity-catenin  RK-33 ++  circ-CTNNB1 + + 1 1 * * * (TOF/FOP flash ratios) -catenin 92 kDa * (TOF/FOP flash ratios) Relative 0 Relative 0 -actin 42 kDa MKN-45 AGS MKN-45 AGS

662) 662) -662) -662) -662) -662) - C 5 0 6 6 -662) -662) 662) ut (1- - t(1 M 662) u 3 (1-662)3 (42 3 (34 3 (31 3 (25 3 M X X X X X X -DD -DD -DDX3 (1-DDX3 (425-DDX3 (340-DDX3 (316-DDX3 (256-662)DDX3 T T- T T T T- Input FLAG-DDFLAG FLAG-DDFLAG-DDFLAG-DDFLAG Input GS GS GS GS GS GS

RT-PCR circ-CTNNB1 RT-PCR circ-CTNNB1

100 kDa 100 kDa 72 kDa WB: FLAG WB:GST 72 kDa 55 kDa 55 kDa 45 kDa

25 kDa 45 kDa

Tag Q I Ia Ib II III IV V VI DDX3 425-662 340-662 316-662 circ-CTNNB1 256-662

1-662 Mut Ia: P T R E L A

DE 2 Mock DDX3 sh-Scb ck o -Scb -circ-CTNNB1 #3 sh-DDX3 #2 M circ-CTNNB1sh sh sh-DDX3 #3 DDX3 73 kDa 1 -actin 42 kDa MKN-45

DDX3 73 kDa levels qRT-PCR) (by Relative circ-CTNNB1 AGS -actin 42 kDa 0 MKN-45 AGS

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 6

A 10 ck o 9 Down Up M circ-CTNNB1 B DDX3 binding TF gene list ACTA2 8 986 1244 ACTC1 proteins ADSSL1 7 APC2 AXIN2 6 BMP4 CDH15 5 CDH17 (p-value) 4 CDH5 151 11 143

10 CDHR2 CELSR3 3 CREBBP 2 CSNK1D -Log CTNNA3 3 1 FZD10 GNG7 0 ITPR1 -4 -3 -2 -1 0 1 2 3 4 MYCL -catenin MYCN activator Log (fold change) MYH1 2 MYH7B MYH8 GO analysis: Panther pathways NKD2 ERG SP1 YY1 PCDH12 Wnt signaling PCDH7 Angiogenesis PCDH9 PCDHB10 C Alzheimer disease-presenilin PCDHB14 6 Mock+sh-Scb PCDHGA6 Nicotinic acetylcholine receptor PRKCB Mock+sh-YY1 #1 Ionotropic glutamate receptor PRKCD PRKCG Mock+sh-YY1 #2 Metabotropic glutamate receptor III TLE1 TLE2 circ-CTNNB1+sh-Scb Muscarinic acetylcholine receptor WNT1 4 WNT10B circ-CTNNB1+sh-YY1 #1 Notch signaling WNT2B WNT3 circ-CTNNB1+sh-YY1 #2 Metabotropic glutamate receptor I WNT4 WNT9A Beta1 adrenergic receptor activity-catenin *

 * 2 0 10 20 30 40 50 -2 -1 0 12 Log 2 Target gene enrichment analysis * (TOF/FOP flash ratios) * * *

DDX3 Relative D 0 Input DMSO RK-33 IgG MKN-45 AGS circ-CTNNB1 + + + + E IB: YY1 68 kDa Mock circ-CTNNB1 Mock circ-CTNNB1 p-VN173-DDX3 ++ + IB: DDX3 73 kDa p-VC155-YY1 +++ YY1 Input DMSO RK-33 IgG circ-CTNNB1 + + + + Venus IB: DDX3 73 kDa

IB: YY1 68 kDa Merged F 6 Mock+sh-Scb 6 sh-Scb+Mock Mock+sh-DDX3 #2 sh-Scb+DDX3 Mock+sh-DDX3 #3 sh-circ-CTNNB1 #2+Mock circ-CTNNB1+sh-Scb sh-circ-CTNNB1 #2+DDX3 circ-CTNNB1+sh-DDX3 #2 sh-circ-CTNNB1 #3+Mock #2 Ratio) 4 Ratio) 4 circ-CTNNB1+sh-DDX3 #3 sh-circ-CTNNB1 #3+DDX3 Scb DDX3 #2 -YY1 #2

Renilla * * Scb DDX3 #2 2 * 2 * H - - CTNNB1+sh- CTNNB1+sh- ck+sh ck+sh ck+sh-YY1 Relative YY1 activity YY1 Relative (Firefly/ Relative YY1activity (Firefly/ Renilla * * * * * o o o * * * M circ- M circ- M circ-CTNNB1+sh 0 0 DDX3 73 kDa MKN-45 AGS MKN-45 AGS YY1 68 kDa G WNT1 WNT3 -catenin 20 Mock+sh-Scb 20 Mock+sh-Scb 92 kDa circ-CTNNB1+sh-Scb circ-CTNNB1+sh-Scb 16 Mock+sh-DDX3 #2 16 Mock+sh-DDX3 #2 WNT1 41 kDa circ-CTNNB1+sh-DDX3 #2 circ-CTNNB1+sh-DDX3 #2 12 12 Mock+sh-YY1 #2 Mock+sh-YY1 #2 WNT3 39 kDa Ratio) circ-CTNNB1+sh-YY1 #2 Ratio) circ-CTNNB1+sh-YY1 #2 8 8 * * AXIN2 95 kDa * 4 * 4 Renilla Renilla FZD10 65 kDa 2 2 BMP4 47 kDa (Firefly/ (Firefly/ 1 1 * * Relative luciferase activity luciferase Relative * activity luciferase Relative * * * * * -actin 42 kDa 0 0 MKN-45 AGS MKN-45 AGS

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 7 MKN-45 A DAPI FITC-peptide Dil Merged C 0 M 15 M 30 M 400 0 μM 15 μM 30 μM 300 24 hr 24 Ctrl

200

Ctrl *

100 * 48 hr 48 DIP-13

Number of colonies per well per colonies of Number 0 Ctrl DIP-13 D MKN-45 400 0 μM 15 μM

24 hr 24 0 M 15 M 30 M 30 μM 300 DIP-13 Ctrl 200 48 hr 48 100 * * DIP-13 B 0 Relative cells invaded per field Ctrl DIP-13

Input R-probe L-probe C-probe DIP-13 + + + + E ) 1.2 Ctrl week 3 DDX3 73 kDa 01 2 3 4 5 DIP-13 0.8 H3 histone 15 kDa 3.0e * +003 0.4 F Ki-67 CD31

Ctrl DIP-13 Ctrl DIP-13 Tumor volume (cm0.0 week 0 1 2 3 4 5 Ctrl

1.2 Ctrl DIP-13 1.0 30 0.8 * * 0.8 20 0.4 0.6 * DIP-13 0.4 (g) weight Tumor 10 0.2 0 0.0

× area Ki-67) by Ctrl DIP-13 count (by CD31) (by count Score(intensity

0.0 Intratumoral vessel 0 Ctrl DIP-13 Ctrl DIP-13 G H Intron week 01 2 3 4567 8 CTNNB1 Exon 9 10 11 12

circ-CTNNB1 DIP-13 DIP-13 Ctrl circ-CTNNB1 circ-CTNNB1 DIP-13 DDX3 TSS DDX3 TSS YY1 0 1.5e+003 YY1

Target gene promoter Target gene promoter 15 P=0.0118 100

Ctrl 75 10 P=0.0297 WNT1 WNT1 50 -catenin activation Ctrl 5 DIP-13 WNT3 WNT3 25 etc. etc. Percent survival Percent Metastasis count Metastasis DIP-13 0 0 Cancer progression Ctrl DIP-13 0 10 20 30 40 50 Days

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 18, 2018; DOI: 10.1158/0008-5472.CAN-18-1559 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Cis-acting circ-CTNNB1 promotes β-catenin signaling and cancer progression via DDX3-mediated transactivation of YY1

Feng Yang, Erhu Fang, Hong Mei, et al.

Cancer Res Published OnlineFirst December 18, 2018.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-18-1559

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2018/12/18/0008-5472.CAN-18-1559.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet Manuscript been edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2018/12/18/0008-5472.CAN-18-1559. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2018 American Association for Cancer Research.