Abstract Background Resistance to poly (ADP-ribose) polymerase inhibitors (PARPi) poses a major challenge to therapeutic efficacy in castration-resistant prostate cancer (CRPC). Although circular RNAs (circRNAs) have emerged as critical regulators in cancer biology, their involvement in PARPi resistance remains largely uncharacterized. Objective This study aims to elucidate the molecular mechanism by which hsa_circ_0038737 modulates PARPi resistance in CRPC through post-transcriptional regulatory pathways. Methods We employed a comprehensive set of in vitro and in vivo approaches, including qRT-PCR, RNA sequencing, RNA-protein pull-down, RNA immunoprecipitation, functional assays, and xenograft/organoid models, to investigate the biological function and mechanistic role of hsa_circ_0038737 in CRPC progression and therapeutic response. Results We identified hsa_circ_0038737 as a nuclear-enriched circRNA significantly upregulated in CRPC, with expression levels correlating with poor prognosis and aggressive clinical features. Mechanistically, hsa_circ_0038737 interacts with RNA-binding protein (RBP) IGF2BP3, enhancing the stability of DNPH1 mRNA, a nucleotide sanitizer critical for DNA repair. The circRNA-RBP-mRNA regulatory axis promotes PARPi resistance by facilitating DNA damage repair capacity. Moreover, we revealed that reverse-complementary Alu elements mediate circRNA biogenesis, with HNRNPDL facilitating this process. Pharmacologic inhibition of DNPH1 effectively restored PARPi sensitivity both in vitro and in vivo. Conclusion Our findings reveal a novel hsa_circ_0038737/IGF2BP3/DNPH1 axis driving PARPi resistance in CRPC, offering promising potential biomarkers and therapeutic targets to overcome resistance and improve treatment outcomes in advanced prostate cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-025-02447-y. Keywords: Castration-resistant prostate cancer (CRPC), Circular RNA (circRNA), PARP inhibitor, IGF2BP3, DNPH1 stabilization Introduction Prostate cancer (PCa) is the second most commonly diagnosed cancer among men, with an estimated 1,466,680 new cases and approximately 396,792 deaths worldwide in 2022 [[44]1, [45]2]. Castration-resistant prostate cancer (CRPC) denotes an advanced stage of PCa characterized by disease progression despite castrate levels of testosterone. Virtually all prostate cancer-related mortalities are attributable to CRPC, frequently with metastatic progression (mCRPC) [[46]3]. Epidemiologically, CRPC constitutes 10%−20% of all PCa cases [[47]4, [48]5]. Upon the onset of metastasis, the median survival duration is less than three years [[49]6]. Approximately 28% of mCRPC patients harbor mutations in homologous recombination repair (HRR) genes, such as BRCA1/2, making them candidates for poly-ADP-ribose polymerase inhibitors (PARPi) [[50]7, [51]8]. PARPi, such as olaparib and rucaparib, exploit synthetic lethality by targeting HRR-deficient tumors, leading to DNA damage accumulation and cell death [[52]9, [53]10]. However, the clinical efficacy of PARPi is often limited by the development of resistance, which remains a significant challenge in CRPC treatment [[54]11]. Mechanisms of PARPi resistance include reversion mutations in BRCA genes, upregulation of drug efflux pumps, and alterations in DNA repair pathways [[55]12–[56]14]. Understanding these resistance mechanisms is crucial for developing strategies to overcome therapeutic limitations and improve patient outcomes. Circular RNAs (circRNAs) have emerged as key regulators of tumorigenesis and cancer progression due to their unique circular structure and functional versatility [[57]15–[58]17]. Unlike linear RNAs, circRNAs are highly stable and resistant to degradation, allowing them to modulate gene expression through interactions with RNA-binding proteins (RBPs) and microRNAs [[59]18]. In PCa, circRNAs have been implicated in regulating critical pathways such as cell proliferation, apoptosis, and drug resistance [[60]16]. For instance, METTL3 enhances the stability of CircGLIS3 through m6A modification, and CircGLIS3 sequesters miR-661 to upregulate MDM2 mRNA levels, thereby regulating the p53 signaling pathway to promote the proliferation, migration, and invasion of PCa [[61]19]. Additionally, circ_0004087 interacts with the transcriptional coactivator SND1, stimulating the transactivation of MYB and enhancing the expression of its downstream target BUB1, which in turn promotes mitosis and increases PCa resistance to docetaxel [[62]20]. These findings underscore the potential of circRNAs as biomarkers and therapeutic targets in PCa. In this study, we investigate the role of hsa_circ_0038737 in mediating PARPi resistance in CRPC. We demonstrate that hsa_circ_0038737 enhances the stability of DNPH1 mRNA by binding to IGF2BP3, a well-characterized RBP involved in mRNA stabilization. This interaction leads to increased DNPH1 expression, which in turn contributes to PARPi resistance in CRPC cells. Our findings provide novel insights into the molecular mechanisms underlying PARPi resistance and highlight the potential of targeting circRNA-RBP interactions as a therapeutic strategy to overcome resistance in CRPC. Materials and methods Human tissue specimens and cell lines Between 2015 and 2022, a total of 78 PCa tissue samples were collected from patients who underwent radical prostatectomy at the Fudan University Shanghai Cancer Center (FUSCC). The collection and use of these tissue samples were approved by the hospital’s ethics committee, and written informed consent was obtained from all participating patients. Clinicopathological features of the 78 PCa patients were subsequently analyzed. The study was conducted in strict accordance with the guidelines of the Declaration of Helsinki. Additionally, five PCa cell lines (LNCaP, DU145, C4-2, PC-3, and 22RV1) and one human normal prostate epithelial cell line (RWPE-1) were obtained from the Chinese Academy of Sciences Type Culture Collection (Shanghai, China) for further experimental investigations. Establishment of patient-derived tumor organoids PCa specimens were obtained from Fudan University Shanghai Cancer Center. Immediately after surgical resection, fresh tumor samples were placed in RPMI 1640 medium containing antibiotics, supplemented with 10 µM Y-27,632 (MCE, USA) and 10% FBS, and transported to the laboratory on ice at 4 °C for immediate processing. Upon arrival, tumors were gently washed at least three times with pre-cooled 1X DPBS containing 3% penicillin-streptomycin solution to remove surface contaminants. The tumor tissue was then minced into small pieces using surgical scissors and subjected to enzymatic digestion using a tissue dissociation system (D1Med, China) to generate a single-cell suspension. The digested suspension was passed through a 70 μm cell strainer to remove any undigested debris, followed by centrifugation at 500 × g for 5 min. The resulting cell pellet was resuspended in Matrigel (D1Med, China) and plated in 24-well culture dishes. After allowing the Matrigel to solidify for 15 min at room temperature, conditioned medium (provided by D1 Medical Technology, China) was added to support cell growth and maintenance. Cell culture and transfection The RWPE-1 cell line was cultured in Keratinocyte Serum-free Medium (K-SFM; Gibco, USA). The prostate cancer cell lines LNCaP, DU145, C4-2, PC-3, and 22RV1 were cultured in Roswell Park Memorial Institute medium (RPMI-1640; Gibco, USA). All cell lines were Maintained in their respective media supplemented with 10% fetal bovine serum (FBS; Gibco, USA), 100 U/mL penicillin, and 100 µg/mL streptomycin (Invitrogen, USA). Cells were incubated at 37 °C in a humidified atmosphere containing 95% air and 5% CO₂. Plasmid construction, transfection and lentivirus infection assay The hsa_circ_0038737 knockdown and overexpression lentiviral vectors were obtained from HANBIO (Shanghai, China). When 22RV1 or PC-3 cells reached approximately 50% confluence, they were transfected with either hsa_circ_0038737 knockdown or overexpression lentiviruses, as well as their respective negative control lentiviruses. Following transfection, cells were selected using puromycin at concentrations of 4–8 µg/mL for two rounds to ensure stable integration and expression. The overexpression plasmids and short hairpin RNAs (shRNAs) targeting IGF2BP3, HNRNPDL, and DNPH1 were acquired from GenePharma (Shanghai, China). Transfections of these constructs into 22RV1 or PC-3 cells were performed using the Lipofectamine 3000 reagent kit (Invitrogen, USA) according to the manufacturer’s instructions. The sequences of the shRNAs used in this study are listed in TableS1. RNA extraction and quantitative real time-PCR (qRT-PCR) Total RNA was isolated from PCa tissues or cell lines using TRIzol reagent (Invitrogen, USA) following the manufacturer’s instructions. The extracted RNA was subsequently reverse-transcribed into cDNA using the HiScript II Reverse Transcriptase Kit (Vazyme, China). qRT-PCR was performed to quantify the expression levels of circRNA or mRNA using the SYBR Green Master Mix (Vazyme, China). The qRT-PCR experiments were conducted on either the StepOne Plus Real-Time PCR System (Applied Biosystems, USA) or the LightCycler 480 (Roche, USA). The primers used in this study were synthesized by TsingKe (China), and their detailed sequences are also provided in TableS1. Protein isolation and Western blot assays Proteins were extracted from tissue or cell samples using RIPA buffer (Sigma, USA) supplemented with protease and phosphatase inhibitors to ensure protein integrity. Protein concentrations were quantified using the BCA Protein Assay Kit (Beyotime, China) according to the manufacturer’s instructions. Equal amounts of protein were separated by SDS-PAGE based on molecular weight and then transferred onto PVDF membranes (Millipore, USA). The membranes were blocked with 5% skim milk in Tris-buffered saline with Tween-20 at room temperature for 1 h to prevent nonspecific binding. Subsequently, the membranes were incubated with primary antibodies overnight at 4 °C. After extensive washing with TBST, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies for 1 h at room temperature. Chemiluminescent signals were detected using an enhanced electrochemiluminescence system (Thermo Fisher Scientific, USA) and visualized using a ChemiDoc MP Imaging System (Bio-Rad, USA). RNase R treatment assay Total RNA was treated with RNase R (0.2 µl/µg, Epicenter) at 37 °C for 30 min. Subsequently, the expression levels of hsa_circ_0038737 and GTF3C1 mRNA were detected by qRT-PCR. Actinomycin D treatment and RNA stability assays The 22RV1 and PC-3 cell lines were treated with actinomycin D (1 µg/ml; Abcam, UK) at designated time points (0, 2, 4, 6, 8, and 10 h). Following treatment, total RNA was extracted from the cells, and qRT-PCR was performed to assess the expression levels of hsa_circ_0038737, GTF3C1, and DNPH1. RNA-protein pulldown, silver staining and mass spectrometry assays RNA-protein pull-down assays were conducted using the Pierce Magnetic RNA-Protein Pull-Down Kit (Thermo Fisher Scientific, USA) according to the manufacturer’s instructions. Biotinylated probes specific to the back-splice junction of hsa_circ_0038737 were synthesized by GenePharma (GenePharma, China). Cells were lysed in IP lysis buffer and incubated with the biotinylated probes at room temperature for 4 h. The RNA-protein complexes were captured using streptavidin-coated magnetic beads. Subsequently, Western blotting was performed to identify the proteins that were pulled down by the probes. Silver staining was carried out using the Quick Silver Staining Kit (Beyotime, China) to visualize the protein bands. Further identification and quantification of the proteins were performed by mass spectrometry analysis at Keygentec (Keygentec, China), utilizing the Proteome Discoverer Software 2.4 (Thermo Fisher Scientific, USA) for data processing. Immunofluorescence and fluorescence in situ hybridization (IF-FISH) assays Cell smears were fixed with 4% paraformaldehyde for 15 min at room temperature and then permeabilized with 0.1% Triton X-100 for 10 min. Cy3-labeled hsa_circ_0038737 probes (Genepharma, China) were hybridized with the cells in hybridization buffer at 37 °C for 16 h. After hybridization, cells were blocked with 5% bovine serum albumin for 1 h at room temperature to reduce nonspecific binding. Subsequently, cells were incubated sequentially with primary antibodies against IGF2BP3 and corresponding secondary antibodies (Beyotime, China) for 1 h each at room temperature. Nuclei were counterstained with DAPI (MCE, USA). Confocal microscopy images were captured using a TCS SPE-II confocal microscopy system (Leica, Germany) to visualize the localization of hsa_circ_0038737 and IGF2BP3 within the cells. RNA immunoprecipitation (RIP) assay For RIP assays, cells were collected and lysed in IP lysis buffer. The Magna RIP RNA-Binding Protein Immunoprecipitation Kit (Millipore, USA) was used according to the manufacturer’s protocol. Specific antibodies against IGF2BP3 (Proteintech, China), FLAG (Proteintech, China), HNRNPL (Proteintech, China), and control IgG (Proteintech, China) were employed to precipitate RNA-protein complexes. Total RNA was extracted from both the input and IP samples using TRIzol reagent (Invitrogen, USA). The levels of specific RNAs were subsequently quantified by quantitative real-time PCR (qRT-PCR). Cell proliferation and cloning formation assays To evaluate the proliferation capacity of PCa cells, 22RV1 or PC-3 cells were plated at a density of 1,000 cells per well in a 96-well plate. Cell proliferation was assessed using the Cell Counting Kit-8 (CCK-8) (MCE, USA), and absorbance values were measured at 450 nm with a microplate reader (Tecan, Switzerland). For colony formation assays, approximately 1,000 22RV1 or PC-3 cells were Seeded into each well of a 6-well plate. After 14 days of incubation, the cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet solution. The colonies were visualized and quantified using ImageJ software (NIH, USA). Cell cycle analysis Cells were harvested and fixed in a 70% ethanol solution at 4 °C for 2 h. After fixation, the cells were washed with cold phosphate-buffered saline and stained with propidium iodide buffer (Beyotime, China) at room temperature for 30 min. PCa cells were then analyzed for cell cycle distribution using a FACS Scan flow cytometer (BD Biosciences, USA). The data were processed and analyzed using ModFit LT 2.0 software (Verity Software House, USA). Half-maximal inhibitory concentration (IC50) assay Cells were Seeded in 96-well plates and treated with varying concentrations of olaparib or niraparib for 7 days. Cell viability was assessed using the CCK8 assay and IC50 values were calculated by plotting dose-response curves and determining the concentration that inhibits 50% cell growth compared to untreated controls. Comet assay Cells were treated, harvested, and mixed with low-melting agarose using the Keygenbio Comet Assay Kit (Keygenbio, China). Lysed cells were subjected to electrophoresis, stained with DNA dye, and visualized under a fluorescence microscope (Olympus FV3000, Olympus Corp.; Japan) to evaluate DNA damage. Proximity ligation assay (PLA) PC-3 and 22RV1 prostate cancer cells were plated in chambered coverslips and cultured for 24 h. Following fixation with 4% paraformaldehyde (15 min) and permeabilization in 0.3% Triton X-100 (10 min), samples were blocked with Duolink blocking solution (Sigma, USA; 60 min, 37 °C). Primary antibodies against PARP1 and γ-H2AX were applied overnight at 4 °C. Species-specific PLA probes (Duolink In Situ PLA Probe Anti-Rabbit PLUS, Sigma; Anti-Mouse MINUS, Sigma) were incubated for 120 min at 37 °C. Oligonucleotide ligation (30 min, 37 °C) and rolling-circle amplification (100 min, 37 °C) were performed using Duolink In Situ Detection Reagents Red (Sigma, USA). Nuclei were counterstained with DAPI (MCE, USA) in ProLong Gold mounting medium (MCE, USA). Imaging was conducted on an Olympus FV3000 confocal microscope with a 63× oil objective. Chromatin fractionation To isolate chromatin-bound proteins, a subcellular fractionation procedure was conducted employing a commercial kit (Thermo Fisher Scientific, USA). Treated cells were harvested, washed with ice-cold PBS, and subjected to sequential extraction. Cytosolic components were first released by resuspending the cell pellet in Cytoplasmic Extraction Buffer (CEB) for a 10-minute incubation on ice, followed by centrifugation (500 × g, 5 min, 4 °C). Subsequent treatment of the resulting pellet with Membrane Extraction Buffer (MEB) for 10 min on ice and centrifugation (3,000 × g, 5 min, 4 °C) yielded the membrane protein fraction. The remaining insoluble material was then exposed to Nuclear Extraction Buffer (NEB) for 30 min on ice. Centrifugation (5,000 × g, 5 min, 4 °C) separated the soluble nuclear proteins in the supernatant. Finally, the chromatin-enriched pellet was digested with NEB containing nuclease for 15 min at ambient temperature. High-speed centrifugation (16,000 × g, 5 min) recovered the chromatin-associated proteins in the supernatant. All steps strictly adhered to the manufacturer’s protocol. Immunohistochemistry (IHC) assay Paraffin-embedded tissue samples were cut into 4 μm sections and mounted onto glass slides. The sections were deparaffinized and rehydrated through a graded ethanol series. Tissue sections underwent heat-mediated antigen retrieval in 10 mM citrate buffer (pH 6.0) for 20 min at 95 °C. The buffer was prepared by dissolving citric acid (MCE, USA) in ultrapure water, adjusted to pH 6.0 with NaOH. Following this, the sections were treated with 3% hydrogen peroxide for 10 min to quench endogenous peroxidase activity. Subsequently, the sections were incubated overnight at 4 °C with primary antibodies targeting Ki-67, IGF2BP3, PSMA, and AR (all from Proteintech, China). After washing, the sections were incubated with secondary antibodies for 30 min at room temperature. Immunostaining was visualized using diaminobenzidine (MCE, USA) as the chromogen, followed by counterstaining with hematoxylin to highlight cell nuclei. The stained sections were examined and imaged under a light microscope. RNA sequencing Total RNA was extracted from three pairs of sh-hsa_circ_0038737 and sh-NC PC3 cells. Following stringent quality control measures, RNA sequencing was performed on the Illumina platform at Oebiotech (Shanghai, China). Differentially expressed genes were identified using the OECloud tool available at Oebiotech Cloud Platform ([63]https://cloud.oebiotech.com), with the criteria of q-value < 0.05 and |log2 fold change (FC)| >0.5. Functional enrichment analysis was subsequently conducted using the ‘clusterProfiler’ R package to elucidate potential pathways associated with the observed gene expression changes. Nucleic and cytoplasmic RNA assay Cellular and nuclear RNA were isolated separately using the Cytoplasmic & Nuclear RNA Purification Kit (Norgen Biotek, Thorold, ON, USA). 22RV1 and PC-3 cells were lysed on ice in Lysis Buffer J, followed by centrifugation at 14,000 × g for 10 min at 4 °C. The supernatant, containing cytoplasmic RNA, was carefully transferred to an RNase-free tube. The pellet, containing nuclear RNA, was retained for further analysis. qRT-PCR was performed to quantify the RNA levels in both cytoplasmic and nuclear fractions. U6 and GAPDH mRNA were used as reference genes for the nuclear and cytoplasmic fractions, respectively, to evaluate the relative distribution of the target RNA between these compartments. Xenograft experiments Male BALB/c nude mice, aged 6 to 7 weeks, were obtained from the Animal Medical Center Affiliated with Fudan University. To establish subcutaneous xenograft models, 22Rv1 cells (vehicle, hsa_circ_0038737 shRNA, or hsa_circ_0038737 overexpression) were mixed 1:1 with Matrigel (Corning, USA) and injected subcutaneously into the dorsal flank of the mice. Experimental treatments included vehicle, olaparib (50 mg/kg), or a combination of olaparib and DNPH1 inhibitor (25 mg/kg). Both drugs were administered orally once Daily. Olaparib was given for 5 consecutive days followed by a 2-day rest period, while the DNPH1 inhibitor was administered continuously for 4 days each week, starting the day after olaparib treatment initiation. Tumor size and volume were measured every 5 days. After 3 weeks of treatment, animals were euthanized, and tumors were excised for further analysis. The animal experimental protocol was approved by the Institutional Animal Care and Use Committee of the Shanghai Veterinary Research Institute (Approval No. FUSCC-IACUC-2024293). Analysis of GO and KEGG enrichment and network-based functional annotations To explore the functional characteristics of the 270 proteins detected by mass spectrometry, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Using clusterProfiler and org.Hs.eg.db, we performed GO enrichment analysis on the gene set, covering three main categories: Biological Process (BP), Cellular Component (CC), and Molecular Function (MF), with the p-value adjustment method being the Benjamini-Hochberg correction. Additionally, we employed the enrichKEGG function to carry out KEGG pathway enrichment analysis on the same gene set. All significantly enriched pathways were ranked based on -log10(p-value) and Rich Factor (defined as the ratio of the number of genes participating in a pathway to the total number of background genes in that pathway). The results were visualized using ggplot2 in a dual-axis bar chart, displaying the significance of p-values (-log10(pvalue)) and the degree of pathway enrichment (Rich Factor), thereby revealing key functional pathway characteristics. Further enrichment analysis based on WikiPathways database and visualization using the ClueGO (version 2.5.9) and CluePedia (version 1.5.9) plugins within the Cytoscape software (version 3.9.1). The enrichment test employed was the two-sided hypergeometric test, with multiple testing correction applied using the Benjamini-Hochberg method (FDR < 0.05). The parameters for network construction were set as follows: a minimum of 3 pathway genes and a minimum gene proportion of 4%, with a functional connectivity (Kappa score) set at 0.4. Cohort validation for the prognostic implications of DNPH1 expression Two independent PCa cohorts were collected from cbioportal database ([64]https://www.cbioportal.org/) in this study. Only samples with complete prognostic information and available RNA-seq transcriptomic data were retained for downstream analysis. The MSKCC cohort (n = 140) comprised patients with primary PCa, while the SU2C 2019 cohort (n = 81) consisted of CRPC cases. Progression-free survival (PFS) and overall survival were used as the index to evaluate prognosis of PCa patients, respectively. All of the expression data was annotated by corresponding platform and expressed as Transcripts Per Million (TPM). To evaluate the prognostic significance of the target gene, we utilized the Survminer package to determine the optimal cutoff value for gene expression based on the maximum selected rank statistic. Subsequently, patients were divided into high-expression and low-expression groups according to this cutoff value. The Kaplan-Meier survival curves were plotted and the differences between groups were compared using the log-rank test. Statistical analysis Continuous variables are presented as mean ± standard deviation (mean ± SD), while categorical variables are described using counts and proportions. Statistical analysis and visualization were conducted using GraphPad Prism 10.0 software. Data were derived from public databases and sequencing results. Statistical comparisons were performed using t-tests, Mann-Whitney U tests, and χ² tests. A p-value of less than 0.05 was considered statistically significant, with significance levels indicated as follows: * for p < 0.05, ** for p < 0.01, *** for p < 0.001, and **** for p < 0.0001. Results Hsa_circ_0038737 is up-regulated in PCa tissues and correlated with aggressive progression of PCa To elucidate the potential clinical relevance of circular RNAs in PCa, we first investigated the expression profile of hsa_circ_0038737, a previously uncharacterized circRNA. Analysis of five independent, publicly available transcriptomic datasets revealed that hsa_circ_0038737 is consistently and significantly upregulated in PCa tissues compared to matched adjacent normal tissues (Fig. [65]1A). This finding was further validated in our cohort using qRT-PCR, and the back-splicing junction characteristic of circular RNAs was confirmed by Sanger sequencing (Fig. [66]1B). Genomic alignment indicated that hsa_circ_0038737 is generated from exons 35 and 36 of the GTF3C1 gene, located on chromosome 16, through a canonical back-splicing mechanism. Fig. 1. [67]Fig. 1 [68]Open in a new tab Upregulation and Oncogenic Association of hsa_circ_0038737 in Prostate Cancer. (A) Detection of differentially expressed circRNAs in prostate cancer through five public sequencing datasets. (B) Identification of the reverse-splicing junction of hsa_circ_0038737 by Sanger sequencing. (C) Detection of hsa_circ_0038737 in cDNA and gDNA of 22RV1 and PC-3 cells using divergent and convergent primers, respectively. (D) The half-life of hsa_circ_0038737 in 22RV1 cells determined by actinomycin D treatment. (E) The relative expression levels of hsa_circ_0038737 in 22RV1 and PC-3 cells with or without RNase R treatment. (F-G) The subcellular localization of hsa_circ_0038737 in 22RV1 and PC-3 cells determined by nuclear and cytoplasmic fractionation and Fish. (H) The expression levels of hsa_circ_0038737 in different prostate cell lines. (I) The expression levels of hsa_circ_0038737 in normal and tumor prostate tissues. (J) The expression levels of hsa_circ_0038737 in different T stage prostate cancer tissues. (K) The expression levels of hsa_circ_0038737 in different Gleason scores prostate cancer tissues. Notes: Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance To verify its circular topology, we designed convergent and divergent primers targeting the splice junction. The circRNA was successfully amplified from complementary DNA (cDNA), but not from genomic DNA (gDNA), confirming the absence of linear genomic contamination and the authenticity of its circular structure (Fig. [69]1C). Functional assays revealed that hsa_circ_0038737 exhibits enhanced stability, with a prolonged half-life under actinomycin D treatment compared to its linear counterpart GTF3C1 mRNA (Fig. [70]1D), and showed marked resistance to RNase R digestion, a hallmark of circRNAs (Fig. [71]1E). Subcellular fractionation and FISH assays demonstrated that hsa_circ_0038737 is predominantly localized in the nucleus (Figs. [72]1F, G), suggesting a potential role in transcriptional or epigenetic regulation. Expression analysis across a panel of PCa cell lines revealed significantly higher levels of hsa_circ_0038737 in malignant cells compared to the normal prostate epithelial cell line RWPE-1 (Fig. [73]1H). Consistently, in a cohort of 78 paired clinical PCa samples, hsa_circ_0038737 was markedly upregulated in tumor tissues (Fig. [74]1I), with expression levels positively correlating with advanced T stage and higher Gleason scores in PCa patients (Fig. [75]1J, K). Collectively, these findings position hsa_circ_0038737 as a stable, nucleus-enriched circRNA with oncogenic potential in prostate cancer. Its strong association with aggressive clinical features and enhanced expression in tumor tissues suggests a functional role in disease progression and highlights its potential utility as a biomarker for prognosis stratification and a candidate target for therapeutic intervention. Hsa_circ_0038737 enhances proliferation of PCa cells via promotion of G2/M cell cycle progression To investigate the functional role of hsa_circ_0038737 in PCa proliferation independent of its parental linear transcript GTF3C1, we established both loss- and gain-of-function models in two representative PCa cell Lines, 22RV1 and PC-3. Lentiviral vectors encoding either shRNAs targeting hsa_circ_0038737 or circRNA-specific overexpression constructs were transduced into the cells. Efficient knockdown and overexpression of hsa_circ_0038737 were confirmed by qRT-PCR (Fig. [76]2A, B; Figure S1 A-D), without significant alteration of GTF3C1 mRNA expression, thereby ensuring circRNA specificity. Fig. 2. [77]Fig. 2 [78]Open in a new tab Hsa_circ_0038737 Enhances Proliferation of Prostate Cancer Cells via Promotion of G2/M Cell Cycle Progression. (A-B) The relative expression levels of hsa_circ_0038737 were detected in 22RV1 and PC-3 cells after transfection with shRNAs (Sh1, Sh2, Sh3) or overexpression constructs. (C-E) CCK-8 and colony formation assays were performed to evaluate the impact of hsa_circ_0038737 knockdown and overexpression on the proliferation of 22RV1 and PC-3 cells. (F-G) Cell cycle analysis was conducted to investigate the effect of hsa_circ_0038737 knockdown and overexpression on cell cycle distribution in PCa cells. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance To assess the impact of hsa_circ_0038737 on tumor cell proliferation, we conducted CCK-8 cell viability assays and colony formation assays. The results consistently demonstrated that silencing hsa_circ_0038737 markedly suppressed cell proliferation, while its overexpression significantly enhanced clonogenic capacity in both cell lines (Fig. [79]2C-E). To further elucidate the underlying mechanism, we performed cell cycle analysis via flow cytometry. Knockdown of hsa_circ_0038737 led to a prominent accumulation of cells in the G2 phase, indicative of a G2/M checkpoint arrest (Fig. [80]2F, G), whereas circRNA overexpression decreased the G2-phase population, suggesting facilitated progression into mitosis. These results imply that hsa_circ_0038737 may regulate tumor growth by accelerating the G2-to-M phase transition, a critical control point in cell division. Collectively, these data demonstrate that hsa_circ_0038737 functions as a positive regulator of PCa cell proliferation through modulation of cell cycle progression, independent of its linear host gene GTF3C1. This proliferative advantage may contribute to the aggressive clinical phenotype observed in tumors with elevated hsa_circ_0038737 expression. Given the essential role of cell cycle control in tumorigenesis, these findings underscore the potential of hsa_circ_0038737 as a mechanistic driver and therapeutic vulnerability in prostate cancer. Hsa_circ_0038737 directly interacts with IGF2BP3 via KH domains to mediate functional activity in prostate cancer Recent evidence suggests that nuclear-localized circRNAs may exert regulatory functions by acting as scaffolds for RNA-binding proteins, thereby modulating post-transcriptional gene regulation. To identify the potential RBP partners of hsa_circ_0038737, we employed a biotinylated RNA pull-down assay using probes specifically designed to target the back-splice junction of the circRNA. Silver staining of the pull-down complexes followed by mass spectrometry revealed a panel of candidate proteins that interact with hsa_circ_0038737, among which IGF2BP3 emerged as a prominent interactor (Fig. [81]3A and Table S2). Fig. 4. [82]Fig. 4 [83]Open in a new tab The hsa_circ_0038737-IGF2BP3 Complex Stabilizes DNPH1 mRNA to Promote DNA Damage Response Signaling. (A) RNA-seq analysis was performed on three pairs of hsa_circ_0038737 knockdown and control 22RV1 cells to identify differentially expressed genes. (B) GO enrichment analysis was conducted to determine the biological processes in which these differentially expressed genes were primarily enriched. (C-D) WikiPathways, Reactome, and KEGG pathway enrichment analyses were carried out to identify significant enrichment of differentially expressed genes in pathways related to DNA repair and DNA damage response. (E) Integrative analysis was performed using downregulated genes from RNA-seq data, publicly available IGF2BP3 CLIP-seq data, and Starbase predictions to identify DNPH1 as a potential target. (F) RIP assays were conducted to confirm the specific binding of IGF2BP3 to DNPH1 mRNA in 22RV1 and PC-3 cells. (G) The effects of hsa_circ_0038737 knockdown or overexpression on DNPH1 mRNA levels were assessed. (H) Changes in DNPH1 protein levels upon IGF2BP3 knockdown or overexpression were observed. (I) The stability of DNPH1 mRNA following knockdown of hsa_circ_0038737 or IGF2BP3 in both cell lines was evaluated. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 Fig. 3. [84]Fig. 3 [85]Open in a new tab Hsa_circ_0038737 Directly Interacts with IGF2BP3 via KH Domains to Mediate Functional Activity in Prostate Cancer. (A) RNA-protein pull-down experiments were performed using biotin-labeled probes targeting the back-splice site of hsa_circ_0038737 in 22RV1 cells. Silver staining was conducted to identify proteins that specifically interact with hsa_circ_0038737. (B) Gene Ontology, KEGG and Wikipathway analysis were carried out to examine the enrichment of these proteins in pathways associated with mRNA binding and regulation of mRNA stability. (C) Data from RBPsuite, CircNet, CSCD2 databases, and mass spectrometry results were integrated to predict IGF2BP3 as a primary protein partner of hsa_circ_0038737. (D-E) RNA-FISH and IF co-localization assays, and RIP experiments using anti-IGF2BP3 antibodies were performed to verify the direct binding between hsa_circ_0038737 and IGF2BP3 in 22RV1 and PC-3 cells. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance To gain insight into the biological relevance of these interactions, we performed Gene Ontology (GO), KEGG, and WikiPathway enrichment analyses, which indicated that the identified proteins were significantly enriched in pathways associated with mRNA binding, mRNA stabilization, and RNA metabolism (Fig. [86]3B). To further prioritize functional interactors, we integrated our mass spectrometry data with computational predictions from three independent circRNA-RBP interaction databases: RBPsuite ([87]http://www.csbio.sjtu.edu.cn/bioinf/RBPsuite), a deep-learning platform for RBP binding site prediction; CircNet ([88]https://awi.cuhk.edu.cn/~CircNet/php/index.php), which maps circRNA-mediated regulatory networks; and CSCD2 ([89]https://geneyun.net/CSCD2/) databases, a cancer-specific circRNA database that annotates RBP interaction motifs. All three resources convergently predicted IGF2BP3 as a high-confidence binding partner of hsa_circ_0038737 (Fig. [90]3C), strongly supporting its functional significance. To experimentally validate this interaction, we performed RNA-FISH combined with immunofluorescence staining. The results revealed co-localization of hsa_circ_0038737 and IGF2BP3 in the nuclear compartment of both 22RV1 and PC-3 cells (Fig. [91]3D). Additionally, RNA pull-down followed by western blot and RIP assays using anti-IGF2BP3 antibodies confirmed the physical binding between the circRNA and the protein (Fig. [92]3E; Figure S1E). To delineate the interaction interface, we generated a series of FLAG-tagged IGF2BP3 truncation constructs corresponding to distinct RNA-binding domains (Figure S2A, B). RIP assays with anti-FLAG antibodies revealed that hsa_circ_0038737 predominantly binds to the KH1 and KH2 domains of IGF2BP3 (Figure S2C), which are known to mediate sequence-specific RNA recognition. Together, these results establish hsa_circ_0038737 as a functional interactor of IGF2BP3, acting through its KH domains to potentially modulate downstream mRNA stability. The circRNA-RBP interaction forms the molecular foundation for subsequent regulatory events, laying the groundwork for the circRNA’s role in therapeutic resistance and cell proliferation in CRPC. IGF2BP3 functions as a proliferative driver and indicator of poor prognosis in prostate cancer To functionally characterize the role of IGF2BP3, a key RNA-binding partner of hsa_circ_0038737, in PCa, we established both gain- and loss-of-function models in 22RV1 and PC-3 cell lines. Lentiviral-mediated overexpression and knockdown of IGF2BP3 were confirmed at the mRNA and protein levels (Figure S3A, B). The CCK-8 assay revealed that IGF2BP3 knockdown significantly suppressed the proliferation rate of both 22RV1 and PC-3 cells, whereas its overexpression markedly enhanced cell proliferation compared to controls (Figure S3C, D). Consistent with these findings, colony formation assays demonstrated that IGF2BP3 depletion substantially decreased the number of viable colonies, while its overexpression resulted in a dramatic increase in clonogenic capacity in both cell lines (Figure S3E, F). These results suggest a negative regulatory role of IGF2BP3 in PCa cell proliferation, which is mechanistically intriguing given its interaction with hsa_circ_0038737. To explore the clinical relevance of IGF2BP3 expression, we performed survival analyses using patient data from two large-scale prostate cancer cohorts: Stand Up to Cancer (SU2C) and Memorial Sloan Kettering Cancer Center (MSKCC). Kaplan–Meier survival curves demonstrated that patients with high IGF2BP3 expression exhibited significantly shorter overall survival (OS) and disease-free survival (DFS) compared to those with low expression levels (Figure S3G, H), underscoring its potential prognostic value. Taken together, these data identify IGF2BP3 as a context-dependent regulator of prostate cancer cell proliferation and a marker of poor clinical outcomes. Although classically described as an RNA stabilizer that enhances oncogenic mRNA expression, our findings suggest that in the presence of specific molecular regulators, such as hsa_circ_0038737-IGF2BP3 may play a dual role, potentially acting as a regulatory node within a broader RNA-protein interaction network. The hsa_circ_0038737-IGF2BP3 complex stabilizes DNPH1 mRNA to promote DNA damage response signaling To elucidate the downstream functional consequences of hsa_circ_0038737 in prostate cancer cells, we performed RNA-seq on 22RV1 cells following knockdown of hsa_circ_0038737, with Matched controls. Differential gene expression analysis identified 101 upregulated and 314 downregulated genes in the knockdown group (Fig. [93]4A and Table S3). Gene Ontology (GO) enrichment revealed that these transcripts were significantly associated with mRNA stability regulation and DNA damage response pathways (Fig. [94]4B). Moreover, pathway enrichment analyses using WikiPathways, Reactome, and KEGG databases consistently highlighted DNA repair, DNA damage checkpoint signaling, and genomic stability maintenance as the most affected biological processes (Fig. [95]4C, D), implicating hsa_circ_0038737 in the modulation of cellular responses to genotoxic stress. Fig. 7. [96]Fig. 7 [97]Open in a new tab Hsa_circ_0038737 Promotes Tumor Growth and Mediates PARP Inhibitor Resistance via DNPH1 in Vivo. (A-D) Subcutaneous xenograft models were established in nude mice to evaluate the effects of hsa_circ_0038737 overexpression or knockdown on tumor growth and Olaparib resistance, with or without DNPH1i co-treatment. Mouse body weight was monitored throughout the experiment. (E-G) IHC analysis was performed on xenograft tissues to detect DNPH1 and Ki-67 levels in the hsa_circ_0038737 overexpression and knockdown groups. (H-I) IHC analysis was conducted on patient-derived tissues to examine the correlation between IGF2BP3 and DNPH1 expression. (J-K) Patient-derived organoids were used to assess Olaparib sensitivity in those with high hsa_circ_0038737 expression, with or without DNPH1i treatment. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance Although manipulation of hsa_circ_0038737 expression did not alter IGF2BP3 protein levels in either 22RV1 or PC-3 cells (Figure S3I), we hypothesized that hsa_circ_0038737 may function as a molecular scaffold, facilitating IGF2BP3-mediated stabilization of specific target mRNAs. To identify these targets, we employed an integrative computational and experimental strategy. Specifically, we intersected (1) the downregulated genes from our RNA-seq dataset, (2) publicly available IGF2BP3 CLIP-seq data [[98]21], and (3) StarBase-predicted mRNA targets of IGF2BP3 (Fig. [99]4E). This analysis pinpointed DNPH1 as a high-confidence target, an mRNA encoding a nucleotide sanitizer enzyme involved in DNA repair and PARPi response modulation [[100]22, [101]23]. To validate this interaction, RIP assays using IGF2BP3 antibodies confirmed direct binding between IGF2BP3 and DNPH1 mRNA in both 22RV1 and PC-3 cells (Fig. [102]4F). Functionally, knockdown of hsa_circ_0038737 significantly reduced DNPH1 mRNA levels, whereas overexpression of hsa_circ_0038737 led to a marked increase in DNPH1 abundance (Fig. [103]4G). Parallel experiments showed that IGF2BP3 knockdown decreased DNPH1 protein expression, while its overexpression elevated DNPH1 levels (Fig. [104]4H), consistent with a post-transcriptional regulatory mechanism. Finally, to determine whether hsa_circ_0038737 and IGF2BP3 cooperatively regulate mRNA stability, we conducted mRNA decay assays following transcriptional inhibition with actinomycin D. Notably, knockdown of either hsa_circ_0038737 or IGF2BP3 substantially decreased the half-life of DNPH1 mRNA, confirming their role in stabilizing DNPH1 transcripts (Fig. [105]4I). Collectively, these findings reveal a novel regulatory axis in which hsa_circ_0038737 serves as a functional adaptor that facilitates IGF2BP3 binding to DNPH1 mRNA, thereby enhancing its stability. Given the established role of DNPH1 in DNA repair and PARPi sensitivity, this mechanism provides important insight into how circRNA-RBP complexes contribute to therapeutic resistance and highlights a potential targetable pathway in castration-resistant prostate cancer. DNPH1 enhances proliferation and confers resistance to PARP inhibitors in CRPC cells Although DNPH1 has been implicated in nucleotide metabolism and DNA repair, its functional role and clinical relevance in CRPC remain largely uncharacterized. To investigate its biological impact, we established DNPH1-overexpressing and DNPH1-silenced 22RV1 and PC-3 cell models. Functional assays demonstrated that DNPH1 overexpression significantly enhanced cell proliferation and colony-forming ability, while DNPH1 knockdown markedly suppressed tumor cell growth (Fig. [106]5A-D), suggesting a positive role in tumor progression. To explore the clinical significance of DNPH1 in PCa, we analyzed transcriptomic and survival data from two independent patient cohorts. In the SU2C cohort, high DNPH1 expression was significantly associated with poorer OS (log-rank P = 0.00012, Figure S4A). Similarly, in the MSKCC cohort, high DNPH1 expression was associated with markedly reduced DFS (log-rank P = 0.037, Figure S4B), highlighting its potential utility as a prognostic biomarker. Fig. 8. Fig. 8 [107]Open in a new tab Proposed model of the hsa_circ_0038737/IGF2BP3/DNPH1 regulatory axis in mediating PARP inhibitor resistance in CRPC. Hsa_circ_0038737 is generated through Alu element-mediated back-splicing, a process facilitated by the RNA-binding protein HNRNPDL. Once formed, nuclear-localized hsa_circ_0038737 interacts with IGF2BP3 via its KH domains, stabilizing DNPH1 mRNA. Elevated DNPH1 expression enhances DNA damage repair capacity, thereby reducing the efficacy of PARP inhibitors in CRPC Fig. 5. [108]Fig. 5 [109]Open in a new tab DNPH1 Enhances Proliferation and Confers Resistance to PARP Inhibitors in Prostate Cancer Cells. (A-D) CCK-8 and colony formation assays were performed to evaluate the effects of DNPH1 overexpression or knockdown on the proliferation of 22RV1 and PC-3 cells. (E-F) IC50 assays were conducted to determine the sensitivity of 22RV1 and PC-3 cells to olaparib and niraparib following DNPH1 knockdown or overexpression. (G-H) Comet assays were used to assess DNA damage levels in 22RV1 and PC-3 cells with DNPH1 knockdown or overexpression, as well as in cells treated with the DNPH1 inhibitor N6-benzyladenosine. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance Given the established role of DNPH1 in maintaining nucleotide pool balance and facilitating DNA repair, we investigated whether it contributes to resistance against PARPi, a class of drugs increasingly used in CRPC therapy. IC₅₀ assays showed that DNPH1 knockdown significantly increased cellular sensitivity to olaparib and niraparib, whereas its overexpression conferred resistance to both agents (Fig. [110]5E, F), suggesting that DNPH1 may function as a modulator of therapeutic response to PARPi. To further elucidate whether DNPH1 influences DNA damage levels, we performed alkaline comet assays. DNPH1 depletion significantly increased DNA strand breaks, while its overexpression reduced DNA damage under the same conditions. Importantly, pharmacologic inhibition of DNPH1 using N6-benzyladenosine phenocopied the effects of gene knockdown, leading to increased DNA damage accumulation (Fig. [111]5G, H), and further supporting its role in DNA repair facilitation. Together, our results demonstrate that DNPH1 promotes PCa cell proliferation and plays a critical role in modulating sensitivity to PARP inhibition by attenuating DNA damage. These findings provide mechanistic insight into how DNPH1 may contribute to therapeutic resistance in CRPC and highlight it as a potential predictive biomarker and therapeutic target for combinatorial strategies with PARPis. Hsa_circ_0038737 induces PARP inhibitor resistance through a DNPH1-Dependent mechanism To directly test the hypothesis that hsa_circ_0038737 modulates sensitivity to PARPis, we conducted a series of in vitro drug response experiments in 22RV1 and PC-3 prostate cancer cell lines. IC₅₀ assays revealed that knockdown of hsa_circ_0038737 significantly enhanced cellular sensitivity to PARPis, including olaparib and niraparib, whereas overexpression of hsa_circ_0038737 conferred marked resistance to both agents (Fig. [112]6A, B). Additionally, co-treatment with DNPH1 inhibitor (DNPH1i) reversed the PARPi resistance induced by hsa_circ_0038737 overexpression. DNPH1 knockdown in two prostate cancer cells abolished the effect of DNPH1i on proliferation (Figure S4C, D), indicating that the compound acts primarily through DNPH1 inhibition rather than off-target mechanisms. Fig. 6. [113]Fig. 6 [114]Open in a new tab Hsa_circ_0038737 Confers Resistance to PARP Inhibitors and Can Be Functionally Antagonized by DNPH1 Inhibition. (A-B) IC50 assays were performed to evaluate the sensitivity of 22RV1 and PC-3 cells to olaparib and niraparib following hsa_circ_0038737 knockdown or overexpression, with or without co-treatment with DNPH1 inhibitor (DNPH1i). (C-D) Comet assays were conducted to assess DNA damage levels in 22RV1 and PC-3 cells subjected to hsa_circ_0038737 knockdown or overexpression, with or without DNPH1i co-treatment. (E-F) γ-H2AX IF assays were carried out to evaluate DNA damage in 22RV1 and PC-3 cells with hsa_circ_0038737 knockdown or overexpression, with or without DNPH1i co-treatment. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance Given our earlier findings that hsa_circ_0038737 stabilizes DNPH1 mRNA via IGF2BP3, and the role of DNPH1 in DNA repair, we next examined whether pharmacologic inhibition of DNPH1 could rescue the drug-resistant phenotype induced by hsa_circ_0038737 overexpression. Strikingly, co-treatment with a DNPH1 inhibitor (DNPH1i, N6-benzyladenosine) effectively reversed the resistance to PARPi conferred by hsa_circ_0038737 overexpression, restoring drug sensitivity to near-baseline levels. To investigate the mechanistic basis of this effect, we performed alkaline comet assays to quantify DNA strand breaks. Knockdown of hsa_circ_0038737 led to a pronounced increase in DNA damage, while its overexpression significantly reduced DNA damage accumulation. Notably, co-treatment with DNPH1i in hsa_circ_0038737-overexpressing cells restored DNA damage levels, confirming that DNPH1 activity is functionally required for the DNA repair advantage conferred by hsa_circ_0038737 (Fig. [115]6C, D). These findings were further validated by γ-H2AX immunofluorescence staining, a marker of double-strand DNA breaks. Consistent with the comet assay results, γ-H2AX signal intensity increased significantly upon hsa_circ_0038737 knockdown, and was reduced upon overexpression. Importantly, DNPH1 inhibition in hsa_circ_0038737-overexpressing cells reactivated DNA damage, as evidenced by elevated γ-H2AX foci formation (Fig. [116]6E, F). In circRNA-overexpressing 22RV1 and PC-3 cells, combined DNPH1i/PARPi treatment significantly increased PARP1 retention in the chromatin-bound fraction, mirroring the effect of PARPi monotherapy in circRNA-knockdown cells (Figure S5A, B). Concurrent PLA analysis revealed pronounced γ-H2AX/PARP1 co-localization foci in both: (1) circRNA-overexpressing cells treated with DNPH1i + PARPi and (2) circRNA-depleted cells receiving PARPi alone (Figure S5C, D), indicating amplified DNA damage signaling. Collectively, these results reveal that hsa_circ_0038737 plays a critical role in modulating the DNA damage response and confers cellular resistance to PARP inhibition via a DNPH1-dependent mechanism. Therapeutically, this resistance can be effectively overcome through pharmacologic targeting of DNPH1, highlighting a potential combinatorial strategy to resensitize resistant tumors to PARPi and improve treatment outcomes in castration-resistant prostate cancer. BRCA mutation enhances DNPH1i-Mediated cytotoxicity in PARPi-Resistant cells To further investigate the therapeutic potential of DNPH1 inhibition in BRCA-mutated contexts, we established BRCA-knockdown models in 22RV1 and PC-3 cell lines. Western blot analysis confirmed that knock down either BRCA1 or BRCA2 did not alter DNPH1 protein expression (Figure S6A, B), demonstrating that DNPH1 regulation occurs independently of BRCA status. Functionally, in both BRCA1- and BRCA2-deficient models, combined PARPi and DNPH1i treatment significantly enhanced suppression of cell proliferation compared to PARPi monotherapy (Figure S6C, D). This increased sensitivity was consistently observed in both prostate cancer cell lines irrespective of whether BRCA1 or BRCA2 was depleted, indicating that DNPH1 inhibition can potentiate PARPi efficacy even in BRCA-deficient settings. Collectively, these results demonstrate that the PARPi/DNPH1i combination remains effective in both BRCA-mutated and wild-type models, with DNPH1 modulation operating independently of BRCA status, supporting the broader therapeutic implications of our findings. Hsa_circ_0038737 Promotes Tumor Growth and Mediates PARP Inhibitor Resistance via DNPH1 in Vivo. To validate the oncogenic role of hsa_circ_0038737 and its impact on PARPi sensitivity in vivo, we established subcutaneous xenograft models using 4-week-old immunodeficient nude mice. Mice were inoculated with 22RV1 prostate cancer cells stably expressing either hsa_circ_0038737 overexpression, knockdown constructs, or control vectors. Ten days post-implantation, mice were randomized to receive vehicle control, olaparib monotherapy, or olaparib combined with the DNPH1i. Tumor growth analysis revealed that overexpression of hsa_circ_0038737 significantly accelerated tumor progression, whereas circRNA knockdown markedly suppressed tumor growth, with no observable difference in body weight across groups, indicating minimal systemic toxicity (Fig. [117]7A-D). Notably, hsa_circ_0038737-overexpressing tumors exhibited resistance to olaparib, while co-treatment with DNPH1i restored drug sensitivity, resulting in significant tumor growth inhibition. Conversely, xenografts with hsa_circ_0038737 knockdown were more responsive to olaparib, further supporting the role of this circRNA in modulating PARPi efficacy. IHC staining of tumor tissues demonstrated that tumors in the overexpression group exhibited elevated levels of DNPH1 and Ki-67, consistent with enhanced proliferation and DNA repair capacity. In contrast, tumors in the knockdown group displayed markedly reduced expression of these markers (Fig. [118]7E-G). Furthermore, to assess the clinical relevance of this regulatory axis, we analyzed human prostate cancer tissues and found a strong positive correlation between IGF2BP3 and DNPH1 expression (R²= 0.5679, P < 0.0001), supporting the existence of a conserved regulatory mechanism in patient samples (Fig. [119]7H, I). To further validate the translational significance of hsa_circ_0038737 in therapeutic resistance, we utilized patient-derived organoid (PDO) models generated from prostate cancer tissues with varying hsa_circ_0038737 expression levels (Figure S7 A, B). Organoids derived from high hsa_circ_0038737 expressing tumors were resistant to olaparib, but this resistance was substantially reversed by co-treatment with DNPH1i. In contrast, PDOs from low hsa_circ_0038737 expressing tumors displayed robust sensitivity to olaparib monotherapy (Fig. [120]7J, K). Furthermore, we performed IHC on tumor specimens from patients with high versus low hsa_circ_0038737 expression (Figure S8A) and observed concordant upregulation or downregulation of HNRNPDL, IGF2BP3, and DNPH1, consistent with our proposed regulatory axis (Figure S8B-D). Taken together, these in vivo and ex vivo findings robustly demonstrate that hsa_circ_0038737 promotes prostate tumor growth and mediates resistance to PARP inhibition through DNPH1 stabilization. Importantly, this resistance is pharmacologically reversible via DNPH1 inhibition, offering a promising therapeutic avenue for overcoming circRNA-driven treatment resistance in advanced prostate cancer. Alu elements and HNRNPDL cooperatively mediate the circularization and biogenesis of hsa_circ_0038737 The biogenesis of circRNAs is often facilitated by reverse-complementary Alu elements located within flanking intronic regions, which promote back-splicing through the formation of RNA duplex structures [[121]24, [122]25]. To explore whether this mechanism underlies the formation of hsa_circ_0038737, we examined the genomic architecture of its parental gene GTF3C1 using the UCSC Genome Browser. We identified two highly complementary Alu repeats (AluSp and AluSq2) positioned in the intronic regions flanking exons 35 and 36, the exonic components of hsa_circ_0038737 (Figure S9A). To functionally assess the contribution of these Alu elements to circRNA formation, we designed and constructed four minigene plasmid sequences: #1 Exon 35–36 and full-length flanking introns containing both AluSp and AluSq2; #2 Deletion of AluSp only; #3 Deletion of AluSq2 only; #4 Deletion of both Alu elements (Figure S9B). These constructs were transfected into 22RV1 and PC-3 cells, and the circularization efficiency of hsa_circ_0038737 was assessed via qRT-PCR. The results revealed that deletion of either Alu element significantly impaired circRNA formation, and removal of both elements resulted in the most pronounced reduction (Figure S9C), confirming AluSp and AluSq2 are essential cis-regulatory elements for the efficient circularization of hsa_circ_0038737. Given emerging evidence that RBPs can facilitate circRNA biogenesis by interacting with Alu elements [[123]25, [124]26], we utilized RBPmap to predict candidate RBPs that may bind to the Alu-containing intronic regions of hsa_circ_0038737. Among the candidates, HNRNPDL emerged as the only RBP predicted to bind both AluSp and AluSq2 simultaneously (Figure S9D). To test its functional involvement, we transfected PCa cells with shRNA targeting HNRNPDL, and observed a significant reduction in hsa_circ_0038737 expression, indicating that HNRNPDL likely facilitates its biogenesis (Figure S9E, F). Interestingly, DNPH1 expression was also downregulated upon HNRNPDL silencing, suggesting a regulatory cascade whereby HNRNPDL promotes circRNA formation, which in turn stabilizes DNPH1 mRNA via IGF2BP3. To further support this regulatory link, we analyzed RNA-seq data from multiple prostate cancer cohorts. A statistically significant positive correlation between HNRNPDL and DNPH1 mRNA levels was observed (Figure [125]S9G), consistent with the hypothesis that HNRNPDL indirectly controls DNPH1 expression through modulation of circRNA biogenesis. Overall, our data identify AluSp and AluSq2 as critical intronic elements and HNRNPDL as a key trans-acting factor that cooperatively facilitate the circularization of hsa_circ_0038737. This, in turn, impacts downstream gene regulation via the hsa_circ_0038737-IGF2BP3-DNPH1 axis, highlighting an additional layer of post-transcriptional control with therapeutic implications in prostate cancer. Comprehensive validation of MYC-Mediated transcriptional control of HNRNPDL Through integrative bioinformatic analyses utilizing six independent platforms (FIMO_JASPAR, PWMEnrich_JASPAR, ChIP-Atlas, ENCODE, CHEA, and GTRD), we identified MYC as the highest-ranking transcription factor candidate regulating HNRNPDL expression (Figure S10A). Cross-platform validation identified evolutionarily conserved MYC binding sites within the proximal promoter region of HNRNPDL (Figure S10B), with the canonical MYC motif < CACGTG > detected as the predominant binding signature (Figure [126]S10C). To experimentally validate this regulatory interaction, we performed chromatin immunoprecipitation quantitative PCR (ChIP-qPCR), which confirmed a significant enrichment of MYC occupancy at the HNRNPDL promoter compared to IgG controls in both 22Rv1 and PC-3 cell lines (Figure S10D). Functionally, MYC knockdown led to a Marked reduction in HNRNPDL protein levels in 22Rv1 and PC-3 cells (Figure S10E, F), while MYC overexpression resulted in a corresponding increase in HNRNPDL expression. These findings establish a bidirectional regulatory relationship, demonstrating that MYC acts as a direct transcriptional activator of HNRNPDL, thereby contributing to prostate cancer pathogenesis. Discussion Our study reveals hsa_circ_0038737 as a critical oncogenic circRNA in prostate cancer that exerts its effects by forming a functional complex with IGF2BP3 to stabilize DNPH1 mRNA, thereby enhancing DNA repair capacity and promoting resistance to PARP inhibitors. The biogenesis of hsa_circ_0038737 is tightly controlled by Alu element-mediated circularization and the RNA-binding protein HNRNPDL, adding a new layer to our understanding of circRNA regulation. Importantly, the pharmacologic inhibition of DNPH1 effectively reverses hsa_circ_0038737-induced PARPi resistance in both cell line-based models and patient-derived organoids model (Fig. [127]8). Our findings uncover a novel circRNA/RBP-mediated regulatory axis, hsa_circ_0038737/IGF2BP3/DNPH1, that promotes CRPC progression and therapeutic resistance, offering new insights into a rationale for combinatorial treatment strategies and identifying a potential combinatorial therapeutic strategy to overcome PARPi resistance. Recent studies have increasingly emphasized the functional versatility of circRNAs in cancer progression, particularly their ability to act as scaffolds for RBPs to regulate mRNA stability and translation [[128]18]. For instance, circITGB6 was recently shown to enhance cisplatin resistance in ovarian cancer by binding to IGF2BP2 and stabilizing FGF9 mRNA [[129]27]. Similarly, our work demonstrates that hsa_circ_0038737 interacts with IGF2BP3 to stabilize DNPH1, a nucleotide sanitizer critical for maintaining genomic integrity under PARPi-induced DNA damage. These parallels underscore a conserved mechanism by which circRNAs exploit RBP partnerships to drive therapy resistance across malignancies. First, we identified hsa_circ_0038737 as a circRNA significantly upregulated in PCa tissues and associated with aggressive clinicopathological features, including higher T stage and Gleason scores. Its nuclear localization and resistance to RNase R digestion align with previous reports demonstrating the stability and functional versatility of circRNAs in cancer progression [[130]28]. Notably, hsa_circ_0038737 promoted PCa cell proliferation and G2/M phase progression, independent of its linear counterpart GTF3C1. These observations reinforce the emerging paradigm that circRNAs can exert oncogenic effects distinct from their parental genes [[131]29]. Recent work by Cheng et al. further supports this notion, showing that METTL3-mediated m6A modification of circGLIS3 drives prostate cancer progression by sequestering miR-661 and activating the MDM2-p53 axis [[132]19]. Our findings expand this landscape by linking circRNA activity to PARPi resistance through DNPH1-mediated DNA repair, a pathway not previously associated with circRNA regulation. Mechanistically, we revealed that hsa_circ_0038737 interacts with IGF2BP3, an RBP known to enhance mRNA stability [[133]21]. This interaction stabilizes DNPH1 mRNA, a nucleotide sanitizer implicated in DNA repair and PARPi resistance [[134]22]. Our RNA-seq and functional assays demonstrated that DNPH1 upregulation reduces DNA damage and confers resistance to olaparib and niraparib, consistent with prior studies linking DNPH1 to PARPi sensitivity [[135]23]. Importantly, the hsa_circ_0038737/IGF2BP3 axis represents a previously unrecognized regulatory layer in PARPi resistance, expanding the repertoire of circRNA-mediated mechanisms in therapy evasion [[136]14, [137]30]. The reversal of PARPi resistance by DNPH1 inhibition underscores the therapeutic potential of targeting this pathway. Recent advancements in DNPH1 inhibitor development, such as N6-benzyladenosine derivatives, have shown promise in sensitizing BRCA-deficient tumors to PARPi in preclinical models [[138]22], aligning with our findings and suggesting translational relevance for combinatorial therapies in CRPC. Furthermore, we identified Alu elements flanking hsa_circ_0038737 as critical drivers of its biogenesis, with HNRNPDL facilitating this process. This aligns with established models of circRNA formation involving complementary Alu sequences and RBPs [[139]31, [140]32]. The correlation between HNRNPDL and DNPH1 expression in clinical cohorts suggests a broader regulatory network Linking circRNA biogenesis to DNA repair pathways. These findings deepen our understanding of circRNA generation and its functional implications in cancer. A 2023 study by Rzechorzek et al. further elucidated the enzymatic mechanism of DNPH1 in hydrolyzing damaged nucleotides, providing structural insights into its role in mitigating PARPi-induced DNA lesions [[141]22]. Our work complements these discoveries by positioning DNPH1 within a circRNA-driven regulatory circuit, highlighting the interplay between non-coding RNA biogenesis and enzymatic DNA repair processes. Clinically, our in vivo xenograft and patient-derived organoid models demonstrated that hsa_circ_0038737 overexpression promotes tumor growth and PARPi resistance, while DNPH1 inhibition restores drug sensitivity. The positive correlation between IGF2BP3 and DNPH1 in patient tissues further supports the translational relevance of this axis. These data suggest that hsa_circ_0038737 expression could serve as a biomarker for predicting PARPi response, while combinatorial targeting of DNPH1 may enhance PARPi efficacy in CRPC patients. Recent clinical trials, such as the PROfound study [[142]33], have established PARPi as a standard therapy for HRR-deficient mCRPC; however, resistance remains a major limitation. Our findings propose a strategy to extend PARPi utility by co-targeting DNPH1, particularly in tumors with elevated hsa_circ_0038737 expression. However, this study has limitations. The sample size of clinical tissues and organoids remains modest, necessitating validation in larger cohorts. Additionally, the functional role of hsa_circ_0038737 in other types of cancer and its potential as a universal biomarker for cancer progression need to be explored in future studies. Recent advances in spatial transcriptomics and single-cell sequencing could further resolve the heterogeneity of circRNA expression within tumor microenvironments [[143]18], offering insights into context-dependent resistance mechanisms. Finally, while DNPH1 inhibitors showed efficacy in preclinical models, their pharmacokinetic and safety profiles in humans require further evaluation. These limitations, however, do not diminish the core findings but highlight directions for future research. Conclusion In conclusion, this study identifies a novel hsa_circ_0038737–IGF2BP3–DNPH1 regulatory axis as a critical driver of PARP inhibitor resistance in castration-resistant prostate cancer (CRPC). By elucidating how circRNA-mediated stabilization of DNA repair–related transcripts contribute to therapeutic escape, our findings expand the current understanding of non-coding RNA function in drug resistance. The demonstrated reversal of resistance via DNPH1 inhibition highlights a promising avenue for combination therapies targeting circRNA-protein interactions. These insights lay the groundwork for future translational studies aimed at optimizing DNPH1-targeted compounds and validating their clinical utility in enhancing PARPi efficacy in advanced prostate cancer. Supplementary Information [144]Supplementary Material 1.^ (192.5KB, pdf) [145]12943_2025_2447_MOESM2_ESM.pdf^ (135.8KB, pdf) Supplementary Material 2: Mapping of the binding domains of IGF2BP3 for hsa_circ_0038737. (A-B) Schematic diagrams illustrate the constructed plasmids containing different fragments of FLAG-tagged IGF2BP3. (C) RIP experiments were performed using FLAG antibody to identify the domains of IGF2BP3 that interact with hsa_circ_0038737 in 22RV1 and PC-3 cells. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance. [146]12943_2025_2447_MOESM3_ESM.pdf^ (342.2KB, pdf) Supplementary Material 3: IGF2BP3 function and prognosis in prostate cancer. (A-B) IGF2BP3 was knocked down and overexpressed in 22RV1 and PC-3 cell lines. (C-D) CCK-8 assays were performed to evaluate the effects of IGF2BP3 modulation on cell proliferation in 22RV1 and PC-3 cells. (E-F) Colony formation assays were conducted to assess the impact of IGF2BP3 modulation on clonogenic survival in 22RV1 and PC-3 cells, with corresponding quantification of colony numbers. (G-H) Kaplan-Meier survival analysis was conducted using data from the SU2C and MSKCC cohorts to assess the relationship between IGF2BP3 expression levels and overall survival (OS) or disease-free survival (DFS). (I) Western Blotting was conducted to verify whether the expression changes of hsa_circ_0038737 affect the protein levels of IGF2BP3 [147]Supplementary Material 4.^ (140.3KB, pdf) [148]12943_2025_2447_MOESM5_ESM.pdf^ (302.3KB, pdf) Supplementary Material 5 : IGF2BP3 function and prognosis in prostate cancer. (A-B) IGF2BP3 was knocked down and overexpressed in 22RV1 and PC-3 cell lines. (C-D) CCK-8 assays were performed to evaluate the effects of IGF2BP3 modulation on cell proliferation in 22RV1 and PC-3 cells. (E-F) Colony formation assays were conducted to assess the impact of IGF2BP3 modulation on clonogenic survival in 22RV1 and PC-3 cells, with corresponding quantification of colony numbers. (G-H) Kaplan-Meier survival analysis was conducted using data from the SU2C and MSKCC cohorts to assess the relationship between IGF2BP3 expression levels and overall survival (OS) or disease-free survival (DFS). (I) Western Blotting was conducted to verify whether the expression changes of hsa_circ_0038737 affect the protein levels of IGF2BP3IGF2BP3 function and prognosis in prostate cancer. (A-B) IGF2BP3 was knocked down and overexpressed in 22RV1 and PC-3 cell lines. (C-D) CCK-8 assays were performed to evaluate the effects of IGF2BP3 modulation on cell proliferation in 22RV1 and PC-3 cells. (E-F) Colony formation assays were conducted to assess the impact of IGF2BP3 modulation on clonogenic survival in 22RV1 and PC-3 cells, with corresponding quantification of colony numbers. (G-H) Kaplan-Meier survival analysis was conducted using data from the SU2C and MSKCC cohorts to assess the relationship between IGF2BP3 expression levels and overall survival (OS) or disease-free survival (DFS). (I) Western Blotting was conducted to verify whether the expression changes of hsa_circ_0038737 affect the protein levels of IGF2BP3 [149]12943_2025_2447_MOESM6_ESM.pdf^ (1MB, pdf) Supplementary Material 6: Molecular and functional analyses in BRCA-deficient prostate cancer models. (A-B) Western blot analysis of DNPH1 protein expression in (A) BRCA1-knockdown and (B) BRCA2-knockdown 22RV1 and PC-3 cells using two independent shRNAs (ShBRCA1-1/ShBRCA1-2; ShBRCA2-1/ShBRCA2-2) with non-targeting shRNA control (NC); GAPDH serves as loading control. (C-D) Cell proliferation assay in BRCA1-knockdown 22RV1 and PC-3 cells under four conditions: NC, BRCA1-targeting shRNA (ShBRCA1), ShBRCA1 with PARP inhibitor (PARPi), and ShBRCA1 with both PARPi and DNPH1 inhibitor (DNPH1i) [150]12943_2025_2447_MOESM7_ESM.pdf^ (5.5MB, pdf) Supplementary Material 7: Immunohistochemical analysis of AR and PSMA in patient-derived organoids from CRPC patients. (A-B) Immunohistochemical staining was performed to detect AR and PSMA expression in patient-derived organoids established from CRPC tissues [151]12943_2025_2447_MOESM8_ESM.pdf^ (3.5MB, pdf) Supplementary Material 8: Immunohistochemical assessment of protein expression in prostate cancer tissues stratified by Hsa_circ_0038737 levels. (A) Representative immunohistochemistry staining images of DNPH1, HNRNPDL, and IGF2BP3 in CRPC tissues with low or high Hsa_circ_0038737 expression. (B-D) Immunoreactive scores of (B) HNRNPDL, (C) IGF2BP3, and (D) DNPH1 protein expression. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns, no significance [152]12943_2025_2447_MOESM9_ESM.pdf^ (1MB, pdf) Supplementary Material 9: Role of Alu elements and HNRNPDL in hsa_circ_0038737 biogenesis. (A) The UCSC Genome Browser was used to identify two highly reverse-complementary Alu elements (AluSp and AluSq2) in the flanking introns of hsa_circ_0038737. (B) Four plasmid constructs were generated: #1 with both Alu elements, #2 lacking AluSp, #3 lacking AluSq2, and #4 lacking both elements, with an empty vector as a control. (C) qRT-PCR analysis was performed after transfecting these plasmids into 22RV1 and PC-3 cells to measure the circularization efficiency of hsa_circ_0038737. (D) RBPmap prediction was conducted to identify RBPs binding to both Alu elements. (E-F) HNRNPDL was knocked down using shRNA, and the effects on hsa_circ_0038737 and DNPH1 levels were assessed. (G) RNA-seq data from multiple prostate cancer cohorts were analyzed to examine the correlation between HNRNPDL and DNPH1 mRNA levels. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 [153]12943_2025_2447_MOESM10_ESM.pdf^ (880.8KB, pdf) Supplementary Material 10: Analysis of MYC-mediated transcriptional regulation of HNRNPDL. (A) Bioinformatics prediction of MYC binding sites within the HNRNPDL promoter region using integrated public ChIP-seq datasets. (B) Publicly available MYC ChIP-seq peak profile at the HNRNPDL promoter region. (C) Consensus MYC binding motif identified from ChIP-seq data. (D) Chromatin immunoprecipitation quantitative PCR validation of MYC occupancy at the HNRNPDL promoter in 22RV1 and PC-3 prostate cancer cells; data presented as fold enrichment relative to input DNA. (E-F) Western blot analysis of HNRNPDL protein expression in (E) 22RV1 and (F) PC-3 cells under MYC modulation: non-targeting shRNA control (NC), MYC knockdown with two independent shRNAs (MYC Sh1/Sh2), empty vector control (Vector), and MYC overexpression (MYC OE); GAPDH serves as loading control [154]Supplementary Material 11.^ (11.5KB, docx) [155]Supplementary Material 12.^ (23.5KB, xlsx) [156]Supplementary Material 13.^ (2.2MB, xlsx) Acknowledgements