Abstract Background Cancer-associated fibroblasts (CAFs) have been reported to play a significant role in the development and metastasis of various tumors; however, research on their role in promoting prostate cancer (PCa) metastasis under castration conditions remains unclear. Methods In this study, we utilized quantitative reverse transcription polymerase chain reaction (qRT-PCR) to detect the expression differences of microRNA-196b-5p (miR-196b-5p) in the exosomes secreted by CAFs before and after castration. We further characterized the transcriptional regulatory landscape through RNA sequencing combined with bioinformatics databases. In vitro and in vivo experiments were conducted to determine the role of miR-196b-5p in promoting tumor migration and metastasis. The dual-luciferase reporter assay, RT-PCR analysis, and Western blot analysis confirmed that miR-196b-5p targets HOXC8 in prostate cancer. Additionally, transwell assays and Western blot analysis were performed to elucidate the role and specific mechanisms of HOXC8 in tumor metastasis. Results By analyzing the expression differences of miRNAs in the exosomes secreted by CAFs before and after castration, along with relevant data from databases, we found that miR-196b-5p is highly secreted by CAFs after castration. miR-196b-5p promotes the migration and metastasis of prostate cancer cells. Subsequently, through RNA sequencing analysis and experimental validation, we determined that miR-196b-5p targets HOXC8. This interaction activates the NF-κB pathway, leading to the upregulation of epithelial-mesenchymal transition (EMT)-related protein expression, thereby driving the metastasis of prostate cancer. Conclusions Our study elucidates a specific mechanism by which CAF-derived exosomes promote prostate cancer metastasis via miR-196b-5p regulation, contributing to the identification of therapeutic targets for managing tumor metastasis following castration. Supplementary Information The online version contains supplementary material available at 10.1186/s13062-025-00667-2. Keywords: Prostate cancer, Exosome, Cancer-associated fibroblasts, miR-196b-5p, HOXC8, NF-κB Background Currently, prostate cancer (PCa) is the most common male malignancy worldwide and the second leading cause of cancer-related deaths in men [[44]1, [45]2]. The majority of PCa cases are classified as adenocarcinomas, characterized by glandular formation, luminal differentiation, and androgen receptor (AR) expression. Androgen deprivation therapy (ADT) is a primary treatment for PCa; however, nearly all patients experience relapse after an initial response, leading to the development of castration-resistant prostate cancer (CRPC) [[46]3]. CRPC is often associated with metastasis and results in poorer prognosis. Recently, increasing evidence underscores the critical role of the tumor microenvironment (TME) and cancer-associated fibroblasts (CAFs) in various cancers [[47]4–[48]6]. In prostate cancer, CAFs play a crucial role in the development of castration resistance; however, the role and mechanisms by which CAFs influence the progression and metastasis of PCa under castration conditions remain unclear [[49]6, [50]7]. In our previous study, we found that exosomes derived from CAFs significantly enhance the metastatic capabilities of PCa cells in a castration environment compared to an androgen-rich environment [[51]8]. Exosomes are extracellular vesicles that can transfer miRNAs, mRNAs, lncRNAs, and proteins between cells, participating in altering the tumor microenvironment, growth, and progression, thereby serving as key regulators of cellular signaling [[52]9–[53]11]. Numerous studies have reported that exosomes derived from CAFs can promote tumor progression [[54]12–[55]15]. Therefore, our research aims to investigate whether exosomes from CAFs post-ADT influence the progression and metastasis of PCa. Our preliminary study revealed that there were no significant changes in the quantity of CAF-derived exosomes after ADT [[56]8]. Consequently, we focused on the components within these exosomes, particularly the most abundant miRNAs. Reanalyzing the miRNA sequencing data from CAF-derived exosomes, We observed a significant elevation of miR-196b-5p in exosomes derived from post-ADT CAFs (Fig. [57]S1A). miR-196b-5p has been reported to promote cell migration and invasion in various malignancies [[58]16–[59]20]; however, its role in PCa remains underexplored. This finding sparked our great interest. We transfected PCa cells with miR-196b-5p mimics and demonstrated that overexpression of miR-196b-5p significantly enhanced the epithelial-mesenchymal transition (EMT), migration, and invasion capabilities of PCa cells. We identified homeobox C8 (HOXC8) as a target gene of miR-196b-5p in PCa cells. HOXC8 encodes a transcription factor containing a homeodomain, and its dysregulation is associated with various cancers [[60]21–[61]24]; however, its role in PCa metastasis remains unexplored. Further in vitro experiments revealed that HOXC8 inhibits EMT by suppressing the activation of the NF-κB pathway. These findings suggest that miR-196b-5p from CAFs promotes cancer cell metastasis through targeting the HOXC8/NF-κB axis. Our results reveal a novel mechanism by which CAFs facilitate tumor metastasis following ADT, highlighting the need for further development of miRNA-based therapies for metastatic castration-resistant prostate cancer (mCRPC). Materials and methods Isolation of primary fibroblasts Neoplastic and normal prostate tissues were obtained from PCa patients treated with radical prostatectomy and patients without PCa undergoing total cystectomy, respectively, at the Department of Urology, Shanghai General Hospital. The method for isolating primary fibroblasts followed previously described protocols [[62]25]. Briefly, prostate tissues were initially minced and subjected to enzymatic digestion with hyaluronidase and collagenase for one hour. Fibroblasts were subsequently isolated via discontinuous gradient centrifugation. Cell culture hTERT PF179T CAF and NCI-H660 cell lines were purchased from American Type Culture Collection (ATCC, USA), while LNCaP, DU145, HEK 293T, PC3, RWPE-1, VCaP, and C4-2B cell lines were obtained from Cell Bank of Shanghai Institute of Cells, Chinese Academy of Science (Shanghai, China). All cell lines were authenticated by (GENEWIZ, China). The isolated primary fibroblasts, hTERT PF179T CAFs, DU145, and HEK 293T cells were cultured in DMEM medium (Gibco, USA) supplemented with 10% fetal bovine serum (FBS, Gibco, USA) and 1% penicillin-streptomycin (Hyclone, USA, Cat# SV30010). LNCaP, VCaP, and C4-2B cells were cultured in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin-streptomycin. NCI-H660 cell line was cultured in RPMI-1640 medium supplemented with 5% FBS, 0.005 mg/mL insulin, 0.01 mg/mL transferrin, 30 nM sodium selenite, 10 nM hydrocortisone, 10 nM beta-estradiol, and 1% penicillin-streptomycin. PC3 cell line was maintained in F-12 K medium containing 10% FBS and 1% penicillin-streptomycin. RWPE-1 cell line was cultured in Keratinocyte-SFM medium (Invitrogen, USA, Cat# 17005-042), supplemented with 5 ng/mL human recombinant epidermal growth factor (EGF) and 0.05 mg/mL bovine pituitary extract (BPE). All cell lines were cultured at 37 °C in a 5% CO₂ incubator. Transfection The lentiviral plasmids encoding miR-196b-5p, HOXC8, and their negative controls were obtained from Genomeditech (Shanghai, Chian). Cells were infected with miR-196b-5p lentivirus according to the manufacturer’s instructions and were selected using puromycin. For HOXC8 lentivirus-infected cells, blasticidin was used for cell selection. The miR-196b-5p mimics, miR-196b-5p negative control, and miR-196b-5p inhibitor were synthesized by RiboBio (Guangzhou, China), with sequences listed in Table [63]S1. The siRNA for HOXC8 and the negative control were designed and synthesized by Genomeditech, with sequences available in Table [64]S2. For miRNA, siRNA, or miRNA transfection, Lipofectamine 3000 Transfection Reagent (Invitrogen, USA) was used according to the manufacturer’s protocol. Immunofluorescence Cells were seeded on coverglasses one day prior to staining. The following day, they were fixed with 4% paraformaldehyde at room temperature for 20 min and then permeabilized with 0.1% Triton X-100 at 4 °C for 15 min. The cells were then blocked with 5% donkey serum at room temperature for 2 h, followed by incubation overnight at 4 °C with primary antibodies: α-SMA (Abcam, UK, Cat# ab7817), FAP (ABclonal, China, Cat# A6349), or β-Actin (Servicebio, China, Cat# GB15001). The cells were then incubated in the dark at room temperature for 30 min with Alexa Fluor 647-conjugated antibody (Abcam, Cat# ab150075) or Alexa Fluor 488-conjugated antibody (Abcam, Cat# ab150109). DAPI (Beyotime, China) was used to stain the nuclei. Fluorescent microscopy (Leica, Germany) was used to observe the cells. Exosome isolation The extraction of exosome was performed as previously described [[65]8]. Briefly, cell culture supernatants were collected and sequentially centrifuged at 300 × g for 10 min, 2,000 × g for 10 min, and 10,000 × g for 30 min. The supernatants were then filtered through a 0.22 μm filter (Millipore, USA) and ultracentrifuged at 100,000 × g for 70 min at 4 °C. Pellets were resuspended in PBS, ultracentrifuged again at 100,000 × g for 70 min, resuspended in PBS, and stored at − 80 °C. Exosome release was inhibited using 20 µM GW4869 (Sigma Aldrich, USA). Dual-luciferase reporter assay Luciferase reporter assays were performed using luciferase reporter constructs containing the NC 3’UTR, wild-type HOXC8 3’UTR, or mutant HOXC8 3’UTR regions, synthesized by Genomeditech, with sequences available in Table [66]S3. In brief, 293T cells were seeded in 24-well plates and co-transfected with the luciferase reporter, followed by transfection with the mimic and luciferase vector. After 48 h, luciferase activity was measured with the dual-luciferase reporter assay kit (Genomeditech). RNA extraction and RT-qPCR Follow the previously mentioned steps to perform RNA extraction and conduct qRT-PCR for mRNA and miRNA. In brief, reverse transcription was performed using the PrimeScript RT reagent kit (Takara, Japan), followed by quantitative real-time PCR with TB Green Premix Ex Taq (Takara) on a QuantStudio 7 Flex system (Applied Biosystems, USA). mRNA levels were quantified using the ΔΔCt method, with β-actin serving as the reference for mRNA and U6 serving as the reference for miRNA. The primers used in this analysis are listed in Table [67]S4. Western blotting Total cellular proteins were extracted using M-PER (Thermo Scientific, USA) with inhibitor of protease and phosphatase (NCM Biotech, China, Cat# P002) and quantified using a BCA assay kit (Thermo Scientific). Approximately 30 micrograms of total protein were loaded onto SDS-PAGE gels and subsequently transferred to 0.22 μm nitrocellulose membranes (Cytiva, USA). The membranes were blocked at room temperature with 5% BSA for 1.5 h, followed by overnight incubation at 4 °C with primary antibodies. The primary antibodies included anti-E-cadherin (Cell Signaling Technology, USA, Cat# 14472 S), anti-N-cadherin (Proteintech, China, Cat# 66219), anti-Vimentin (Abcam, UK, Cat# ab92547), anti-Phospho-IκBα (Cell Signaling Technology, Cat# 2859), anti-IκBα (Cell Signaling Technology, Cat# 4812), anti-p65 (Cell Signaling Technology, Cat# 8242), anti-Histone H3 (Cell Signaling Technology, Cat# 9715), anti-Snail (Cell Signaling Technology, Cat# 3879 S), anti-Slug (Cell Signaling Technology, Cat# 9585 S), anti-HOXC8 (Proteintech, Cat# 15448), and anti-β-actin (Servicebio, China, Cat# GB15001). Secondary antibodies were applied at room temperature for 1 h the following day, followed by visualization. Cell viability test Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay. Ten microliters of CCK-8 reagent (NCM Biotech, China, Cat# C6005) were added to each well of a 96-well plate. The plate was incubated at 37 °C, 5% CO₂ incubator for 1.5 h, after which the absorbance was measured at 450 nm using a spectrophotometer. Transwell assay and wound healing assay For the Transwell migration assay, DU145, PC3, or LNCaP cells were seeded in the upper chamber containing serum-free medium, while the lower chamber was filled with medium supplemented with 10% FBS. After incubating the plates at 37 °C, 5% CO₂ incubator for 48 h, the cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. Images were then captured using a fluorescence microscope (Leica, Germany). DU145 or PC3 cells were seeded in six-well plates one day prior to the wound healing assay. After incubating for 24 h, the cells were washed with PBS, and a linear scratch wound was created. The plates were then incubated at 37 °C, 5% CO₂ for 24 h, and images were captured using a fluorescence microscope (Leica, Germany). Colony forming Cells were seeded in six-well plates at a density of 400 to 1,000 cells per well in each experimental group and incubated for 7 days. After incubation, cells were washed once with PBS, followed by the addition of 1 mL of 4% paraformaldehyde to fix the cells for 60 min. The cells were then washed again with PBS, and 1 mL of crystal violet solution was added to stain the cells for 15 min. After staining, the cells were washed several times with PBS and photographed. Xenograft models and bioluminescence Male BALB/c nude mice aged 4 to 6 weeks underwent orchiectomy. DU145 luciferase cells were injected via the tail vein. Tumor metastasis was monitored using a bioluminescent in vivo imaging system (IVIS, USA) at 4 weeks post-injection. Mice were anesthetized with 1.5% isoflurane in air and received an intraperitoneal injection of D-luciferin (200 µL, 15 mg/mL in PBS) prior to imaging. After 8 weeks post-tumor injection, the mice were euthanized, and lung tissues were excised for imaging. All animal procedures were conducted by laboratory animal guidelines and were approved by the Animal Care and Use Committee of Shanghai General Hospital. Immunohistochemistry Tumors from mice were collected and fixed in 4% paraformaldehyde, followed by dehydration using a gradient of ethanol concentrations. Subsequently, tumors were embedded in paraffin and processed for sectioning. Immunohistochemical staining was performed using anti-HOXC8 antibody (Proteintech, China, Cat# 15448), and the expression of HOXC8 in the specimens was quantified using the immunohistochemical IRS scoring system. Bioinformatic data acquisition Transcriptomic sequencing data and prognostic information for TCGA-PRAD were obtained from the TCGA database ([68]https://portal.gdc.cancer.gov/). Sequencing data for [69]GSE70770 and [70]GSE64333 were sourced from the GEO database ([71]https://www.ncbi.nlm.nih.gov/geo/) [[72]26, [73]27]. The expression of MIR196B in neuroendocrine prostate cancer (NEPC) versus castration-resistant prostate cancer (CRPC) was compared using Beltran’s dataset [[74]28]. Transcriptome sequencing and differential gene expression analysis Total RNA was isolated using TRIzol Reagent (Invitrogen, USA) following the manufacturer’s protocol. RNA purity and concentration were assessed using a NanoDrop 2000 spectrophotometer (Thermo Scientific, USA), while RNA integrity was evaluated with an Agilent 2100 Bioanalyzer (Agilent Technologies, USA). Transcriptomic analyses were performed by OE Biotech (Shanghai, China). Differential gene expression analysis was conducted using the DESeq2 package (Version 1.36.0) in R, with a significance threshold of P < 0.05 and|log2 fold change| > 1.” Statistical analysis Statistical analyses were conducted using Prism version 9.5.0. Differences between two groups were assessed using the Student’s t-test, with a P-value less than 0.05 considered statistically significant. Data are presented as mean ± standard deviation (SD). Results Expression of miR-196b-5p in primary CAFs and PCa cell lines We successfully isolated and cultured primary CAFs from three PCa samples, as well as primary normal fibroblasts from three different normal prostate samples. Immunofluorescence was utilized to validate the phenotype of the primary CAFs and primary normal fibroblasts. The expression of fibroblast activation protein-α (FAP) is a key characteristic of CAF activation [[75]29]. Notably, while detectable in activated fibroblasts participating in wound healing processes, FAP exhibits no expression in other healthy tissues [[76]30]. FAP-positive CAFs contribute to shaping an immunosuppressive tumor microenvironment by secreting various cytokines [[77]31, [78]32]. Currently, FAP is one of the most promising therapeutic targets in CAFs. The staining demonstrated that both the primary CAFs and the hTERT PF179T CAF had significantly higher FAP expression compared to primary normal fibroblasts, highlighting the activated fibroblast phenotype in these cells (Fig. [79]1A). Following total RNA extraction, we observed that miR-196b-5p expression was significantly elevated in both primary CAFs and the hTERT PF179T CAF compared to primary normal fibroblasts (Fig. [80]1B). Further analysis of baseline miR-196b-5p expression in multiple PCa cell lines and RWPE-1 cell line revealed that miR-196b-5p levels were significantly higher in various prostate cancer cell lines, especially in androgen-insensitive cell lines such as DU145, PC3 and NCI-H660 (Fig. [81]1C). Fig. 1. [82]Fig. 1 [83]Open in a new tab Identification of miR-196b-5p expression levels in primary fibroblasts and prostate cell lines. (A) Immunofluorescence staining of α-SMA and FAP in primary normal prostate fibroblasts, primary prostate CAFs, and hTERT PF179T CAFs (scale bar = 100 μm). (B) The expression levels of miR-196b-5p in primary normal prostate fibroblasts, primary prostate CAFs, and hTERT PF179T CAFs measured by RT-PCR. (C) The expression levels of miR-196b-5p in the normal prostate epithelial cell line RWPE-1 and various prostate cancer cell lines measured by RT-PCR. Data are presented as mean ± SD, representing triplicate measurements. (Student’s t-test, ***P < 0.001) miR-196b-5p significantly elevated in CAFs exosomes after ADT and transferred to PCa cells To investigate how castration conditions affect the secretion of miR-196b-5p by CAFs, we treated CAFs with media containing DHT as well as control media without DHT. The results showed that CAF-derived exosomes treated with ETOH had significantly higher miR-196b-5p levels compared to DHT-treated CAF-derived exosomes (Fig. [84]2A). We then treated DU145 and LNCaP cell lines with the aforementioned exosomes and observed that the ETOH-treated CAF-derived exosomes significantly elevated miR-196b-5p levels in PCa cells compared to DHT-treated CAF-derived exosomes (Fig. [85]2B). To confirm whether miR-196b-5p was transferred from CAFs to PCa cells via exosomes, we transiently transfected CAFs with Cy3-labeled miR-196b-5p and co-cultured them with DU145 cells for 24 h. Fluorescence microscopy revealed red signals (Cy3-labeled miR-196b-5p) and green signals (β-actin) in DU145, with the red signal disappearing after CAFs were treated with the exosome inhibitor GW4869 (Fig. [86]2C). Additionally, we extracted exosomes from CAFs transfected with Cy3-labeled miR-196b-5p as well as from control CAFs, and co-cultured them with DU145 cells for 24 h. The results showed that Cy3 signals was detectable in DU145 cells. And we obtained similar results in LNCaP cells (Fig. [87]S2A-B). These results indicate that CAFs secrete significant amounts of miR-196b-5p via exosomes under ADT, and transfer it to PCa cells. Fig. 2. [88]Fig. 2 [89]Open in a new tab Exosomes transfer miR-196b-5p from CAFs to PCa cells. (A) Expression of miR-196b-5p in exosomes from DHT-treated CAFs and matched ETOH-treated CAFs measured by RT-PCR. (B) Expression of miR-146a-5p in LNCaP and DU145 cells measured by RT-PCR at 24 h after treating with exosomes (25 µg/mL) derived from DHT-treated CAFs or ETOH-treated CAFs. (C) CAFs were transiently transfected with Cy3-labeled miR-control or miR-196b-5p and co-cultured with DU145 cells for 24 h following GW4869 treatment. Fluorescence microscopy was used to observe green signals (β-actin) and red signals (Cy3) in DU145 cells (scale bar = 50 μm). (D) Exosomes were isolated from the supernatant of CAFs transfected with Cy3-labeled miR-196b-5p or miR-control, and exosomes (25 µg/mL) were incubated with DU145 cells for 24 h. Fluorescence microscopy was used to detect green signals (β-actin) and red signals (Cy3) in DU145 cells (scale bar = 50 μm). Data are presented as mean ± SD, representing triplicate measurements. (Student’s t-test, **P < 0.01, ***P < 0.001, ****P < 0.0001) Transcriptome analysis revealed an elevated expression of miR-196b-5p in PCa samples We conducted bioinformatics analysis to assess the expression levels of miR-196b-5p across multiple PCa datasets. The results indicated that tumor samples in the TCGA-PRAD database exhibited significantly higher levels of miR-196b-5p expression compared to normal samples (Fig. [90]3A). Similarly, the [91]GSE64333 dataset also indicated that miR-196b (precursor of miR-196b-5p) levels were significantly elevated in tumor samples (Fig. [92]3B). Additionally, the [93]GSE70770 dataset, which includes CRPC samples, showed that the expression of miR-196b was significantly higher in CRPC samples than in HSPC samples (Fig. [94]3C). Furthermore, in the [95]GSE70770 dataset, the expression level of miR-196b was correlated with the patients’ pathological staging, showing higher expression in samples from patients at the T3-T4 stage compared to those at the T2 stage (Fig. [96]3D). Although not statistically significant (p = 0.067), the high miR-196b-5p expression group was associated with poorer biochemical recurrence-free survival (BCRFS) based on the TCGA-PRAD dataset (Fig. [97]S1B). Interestingly, in the Beltran (2016) dataset, the levels of miR-196b were significantly higher in NEPC samples compared to those in CRPC, suggesting that miR-196b-5p may play a role not only in the progression from HSPC to CRPC but also potentially from CRPC to NEPC. Fig. 3. [98]Fig. 3 [99]Open in a new tab Expression landscape of miR-196b-5p across multiple prostate cancer datasets. (A) Differential expression of miR-196b-5p between tumor and normal samples from the TCGA-PRAD database. (B) Analysis of differential expression of miR-196b in tumor and normal samples from the [100]GSE64333 dataset. (C) Differential expression of miR-196b between CRPC and HSPC samples from the [101]GSE70770 dataset. (D) Comparative differential expression analysis of miR-196b between tumor samples at stages T3–T4 and T2 based on the [102]GSE70770 dataset miR-196b-5p promotes cell migration and invasion in PCa cells by activating EMT Our previous research has confirmed that exosomes derived from CAFs under ADT can enhance the migration and invasion of PCa cells by activating EMT both in vivo and in vitro [[103]8]. Notably, various miRNAs, especially miR-196b-5p, were upregulated in exosomes secreted by CAFs after castration. To examine whether the increased levels of miR-196b-5p influence the migration and invasion of PCa cells, we transfected PCa cell lines with miR-196b-5p mimics and conducted Transwell and wound healing assays. The results indicated that the expression of miR-196b-5p facilitated the migration and invasion of both DU145 and LNCaP cells (Figs. [104]4A-D, and [105]S4A). Subsequently, We performed Western blot analysis of EMT proteins in DU145 and LNCaP cell lines infected with lenti-miR-196b-5p. The results revealed that, in the group infected with lenti-miR-196b-5p, the relative expression of N-cadherin, Snail, and Slug was increased, while the relative expression of E-cadherin decreased, with no significant changes observed in Vimentin expression (Fig. [106]4G-H). Furthermore, we transfected miR-196b-5p inhibitor into the PC3 cell line, which has a high baseline expression of miR-196b-5p (Fig. [107]4I). The results from Transwell and wound healing assays demonstrated that the reduced expression of miR-196b-5p inhibited the migration and invasion of PC3 cells (Figs. [108]4J-K, Fig. [109]S4B), with Western blotting showing increased expression of E-cadherin and decreased expression of N-cadherin, Snail, and Slug. In summary, miR-196b-5p promotes migration and invasion in PCa cells by activating EMT. Fig. 4. [110]Fig. 4 [111]Open in a new tab miR-196b-5p promotes cell migration and invasion in PCa cells via activating EMT. (A) and (B) Transwell assays performed to assess the effect of miR-196b-5p on migration (A) and invasion (B) in DU145 cells (scale bars = 25 μm). (C) and (D) Transwell assays performed to assess the effect of miR-196b-5p on migration (C) and invasion (D) in LNCaP cells (scale bars = 25 μm). (E) and (F) Efficiency of miR-196b-5p overexpression in DU145 and LNCaP cells was measured by RT-PCR. (G) and (H) Western blot utilized to assess the effect of miR-196b -5p on EMT marker protein in DU145 and LNCaP cells, respectively. Each set of three lanes in the figure represents three biological replicates. (I) Knockdown efficiency of miR-196b-5p in PC3 cells by miR-196b-5p inhibitor measured using RT-PCR. (J) and (K) Transwell assays performed to assess the effect of miR-196b-5p inhibitor on migration and invasion, respectively (scale bars = 25 μm). (L) Western blot analysis used to detect EMT marker protein in PC3 cells. Data are presented as mean ± SD, representing triplicate measurements. (Student’s t-test, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001) Identification of transcripts targeted by miR-196b-5p in PCa To clarify the impact of miR-196b-5p on the gene expression profile, we conducted RNA sequencing analysis on DU145 cells transfected with lenti-miR-196b-5p or lenti-miR-control. Our result indicated significant changes in the transcriptome due to miR-196b-5p, identifying a total of 1,009 differentially expressed genes (DEGs), including 519 upregulated genes and 490 downregulated genes (Fig. [112]5A). The heatmap also demonstrated that the DEGs could effectively differentiate between miR-196b-5p overexpressing samples and control samples (Fig. [113]5B). To identify potential targets of miR-196b-5p in PCa cells, we employed multiple public databases for microRNA target gene prediction. This analysis revealed that HOXC8, HOXA9, RDX, IGF2BP1, and GATA6 were highly correlated target genes (Fig. [114]5C). We intersected these five genes with the 490 downregulated DEGs identified in our RNA sequencing data, resulting in HOXC8 (homeobox C8) being the only highly relevant gene (Fig. [115]5D). HOXC8 encodes a transcription factor containing a homeobox domain, and its dysregulation is associated with various tumors; however, its role in PCa remains unexplored. To verify whether miR-196b-5p can directly target the 3’ UTR of HOXC8, we designed luciferase reporter vectors containing either wild-type or mutated HOXC8 3’ UTR regions for dual-luciferase reporter assays (Fig. [116]5E). The results showed that co-transfection with miR-196b-5p mimics significantly reduced the luciferase activity of the HOXC8 WT 3’ UTR compared to co-transfection with miR-NC mimics in HEK-293T cells. In contrast, no significant direct interaction was observed between miR-196b-5p and the HOXC8 MUT 3’ UTR-containing vector (Fig. [117]5F). Subsequently, we assessed the RNA and protein levels of HOXC8 in DU145 and LNCaP cell lines, revealing a notable downregulatory effect of miR-196b-5p on HOXC8 expression. (Fig. [118]5G-J). In conclusion, we demonstrated that miR-196b-5p targets the particularly interesting gene HOXC8 in PCa, which is a focus point of our study. Fig. 5. [119]Fig. 5 [120]Open in a new tab HOXC8 is a direct target of miR-196b-5p in PCa cells. (A) Volcano plot of DEGs. (B) Heatmap of DEGs. (C) miR-196b-5p target genes predicted by the MIRWalk, TargetScan, and miRDB databases. (D) Intersection of significantly downregulated DEGs with predicted miR-196b-5p target genes. (E) Predicted target sequence of miR-196b-5p in the 3′-UTR of HOXC8, along with the construction of wild-type and mutant HOXC8 3′-UTR luciferase reporter constructs in HEK-293T cells. (F) The relative luciferase activities were measured after co-transfection of miR-NC mimics or miR-196b-5p mimics and a luciferase vector encoding NC, wild-type, or mutant HOXC8 3′-UTR region. (G) and (H) HOXC8 mRNA levels in DU145 and LNCaP cells transfected with lenti-miR-196b-5p. (I) and (J) HOXC8 protein levels transfected with lenti-miR-196b-5p in DU145 and LNCaP cells. Data are presented as mean ± SD, representing triplicate measurements. (Student’s t-test, *P < 0.05, **P < 0.01, ****P < 0.0001) HOXC8 inhibits EMT in PCa cells To investigate whether HOXC8 expression is also associated with cell migration and invasion, we transfected PCa cell lines with lenti-HOXC8-OE or lenti-HOXC8-control. The expression efficiency of HOXC8 was thereafter validated using RT-PCR and Western blot analysis (Fig. [121]6A-D). We subsequently examined the impact of HOXC8 expression on the levels of EMT proteins and found that overexpression of HOXC8 in both DU145 and LNCaP cell lines led to a significant increase in E-cadherin expression (Fig. [122]6E-F). Through Transwell assays and wound healing assays, we discovered that overexpression of HOXC8 significantly inhibited the migration and invasion of PCa cells(Fig. [123]6G-J). Considering the important role of HOXC8 as a transcription factor, we performed RNA sequencing analysis on DU145 cells transfected with lenti-HOXC8-OE or lenti-HOXC8-control, with relevant DEGs and transcription factor prediction analyses presented in Fig. [124]S6. To further investigate whether miR-196b-5p exerts its EMT activating effects by inhibiting HOXC8 expression in PCa cells, we restored HOXC8 expression in PCa cells transfected with lenti-miR-196b-5p. Western blot analysis indicated that HOXC8 overexpression could reverse the effects of miR-196b-5p in DU145 and LNCaP cells (Fig. [125]7A-B). Importantly, similar results were obtained when HOXC8 was knocked down using siRNA in PCa cells, resembling the effects of miR-196b-5p transfection (Fig. [126]7C-F). These experiments suggest that miR-196b-5p activates EMT in PCa cells by downregulating HOXC8. Fig. 6. [127]Fig. 6 [128]Open in a new tab HOXC8 inhibits EMT in PCa cells. (A) and (B) Western blot analysis to assess the efficiency of HOXC8 overexpression in DU145 and LNCaP cells, respectively. (C) and (D) RT-PCR analysis to assess the efficiency of HOXC8 overexpression in DU145 and LNCaP cells, respectively. (E) and (F) Western blot assessment of EMT marker protein in DU145 and LNCaP cells transfected by HOXC8-control or HOXC8-OE. (G) and (H) Evaluation of the effects of HOXC8 on migration (G) and invasion (H) in DU145 cells by Transwell assays (scale bars = 25 μm). (I) and (J) Evaluation of the effects of HOXC8 on migration (I) and invasion (J) in LNCaP cells by Transwell assays (scale bars = 25 μm). Data are presented as mean ± SD, representing triplicate measurements. (Student’s t-test, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001) Fig. 7. [129]Fig. 7 [130]Open in a new tab miR-196b-5p promotes EMT in PCa cells by targeting HOXC8. (A) and (B) Western blot analysis to assess the protein levels of E-cadherin, N-cadherin, Snail, Slug, and HOXC8 in DU145 and LNCaP cells transfected with lenti-miR-control, lenti-miR-196b-5p, lenti-miR-196b-5p HOXC8-control, or lenti-miR-196b-5p HOXC8-OE. (C) and (D) Relative expression levels of HOXC8 mRNA in DU145 and LNCaP cells transfected with siHOXC8. (E) and (F) Western blot analysis to assess the protein levels of E-cadherin, N-cadherin, Snail, Slug, and HOXC8 in DU145 and LNCaP cells following transfected with siHOXC8 ADT-induced miR-196b-5p activates EMT and suppresses HOXC8 expression in vivo To confirm the effect of miR-196b-5p on PCa metastasis in vivo following castration, we induced androgen deprivation by performing surgical castration on BALB/c nude mice. DU145 luciferase cells were transfected with miR-196b-5p and injected via the tail vein. Tumor metastasis was monitored using small animal in vivo imaging. Consistent with in vitro experiments, the luciferase signal and the number of lung metastases in the miR-196b-5p group were significantly higher than those in the control group (Fig. [131]8A-B). Additionally, IHC analysis confirmed that the tumors in the control group exhibited significantly stronger HOXC8 staining compared to the miR-196b-5p group (Fig. [132]8C). Fig. 8. [133]Fig. 8 [134]Open in a new tab miR-196b-5p promotes metastasis of PCa cells in vivo. (A) Bioluminescence images of BALB/c nude mice that injected with DU145 luciferase cells treated with either miR-196b-5p mimics or miR-NC mimics. The color scale bar depicts the photon flux emitted from the mice. (B) Tumor foci in lungs of BALB/c nude mice after injecting DU145 cells treated with either miR-196b-5p mimics or miR-NC mimics. (C) Immunohistochemical staining of HOXC8 in tumors from mice after injecting DU145 cells treated with either miR-196b-5p mimics or miR-NC mimics. (Student’s t-test, *P < 0.05, **P < 0.01) miR-196b-5p activates NF-κB pathway by targeting HOXC8 To further investigate the downstream mechanisms of miR-196b-5p targeting HOXC8, we performed KEGG pathway enrichment analysis and GO enrichment analysis on the DEGs in DU145 cells transfected with miR-196b-5p (Figs. [135]9A and [136]S5A-B). The KEGG pathway enrichment result indicated that the DEGs were primarily enriched in the NF-κB signaling pathway. The GSEA enrichment analysis of the NF-κB signaling pathway is shown in Fig. [137]S5C. Considering the increasing evidence suggesting that the HOX protein family plays an important role in regulating the NF-κB pathway [[138]33], we conducted a series of experiments to investigate the effects of miR-196b-5p and HOXC8 on this pathway. The expression of IκBα (NF-κB inhibitor α) and p-IκBα (phosphorylated IκBα) were assessed through western blot analysis. The results showed that the expression of p-IκBα protein was upregulated in DU145 and LNCAP cells transfected with miR-196b-5p, and this change was reversed in cells restored HOXC8 (Fig. [139]9B-C). To further confirm the activation of the NF-κ pathway, we evaluated the expression of nuclear protein P65 in DU145 and LNCAP cells. In prostate cancer (PCa) cells transfected with miR-196b-5p, nuclear P65 expression was significantly elevated; however, the restoration of HOXC8 notably decreased nuclear P65 protein levels (Fig. [140]9D-E). Fig. 9. [141]Fig. 9 [142]Open in a new tab miR-196b-5p exhibits its functions by activating NF-κB signaling pathway in PCa cells (A) KEGG enrichment analysis of DEGs. (B-E) Western blot analysis of IKBα, p-IKBα, and nuclear protein P65 levels in DU145 and LNCaP cells transfected with lenti-miR-control, lenti-miR-196b-5p, lenti-miR-196b-5p HOXC8-control, or lenti-miR-196b-5p HOXC8-OE. (F-I) Western blot analysis of E-cadherin, N-cadherin, Snail, Slug, IKBα, p-IKBα, and nuclear protein P65 levels in PCa cells treated with NF-κB inhibitor after transfection with lenti-miR-control, lenti-miR-196b-5p, lenti-miR-196b-5p HOXC8-control, or lenti-miR-196b-5p HOXC8-OE Furthermore, the addition of BAY 11-7082 (an NF-κB pathway inhibitor) to PCa cells transfected with miR-196b-5p revealed that BAY 11-7082 exhibited effects similar to those of HOXC8, inhibiting IκBα phosphorylation and P65 nuclear translocation (Fig. [143]9F-I). Surprisingly, although BAY 11-7082 has been reported as an IκBα phosphorylation inhibitor [[144]34], our findings suggest that it appears to inhibit total IκBα protein. Notably, the utilization of BAY 11-7082 to inhibit the NF-κB signaling pathway counteracted the effects of miR-196b-5p on EMT activation, as evidenced by an increase in E-Cadherin expression and a decrease in N-Cadherin, Snail, and Slug expression (Fig. [145]9F-G). The expression levels of nuclear P65 in DU145 and LNCaP cell lines are shown in Figs. [146]9H and I, respectively, with the corresponding quantification presented in Fig. [147]S7. Collectively, these experimental results strongly indicate that miR-196b-5p activates EMT by inhibiting HOXC8, thereby activating the NF-κB signaling pathway (Fig. [148]10). Fig. 10. [149]Fig. 10 [150]Open in a new tab The flowchart of this study, created with BioRender.com Discussion Increasing evidence suggests that the biological behaviors of cancer cells, such as proliferation, invasion, and metastasis, are strongly influenced by the characteristics of stromal cells in the microenvironment [[151]35–[152]37]. CAFs, as the most abundant component in the tumor microenvironment, play a crucial role in the progression to CRPC following ADT for prostate cancer [[153]38, [154]39]. Studies have shown that CAFs secrete growth factors and cytokines that promote the proliferation, migration, and invasion of tumor cells [[155]40, [156]41]. Additionally, CAFs synthesize and remodel the extracellular matrix, creating a barrier that hinders the penetration of drugs and immune cells, thereby reducing the effectiveness of tumor therapy [[157]42]. The extensive heterogeneity of CAFs in prostate cancer is a challenge that limits their clinical therapeutic translation. ScRNA-seq analysis can categorize CAFs into different molecular subtypes, which may have distinct biological and clinical significance in tumor progression and immune-suppressive microenvironments [[158]43]. Research indicates that androgen deprivation therapy-induced SPP1^+ myCAFs are key stromal components driving the development of castration-resistant prostate cancer. These myofibroblasts can paracrine SPP1 to activate the ERK signaling in tumor cells, promoting castration resistance in prostate cancer [[159]39]. Furthermore, the interaction between CAFs and prostate cancer stem cells (PCSCs) is also important for the progression and therapeutic resistance of prostate cancer. CAFs promote the growth and survival of PCSCs by releasing signaling molecules and altering the surrounding environment, and vice versa, PCSCs may also influence the characteristics and behavior of CAFs through the production of various molecules [[160]44]. This crosstalk mechanism may be crucial for the development of progression and therapeutic resistance in prostate cancer. In our research, we found that CAFs post-castration treatment have an exuberant secretory function and can mediate cancer cell metastasis through the exosomal pathway. This finding helps explain the clinical phenomenon of metastasis in prostate cancer patients after ADT and is significant for targeting CAFs-derived exosomes in combination with ADT treatment to improve the prognosis of advanced prostate cancer patients. MiRNAs are the key component of exosomes derived from CAFs and have been demonstrated to play a critical role in the progression of various cancers, including prostate cancer [[161]45–[162]47]. MiRNAs are widely dysregulated in CRPC and have the potential to serve as therapeutic targets and biomarkers [[163]48–[164]51]. In our study, we identified that miR-196b-5p as a crucial factor promoting metastasis in PCa. It has been reported that in non-small cell lung cancer, miR-196b-5p promotes tumor proliferation, migration, and invasion by directly targeting the tumor suppressor genes FAS, GATA6, and TSPAN12 [[165]18, [166]52]. In laryngeal squamous cell carcinoma, miR-196b enhances tumor cell proliferation and invasion by targeting SOCS2 [[167]53]. Additionally, the entire miR-196 family has been shown to promote cell migration and invasion in oral cancer cells [[168]54]. These findings indicate that miR-196b-5p plays a pro-tumorigenic role across various cancers. HOX transcription factors are among the most widely studied target genes of the miR-196 family [[169]55–[170]57]. The HOX family comprises a group of evolutionarily conserved genes that regulate crucial embryonic developmental processes, such as anterior-posterior axis formation and organ morphogenesis, while also playing significant roles in cell apoptosis, proliferation, and migration [[171]58]. Different HOX proteins exhibit varying functions in different tumors, acting as either oncogenes or tumor suppressors. HOXA5 is well-known as a tumor suppressor in breast cancer and solid tumors like colon cancer [[172]59], yet it does not exhibit the same effect in leukemia [[173]60]. Conversely, HOXA9 is an important oncogene in leukemia [[174]61], while its role in breast cancer is contrary [[175]62]. In our study, we employed luciferase assays and western blot analysis to identify HOXC8 as a direct target of miR-196b-5p, demonstrating that it regulates the expression of EMT-related proteins through the NF-κB pathway. There is a well-documented connection between the expression of HOX family proteins and NF-κB signaling, where HOX transcription factors and NF-κB mutually regulate each other via various mechanisms [[176]63]. For instance, a study highlighted that HOXA9 can inhibit NF-κB signaling by reducing its ability to bind to DNA [[177]64]. Our study is the first to elucidate the role of HOXC8 protein in promoting tumor metastasis and regulating the NF-κB signaling pathway, offering novel insights that may contribute to the development of effective therapies for CRPC. Our study also has some limitations. First, the potential mechanism behind the increase in miR-196b-5p in CAF-derived exosomes following ADT remains unclear. Second, the precise mechanism by which HOXC8 influences NF-κB is yet to be elucidated. HOX transcription factors have been reported to interact with NF-κB through various mechanisms, including direct physical interactions, transcriptional regulation, and modulation of upstream regulators [[178]65–[179]67]. The specific regulatory mechanism between HOXC8 and NF-κB requires further investigation. Conclusion In conclusion, we conducted an in-depth investigation into the oncogenic role of miR-196b-5p derived from CAF exosomes following ADT, along with its specific mechanism of action. Our study revealed that its target, HOXC8, is closely associated with the NF-κB pathway and EMT regulation. These novel findings may pave the way for further exploration of the pro-carcinogenic mechanisms of CAFs in prostate cancer and the identification of potential therapeutic targets. Electronic supplementary material Below is the link to the electronic supplementary material. [180]13062_2025_667_MOESM1_ESM.tif^ (1.4MB, tif) Supplementary Material 1: Supplementary Fig. 1 (A) The volcano plot illustrates the highly expressed miRNAs in exosomes derived from CAFs treated with DHT. (B) Kaplan-Meier analysis comparing BCRFS differences between the high miR-196b-5p group and the low miR-196b-5p group. (The high-expression and low-expression groups were categorized by the optimal cutoff value.) (C) The differential expression of miR-196b between NEPC and CRPC samples in the Beltran (2016) cohort. [181]13062_2025_667_MOESM2_ESM.tif^ (9.9MB, tif) Supplementary Material 2: Supplementary Fig. 2 (A) CAFs were transiently transfected with Cy3-labeled miR-control or miR-196b-5p, as well as with Cy3-labeled miR-196b-5p treated with GW4869, and then co-cultured with DU145 cells for 24 h. Fluorescence microscopy was used to observe the green signals (β-actin) and red signals (Cy3) in LNCaP cells (scale bar = 50 μm). (B) Exosomes were isolated from the supernatant of CAFs transfected with Cy3-labeled miR-196b-5p or miR-control, and exosomes (25 µg/mL) were added to DU145 cells and incubated for 24 h. Fluorescence microscopy was used to detect the green signal (β-actin) and red signal (Cy3) in LNCaP cells (scale bar = 50 μm). [182]13062_2025_667_MOESM3_ESM.tif^ (3.8MB, tif) Supplementary Material 3: Supplementary Fig. 3 (A) and (B) Cell viability levels were assessed using the CCK-8 assay in DU145 and LNCaP cells following transfection with lenti-miR-control or lenti-miR-196b-5p. (C) and (D) Cell proliferation levels were evaluated through a colony-forming assay in DU145 and LNCaP cells after transfection with lenti-miR-control or lenti-miR-196b-5p. [183]13062_2025_667_MOESM4_ESM.tif^ (4.9MB, tif) Supplementary Material 4: Supplementary Fig. 4 (A) Cell migration ability was evaluated in DU145 cells using a wound healing assay following transfection with miR-NC mimics or miR-196b-5p mimics. (B) Cell migration ability was assessed in PC3 cells using a wound healing assay after transfection with a miR-196b-5p inhibitor or control. (C) Cell migration ability was measured in DU145 cells using a wound healing assay after transfection with HOXC8-OE or control. [184]13062_2025_667_MOESM5_ESM.tif^ (2.6MB, tif) Supplementary Material 5: Supplementary Fig. 5. (A) Chord diagram of GO enrichment analysis for upregulated genes in DU145 cells overexpressing miR-196b-5p. (B) Chord diagram of GO enrichment analysis for downregulated genes in DU145 cells overexpressing miR-196b-5p. (C) Gene Set Enrichment Analysis of the NF-kB signaling pathway. [185]13062_2025_667_MOESM6_ESM.tif^ (5MB, tif) Supplementary Material 6: Supplementary Fig. 6. (A) Volcano plot of differentially expressed genes in DU145 cells overexpressing HOXC8. (B) Heatmap of differentially expressed genes in DU145 cells overexpressing HOXC8. (C) Sankey diagram of differential transcription factor (TF)-target gene interactions: from left to right, the first column represents transcription factor families, the second column represents differentially expressed transcription factors, and the third column represents differentially expressed target genes. Lines indicate relationships between TF families, TFs, and target genes. (D) Distribution of differential TF families: the x-axis represents TF families, and the y-axis represents gene counts. Dark blue bars represent all genes regulated by each TF family; yellow bars represent differentially expressed TFs regulated by each family; pink bars represent upregulated differentially expressed TFs; and light blue bars represent downregulated differentially expressed TFs. (E) Circular plot of the top 30 genes in the protein-protein interaction (PPI) network. Red nodes represent upregulated differentially expressed genes, and blue nodes represent downregulated differentially expressed genes. The size of each gene node reflects the number of associated genes. [186]13062_2025_667_MOESM7_ESM.tif^ (1.2MB, tif) Supplementary Material 7: Supplementary Fig. 7. (A) The expression levels of nuclear P65 in each group of DU145 cells. (B) The expression levels of nuclear P65 in each group of LNCaP cells. Data are presented as mean ± SEM, representing triplicate measurements. [187]13062_2025_667_MOESM8_ESM.docx^ (14.1KB, docx) Supplementary Material 8: Table S1 The miR-196b-5p mimics and matched control sequences used in this study. [188]13062_2025_667_MOESM9_ESM.docx^ (14.2KB, docx) Supplementary Material 9: Table S2 siRNA sequences used in the study. [189]13062_2025_667_MOESM10_ESM.docx^ (14.4KB, docx) Supplementary Material 10: Table S3 The plasmid sequences used for the dual-luciferase reporter assay. [190]13062_2025_667_MOESM11_ESM.docx^ (14.2KB, docx) Supplementary Material 11: Table S4 Primer sequences used in our study. Acknowledgements