Abstract Background Emerging evidence underscores the pivotal role of M2-polarized tumor-associated macrophages (M2-TAMs) in orchestrating immunosuppressive tumor microenvironments that fuel metastatic dissemination in prostate cancer (PCa), yet the fundamental mechanisms governing M2-TAM trafficking in lethal PCa progression remain poorly understood. Methods Multi-cohort transcriptomic analyses were performed to identify metastasis-associated genes, with CTSZ prioritized as a key cathepsin linked to prostate cancer progression. Circulating tumor and bone metastatic mouse models were employed to investigate CTSZ-driven M2-TAM infiltration and metastatic behavior. Mechanistic studies included proteasomal degradation assays, IL32 pre-mRNA splicing analysis, and IL-32 binding experiments using RGD motif-dependent interactions. Therapeutic efficacy was tested with the ITGA5 inhibitor GLPG0187 in preclinical models. Results Elevated CTSZ expression shows strong clinical association with advanced pathological progression. CTSZ overexpression in PCa cells drives lung metastasis dissemination and bone metastatic in vivo model but fails to enhance cell-intrinsic oncogenic behaviors in vitro systems. Overexpression of CTSZ promotes M2-TAM infiltration and metastasis by inducing TRA2A degradation via the proteasome pathway, which alleviates TRA2A-mediated suppression of IL32 alternative splicing. Enhanced IL-32 secretion facilitates M2-TAM recruitment through binding to macrophage integrin ITGA5. Pharmacological inhibition of ITGA5 with GLPG0187 significantly reduced metastatic burden and M2-TAM infiltration in vivo. Conclusions The CTSZ/TRA2A/IL-32/ITGA5 axis orchestrates protumoral immunity in PCa metastasis by driving M2-TAM recruitment. Targeting this pathway, particularly through ITGA5 blockade, represents a promising therapeutic strategy to inhibit metastatic progression and remodel the immunosuppressive tumor microenvironment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-025-06865-w. Keywords: CTSZ, IL-32, TRA2A, ITGA5, Macrophage, Tumor microenvironment Introduction Prostate cancer (PCa) represents a formidable clinical challenge as the second most lethal malignancy in men globally, with over 1.4 million new cases diagnosed annually. The 5 year survival rate exhibits a dramatic decline—from nearly 90% in localized stages to below 30% in metastatic disease [[48]1, [49]2]. The immunosuppressive tumor microenvironment (TME), characterized by dysfunctional cytotoxic T cells and paradoxical accumulation of protumoral M2-polarized macrophages, has emerged as a key driver of therapeutic resistance and metastatic progression [[50]3, [51]4]. Recent single-cell analyses reveal that M2 tumor-associated macrophages (TAMs) constitute the predominant immune infiltrate in advanced PCa. These cells form feedforward regulatory loops with cancer cells through androgen receptor co-activation and suppress natural killer (NK) cell function via the IL-10/PD-L1 signaling axis [[52]5–[53]7]. However, the molecular underpinnings governing their preferential recruitment to the TME remain poorly characterized, creating a critical knowledge gap in developing macrophage-targeted therapies. Cathepsin Z (CTSZ), a lysosomal cysteine protease with unique RGD-motif dependent extracellular signaling capacity, has emerged as a key mediator of tumor-stroma crosstalk [[54]8, [55]9]. The catalytically mature CTSZ isoform is restricted to carboxypeptidase activity via its C-terminal monopeptidase domain, which mediates proteolytic processing of substrate C-terminal residues [[56]10–[57]12]. In addition, relevant evidence shows that in the studies of triple negative breast cancer and hepatocellular carcinoma, the overexpression of CTSZ is correlates with advanced disease and low patient survival rate [[58]13, [59]14]. However, the functional landscape of CTSZ in metastatic PCa remains unknown: its mechanistic contribution to the metastatic cascade, crosstalk with TAMs, and immunomodulatory role within the TME require experimental dissection. This metastasis-associated expression pattern, coupled with its undefined role in PCa immunology, prompted investigation into CTSZ-mediated TME reprogramming. Our findings establish CTSZ as a non-cell-autonomous driver of prostate cancer metastasis, mediating M2-polarized TAM recruitment through proteolytic regulation of the TRA2A/IL-32/ITGA5 axis. Mechanistically, CTSZ-mediated degradation of TRA2A liberates IL32 mRNA from splicing repression, leading to increased secretion of IL-32 that engages ITGA5 receptors on macrophages. This ligand-receptor interaction activates PI3K/AKT signaling in macrophages, driving chemotaxis and M2 polarization. Pharmacological inhibition of ITGA5 using the antagonist GLPG0187 significantly attenuated tumor metastasis in syngeneic mouse models. These results decode a tumor cell-macrophage crosstalk circuit and propose a dual therapeutic strategy targeting both CTSZ protease activity and IL-32/ITGA5 signaling to dismantle the immunosuppressive TME in advanced PCa. Materials and methods Online bioinformatics analysis and transcriptome sequencing Transcriptome data from [60]GSE32269, [61]GSE101607, and [62]GSE210729 datasets were analyzed through GEO database. Clinical correlations of CTSZ with nodal metastasis and Gleason score in PCa patients were evaluated using UALCAN platform. Genomic profiling and survival analysis across nine prostate cancer cohorts were performed via cBioPortal, where SU2C mPCa cohort data further revealed CTSZ correlations with macrophage markers and IL-32/TRA2A interactions. Immune infiltration patterns were assessed using Time2.0 database, while CTSZ expression profiles across disease stages were retrieved from Cambridge CRUK dataset.For experimental validation, TRIzol-extracted RNA from PC3 cells (sh-CTSZ vs sh-NC groups) underwent RNA-seq analysis (Majorbio Co.) using SMARTer mRNA-seq library prep kit per manufacturer’s protocol. Single-cell sequencing and immune infiltration analysis Single-cell RNA sequencing data from [63]GSE143791 (10 bone metastatic cases) were processed through the Seurat pipeline (v3.1.2) [[64]15]. This was followed by doublet removal via the DoubletFinder R package, with a 10% doublet formation rate [[65]16]. After doublet removal using DoubletFinder, datasets underwent anchor-based integration with subsequent scaling and PCA dimensionality reduction. Graph-based clustering analysis was performed before UMAP visualization of cell populations. The cell type enrichment analysis tool CIBERSORT was used to calculate the gene set enrichment scores of the 22 immune cell subtypes via the GSVA package. Cell acquisition and culture All the cell lines used in this study were sourced from the American Type Culture Collection (ATCC) and were accompanied by species verification and mycoplasma contamination detection certificates. The cell cultures were meticulously maintained in a CO2 incubator at 37 °C with either RPMI 1640 medium or DMEM supplemented with 10% FBS and 1% penicillin‒streptomycin solution. Cell transfection CTSZ-, IL-32-, and TRA2A-targeting siRNAs (sequences in Table S1) and lentiviral constructs were obtained from Tsingke Biotech. siRNA transfection in PC3/22RV1 cells followed Lipo3000 protocol, with RNA/protein harvested at 24/48 h post-transfection. For stable CTSZ overexpression, 22RV1/RM1 cells were transduced with pLenti-CMV-Puro-Luciferase-CTSZ (vector-modified by CTSZ insertion) followed by titer-optimized puromycin selection. Empty vector served as negative control. Lung metastasis model and bioluminescence imaging All procedures complied with AAALAC (Association for Assessment and Accreditation of Laboratory Animal Care, International) guidelines under Army Medical University IACUC (Institutional Animal Care and Use Committee) approval (No. AMUWEC20232943). Male BALB/c nude mice (4–5 weeks, 18–22 g; n = 6/group) were injected via tail vein with 1 × 10^6 PC3 cells suspended in PBS. Metastatic progression was monitored by bioluminescence imaging (IVIS Spectrum CT) at days 3 and 21 post-injection following intraperitoneal D-luciferin administration (150 mg/kg; 10 μL/g, Beyotime ST198). Lungs harvested at endpoint were fixed in bitter acid for 10 min, rinsed in PBS, and metastatic nodules (> 1 mm) quantified macroscopically before 4% PFA fixation and H&E validation. Bone metastasis model Ten nude mice, aged 5–6 weeks, were prepared for the experiment using CTSZ-OE and Vector cells. The mice were sedated with isoflurane gas anesthesia throughout the procedure. Tumor cells, 10^5 in number and suspended in 20 μl of PBS, were injected into the tibia of each mouse after disinfecting the injection site. An insulin needle was inserted gently along the tibial plateau to reach the bone marrow cavity. After the injection, the area was disinfected. On day 21, the mice were euthanized, and their tibiae were collected. The samples were fixed for at least 24 h and subjected to a bone micro-CT scan in Kontich, Belgium. Relevant indicators were assessed via H&E staining, immunohistochemical staining, and trap staining after decalcification. Isolation of macrophages from tumor tissues Fresh tumor tissue was dissected into small 1 mm pieces in a 10 cm dish and then digested with a solution containing 1 g/L type IV collagenase (#[66]LS004188, Worthington), 0.1 g/L hyaluronidase (Sigma), and 0.01 g/L DNase I (#D8071, Solarbio, China) in a cell incubator for 2 h. After dispersion, the cell suspensions were collected and centrifuged at 600 × g for 5 min, the supernatant was discarded, ACK lysis buffer was added for cell lysis, and the mixture was washed through centrifugation at 600 × g for 5 min, which was repeated three times. The cell pellet was then resuspended in complete culture medium and filtered through a 100-μm filter to collect the cells for further isolation of the macrophages. Transwell assay Invasion/migration assays were performed in triplicate using Matrigel-coated Transwell chambers (8 μm pores). PCa cells (3 × 10^3/well, counted via automated cell counter) in serum-free medium were seeded in upper chambers, with complete medium as chemoattractant in lower chambers. After 18 h incubation, traversed cells were fixed in 4% PFA, stained with crystal violet, and quantified by microscopic examination of five random fields (20 × objective). Parallel migration assays omitted Matrigel coating. Three biological replicates with duplicate technical repeats were conducted. Macrophage migration assays Vector- or CTSZ-OE-transfected 22RV1 cells were cultured to 90% confluence in T25 flasks. Conditioned medium (CM) was collected after 48 h, centrifuged (300 × g, 5 min), and used for Transwell assays. THP-1-derived macrophages were polarized with PMA (100 nM) followed by LPS (100 ng/ml, M1) or IL-4/IL-13 (20 ng/ml each, M2). Polarized macrophages (5 × 10^4 cells/well) were seeded in 8-μm Transwells with CM in lower chambers. Migrated cells were fixed (4% paraformaldehyde) and stained (0.1% crystal violet) after 24 h. Real-time PCR Total RNA was extracted via TRIzol reagent (Ambion 317908) and then converted into cDNA using PrimeScript (DRR047A; TaKaRa, China). Gene expression analysis was conducted via real-time qPCR in a Bio-Rad CFX Connect real-time system utilizing SYBR Green Master Mix from Thermo Fisher Scientific (A25742, China) and the primers listed in Supplementary Table S2 from Sangon Biotech. The gene expression levels were determined via the 2^−ΔΔCt method. Western blot Cell proteins were extracted via radioimmunoprecipitation assay (RIPA) lysis buffer (#P0013, Beyotime, China) and quantified using a BCA kit (#P0009, Beyotime, China). For SDS‒PAGE analysis, 2 µg of protein was added to each well of a 6/10/15% SDS‒PAGE gel after BCA quantification. The proteins were then transferred to Immune-Blot PVDF membranes (#1620177, Bio-Rad, USA). The membranes were blocked with protein-free rapid sealing solution (#G2052, Servicebio, China) for 10 min, followed by overnight incubation with the primary antibody at 4 °C. After the membranes were washed three times for 15 min each with PBST, they were incubated at room temperature for 1 h with the corresponding secondary antibodies. The signals were detected via the Ultrasensitive ECL Western HRP Substrate (#17047, Zenbio, China) and a Bio-Rad ChemiDoc MP System (170–8280). The primary antibodies used are listed in Table S3. Immunohistochemistry and immunofluorescence The tumor tissue microarray of patients with prostate cancer used in this study was provided by Changsha Yaxiang Biotechnology Co., Ltd. Mouse orthotopic, lung, and tibial tumor tissues were sectioned into 5-mm-thick slices. The tissue microarrays and mouse tumor tissue slices were deparaffinized at 70 °C for 2 h. EDTA repair was then performed at a high temperature for 3 min, followed by washing with PBS and blocking with serum for 1 h. The primary antibody was added to the samples in the dark and stored overnight at 4 °C. After the samples were washed with PBS, secondary antibodies were added, and the samples were incubated at room temperature for 30 min. The immunofluorescence procedure was similar to that used for immunohistochemistry, with the addition of DAPI for nuclear staining. Finally, the slides were sealed with mounting medium containing an anti-fluorescence quencher, and all the slides were scanned via a whole-slide scanner. All tumor samples were processed in the same manner. EMSA Biotin labels were introduced into the 5′-TCTGCTGGAAACGACTCGGAG-3′ sequence of IL-32 immature mRNA, which includes the GAAARGARR sequence and has been confirmed as the binding site for TRA2A. In the sequence from positions 602 to 767, the GAAACGA sequence was subsequently mutated to AGGCCTA, whereas in the sequence from positions 2297 to 2489, the GAAACAA sequence was subsequently mutated to TAGCGAT. To assess whether TRA2A binds to this probe and whether the corresponding sequence mutation affects binding, a chemiluminescent EMSA kit (#GS009, Beyotime, China) was used. The sample was loaded onto a BeyoGel™ EMSA Precast PAGE gel (#GS301S, Beyotime, China) for electrophoresis. The supershift group consisted of 1 μg of anti-TRA2A antibody and 20 pmol/μL wild-type probe. The experimental procedures were conducted according to the Chemiluminescent EMSA Kit protocol. Signal detection was performed via a Bio-Rad ChemiDoc MP System after exposure to the nylon membrane. RNA immunoprecipitation An RNA immunoprecipitation (RIP) kit (#p0102, Geneseed, China) was used to obtain RNA bound to TRA2A according to the manufacturer’s protocol. The RNA concentrations were determined via Nanodrop Lite (Thermo Science, Serial Number: LT1613). Because TRA2A may bind to the intron region of IL-32 pre-mRNA, it is necessary to use random primers to complete reverse transcription (#151600, Toyobo, China). qPCR was performed to determine the expression of IL-32 in the reverse-transcribed IgG and IP group cDNAs. The relative expression of the target genes was calculated according to the 2^−ΔΔCt formula, with ΔΔCt = (Ct[IP]–Ct[Input])–(Ct[IgG]–Ct[Input]). Using the Ct value of the input group as the reference CT value, the gene expression level of the IP group of the target gene was 2^−X greater than that of the IgG group. RNA pulldown Biotin-labeled RNA probes (602-767, 602-767-M, 2297-2489, 2297-2489-M) were synthesized by Shanghai Shenggong Biotechnology. PC3 lysates (> 2 mg/mL) were prepared for RNA pull-down using a commercial kit (#DW1127, Dowobio, China). Magnetic beads were washed twice with 20 mM Tris (50 μL), resuspended in 1 × RNA capture buffer (50 μL), and incubated with RNA pull-down mix (100 μL, 4 °C rotation, 3 h). Beads were further incubated in capture buffer (50 μL, RT, 2 h), washed thrice with wash buffer, eluted (50 μL elution buffer, 1 h rotation), and supernatants collected for Western blot. ELISA The supernatants were collected from PCa cells with either knockdown or overexpression of CTSZ. After centrifugation at 2000 × g, the supernatant was collected for further use. Next, we prepared the standard gradient working solution, 1 × Biotin-antibody, streptavidin-HRP, and washing buffer. The prepared samples (100 μL) were added to each well of a 96-well plate and incubated for 1 h in a cell incubator. After the liquid was removed, 100 μL of 1 × biotin-antibody was added to each well and incubated at 37 °C for 1 h. The wells were washed three times with washing buffer, and 100 μL of streptavidin-HRP solution was added to each well and incubated at 37 °C for 1 h. After washing three times, 90 μL of substrate (TMB) was added to each well and incubated at 37 °C for 15 min in the dark. After incubation, the plate was removed, and 50 μL of stop solution was added to each well. The optical density (OD) of each well was measured immediately at 450 nm. The purchased reagent kits included the following products (#JONLOBIO, China): Cat No. JL19279 (IL32), JL14142 (CTSZ), JL19334 (CCL2), and JL1102 (CXCL10). Protease degradation reaction CTSZ protease activity was measured via Octet 2.0. The sensor (Biosensors, NI–NTA, Tray) was prewetted with PBST for 10 min and then successfully immobilized with recombinant TRA2A protein (#TP762583, ORIGENE, USA) containing a histidine tag. The reaction was performed at 37 °C with varying concentrations of recombinant CTSZ (#934-CY, R&D Systems, Minneapolis, MN, USA) to detect its ability to hydrolyze the immobilized TRA2A protein in 0.20 ml per working volume. Hydrolytic events were quantified by measuring the shift in the optical interference pattern. Mass spectrometry analysis Matrix-assisted laser desorption and ionization time-of-flight (MALDI-TOF) was used to analyze pure TRA2A and enzymatically cleaved TRA2A. Recombinant CTSZ (#934-CY; R&D Systems, Minneapolis, MN, USA) was generated according to the manufacturer’s instructions. A mixture of CTSZ and TRA2A (#TP762583, ORIGENE, USA) at a final concentration of 1 μg/μL was incubated at 37 °C for 2 h. Subsequently, the reaction was terminated at − 20 °C and analyzed via a Bruker Daltonics Ultraflex MALDI‒TOF mass spectrometer (Bremen, Germany). Calibration was performed using Bruker protein standards. Fluorescence in situ hybridization (FISH) PC3 cells were fixed on coverslips with 4% paraformaldehyde for 20 min and then washed three times with PBS for 5 min each. Proteinase K (20 μg/mL) was added, and the mixture was digested for 8 min. The cells were subsequently washed three times with PBS for 5 min each. Prehybridization solution was added and incubated at 37 °C for 1 h. The prehybridization solution was removed, and the hybridization solution containing the IL-32 probe was added and incubated overnight at 37 °C. The hybridization mixture was washed with 2 × SSC at 37 °C for 10 min, followed by 1 × SSC at 37 °C for 10 min and then 0.5 × SSC at 37 °C for 10 min. The hybridization mixture was removed, and the TRA2A antibody was added and incubated overnight at 37 °C. The DAPI staining solution was added to the slides, which were subsequently incubated for 8 min in the dark. After washing, the slides were sealed with anti-fade mounting medium. The slides were examined under a fluorescence microscope, and images were captured. Statistical analysis All data presented in this study are representative of similar results obtained from a minimum of three replicates unless stated otherwise. The Shapiro‒Wilk normality test was used to assess the data distribution. The quantitative data are expressed as the means ± standard deviations (SDs) and were analyzed via GraphPad Prism 8 software. Unpaired t tests were used to compare differences between two groups, whereas one-way ANOVA and log-rank trend tests were employed to determine statistically significant differences among three or more groups. A p value < 0.05 was considered statistically significant. Results Aberrant CTSZ upregulation correlates with adverse prognosis in metastatic PCa To identify molecular drivers of PCa metastasis, we analyzed three independent GEO datasets ([67]GSE32269, [68]GSE101607, [69]GSE210729) using stringent criteria (adjusted p < 0.05, |log2FC|> 1). Intersectional analysis revealed two candidate genes: CTSZ and COCH (Fig. [70]1A). Notably, CTSZ was the only gene showing consistent upregulation across all metastatic cohorts (Fig. [71]1B, S1A). Moreover, the expression of CTSZ was increased in metastatic PCa (mPCa) cell lines such as PC3, LNCAP, C4-2B, and DU145 than in non-metastatic PCa cell lines 22RV1 (Fig. [72]1C). Immunohistochemical validation using tissue microarrays (42 metastatic vs. 40 localized PCa cases) showed markedly elevated CTSZ protein abundance in metastatic specimens (positive cell proportion: 5.911 ± 4 vs 10.44 ± 8; p < 0.001, unpaired t-test), demonstrating progressive CTSZ upregulation through disease advancement (Fig. [73]1D). Clinical relevance analyses using GEPIA-based TCGA datasets revealed that CTSZ overexpression strongly correlated with advanced lymph node metastasis and higher Gleason scores (Fig. [74]1E, F). Critically, quantitative analysis of CTSZ expression in 42 mPCa specimens versus 40 localized PCa controls revealed that elevated CTSZ levels were significantly associated with diminished clinical outcomes, demonstrating a median reduction of survival times in both overall survival (OS) and disease-free survival (DFS) (log-rank p < 0.05) (Fig. [75]1G, H). This association was validated in the TCGA-PRAD cohort (n = 498), where CTSZ overexpression correlated with reduced survival duration (HR = 2.7) (Fig. [76]1I). Genomic profiling in the SU2C/PCF metastatic cohort identified increased CTSZ amplification frequency in metastatic lesions (Figure S1B), with amplified cases showing poorer survival outcomes (p < 0.05, Figure S1C). Collectively, multi-cohort analyses establish CTSZ overexpression as a robust biomarker of metastatic progression in PCa, underscoring the need for functional studies to dissect its mechanistic role. Fig. 1. [77]Fig. 1 [78]Open in a new tab CTSZ overexpression correlates with metastatic progression and poor prognosis in prostate cancer. A Venn diagram of differentially expressed genes (DEGs) across three PCa progression datasets ([79]GSE32269, [80]GSE101607, [81]GSE210729). B CTSZ mRNA expression in PCa progression datasets. C CTSZ protein (Western blot) and mRNA (qRT-PCR) levels in PCa cell lines (DU145, PC3, LNCaP, 22Rv1, C4-2B). D Representative H&E and CTSZ IHC staining in localized (n = 42) versus metastatic (n = 40) PCa specimens (scale bar: 50 μm). E Expression of CTSZ in PRAD based on nodal metastasis status in the UALCAN database. F CTSZ expression in PRAD patients on the basis of the patient Gleason score in the UALCAN database. G, H Kaplan–Meier analysis of overall survival (OS) and disease-free survival (DFS) stratified by CTSZ expression (high vs low, log-rank test). I TCGA-PRAD cohort analysis of CTSZ-associated DFS (Cox proportional hazards model). Data represent mean ± SD. Statistics: unpaired t-test (B, D); one-way ANOVA (C, E, F). Statistical significance: *p < 0.05, **p < 0.01, ***p < 0.001 CTSZ overexpression drives prostate cancer metastasis independent of cell-autonomous invasion and migration To determine the role of cancer cell-derived CTSZ in PCa metastasis, we modulated CTSZ expression in human PCa cell lines (PC3, 22RV1) and a mouse PCa line (RM1). Transcriptional and translational validation confirmed successful CTSZ knockdown/overexpression (Fig. [82]2A, B and S1A). Notably, CTSZ manipulation in PC3 (knockdown) and 22RV1 (overexpression) did not significantly affect cellular invasion/migration capacities in Transwell assays (Fig. [83]2C) or alter epithelial-mesenchymal transition (EMT) markers (CDH1, CDH2, VIMENTIN) as assessed by western blot analysis. (Fig. [84]2D, E). These in vitro findings collectively demonstrate that CTSZ manipulation does not directly regulate malignant cellular phenotypes. Fig. 2. [85]Fig. 2 [86]Open in a new tab Overexpression of CTSZ in PCa cells promotes PCa metastasis in vivo. A, B Validation of CTSZ knockdown (PC3) and overexpression (22Rv1) efficiency by qRT-PCR and immunoblotting (mean ± SD; one-way ANOVA). C Transwell migration/invasion assays (representative images and quantification; three biological replicates). D, E EMT marker expression (CDH1, CDH2, VIMENTIN) in CTSZ-modulated cells (immunoblots). F Longitudinal bioluminescence imaging (BLI) of systemic metastasis (n = 6/group; days 3–21). G, H Ex vivo and H&E stain of lung metastasis (H&E; scale bars: 2.5 mm). I Quantification of metastatic nodules (mean ± SD; unpaired t-test). J Micro-CT reconstruction of proximal tibiae (red box: ROI; 3D trabecular analysis). K Histomorphometry (H&E, TRAP, CD206 staining; scale bars: 50 μm). L Bone structural parameters: BMD, BV/TV, Tb.Th, Tb.N, Tb.Sp, SMI (n = 3/group; one-way ANOVA). *p < 0.05; **p < 0.01. ns, not significant. BLI: Bioluminescence imaging Given the lack of in vitro phenotypic effects, we subsequently employed xenograft models to investigate CTSZ's potential role in metastatic dissemination. Bioluminescence imaging of the tail vein-injected mice revealed that substantially higher metastatic burden was observed in the mice inoculated with CTSZ-OE cells than in the mice inoculated with Vector cells at day 21 (Fig. [87]2F). Picric acid staining confirmed increased tumor burden in CTSZ-OE tissues (red arrows, Fig. [88]2G). Histopathological analysis of H&E-stained lung sections revealed an 8.5-fold increase in metastatic foci density in CTSZ-OE mice (red arrows, p < 0.001, Fig. [89]2H, I). Since bone metastasis is an important cause of mortality in patients with advanced PCa, we inoculated RM1 (vector control and CTSZ-OE) cells into the tibia of male nude mice. Micro-CT analysis (2D/3D reconstruction) showed that CTSZ-OE inoculation significantly reduced trabecular bone number (dashed red boxes, Fig. [90]2J). H&E and TRAP staining confirmed decreased trabecular bone mass and increased osteoclast formation in CTSZ-OE tibiae (red arrows, Fig. [91]2K). Parameters representing the structural integrity of trabecular bone, such as percent trabecular area (BV/TV), trabecular thickness (Tb.Th), and trabecular number (Tb.N) were significantly decreased in the CTSZ-OE group than that in the Vector group, whereas parameters representing trabecular separation (Tb.Sp) and the structural model index and (SMI) were notably elevated in the CTSZ-OE group (Fig. [92]2L). Collectively, these findings suggest that overexpression of CTSZ in tumor cells facilitates PCa metastasis in vivo. CTSZ correlates with M2 macrophage enrichment in PCa microenvironment Prostate cancer is increasingly recognized as a process driven by immunosuppressive microenvironment reprogramming, characterized by tumor-associated macrophage (TAM) enrichment and cytotoxic T cell depletion [[93]3]. To investigate which cell subtype drives the immune-evasive phenotype, we performed single-cell transcriptomic profiling on metastatic PCa lesions ([94]GSE143791). This analysis revealed significantly higher macrophage infiltration in metastatic versus primary tumors (Fig. [95]3A and B). Strikingly, M2-polarized TAMs dominated these infiltrates (78% vs. 22% M1; Fig. [96]3C). Fig. 3. [97]Fig. 3 [98]Open in a new tab CTSZ correlates with M2 macrophage enrichment in PCa microenvironment. A Single-cell transcriptomic analysis ([99]GSE143971) of immune cell heterogeneity in localized vs metastatic PCa (Seurat v4.0). B, C Proportional distribution of 16 immune subsets (left) and M1/M2 macrophage ratios (right; ***p < 0.001, χ^2 test). D ssGSEA enrichment scores for M1/M2 signatures in [100]GSE32269 cohort (n = 21 localized vs 29 metastatic; two-way ANOVA). E, F Top CTSZ-correlated genes (E) and M1/M2 marker correlations (F) in SU2C-mPCa cohort (cBioPortal). G CIBERSORT-ABS quantification of CTSZ-M2 macrophage association in TCGA-PRAD (TIMER2.0). H Multiplex IF staining of CTSZ^+ (magenta), CD206^+ (M2, red), and CD86^+ (M1, green) cells in clinical specimens (H&E: 100 μm; IF: 25 μm). Data presented as mean ± SD. Statistics: unpaired t-test (D, H), Pearson correlation (E, F). *p < 0.05, **p < 0.01; ns = not significant Using RNA-seq data from localized/metastatic PCa tissues ([101]GSE32269), we quantified 22 immune cell subsets and found M2 macrophages to be the most differentially enriched cell type in metastatic specimens (Fig. [102]3D). The CTSZ-M2 axis was further validated in the SU2C metastatic PCa cohort, where CTSZ expression exhibited strong correlation with pan-macrophage marker CD68 (P < 0.001) and preferential associations with canonical M2 markers (CD163, CD115, CD301, CSF1R, TGFβ1) over M1 signatures (TLR2, TNF, NOS2, CD80, CD86; Fig. [103]3E, F). CIBERSORT-ABS analysis of TCGA-PRAD data further confirmed that M2 macrophages showed the strongest correlation with CTSZ among all immune subsets (Fig. [104]3G, S2B). Spatial histopathological analysis (n = 38 localized vs 40 metastatic) demonstrated that metastatic PCa lesions exhibited elevated CTSZ expression concomitant with CD206^+ M2 macrophage enrichment, contrasting with concurrent reduction of CD86^+ M1 populations (Fig. [105]3H). These findings collectively position CTSZ as a molecular orchestrator of M2-dominated immunosuppressive niches in advanced PCa. CTSZ-mediated M2 macrophage infiltration facilitates tumor metastasis To delineate the tumor cell-macrophage crosstalk mediated by CTSZ, we evaluated the impact of CTSZ-OE in tumor cells on the behavior of macrophages. In vitro co-culture models revealed that conditioned media from 22RV1-CTSZ-OE cells preferentially chemoattracted M2-polarized macrophages, with no significant effect on M1 subsets (Fig. [106]4A). To validate these findings in vivo, we investigated whether prostate cancer cells overexpressing CTSZ could recruit M2 macrophages. Western blot analysis of metastatic lesions confirmed elevated expression of the M2 macrophage marker arginase-1 (ARG1) and suppressed levels of the M1 marker TNF-α. These results suggest that CTSZ induces metabolic reprogramming favoring an M2-like polarization in the tumor microenvironment (Fig. [107]4B). Similarly. Immunofluorescence and immunohistochemical analyses of tumor tissues confirmed that CTSZ-OE tumors exhibited increased CTSZ expression, enhanced CD206 + M2 macrophage infiltration, and reduced CD86 + M1 macrophage density compared to vector controls (Fig. [108]4C, E). To further validate the pivotal role of CTSZ-mediated M2 macrophages in promoting tumor metastasis, we established a lung metastasis model using CTSZ-OE PCa cells. Bioluminescence imaging (BLI) quantification revealed a significant reduction in pulmonary tumor fluorescence intensity in the clodronate-treated group compared to both untreated controls and liposome-only controls (Fig. [109]4F). Picric acid staining of isolated lungs confirmed fewer metastatic foci in the clodronate group (red arrows, Fig. [110]4G). The number of metastatic lesions is indicated by the red arrow and quantified in Fig. [111]4H for statistical analysis. Immunohistochemical analysis revealed that clodronate treatment attenuated epithelial-mesenchymal transition (EMT) markers (reduced CDH2, VIMENTIN) and Ki67 proliferation index, accompanied by restored E-cadherin (CDH1) expression (Fig. [112]4I). Collectively, these results demonstrate that CTSZ-driven PCa metastasis is critically dependent on M2 macrophage infiltration. Fig. 4. [113]Fig. 4 [114]Open in a new tab CTSZ-mediated M2 macrophage infiltration facilitates tumor metastasis. A Transwell migration of THP-1 cells toward PCa-conditioned media (CM: Vector vs CTSZ-OE; n = 3; mean ± SD; scale bar = 50 μm). B Immunoblot analysis of M2 (ARG1) and M1 (TNFα) markers in xenograft tumors. C -E Representative images of H&E staining, immunohistochemistry, and immunofluorescence staining for CD86 and CD206 in the CTSZ-OE and Vector groups. CD206 (Red), and CD86 (green) staining in the CTSZ-OE and Vector groups. Scale bar = 50 μm. F Longitudinal bioluminescence imaging (BLI) of systemic metastasis (n = 3/group). G Ex vivo lung metastasis. H Quantification of metastatic nodules (one-way ANOVA; unpaired t-test). I Representative image of immunohistochemistry for CDH1, CDH2, VIMENTIN and Ki67 in the Li-Clodronate, Liposome and CTSZ-OE groups. Scale bar = 50 μm. *p < 0.05; **p < 0.01. CM, conditioned medium CTSZ^+PCa cells promote the recruitment of M2 macrophages via IL-32 secretion To investigated the molecular mechanisms by which CTSZ orchestrates M2 macrophage recruitment, we performed transcriptomic profiling of CTSZ-knockdown (CTSZ-KD) versus control PC3 cells. RNA-seq analysis revealed distinct transcriptional alterations in cytokine-mediated signaling pathways (Fig. [115]5A, B). In the cytokine signaling pathway, ten secreted proteins were identified. Heatmap analysis of transcriptome sequencing data revealed that IL-32 exhibited the great fold change in expression and the lowest p-value among these proteins. (Fig. [116]5C, S3A). Simultaneously, qPCR and Western Blot experiments confirmed that knocking down CTSZ resulted in downregulation of IL-32 expression in PC3 cells, while overexpression of CTSZ resulted in upregulation of IL-32 expression in 22RV1 cells (Fig. [117]5D–G). ELISA assays confirmed that IL-32 secretion was significantly altered in the supernatants of CTSZ-knockdown or -overexpressing cells compared to controls. There was no difference in secretion between CTSZ and known molecules that regulate macrophage infiltration, such as CXCL10 and CCL2 (Fig. [118]5H). Functional rescue experiments validated the necessity and sufficiency of IL-32 in CTSZ-driven macrophage chemotaxis. Conditioned media from CTSZ-OE cells enhanced macrophage migration, an effect abolished by IL-32 neutralization. Conversely, impaired macrophage recruitment in CTSZ-KD models was rescued by exogenous IL-32 supplementation (Fig. [119]5I, J). Collectively, these findings define a CTSZ-IL-32 paracrine axis that governs protumoral M2 macrophage infiltration. Although IL-32 is established as the key mediator, the precise mechanisms underlying CTSZ-regulated IL-32 secretion, including post-transcriptional modification or secretory pathway modulation, require further investigation. Fig. 5. [120]Fig. 5 [121]Open in a new tab CTSZ + PCa cells promote the recruitment of M2 macrophages via IL-32 secretion. A Volcano plot of differentially expressed genes in PC3 cells following CTSZ knockdown. B KEGG pathway enrichment analysis comparing shCTSZ and shNC groups in PC3 cells. C Hierarchical clustering heatmap of cytokine signaling-related genes identified in KEGG analysis. D–G Validation of CTSZ-mediated IL-32 regulation: qRT-PCR (D, E) and immunoblotting. H ELISA quantification of secreted IL-32, CTSZ, CCL2, and CXCL10 in CM from PC3 cells with CTSZ knockdown (shCTSZ) or 22RV1 overexpression (CTSZ-OE). I THP-1 macrophage migration assay: Comparative effects of CM from shNC, shCTSZ, and shCTSZ + IL-32 rescue conditions. J Transwell migration assay demonstrating IL-32-dependent chemotaxis: CM from Vector, CTSZ-OE, and CTSZ-OE + siIL32 groups. Data expressed as mean ± SD (n = 3 biological replicates). Significance determined by one-way ANOVA with post hoc Tukey test. *p < 0.05; **p < 0.01. CM: conditioned media CTSZ degrades TRA2A protein expression through peptidase activity While intracellular CTSZ is established as a peptidase through C-terminal protein cleavage, our study uncovers a novel role for CTSZ in upregulating IL-32 expression and secretion in tumor cells. The positive correlation between CTSZ and IL-32 production implies an indirect regulatory mechanism. To investigate CTSZ-mediated proteolytic targets, comparative proteomics of 22RV1 Vector vs. CTSZ-OE groups identified 31 downregulated and 22 upregulated differential expressed proteins (Fig. [122]6A). Notably, TRA2A, a top-ranked CTSZ-downregulated RNA-binding protein, is a known suppressor of pre-mRNA splicing and mature mRNA biogenesis [[123]17]. CatRAPID analysis predicted TRA2A-IL-32 regulatory interactions (Figure S3C). Additionally, SU2C analysis revealed that negative correlations between TRA2A and both IL-32/CTSZ (Figure S3D, E, ). Functional validation in PCa cells showed that CTSZ knockdown increased TRA2A protein levels without altering mRNA expression (Fig. [124]6B, C), indicating post-transcriptional regulation. MALDI-TOF analysis of TRA2A-CTSZ co-incubation revealed enhanced proteolytic cleavage, with recombinant CTSZ (500 μg/mL) generating 31 specific peptide peaks versus 20 in controls (Fig. [125]6D). Octet-based dose–response assays showed concentration-dependent TRA2A degradation, with 10–50 nM activated CTSZ efficiently cleaving sensor-immobilized TRA2A (Fig. [126]6E). These findings conclusively establish CTSZ as a direct protease catalyzing TRA2A degradation. Fig. 6. [127]Fig. 6 [128]Open in a new tab CTSZ promotes the splicing and expression of IL-32 by degrading TRA2A. A Volcano plot of proteomic sequencing comparing 22RV1-Vector vs. 22RV1-CTSZ-OE cells. B, C CTSZ knockdown validation: TRA2A mRNA (B) and protein (C) levels in PC3 cells. D MALDI-TOF MS analysis of CTSZ-mediated TRA2A proteolytic cleavage products. E Reflectometric interference spectroscopy quantification of CTSZ concentration-dependent TRA2A hydrolysis efficiency (Δλ shift). F In silico prediction of IL-32-TRA2A binding motifs via CatRAPID algorithm. G RIP-qPCR validation of TRA2A binding to IL-32 pre-mRNA regulatory sequences. H RNA pulldown assay demonstrating TRA2A interaction with IL-32 pre-mRNA probes. I Dual-color FISH co-localization of TRA2A (green) and IL-32 pre-mRNA (red). Scale bar = 10 μm. J EMSA showing nuclear protein binding affinity to IL-32 RNA probes: wild-type (WT), cold competitor (CC), and mutant (Mut) probes. K, L TRA2A knockdown effects on IL-32 expression: qRT-PCR K and immunoblot (L). The data are presented as the means ± SDs and were analyzed via two-tailed unpaired Student’s t test. Figure K was analyzed via one-way ANOVA; * p < 0.05; ** p < 0.01. ns: not significant TRA2A binds to IL-32 precursor mRNA and inhibits its splicing To define the specific TRA2A binding sites on IL-32 pre-mRNA, computational prediction using CatRAPID identified three potential interaction regions (nt 602–767, 1668–1833, 2297–2489) in IL-32 pre-mRNA (Fig. [129]6F). RNA immunoprecipitation (RIP) assays in PC3 cells confirmed preferential enrichment of nt 602–767 and 2298–2489 regions by TRA2A-specific antibodies compared to isotype controls (Fig. [130]6G). These regions contain a conserved GAAARGARR motif, a known recognition element for TRA2A’s RNA recognition motif (RRM) and arginine-rich domains [[131]17, [132]18]. Subsequently, RNA pull-down assays using biotinylated probes further validated direct interaction: TRA2A bound to wild-type nt 602–767 and 2297–2489 sequences but showed negligible binding to motif-mutated probes (Fig. [133]6H). Consistently, RNA fluorescence in situ hybridization (RNA FISH) revealed nuclear co-localization of TRA2A with IL-32 pre-mRNA at these motifs, with binding significantly reduced upon motif mutation (Fig. [134]6I). Moreover, Electrophoretic mobility shift assays (EMSA) showed stronger TRA2A binding to wild-type versus mutated or cold probes for both regions (Fig. [135]6J). Functional validation in PC3 cells showed that TRA2A silencing significantly upregulated IL-32 mRNA and protein levels (Fig. [136]6K, L). Collectively, these results demonstrate that TRA2A binds to the GAAARGARR motif in IL-32 pre-mRNA to suppress splicing. Elevated CTSZ promotes IL-32 maturation and secretion by proteolytically degrading TRA2A, thereby relieving translational repression. Targeting IL-32/ITGA5-PI3K/AKT axis disrupts protumoral M2 macrophage recruitment While the cellular receptor for IL-32 remains undefined, previous literature indicates that all IL-32 isoforms contain an RGD motif. Given the canonical role of RGD motifs in integrin binding, we performed integrin interaction screening in SU2C-metastatic PCa cohorts. This analysis identified ITGA5 as the strongest correlate of IL-32 expression, establishing a potential ligand-receptor relationship (Spearman = 0.43; Fig. [137]7A). Biochemical validation via co-IP confirmed direct IL-32/ITGA5 binding, which was RGD-dependent as GRGDNP treatment reduced affinity (Fig. [138]7B, C). Molecular docking (Gram-X) identified high-affinity interaction (ΔG = − 18.5kcal/mol) through LEU706/THR38 hydrogen bonds (Fig. [139]7D). Fig. 7. [140]Fig. 7 [141]Open in a new tab Targeting IL-32/ITGA5-PI3K/AKT axis disrupts protumoral M2 macrophage recruitment. A Correlation analysis of IL-32 and RGD-binding integrin receptors in the SU2C metastatic prostate cancer cohort (cBioPortal). B, C IL-32/ITGA5 interaction validation: (B) Co-immunoprecipitation (Co-IP) of IL-32 and ITGA5 in THP-1 macrophage lysates. C RGD motif dependency confirmed via GRGDNP inhibitor treatment. D Computational docking model of IL-32(Blue)/ITGA5(Yellow) complex. E KEGG pathway enrichment of ITGA5-associated genes in TCGA-PRAD cohort. F GLPG0187 dose-dependent effects on macrophage polarization: Immunoblot analysis of M1 (iNOS) and M2 (CD206) markers, PI3K/AKT pathway inhibition assessed by p-AKT (Ser473) levels. G Bioluminescence imaging (BLI) of lung metastasis in mice injected with 22RV1-CTSZ-OE cells ± GLPG0187 (n = 4/group). H Metastatic burden quantification: (H) Sirius Red-stained lung sections showing metastatic nodules (yellow arrows). I H&E quantification of lung metastases (blue arrows). J Statistical of tumor metastasis numbers. K Multiplex immunofluorescence of CD206 (M2, red), CD86 (M1, green), and DAPI (blue) in lung metastases. L IHC validation of CD206/CD86 expression across treatment groups. The data are presented as the means ± SDs and were analyzed via one-way ANOVA; *p < 0.05; **p < 0.01. ***p < 0.01; ns: not significant. 50 μm ( K, L); 2.5 mm (I). BE: binding energy Activation of the ITGA5-PI3K/AKT signaling axis has been established to promote tumor progression [[142]19–[143]22]. KEGG pathway analysis of ITGA5-associated differentially expressed genes in TCGA-PRAD confirmed PI3K/AKT as the top enriched signaling pathway (Fig. [144]7E). Pharmacological inhibition with GLPG0187 demonstrated dose-dependent suppression of PI3K/AKT phosphorylation and M2 macrophage polarization, with maximal inhibitory efficacy observed at a concentration of 40 ng/mL (Fig. [145]7F). To explore the effect of GLPG0187 on ITGA5 and downstream signaling in PCa cells. Western blot analysis revealed no significant changes in ITGA5 protein expression across all treatment groups compared to the wildtype cells (Figure S4A). Concomitantly, phosphorylation levels of PI3K/AKT (pPI3K/AKT) remained unchanged in GLPG0187-treated cells, as confirmed by densitometric quantification (Figure S4B). In vivo validation using a tail vein injection model with CTSZ-OE PCa cells showed that GLPG0187(100 mg/kg) treatment significantly reduced lung metastasis burden, as confirmed by bioluminescence imaging (Fig. [146]7G). Compared with the untreated and PBS-treated groups, the GLPG0187-treated group exhibited significantly reduced PCa lung metastasis with bitter acid and HE staining (Fig. [147]7H–J). Additionally, immunofluorescence and immunohistochemical analyses of lung tumor sections revealed that GLPG0187 markedly decreased CD206^+ M2 macrophage infiltration while increasing CD86^+ M1 macrophage populations (Fig. [148]7K, L). Collectively, our data suggest that the ITGA5/PI3K axis in macrophages mediates CTSZ-induced M2 macrophage infiltration and PCa metastasis. Discussion Our findings elucidate a non-canonical role for CTSZ in driving prostate cancer metastasis through orchestration of M2-polarized macrophage recruitment. Multi-omics analyses established CTSZ as a metastasis-associated protease that remodels the immunosuppressive niche without intrinsic tumor cell effects. Mechanistically, CTSZ-mediated degradation of the RNA splicing repressor TRA2A licenses IL-32 isoform maturation, which engages macrophage ITGA5 via its RGD motif. This interaction triggers PI3K/AKT-dependent chemotaxis and M2 macrophage infiltration. Therapeutic disruption of this axis using ITGA5 inhibitor GLPG0187 significantly attenuated lung metastasis in vivo, validating the CTSZ/TRA2A/IL-32/ITGA5 pathway as a druggable effector of metastatic progression. While the cathepsin family has been extensively studied for its matrix metalloproteinase (MMP)-dependent roles in extracellular matrix (ECM) remodeling and tumor invasion [[149]8], CTSZ represents a functional dichotomy. As a carboxypeptidase lacking direct ECM degradation capacity [[150]23], CTSZ exerts dual functionality through its unique structural determinants: (1) The extracellular RGD motif facilitates integrin receptor binding to modulate cell adhesion/migration; (2) Intracellularly, it serves as a key lysosomal enzyme mediating proteolytic processing of PolyQ peptides, chemokines (CXCL12), and pathogenic proteins (huntingtin) in disease contexts [[151]12, [152]24–[153]27]. This dual nature positions CTSZ as a molecular rheostat coordinating extracellular signaling and intracellular proteostasis. Despite documented CTSZ upregulation in multiple malignancies, its tumor-intrinsic regulatory mechanisms remain incompletely understood. Current paradigms emphasize CTSZ-integrin interactions in regulating angiogenesis and metastatic dissemination, yet critical knowledge gaps persist regarding its tumor microenvironment (TME) remodeling capacity. Seminal work by Akkari et al. revealed macrophage-derived CTSZ promotes pancreatic cancer invasion via tumor cell integrin engagement [[154]9]. Contrastingly, our findings establish an inverse regulatory axis: Tumor cell-intrinsic CTSZ orchestrates TME immunosuppression through TRA2A/IL-32/ITGA5-mediated M2 macrophage recruitment—a mechanism substantiated by three lines of evidence. First, CTSZ-mediated TRA2A degradation relieves suppression on IL-32 mRNA splicing. Second, secreted IL-32 activates macrophage ITGA5/PI3K signaling to drive M2 macrophage infiltration. Third, pharmacological ITGA5 inhibition reverses CTSZ-driven metastasis in vivo. The TRA2A/IL-32/ITGA5 signaling axis uncovered in this study introduces new dimensions to cancer immunology, yet warrants deeper mechanistic interrogation. While IL-32 exists as nine isoforms with divergent tumor-modulatory roles [[155]28], our focus on its macrophage-polarizing function aligns with consensus findings in esophageal cancer and myeloma, where IL-32β/γ isoforms drive M2 polarization [[156]29]. Importantly, we identified TRA2A as a gatekeeper of IL-32 mRNA maturation—a regulatory layer distinct from canonical transcriptional control. Three lines of evidence substantiate this paradigm: (1) Transcriptome profiling of CTSZ-knockdown PCa cells revealed IL-32 downregulation, implicating post-transcriptional regulation; (2) Proteomic analysis pinpointed TRA2A among top CTSZ-downregulated RBPs (RNA-binding proteins); (3) Biochemical validation confirmed CTSZ-mediated TRA2A proteolysis and its steric hindrance on IL-32 pre-mRNA splicing via conserved GAAARGARR motif binding. We detected IL-32mRNA levels in non-tumoral prostate epithelial cells (RWPE-1) and prostate cancer cells (PC3, DU145) (Figure S3B). The results revealed that IL-32 expression was significantly lower in RWPE-1 cells compared to PC3 and DU145 cells. While this confirms minimal IL-32 contribution from non-tumoral epithelium, we did not assess IL-32 expression in other TME components, such as immune or stromal cells. This finding helps to preliminarily exclude the potential influence of non-cancerous IL-32 on our experimental results. Current research on ITGA5 predominantly explores its translational potential in clinical therapeutics, with GLPG0187 and volociximab emerging as promising targeted agents. Preclinical evaluation through a phase I clinical trial demonstrated GLPG0187’s inhibitory effects on tumor progression and metastasis across multiple solid tumor models [[157]30]. Notably, while volociximab has exhibited therapeutic efficacy in metastatic lung and breast cancer management [[158]31, [159]32], emerging evidence highlights GLPG0187’s specific potency in suppressing osseous metastasis of prostate carcinoma [[160]33]. Our novel finding through orthotopic prostate tumor analysis reveals M2 macrophages as the primary cellular target of GLPG0187, providing a distinct mechanistic perspective. It is noteworthy that neither compound has entered clinical application for prostate cancer treatment, thereby limiting direct assessment of their therapeutic efficacy in this malignancy. This evidence gap underscores the significance of our study in establishing theoretical foundations for future clinical translation of these agents in prostate cancer management. Our findings position the CTSZ/IL-32/ITGA5 axis as a novel immunomodulatory target distinct from current macrophage-focused therapies. While CSF1R inhibitors aim to deplete TAMs broadly, their clinical efficacy in solid tumors has been limited by compensatory immune evasion and stromal resistance [[161]34]. In contrast, targeting ITGA5 specifically disrupts CTSZ-driven M2-TAM recruitment without globally suppressing myeloid populations, potentially preserving antitumor immunity mediated by dendritic cells and M1 macrophages. Furthermore, the observed immunotherapy resistance in prostate cancer, characterized by diminished immune cell infiltration (excluding macrophages) as evidenced by our database analysis, suggests macrophage-mediated immune evasion mechanisms. This finding reinforces the therapeutic rationale for targeting tumor-associated macrophages within the tumor microenvironment to overcome treatment resistance. From a diagnostic perspective, the potential utility of CTSZ as a circulating biomarker for metastatic prostate cancer warrants further investigation. We propose that integrating macrophage-targeted therapeutic strategies with existing treatment modalities may yield synergistic antitumor effects, offering new avenues for combinatorial therapy development. Conclusion This study elucidates a CTSZ-driven metastatic axis in cancer. Tumor cell–expressed CTSZ promotes TRA2A degradation, thereby unleashing IL-32-mediated activation of macrophage ITGA5/PI3K signaling to orchestrate protumoral M2 macrophage infiltration. Therapeutically, targeting this axis with GLPG0187 specifically disrupts tumor microenvironment (TME) immunosuppression by blocking ITGA5 in M2 macrophages. This mechanism differs from conventional tumor cell–directed therapies. These findings not only reveal a novel proteolytic–immune crosstalk pathway governing metastasis but also establish ITGA5 inhibition as a clinically actionable strategy for reprogramming the metastatic niche. Supplementary Information [162]12967_2025_6865_MOESM1_ESM.jpg^ (2MB, jpg) Supplementary Material 1: Figure S1..Expression of COCH in GSE32269 and GSE101607 datasets.Frequency of genomic alterations in the CTSZ in nine prostate cancer genomic sequencing datasets from the cBioPortal database. Relationships between CTSZ genomic alterations and overall patient survival.Survival analysis of patients in gene amplification group and non- amplification group. Data presented as mean ± SD. Statistics: unpaired t-test; *p<0.05 [163]12967_2025_6865_MOESM2_ESM.jpg^ (2.8MB, jpg) Supplementary Material 2: Figure S2..Western Blot assay to verify the efficiency of CTSZ overexpression in RM1 cells.Validation of CTSZ expression and its correlation with T cell, B cell, and Treg cell infiltration in the TIMER2.0 database [164]12967_2025_6865_MOESM3_ESM.jpg^ (1.9MB, jpg) Supplementary Material 3: Figure S3. TRA2A may be a key protein that negatively regulates IL-32 expression.Original expression matrix of 10 differentially secreted proteins in the cytokine signaling pathway of transcriptome sequencing.Top ten ranked RNA-binding protein prediction scores for IL-32.Correlation between IL-32 and TRA2A levels in the SU2C-mPCa database.Correlation between TRA2A and CTSZ expression in the SU2C-mPCa database.Transwell experiment detects the effect of conditioned medium overexpressing TRA2A on M2 macrophage recruitment in cells overexpressing CTSZ [165]12967_2025_6865_MOESM4_ESM.jpg^ (850.6KB, jpg) Supplementary Material 4: Figure S4. GLPG0187 did not affect the expression of ITGA5 and activation of the PI3K/AKT pathway in PC3 cells.Western Blot experiment verifies the expression of ITGA5 in PC3 treated with different concentrations of GLPG0187.Western Blot experiment verifies the changes in PI3K/AKT phosphorylation levels of PC3 treated with different concentrations of GLPG0187 Acknowledgements