Clinical characteristics of the study samples
A summary of the participants' baseline characteristics is shown in Table 1, which encompassing age, Gleason score, TNM stage, lymph node metastasis, and PSA serum levels. Serum and urine samples were collected from 93 participants, including control (n = 45), and PCa (n = 48) cases.
Table 1
Associations between urine-derived exosomal PSM-E expression and clinicopathological characteristics of PCa patients
Characteristic | Cases (%) (n = 48) | PSM-E expression (%) | p |
high | low |
Age |
< 66 | 16 (33.3) | 8 | 8 | 0.838 |
≥ 66 | 32 (66.7) | 17 | 15 |
Gleason score |
≤ 6 | 6 (12.5) | 2 | 4 | 0.028 * |
3 + 4 | 12(25) | 5 | 7 |
4 + 3 | 11(22.9) | 8 | 3 |
≥ 8 | 19 (39.6) | 10 | 9 |
TNM stage |
I/II | 17 (35) | 5 | 12 | 0.003 ** |
III/IV | 31 (65) | 20 | 11 |
Lymph node metastasis |
Yes | 6 (12.5) | 2 | 4 | 0.383 |
No | 42 (87.5) | 20 | 22 |
fPSA |
< 4 ng/ml | 40 (83.3) | 19 | 21 | 0.113 |
≥ 4 ng/ml | 8 (16.7) | 6 | 2 |
p value < 0.05 was considered significant. *p < 0.05, **p < 0.01, and ***p < 0.001 |
PSM-E is encapsulated within serum and urine-derived exosomes from PCa patients
Exosomes were isolated from serum or urine samples obtained from the participants using the corresponding commercial kits. Subsequently, exosomes purified from 5 samples underwent transmission electron microscopy and Western blotting analysis to detect PSM-E protein levels. The electron micrographs of exosomes revealed vesicle sizes primarily around 100 nm (Fig. 1A, B). Notably, PSM-E was enriched in serum-derived exosomes from BPH and PCa patients, and abundant in those isolated from urine samples of BPH and PCa patients (Fig. 1C, D). Importantly, PSM-E levels in PCa patients were significantly higher than those in control group. In contrast, exosomes from the control group did not contain detectable PSM-E, with CD63 and flotillin-2 as exosomal control markers. In summary, these findings demonstrate that PSM-E is found in both the serum and urine of BPH and PCa patients.
Urine-derived exosomal PSM-E yielded a good diagnostic performance for PCa
In an attempt to investigate the value of urine-derived exosomal PSM-E in PCa, LC-MS/MS analysis was used to detect the protein level of PSM-E in the exosomes of each group by recognizing the specific peptide FLAAYACTGCLAER of PSM-E. As shown in Fig. 1E, urine-derived exosomal PSM-E concentration was significantly elevated in the PCa patients compared to the control group (p < 0.001). Exosomal PSM-E expression exhibited significantly higher levels in patients with PCa than those with BPH (p < 0.01). Remarkably, high level of exosomal PSM-E positively correlated with a high Gleason score (≥ 8, p < 0.05) and advanced pathological tumor stage (III/IV TNM stage p < 0.01) (Fig. 1F, G,).
To analyze the diagnostic specificity and sensitivity of exosomal PSM-E for PCa compared with those of tPSA, ROC curves for the detection of PCa were applied. As shown in Fig. 1H, I, in all PCa subjects, the values of AUC were 0.8904 (95% CI, 0.8311 to 0.9497) for exosomal PSM-E, 0.5295(0.3812 to 0.6779) for tPSA and 0.7189 (95% CI, 0.5890 to 0.8487) for f/t PSA. These differences between exosomal PSM-E and f/t PSA were statistically significant in all PCa subjects (p < 0.0001 and p < 0.001). Using a cutoff value of 42.08 ng/ml for exosomal PSM-E, exosomal PSM-E level significantly differentiated PCa patients from control cases among consecutive patients with 75% sensitivity and 80% specificity for diagnosing PCa. These results suggest that exosomal PSM-E exhibited good performance in distinguishing PCa and outperformed other traditional clinical biomarkers tPSA or f/t PSA.
PSM-E can be exported from cells by exosomes
We next sought to investigate whether PSM-E is present in cell-derived exosomes. Specifically, PSM-E was overexpressed in 293T and PC3 cells, which had low basal levels of PSM-E (Supplementary Figure S1A), through transfection with the PSM-E-Flag construct. Subsequently, culture medium supernatants were collected and processed through a series of steps, which included two rounds of centrifugation at 4°C so as to remove cells and cell debris. The resulting supernatants were further passed through 0.22-µm filters. After this, using a centrifugal filter unit (Amicon Ultra-15) with a 3 kDa cutoff value, the supernatants were concentrated ten-fold. As depicted in Fig. 2A and 2B, our findings demonstrated the presence of PSM-E-Flag protein both in the cytoplasm and the in culture medium supernatants. Additionally, Western blotting analysis, utilizing a PSM-E-specific antibody, confirmed the presence of PSM-E protein in the supernatant derived from LNCaP cells with high basal levels of endogenous PSM-E (Fig. 2C). Meanwhile, we constructed siRNAs targeting the back-splice region of PSM-E (PSM-E siRNA) to depleted basal-level PSM-E expression in LNCaP cells (Supplementary Figure S1B).
Since a putative signal peptide is not present in the amino acid sequence of PSM-E, we hypothesized that PSM-E is exported via an exosome-mediated mechanism. To validate this hypothesis, we isolated exosomes from the culture media of various cell lines, including 293T cells transfected with PSM-E (293T-PSM-E), 293T cells transfected with control vector (293T-Vector), PSM-E-transfected PC3 cells (PC3-PSM-E), control vector-transfected PC3 cells (PC3-Vector), LNCaP cells, PSM-E siRNA transfected LNCaP cells (LNCaP-siRNA), and a non-targeting control siRNA transfected LNCaP cells (LNCaP-siNC). As shown in Fig. 2D-F, electron microscopy revealed the typical double-layer membrane structure of exosomes with an approximate diameter of 100 nm. Notably, PSM-E was also abundant in those exosomes derived from 293T-PSM-E, PC3-PSM-E and LNCaP-siNC cells. In contrast, exosomes obtained from 293T-Vector, and PC3-Vector did not exhibit detectable levels of PSM-E, and transfection of PSM-E-siRNA efficiently depleted PSM-E expression level in these exosomes (Fig. 2G-I). These results were validated using CD63 and flotillin-2 as exosomal control markers (Fig. 2G-I). The presence of PSM-E in the exosome composition was further confirmed using mass spectrometry (Fig. 2J). Collectively, these data demonstrate that exosome secretion of PSM-E is not limited to LNCaP cells, which express high levels of endogenous PSM-E, but also occurs in 293T and PC3 cells transiently expressing exogenous PSM-E.
Exosomal PSM-E can be transferred to recipient cells
Exosomes are protein-containing extracellular vesicles known to play a major role in protein transport among cells. In light of the discovery that PSM-E is contained in exosomes and can be exported from cells expressing it, we investigated whether PSM-E can be transferred to recipient cells. To this end, we tested such a possibility by employed a co-culture system. After transfecting PC3 and 293T cells with a Flag epitope-tagged PSM-E plasmid, these cells were co-cultured with M0 macrophages in the transwell plates (Fig. 3A). Our data demonstrated that the Flag epitope-tagged PSM-E from the upper transwells was delivered into the recipient M0-type THP-1 cells, which were seeded in the lower wells (Fig. 3A), confirming that cells can indeed secrete extracellular PSM-E that is taken up by M0 macrophages.
Next, we tested whether recipient cells could take up exosomal PSM-E when treated with purified exosomes derived from PSM-E-expressing cells. We differentiated the human monocyte cell line THP-1 into M0-like macrophages using PMA, characterized by their adherent morphology and elevated expression of the macrophage marker CD68 (Supplementary Figure S2A). Subsequently, these M0 macrophages were incubated with exosomes purified from different sources, including PC3-Vector, PC3-PSM-E, LNCaP-siNC, and LNCaP-siPSM-E. As shown in Fig. 3B, an increased abundance of PSM-E in the recipient M0 macrophages treated with exosomes from PC3-PSM-E (PC3-Vector-Exos and PC3-PSM-E-Exos, 80 µg/ml). Additionally, the identification of PSM-E proteins in the lysates of recipient M0 macrophages treated with exosomes from LNCaP-siNC and LNCaP-siPSM-E further confirmed the transfer of PSM-E (Fig. 3C). A fluorescent dye PKH67 was added to the culture medium of M0 macrophages to identify these exosomes derived from cells. After 12 hours, the THP-1 cells exhibited efficient uptake of the cell-secreted exosomes, as indicated by the presence of green fluorescence staining in M0 macrophages (Fig. 3D).
Furthermore, to validate that exosomal transfer of PSM-E was responsible for the increase in PSM-E protein levels in recipient cells, PSM-E levels was measured in the recipient cells treated with various concentrations of PSM-E-containing exosomes. As shown in Fig. 3E, F, the finding demonstrated that the levels of PSM-E proteins in the lysates of recipient M0 macrophages and PC3 cells treated with PSM-E-laden exosomes derived from 293T-PSM-E cells (at 40 µg or 80 µg) were dose-dependent, as confirmed by immunoblotting. Importantly, when an extracellular vesicle secretion inhibitor, GW4869, was added to the cells 24 hours prior, it blocked exosome production and PSM-E delivery into the recipient M0-THP-1 cells, indicating that cells primarily secrete extracellular PSM-E in an exosome-dependent manner (Fig. 3G). Overall, these results demonstrate that prostate cancer cells can secrete PSM-E-containing exosomes efficiently taken up by recipient cells.
Exosomal PSM-E inhibits PCa cell migration, invasion, and the polarization of M0 macrophage to the M2 phenotype
Our previous studies revealed that PSM-E could suppress the proliferation, migration, and invasiveness of prostate cancer cells18,19. To explore the effects of exosomal PSM-E on tumor migration and invasion, we conducted wound healing assays and transwell invasion experiments on recipient PC3 cells treated with exosomes derived from 293T-PSM-E and 293T-Vector cells at different concentrations (40 µg or 80 µg/ml). The results indicated that 293T-PSM-E-Exosomes inhibited PC3 cell migration and invasion compared to 293T-Vector-Exosomes (Fig. 4A, B). These results suggest that exosomal PSM-E can also suppress the invasive and metastatic abilities of PC3 cells.
Our previous findings revealed that PSM-E was specifically overexpressed in PCa and its expression is negatively correlated with the expression of CD206 (a specific marker of M2-type macrophages), which suppressing the secretion of inflammatory cytokines in the PCa microenvironment and inhibiting the chemotaxis of monocytes 23. In this study, we focused on investigating PSM-E's potential impact on the tumor microenvironment in the context of PCa progression 23. Additionally, M0 macrophages were treated with IL-4 to induce their differentiation into M2-like macrophages (Supplementary Figure S2B). Real-time RT-PCR analysis revealed a significant increase in the levels of M2 markers, including CCL17, CCL18, and CCL22, in M2 macrophages (Supplementary Figure S2C). Subsequently, we examined the effects of 293T cell-derived exosomal PSM-E on the polarization of M0 macrophages towards the M2 phenotype. The expression levels of M2 markers (CCL17, CCL18, and CCL22) markedly decreased in M0 macrophages co-treated with 293T-PSM-E-Exos and IL-4, compared to those in the 293T-vector-Exos and IL-4 treatment groups (Fig. 4D), suggesting that the proportion of M2-type macrophages in THP-1 cells was reduced following treatment with PSM-E exosomes. In contrast, there was no change in the expression of M1 macrophage markers (iNOS and IL-1β) after similar treatment with 293T cell-derived exosomal PSM-E (Fig. 4E). As shown in Fig. 4F, our results demonstrated that the levels of CD206 proteins in the lysates of recipient THP-1 macrophages (THP-1 treated with PMA and IL-4) exposed to PSM-E-laden exosomes derived from 293T-PSM-E-cells (40 µg, 80 µg) exhibited a dose-dependent decrease, as determined by immunoblotting. In summary, the above findings confirm that cell-derived exosomal PSM-E can suppress the M2 polarization of macrophages.
PMS-E colocalizes and interacts with RACK1
We further sought to explore the mechanisms underlying the inhibitory effect of PCa-derived exosomal PSM-E on the M2 polarization of macrophages. To this end, using the protein G-conjugated IgG antibody or anti-Flag monoclonal antibody, lysates from PC3-PSM-E-Flag cells were immunoprecipitated (Fig. 5A), followed by a pull-down experiment, and subsequently analyzed the immunoprecipitates by LC-MS/MS (Fig. 5A). We screened the putative interacting proteins with PSM-E obtained from IP-MS analysis based on the unique peptides (unique peptides ≥ 2) and peptide spectrum scores ranking (Fig. 5A). Notably, RACK1 was picked out as a potential candidate protein, and the mass spectrometer of the RACK1 peptide segment was depicted in Fig. 5B. To validate that PSM-E interacts with RACK1, we performed co-immunoprecipitation (co-IP) assays after co-transfection of PSM-E-Flag and RACK1-HA expression plasmids in 293T cells. Significantly, as shown in Fig. 5C and 5D, our results showed that RACK1-HA was precipitated by the Flag antibody, meanwhile, PSM-E-Flag was precipitated by the HA antibody, revealing the formation of a complex between PSM-E-Flag and RACK1-HA. Furthermore, the interaction between Flag-tagged PSM-E and RACK1 was investigated using an IP assay using endogenous RACK1 as a precipitate, which resulted in the successful pull-down of endogenous RACK1 together with Flag-tagged PSM-E (Fig. 5E).
To identify the specific domains of PSM-E responsible for its interaction with RACK1, we generated two deletion mutants that removed the protease-associated domain (PA) and the peptidase M28 domain (M28) of PSM-E. Co-IP assays revealed that the protease-associated domain of PSM-E was crucial for its interaction with RACK1 (Fig. 5F). It is well-established that RACK1 belongs to the tryptophan- aspartate repeat (WD-repeat) proteins family which share significant homology with the β subunit of G-proteins. We generated five serial WD domain deletion constructs, including full-length (FL), WD2-7, WD3-7, WD4-7, and WD5-7 (Fig. 5G). Co-IP assays using an anti-HA antibody and subsequent immunoblotting with an anti-Flag antibody showed that the fourth WD domain of RACK1 was essential for its interaction with PSM-E (Fig. 5G). Taken together, these results indicate that the protease-associated domain of PSM-E and the fourth WD domain of RACK1 are crucial for the interaction between PSM-E and RACK1.
Exosomal PSM-E PCa growth and correlates with low infiltration of M2 macrophage in vivo
To assess the anti-PCa activity of exosomal PSM-E in vivo, we established a subcutaneous PCa tumorigenesis model in immunocompetent C57BL/6J mice. In this model, we subcutaneously injected RM-1 cells into C57BL/6J mice to develop solid tumors until the tumors reached a volume of approximately 50 mm3 (Supplementary Figure S3A). We then monitored the tumor growth after intraperitoneal injections of exosomes derived from PC3 cells with exogenously overexpressed PSM-E (PC3-Vector-Exos, PC3-PSM-E-Exos) or LNCaP cells with high basal levels of endogenous PSM-E (LNCaP-siNC-Exos and LNCaP-siPSM-E -Exos) every three days for four weeks (Supplementary Figure S3A). As shown in Supplementary Fig. 6A and Figure S3B, the volume of the homograft PCa tumors was significantly reduced by 43.6in mice treated with PC3-PSM-E-Exos. The weight of the homograft tumors at the experimental endpoint was also notably reduced by 49.12 ± 3.81% in C57BL/6 mice treated with PC3-PSM-E-Exos (Fig. 6B). Conversely, compared to the LNCaP-siNC-Exos group, the volume and weight of tumors derived from the LNCaP-siPSM-E-Exos group were significantly increased (Fig. 6C, D and Figure S3C). IHC analysis of mouse homograft tumors revealed a significantly reduced expression of CD206+ (a specific marker of M2-type tumor-associated macrophages) in the PC3-PSM-E-Exos treatment group, while a higher expression of CD206+ was observed in the LNCaP-siPSM-E-Exos treatment group (Fig. 6E). Furthermore, the results revealed higher expression of the proliferation marker Ki67 in the tumors derived from the LNCaP-siPSM-E-Exos group compared to those in the PC3-PSM-E-Exos group (Fig. 6E). These findings suggest that exosomal PSM-E can suppress tumor growth and correlate with a lower infiltration of M2 macrophages in vivo.
Exosomal PMS-E inhibits M2 polarization of macrophages via suppression of the ERK and FAK signaling pathways
Numerous studies have indicated that the activation of the ERK signaling pathway in macrophages is involved in the M2 polarization of macrophages [27]. Moreover, RACK1 has been reported to regulate the ERK and FAK pathway in various cellular activities [28]. To gain a deeper understanding of the RACK1 signaling cascades that mediate the inhibitory effect of exosomal PMS-E on M2 macrophage polarization, we examined changes in the phosphorylation status of RACK1 downstream substrates, such as ERK and FAK, using Western blotting. As shown in Fig. 7A and 7B, exosomal PSM-E led to a significant decrease in the levels of phosphorylated FAK and ERK (p-FAK and p-ERK) without affecting the expression of total ERK and FAK in both PC3 and M0 macrophages (THP-1 macrophage cells induced by PMA) in a dose-dependent manner. In contrast, the attenuation of PSM-E expression in LNCaP cells through transfection with PSM-E siRNA (siPSM-E) reversed the activation of the ERK/FAK pathway (Fig. 7C). Notably, M2-like macrophages treated with FR180204 (an ERK inhibitor) for 12 hours exhibited a significant decrease in phosphorylated ERK and FAK levels without affecting the expression of total ERK and FAK (Fig. 7D). As expected, CD206 expression decreased in M2 macrophages treated with 293T-PSM-E-Exos (Fig. 7E). In summary, these results suggest that exosomal PSM-E could inhibit M2 macrophage polarization via suppressing the RACK1-ERK/FAK signaling pathway.