3.1 CAFs-Derived Exosomes Enhance OSCC Cell Proliferation, Migration, as well as Invasion
CAFs as well as NFs were isolated from oral squamous cell carcinoma and surrounding non-tumor tissues, respectively. They exhibited a spindle shape and were confirmed as fibroblasts through positive immunofluorescence staining for fibroblast-specific markers (Fig. 1A). Compared to NFs, CAFs showed elevated levels of α-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP) (Fig. 1A-C). Exosomes, pivotal for cellular communication within the tumor microenvironment, were isolated from CAFs and NFs-conditioned mediums (CM) via ultracentrifugation[39]. Their presence was validated by western blotting and electron microscopy, revealing an average diameter of 219 nm (range 30–220 nm) and characteristic double-layered membranes (Fig. 1D,F). Elevated levels of exosomal markers CD63 and TSG101 and the absence of calnexin, an endoplasmic reticulum marker, were noted in fibroblast-derived exosomes (Fig. 1E). The incorporation of exosomes derived from fibroblasts into OSCC cells was demonstrated through PKH26 fluorescent dye labeling; red fluorescence observed in SAS and CAL27 cells after three hours of co-culture indicated successful absorption of these exosomes by OSCC cells (Fig. 1G).
An investigation into the effects of CAFs-secreted exosomes on OSCC cell proliferation, invasion, and migration was conducted. Experiments with SAS and CAL27 cell lines demonstrated that CAFs-derived exosomes significantly augmented cell proliferation compared to exosomes from NFs, as shown by CCK8 and EdU assays (Fig. 2A,B). Furthermore, the presence of CAFs-derived exosomes considerably elevated the migratory and invasive capabilities of both SAS and CAL27 cells (Fig. 2C). Treatment with CAFs-secreted exosomes resulted in the upregulation of mesenchymal markers Twist1 and Vimentin and a decrease in the epithelial marker E-cadherin in both cell lines (Fig. 2D).
To further delineate the contributions of exosomes to OSCC cell dynamics, particularly focemploying on growth, migration, as well as invasion, supernatants devoid of exosomes were obtained through ultracentrifugation. It was determined that CAFs facilitated oral cancer cell proliferation and migration via exosomes rather than via co-purifying complexes found in ultracentrifuged pellets (Fig. S1A,B), indicating a pivotal role of CAFs-secreted exosomes in OSCC cell dynamics.
3.2 piR-35462 Enrichment in CAFs-Derived Exosomes Correlates with OSCC Patient Survival
Exosomes are known to be rich in small non-coding RNAs, crucial for intercellular communication[40]. Through small RNA-sequencing of exosomes extracted from three pairs of CAFs and NFs, numerous small RNAs, particularly piRNAs, exhibited differential expression (adjusted p-value less than 0.05 and fold change more than 2) between the two groups (Fig. 3A). Further analysis of the highest ranked ten piRNAs from 20 pairs of NFs or CAFs-derived exosomes revealed a significant increase in piR-35462 and piR-60565 levels in CAFs-derived exosomes (Fig. 3B). Additionally, the assessment of piRNA expressions was carried out in 76 matched pairs of OSCC and adjacent normal tissues. qPCR analysis showed elevated levels of piR-35462, but not piR-60565, in OSCC tissues (Fig. 3C, Figure S2A). Elevated piR-35462 expression was also significant in both recipient OSCC cells and CAFs (Fig. 3D,E, and S2B,C). PiR-35462 expression was notably associated with the N stage, with no significant correlation to age, gender, T stage, or pathological stage (Table 4). High levels of piR-35462 were indicative of reduced overall survival in OSCC patients (Fig. 3E), suggesting a potential oncogenic role of piR-35462 in OSCC progression.
Table 4
Baseline data table of 76 patients
| constitute(%) |
---|
Category | piR-35462 high expression (n = 38) | piR-35462 low expression (n = 38) | P value |
Age | 45.3 (32–81) | 51.4 (30–86) | 0.562 |
Gender | | | |
Male | 20 (52.63) | 25 (65.22) | |
Female | 18 (47.37) | 13 (34.21) | 0.243 |
Smoking or not | | | |
Non-smoking | 17 (44.74) | 20 (52.63) | |
Smoking | 21 (55.26) | 18 (47.37) | 0.491 |
N stagea | | | |
N0 | 13 (34.21) | 26 (69.57) | |
N1 + N2 | 25 (65.79) | 12 (31.58) | 0.003 |
Stagesa | | | |
I | 13 (34.21) | 15 (39.47) | |
II | 8 (21.05) | 10 (26.32) | |
III | 10 (26.32) | 7 (18.42) | |
IV | 7 (18.42) | 6 (15.79) | 0.808 |
aAmerican Joint Committeeon Cancer,7th Edition staging. |
To ascertain if the elevation of piR-35462 in OSCC cells resulted from transfer via CAFs-derived exosomes, actinomycin D was introduced into the co-culture system. This intervention did not alter piR-35462 levels in OSCC cells, negating the theory of an endogenous source for the increase (Figure S2D). Reduction of exosome secretion was achieved through treatment with GW4869 (an nSMase inhibitor) or knockdown of Rab27a via shRNAs, which predictably decreased piR-35462 and piR-60565 levels in CM from CAFs (Fig. 3G, S2E). RNA fluorescence in FISH analysis disclosed the presence of piR-35462 within both the nucleus and cytoplasm of OSCC cells following exosome uptake (Fig. 3H). Collectively, these findings demonstrate that piR-35462 is upregulated in exosomes from CAFs and in OSCC tissues, correlating with an adverse prognosis for OSCC patients.
3.3 Augmentation of Exosomal piR-35462 in CAFs Enhances Oral Cancer Progression
The impact of CAFs-released exosomal piR-35462 on OSCC advancement was further investigated. To assess the capability of CAFs to transfer exosomal piR-35462 to OSCC cells, exosomes from CAFs, either transfected with piR-35462 mimics or a control, were introduced to SAS and CAL27 cell cultures. Notably, after 24 hours of exposure to exosomes from CAFs genetically modified to express Cy3-tagged piR-35462 mimics, both CAL27 and SAS cells displayed red fluorescence (Figure S3A). Subsequent qRT-PCR analysis post-24-hour exosome exposure revealed a significant increase in piR-35462 within the recipient cells (Fig. 4A). The addition of exosomes from CAFs overexpressing piR-35462 to the OSCC cell culture media led to a marked enhancement in cell proliferation, as evidenced by CCK-8 and EdU assays (Fig. 4B,C, and S3B). Furthermore, exosomal piR-35462 positively influenced OSCC cell migration and invasion under co-culture conditions (Fig. 4D). These findings validate the ability of OSCC cells to internalize exosomes laden with piR-35462 mimics, promoting OSCC progression.
Subsequent experiments involved transfecting CAL27 and SAS cells with piR-35462 mimics or antagomir-35462 to investigate the influence of piR-35462 on OSCC growth and metastatic capabilities (Figure S3C). Proliferation assays, including CCK-8 and EdU, revealed that piR-35462 significantly accelerated cellular proliferation in CAL27 and SAS cells compared to controls, an effect that was counteracted by antagomir-35462 (Fig. 5A-C). Moreover, enhanced cell migration and invasion were observed following piR-35462 overexpression, whereas its suppression notably curtailed OSCC cell metastasis (Fig. 5D). Given the pivotal role of EMT in cancer metastasis, we assessed the impact of altering piR-35462 levels on EMT marker expression. Western blot results indicated that elevating piR-35462 levels resulted in upregulation of mesenchymal markers and downregulation of epithelial markers (Figure S3D). Collectively, these findings underscore the potent role of piR-35462 in fostering OSCC cell proliferation, invasion, and migration.
3.4 Exosomal piR-35462 from CAFs facilitates epithelial-mesenchymal transition (EMT) in OSCC cells through the FTO/Twist1 signaling pathway
To explore the influence of piR-35462, carried by exosomes from CAFs, on the behavior of OSCC cells, sequencing of RNA was conducted on CAL27 cells treated with exosomes from CAFs and NFs. This analysis identified distinct gene expression profiles between the groups, with a notable upregulation of genes associated with EMT in cells treated with CAFs-derived exosomes, particularly highlighting Twist1 as significantly elevated (Fig. 6A-C). This led to the hypothesis that piR-35462 from CAFs-derived exosomes might facilitate metastasis via Twist1-mediated EMT processes. Subsequent investigations confirmed that CAFs-derived exosomes carrying piR-35462 indeed augmented the expression of Twist1 as well as Vimentin within OSCC cells, a process mitigated by inhibiting piR-35462 (Fig. 6D). Given piRNA's lack of complete complementarity to its mRNA targets, further analysis revealed no direct binding sites between piR-35462 and Twist1 mRNA, suggesting an indirect regulatory mechanism.
The literature suggests piRNAs influence m6A methylation, prompting an examination of m6A levels post-piR-35462 inhibition[21, 24, 41], revealing a reduction in m6A methylation in OSCC cells (Figure S3F). Analysis of m6A regulatory proteins, which could potentially interact with piR-35462, showed decreased FTO expression following piR-35462 inhibition (Fig. 6E). Notably, a potential interaction site between piR-35462 and the 3’UTR of FTO mRNA was identified, indicating that piR-35462 may indirectly modulate Twist1 expression via FTO and m6A methylation adjustments (Fig. 6F). Dual luciferase reporter assays further verified this regulatory pathway, showing reduced luciferase activity in cells with piR-35462 inhibition, an effect reversible by mutating the predicted binding site (Fig. 6G). Additionally, piR-35462 overexpression was found to stabilize FTO mRNA, as evidenced by actinomycin D assays (Fig. 6H), and Western blot analysis confirmed that CAFs-derived exosomes upregulated FTO expression, an effect nullified by piR-35462 depletion (Fig. 6D). Furthermore, downregulating FTO via shRNAs counteracted the EMT gene expression changes induced by CAFs-derived exosomes (Figure S3G), with a positive correlation between FTO and Twist1 expressions observed within the TCGA database (Figure S3H). Collectively, these findings suggest that exosomal piR-35462 from CAFs promotes OSCC cell EMT through the FTO/Twist1 signaling pathway, presenting a novel mechanism of OSCC progression and a potential target for therapeutic intervention.
3.5 piR-35462-containing Exosomes Derived from CAFs Promote OSCC Progression in a Xenograft Animal Model
In an in vivo study, we validated the impact of CAFs-derived exosomal piR-35462 on OSCC progression by administering a lateral dorsal subcutaneous injection of a SAS cell mixture with either NFs, CAFs, or CAFs with shRab27a knockdown into nude mice. Tumors co-injected with CAFs exhibited significantly accelerated growth compared to those injected with NFs. Conversely, tumors from the CAFs-shRab27a group demonstrated reduced growth rates (Figs. 7A–C). Further analysis via qPCR revealed elevated levels of piR-35462 and Twist1 in tumors co-cultured with CAFs compared to those with CAFs-shRab27a (Fig. 7D,E). Moreover, tumors from SAS cells mixed with CAFs showed decreased E-cadherin expression and increased Vimentin and Twist1 levels, indicative of enhanced EMT, a trend reversed in the CAFs-shRab27a group (Fig. 7F). These findings underscore the function of CAFs-derived exosomal piR-35462 in promoting OSCC tumor growth and metastasis by facilitating EMT in a xenograft mouse model.