Skip to main content
World Journal of Pediatric Surgery logoLink to World Journal of Pediatric Surgery
. 2025 Jun 24;8(3):e000993. doi: 10.1136/wjps-2024-000993

Exosome encapsulation of miR-205-5p suppresses neuroblastoma progression by targeting RUNX2

Jiaxiang Tang 1, Qi Liu 1, Binyi Yang 1, Hongting Lu 1,
PMCID: PMC12198801  PMID: 40575351

Abstract

Objective

This study investigates the tumor-suppressive role of microRNA (miR)-205-5p in neuroblastoma (NB) and evaluates exosome-mediated delivery of miR-205-5p as a therapeutic strategy.

Methods

miR-205-5p expression in NB cells was quantified via quantitative reverse transcription PCR. Functional assays (CCK-8, colony formation, wound healing, Transwell) assessed proliferation, migration, and invasion. Bioinformatic tools and dual-luciferase assays identified miR-205-5p/Runt-related transcription factor 2 (RUNX2) binding. RUNX2 rescue experiments reversed miR-205-5p effects. Exosomes from SH-SY5Y cells transfected with miR-205-5p mimics/NC (negative control) lentiviruses were isolated, characterized, and co-cultured with recipient cells. In vivo, subcutaneous NB xenografts in nude mice were established using OE-miR-205-5p, sh-miR-205-5p, or NC lentiviral cells, followed by exosome injections to evaluate tumor growth.

Results

miR-205-5p was downregulated in NB cells. Its overexpression suppressed proliferation, migration, invasion, and tumor growth in vitro and in vivo. RUNX2 was confirmed as a direct target; its restoration reversed miR-205-5p-mediated inhibition. Exosomes efficiently delivered miR-205-5p to recipient cells, downregulating RUNX2 and impairing malignant behaviors. In mice, miR-205-5p-enriched exosomes significantly inhibited tumor progression.

Conclusions

Exosome-encapsulated miR-205-5p inhibits NB progression by targeting RUNX2, highlighting its potential as a novel therapeutic modality.

Keywords: Animal Experimentation; Biochemistry; Hospitals, Pediatric; Medical Oncology


WHAT IS ALREADY KNOWN ON THIS TOPIC

  • Exosomes and microRNAs (miRNAs) are implicated in cancer progression, but the mechanistic role of miR-205-5p in neuroblastoma, particularly its exosome-mediated transfer and downstream targets, remained undefined.

WHAT THIS STUDY ADDS

  • This study reports that exosomal miR-205-5p suppresses neuroblastoma cell proliferation, migration, and invasion by directly targeting Runt-related transcription factor 2 (RUNX2). It further demonstrates that exosome-mediated delivery of miR-205-5p inhibits tumor growth in vivo, establishing a functional link between exosomal miRNA cargo and neuroblastoma progression.

HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY

  • These findings propose exosome-based delivery of miR-205-5p as a novel therapeutic strategy for neuroblastoma and highlight RUNX2 as a potential biomarker or therapeutic target for progressing research on extracellular vesicle-mediated miRNA therapies in oncology.

Introduction

Neuroblastoma (NB) is an embryonic malignancy originating from primitive neural crest cells in the sympathetic ganglia and adrenal medulla. It represents the most common extracranial pediatric tumor,1 comprising 6%–10% of all childhood malignant tumors and accounting for approximately 15% of pediatric cancer-related deaths globally.2 3 MicroRNAs (miRNAs) are small, noncoding RNAs (18–25 nucleotides) that regulate post-transcriptional gene expression. Emerging evidence indicates that miRNA expression is often dysregulated in tumors, with these changes promoting tumorigenesis by influencing processes such as differentiation, proliferation, invasion, migration, and apoptosis of cancer cells. While the roles of various miRNAs in NB have been studied, the specific effects of miR-205-5p on NB progression and the underlying molecular mechanisms remain poorly understood. This study aims to elucidate the functional role of miR-205-5p in NB by manipulating its expression and investigating how this modulates NB cell malignancy.

Exosomes are nanovesicles, typically 30–150 nm in diameter, enclosed by a phospholipid bilayer. They are secreted into the extracellular space via the endosomal pathway following fusion of multivesicular bodies with the cell membrane.4 These vesicles carry a diverse cargo, including nucleic acids, lipids and proteins, and have been implicated in intercellular communication, tumor progression, immune evasion, and metastasis.5 6 Notably, exosomes can deliver miRNAs that regulate recipient cell behavior.7 Despite growing evidence linking exosomes to tumor biology, the relationship between exosomes, miR-205-5p, and NB has yet to be fully explored.

In this study, we put forward a core hypothesis that miR-205-5p may target and regulate Runt-related transcription (RUNX) family transcription factor 2 (RUNX2) through the exosome-mediated transfer mechanism and, thereby, inhibit malignant progression of NB. First, we confirmed the low expression of miR-205-5p in NB cells through quantitative reverse transcription PCR (qRT-PCR) experiments. Then, we overexpressed or inhibited miR-205-5p by transfection to explore the impact on tumor cell function. We used bioinformatic prediction and a dual luciferase reporter system to verify the targeting relationship between miR-205-5p and RUNX2, where we showed that RUNX2 overexpression could relieve the inhibitory effects of miR-205-5p. Next, we extracted and identified exosomes secreted by SH-SY5Y cells and confirmed that these could be taken up by NB cells. We verified that miR-205-5p carried by exosomes could inhibit the malignant behavior of receiver tumor cells. Finally, using a nude mouse subcutaneous tumor transplantation model, we further verified the direct tumor-suppressing effect of miR-205-5p and the therapeutic potential of exosome delivery.

Materials and methods

Cell culture

The human embryonic kidney cell line HEK-293 and human NB cell lines SK-N-SH, SK-N-DZ, SK-N-AS, SK-N-BE (2), SH-SY5Y were obtained from Wuhan Prirui Biological Technology Co., on May 16, 2023 and were authenticated by STR profiling. Cells were cultured in DMEM supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin in a 5% CO₂ atmosphere at 37°C. On reaching 80%–90% confluence, cells were harvested using trypsin and passaged.

miRNA transfection

SH-SY5Y and SK-N-BE (2) cells, at 60%–70% confluence, were seeded in 6-well plates 1 day prior to transfection. The medium was replaced with 1 mL Opti-MEM before starting the transfection. In a 1.5 mL enzyme-free EP tube, 250 µL of Opti-MEM was mixed with 5 µL of miRNA (miR-205-5p mimics, inhibitor, or control). In a separate tube, 250 µL of Opti-MEM was combined with 5 µL of Lipofectamine 2000 transfection reagent, gently mixed, and incubated for 5 min at room temperature. The contents of the two tubes were combined, mixed gently, and incubated for 20 min in the dark at room temperature. The transfection mixture was then added to the cell culture plates. After gentle mixing, the cells were cultured in an incubator. After 6 hours, the medium was replaced with complete medium. After transfection with miRNA, all the cells were subjected to cell functional analysis within 24–48 hours. Transfection material sequences are shown in table 1. Plasmid transfection and lentiviral transfection are described in online supplemental appendix 1.

Table 1. Transfection material sequences.

Name Sense Antisense
NC mimics 5′-UUCUCCGAACGUGUCACGUTT-3′ 5′-ACGUGACACGUUCGGAGAATT-3′
miR-205-5p mimics 5′-UCCUUCAUUCCACCGGAGUCUG-3′ 5′-GACUCCGGUGGAAUGAAGGAUU-3′
NC inhibitor 5′-CAGUACUUUUGUGUAGUACAA-3′ \
miR-205-5p inhibitor 5′-CAGACUCCGGUGGAAUGAAGGA-3′ \

miR, microRNA; NC, negative control.

Exosome characterization

When SH-SY5Y cells stably transfected with NC and miR-205-5p mimics lentiviruses reached 60%–70% confluence the medium was replaced with DMEM containing 10% FBS (Exosome-depleted). After an additional 48 hours of culture, exosomes were isolated using ExoQuick-TC (SBI), following the manufacturer’s protocol (10 min at 300×g; 15 min at 3000×g, 4°C), followed by the addition of EXOquick-TC. Samples were incubated at 4°C for 12 hours, centrifuged at 1500×g for 30 min and the exosome-enriched pellet was collected. Exosomes were then resuspended in phosphate-buffered saline (PBS) and used for downstream experiments. The extracted exosomes were quantified by determining protein concentration using the BCA Protein Assay Kit. Exosome morphology was assessed using transmission electron microscopy (TEM), and particle size distribution was analyzed by nanoparticle tracking analysis (NTA). The details of exosome-related experiments are described in online supplemental appendix 1.

Quantitative reverse transcription PCR

Total RNA was extracted from cells and exosomes using Trizol, following the manufacturer’s instructions. RNA purity was confirmed by the A260/A280 ratio (1.8–2.0). Reverse transcription experiments were performed using a miRNA first Strand cDNA Synthesis Kit and HiScript II QRT SuperMix for qPCR kit. Gene-specific primers for miR-205-5p, RUNX2, GAPDH, and U6 were obtained from Shanghai Sangon Biological Co. (table 2). U6 and GAPDH were used as internal controls for miRNA and mRNA normalization respectively. qPCR was performed using the ChamQ Universal SYBR qPCR Master Mix kit. The qPCR conditions were as follows: 95°C for 3 min, followed by 45 cycles of 95°C for 10 s and 60°C for 30 s. Data were analyzed using the 2−△△Ct method, with four technical replicates for each sample.

Table 2. qRT-PCR primer sequences.

Gene name Forward Reverse
miR-205-5p 5′-CGTCCTTCATTCCACCGG-3′ 5′-AGTGCAGGGTCCGAGGTATT-3′
RUNX2 5′-AGTAGATGGACCTCGGGAACC-3′ 5′-ACTGAGGCGGTCAGAGAACAA-3′
U6 5′-CTCGCTTCGGCAGCACA-3′ 5′-AACGCTTCACGAATTTGCGT-3′
GAPDH 5′-CCTCGTCTCATAGACAAGATGGT-3′ 5′-GGGTAGAGTCATACTGGA ACATG-3′

miR, microRNA; qRT-PCR, quantitative reverse transcription PCR; RUNX2, Runt-related transcription factor 2.

CCK-8 assay

SH-SY5Y cells transfected with miRNA (miR-205-5p mimics, inhibitors, or controls) and SH-SY5Y cells co-cultured with exosomes extracted from SH-SY5Y cells that stably expressed NC or miR-205-5p mimics by lentiviruses were used. Logarithmically growing cells (80%–90% confluent) were resuspended at 1000 cells/mL and plated in 96-well plates (100 µL/well). After incubation for 0, 24, 48, 72, and 96 hours, 10 µL of CCK-8 reagent was added to each well, and cells were incubated for an additional 2 hours. Absorbance was measured at 450 nm, and growth curves were generated.

Colony formation assay

SH-SY5Y cells transfected with miRNA (miR-205-5p mimics, inhibitors, or controls) and SH-SY5Y cells co-cultured with exosomes extracted from SH-SY5Y cells that stably expressed NC or miR-205-5p mimics by lentiviruses were used. Logarithmically growing cells were seeded into 6-well plates (800 cells/well) in complete medium and incubated at 37°C with 5% CO₂ for 14 days. Colonies were then fixed with 4% paraformaldehyde, stained with 0.1% crystal violet, and imaged. Colonies in five randomly selected fields (identified using a random number table) were counted.

Wound healing assay

SH-SY5Y cells transfected with miRNA (miR-205-5p mimics, inhibitors, or controls) and SH-SY5Y cells co-cultured with exosomes extracted from SH-SY5Y cells that stably expressed NC or miR-205-5p mimics by lentiviruses were used. SH-SY5Y cells were plated in 6-well plates and cultured to 80%–90% confluence. A straight scratch wound was made in the monolayer using a 10 µL pipette tip. After washing with PBS, serum-free medium was added, and cells were incubated at 37°C with 5% CO₂. Wound closure was assessed and imaged at 0 and 24 hours using an inverted microscope.

Transwell assays

SH-SY5Y cells transfected with miRNA (miR-205-5p mimics, inhibitors, or controls) and SH-SY5Y cells co-cultured with exosomes extracted from SH-SY5Y cells that stably expressed NC or miR-205-5p mimics by lentiviruses were used. For migration and invasion assays, Transwell chambers with an 8.0 µm pore size were used. Cells were resuspended in serum-free DMEM at 1×10⁵ cells/mL and plated in the upper chamber (200 µL) of a Transwell insert, while the lower chamber contained 600 µL of culture medium supplemented with 20% FBS. After 48 hours incubation, non-migrated cells were removed, and the migrated cells were fixed with 4% paraformaldehyde for 20 min, stained with 0.1% crystal violet for 20 min and imaged in five randomly selected fields. For invasion assays, the chambers were precoated with Matrigel. The details of protein extraction and purification can be found in online supplemental appendix 1.

Western immunoblotting

Protein concentrations were determined using a BCA assay kit (Wuhan Eilerite Biotechnology Co.) and samples were boiled at 95°C for 10 min before separation by 10% SDS-PAGE (Yase Biological Co.). Proteins were transferred onto PVDF membranes which were blocked with 5% non-fat milk for 2 hours, followed by incubation with primary antibodies against CD9 (1:2000), CD63 (1:2000), TSG101 (1:2000), RUNX2 (1:1000) and GAPDH (1:1000). After washing, the membranes were incubated with a secondary antibody (1:1000) for 1 hour at room temperature. Protein bands were visualized using an Enhanced Chemiluminescence solution, and image quantification was performed using ImageJ software.

Luciferase reporter assay

The pmiRGLO dual-luciferase vector (150 ng) was subcloned with wild-type or mutant RUNX2 3' untranslated region (3' UTR), generating pmiRGLO-RUNX2 WT/MUT. These constructs were co-transfected into cells along with miR-205-5p mimics or control constructs. After 48 hours, luciferase activity was measured using a dual-luciferase reporter system.

Subcutaneous tumor model in nude mice

Nude mice (4 weeks old) were purchased from VitonLihua and allowed to acclimatize for 1 week before experiments. SH-SY5Y cells transfected with lentiviruses were resuspended in PBS and Matrigel (5×10⁷ cells/mL) and injected subcutaneously into the right forelimb of nude mice (200 µL per mouse). Tumor growth was monitored every 5 days by measuring tumor volume (V=1/2×length×width²) and body weight. After 35 days, mice were killed, tumors were harvested, imaged, and weighed.

Multipoint injection of exosomes adjacent to the tumor

Nude mice were used as above. SH-SY5Y cells transfected with lentiviruses were resuspended in PBS and Matrigel (5×10⁷ cells/mL) and injected subcutaneously into the right forelimb of nude mice (200 µL per mouse). 3 days after the injection of tumor cells, mice were randomly divided into two groups: NC-exo and miR-205-5 p mimics-exo. The NC group and the miR-205-5p mimics group, respectively, were injected with exosomes extracted from SH-SY5Y cells that were stably transfected with NC and miR-205-5p mimics lentiviruses. Injection was performed at multiple sites around the tumor every 3 days at a dose of 20 µg exosomes+100 µL PBS. The injection was carried out continuously for 30 days. Tumor size and body weight were monitored every 5 days. After 30 days, mice were killed, tumors were collected, imaged, and weighed. The immunohistochemistry process is described in online supplemental appendix 1.

Statistical analysis

All data were analyzed using SPSS V.29 and figures were constructed using GraphPad Prism V.10. Data are presented as the mean±SD from at least three independent experiments. Differences between groups were assessed using student t-tests or one-way analysis of variance, as appropriate. A p<0.05 was considered statistically significant.

Results

NB cells exhibit characteristic miR-205-5p downregulation

Previous studies from our group have shown miR-205-5p expression is strongly associated with NB. The expression of miR-205-5p in five NB cell lines SK-N-SH, SK-N-DZ, SK-N-AS, SK-N-BE(2) and SH-SY5Y was compared with that of human embryonic kidney cell line HEK-293 by qPCR (figure 1A). The results showed that the expression of miR-205-5p in all NB cell lines was significantly lower than that in HEK-293 cells. NB cells express lower miR-205-5p than normal tissue cells, and miR-205-5p may, therefore, act as a tumor suppressor.

Figure 1. miR-205-5p overexpression inhibits NB cell proliferation, migration, and invasion. (A) miR-205-5p expression in NB (SK-N-SH, SK-N-DZ, SK-N-AS, SK-N-BE (2), SH-SY5Y) versus EK-293cells (qRT-PCR). (B) Transfection efficiency of miR-205-5p mimics/inhibitor versus controls in SH-SY5Y/SK-N-BE (2) (qRT-PCR) (C–G) Functional effects of miR-205-5p mimics/inhibitor on viability (CCK-8), proliferation (colony formation), migration (scratch, Transwell), and invasion (Transwell) in SH-SY5Y/SK-N-BE (2). ns: no significant; *p<0.05, **p<0.01, ***p<0.001. miRNA: microRNA; NB: neuroblastoma; NC: negative control.

Figure 1

miR-205-5p overexpression inhibits NB cell proliferation, migration, and invasion

SH-SY5Y and SK-N-BE(2) cells were successfully transfected with miR-205-5p mimics or inhibitors (figure 1B). The effects of miR-205-5p overexpression or knockdown on NB cell proliferation and survival were assessed through CCK-8 and colony formation assays. Overexpression of miR-205-5p significantly reduced cell viability and proliferative activity, while knockdown of miR-205-5p had the opposite effect (figure 1C-D). Additionally, Transwell and wound healing assays revealed that miR-205-5p overexpression decreased NB cell migration and invasion, while its inhibition enhanced these metastatic activities (figure 1E-G).

miR-205-5p and RUNX2 interactions

To explore the molecular mechanisms underlying the role of miR-205-5p in NB, we investigated potential interactions with target genes. Bioinformatic analysis using the TargetScan website (https://www.targetscan.org/) predicted that RUNX2 could be a target of miR-205-5p. Further analysis revealed a correlation between the expression of miR-205-5p and RUNX2 in NB cells (figure 2A). This interaction was confirmed in dual-luciferase reporter assays, which demonstrated that miR-205-5p directly binds to the UTR of RUNX2 mRNA (figure 2B) (). Furthermore, qPCR (figure 2C) and Western immunoblotting analyses (figure 2D) revealed that overexpression of miR-205-5p led to a significant downregulation of RUNX2 at both the mRNA and protein levels, confirming RUNX2 as a direct target of miR-205-5p in NB cells.

Figure 2. miR-205-5p and RUNX2 interactions. (A) Predicted miR-205-5p binding site in RUNX2 3’UTR (TargetScan); (B) miR-205-5p/RUNX2 3’UTR interaction confirmed by dual-luciferase assay; (C-D) RUNX2 mRNA/protein levels post miR-205-5p mimics/inhibitor transfection (qRT-PCR, WB). ns: no significant, *p<0.05, **p<0.01, ***p<0.001. miRNA: microRNA; NC: negative control; RUNX2: Runt-related transcription family transcription factor 2.

Figure 2

RUNX2 restores miR-205-5p inhibition of NB cell function

The expression of RUNX2 in the blank control group (NC+pcDNA3.1), miR-205-5p mimics and pcDNA3.1 co-transfection group (miR-205-5p mimics+pcDNA3.1), miR-205-5p mimics and RUNX2 overexpression plasmid co-transfection group (miR-205-5p mimics+RUNX2) and NC and RUNX2 overexpression plasmid co-transfection group (NC+RUNX2) was detected by qRT-PCR and WB. Transfection of RUNX2 overexpression plasmid could relieve the inhibitory effect of miR-205-5p mimics on the expression of RUNX2, and the expression of RUNX2 increased (figure 3A-B) . CCK-8 assay and cell colony formation assay detected the effect of restoring the expression of RUNX2 on the proliferation of NB cells induced by miR-205-5p (figure 3C-D). Compared with the miR-205-5p mimics and pcDNA3.1 co-transfection group, the miR-205-5p mimics and RUNX2 overexpression plasmid co-transfection group significantly increased the proliferation. In addition, the scratch assay and Transwell migration assay were used to evaluate the effect of restoring RUNX2 on the migration of NB cells induced by miR-205-5p (figure 3E-F). showed that increasing the expression of RUNX2 enhanced the migration ability of NB cells and relieved the inhibitory effect of miR-205-5p. Transwell invasion assays verified invasion ability. As shown in figure 3G, the RUNX2 overexpression plasmid significantly reversed the inhibition of miR-205-5p on cellular invasion ability. In conclusion, RUNX2 overexpression can restore proliferation, migration, and invasion abilities of NB cells inhibited by miR-205-5p.

Figure 3. RUNX2 restores miR-205-5p inhibition of neuroblastoma cell function. (A-B) RUNX2 rescue: mRNA/protein levels in cells co-transfected with miR-205-5p mimics + RUNX2 plasmid (qRT-PCR, WB); (C-G) Functional rescue: Cell viability (CCK-8), proliferation (colony), migration (scratch, Transwell), and invasion (Transwell) post miR-205-5p mimics + RUNX2 plasmid co-transfection. Ctr: NC+pcDNA3.1; miR+Ctr: miR-205-5p mimics+pcDNA3.1; miR+R: miR-205-5p mimics+RUNX2; Ctr+R: NC+RUNX2. ns: no significant, *p<0.05, **p<0.01, ***p<0.001. miRNA: microRNA; NC: negative control; RUNX2: Runt-related transcription family transcription factor 2.

Figure 3

Exosomal characterization

TEM analysis of the vesicles in the supernatants of NB cells revealed that they were predominantly ovoid in shape (figure 4A). NTA indicated that the exosome diameter was approximately 100 nm (figure 4B), consistent with the expected size range for exosomes. Western immunoblotting further confirmed the presence of exosomal markers, including CD9, TSG101, and CD63 (figure 4C), further validating the successful isolation of exosomes.

Figure 4. Characterization and internalization of Exosomes. (A) Exosome morphology (TEM); (B) Exosome size distribution (nanoparticle tracking); (C) Exosome marker detection (WB: supernatant vs. exosomes); (D) Exosome internalization by SH-SY5Y cells (confocal microscopy: PKH26/PKH67-labeled exosomes, DAPI nuclei).

Figure 4

NB cells can internalize exosomes

To assess internalization of exosomes by NB cells, exosomes were labeled with PKH26 (red) and PKH67 (green) fluorescent dyes. The washed and centrifuged supernatant (poststaining) was used as a control. SH-SY5Y cells were co-cultured with these labeled exosomes for 12 hours. Following DAPI nuclear staining, confocal imaging revealed that PKH26-labeled exosomes exhibited a red fluorescent signal around the perinuclear region, and PKH67-labeled exosomes exhibited a green fluorescent signal within SH-SY5Y cells (figure 4D), confirming exosome uptake.

Exosomes deliver miR-205-5p to suppress NB cell proliferation, migration, and invasion

Exosomes were extracted from the supernatants of SH-SY5Y cells stably transfected with either NC or miR-205-5p mimic lentiviruses, and miR-205-5p levels in the exosomes were quantified by qRT-PCR. The results showed that the miR-205-5p content in exosomes secreted by SH-SY5Y cells transfected with miR-205-5p mimics was approximately twice that in exosomes secreted by NC-transfected SH-SY5Y cells, indicating that overexpressed miR-205-5p can be carried by exosomes (figure 5A). Co-culturing SH-SY5Y cells with exosomes from miR-205-5p mimic-transfected cells resulted in significantly higher levels of miR-205-5p in the recipient cells compared with exosomes from NC-transfected cells (figure 5B), confirming the transfer of miR-205-5p between cells. Next, the effects of exosomal miR-205-5p were evaluated on proliferative and migratory abilities using CCK-8 and colony formation assays. Exosomes containing miR-205-5p significantly inhibited both cell viability and colony formation compared with the NC-exosome group (figure 5C-D)). Additionally, exo-miR-205-5p inhibited NB cell migration as measured in wound healing and Transwell migration assays (figure 5E-F) and reduced invasion in Transwell assays (figure 5G). Exosomes with lower miR-205-5p levels resulted in higher RUNX2 expression in co-cultured NB cells (figure 5H). Collectively, these findings indicate that exosomes can deliver miR-205-5p between cells, leading to the downregulation of RUNX2 and the inhibition of cell proliferation, migration, and invasion.

Figure 5. Exosomes deliver miR-205-5p to suppress neuroblastoma cell proliferation, migration, and invasion. (A) miR-205-5p levels in exosomes from NC/miR-205-5p mimics-transfected cells (qRT-PCR);(B) Recipient SH-SY5Y cell miR-205-5p levels after co-culture with NC-EXO/miR-205-5p-EXO (qRT-PCR);(C-G) Functional effects of exosome-delivered miR-205-5p: viability (CCK-8), proliferation (colony), migration (scratch, Transwell), invasion (Transwell);(H) RUNX2 protein expression post exosome treatment (WB). ns: no significant, *p<0.05, **p<0.01, ***p<0.001. EXO: exosomes; miRNA: microRNA; NC: negative control; RUNX2: Runt-related transcription family transcription factor 2.

Figure 5

Overexpression of miR-205-5p inhibits NB cell proliferation in vivo

To investigate the in vivo effect of miR-2055p on the tumorigenic potential of NB, 15 nude mice, aged 4 weeks, were selected and randomly divided into three groups. SH-SY5Y cells stably transfected with miR-205-5p mimics, miR-205-5p inhibitor lentiviruses, and control SH-SY5Y cells were subcutaneously inoculated into the axillae of the three groups to establish xenograft tumor models. The groups were designated as the OE-miR-205-5p group, sh-miR-205-5p group, and BLANK group. Tumor volume was monitored starting 5 days postinjection, and measurements were taken every 5 days until the mice were killed (figure 6A). Postmortem, tumors were collected (figure 6B) and weighed (figure 6C). Tumor growth was inhibited in the OE-miR-205-5p group and promoted in the sh-miR-205-5p group, as reflected in tumor volume and weight (figure 6A-D). IHC for RUNX2 revealed that miR-205-5p overexpression resulted in decreased RUNX2 expression in tumor tissue (figure 6E). These findings suggest that miR-205-5p suppresses the growth of NB tumors in vivo and downregulates RUNX2 expression.

Figure 6. Overexpression of miR-205-5p inhibits NB cell proliferation in vivo. (A-C) Subcutaneous SH-SY5Y tumor size/weight/volume in NC, miR-205-5p mimics, and inhibitor lentiviral groups; (D) RUNX2 expression in tumors (IHC); (E) Immunohistochemical RUNX2 tumor staining of tumor in nude mice. ns: no significant, *p<0.05, **p<0.01, ***p<0.001. EXO: exosomes; miRNA: microRNA; NC: negative control; RUNX2: Runt-related transcription family transcription factor 2.

Figure 6

Exosomes carrying miR-205-5p suppress tumor growth in nude mice

To further investigate the role of exosomes in the transport of miR-205-5p, 10 nude mice, aged 4 weeks, were randomly divided into two groups: NC-exosome and miR-205-5p mimics-exosome. SH-SY5Y cell-derived tumors were established in the axillary region of the mice, and exosomes (20 µg exosomes+100 µL PBS) from the NC mimics and miR-205-5p mimics groups were injected into the tumors every 3 days for 30 days. Tumor growth was monitored starting 5 days postinjection (figure 7A), and tumor volumes were measured every 5 days until the mice were killed (Figure 7B). Tumors were collected and weighed (figure 7C). Exosomes from the miR-205-5p mimics group significantly inhibited tumor growth; there was decreased tumor volume and weight compared with the NC-exosome group (figure 7A-D). IHC showed that exosomes carrying miR-205-5p led to reduced RUNX2 tumor expression (figure 7E). These results indicate that exosomes can successfully deliver miR-205-5p to NB cells, inhibiting tumor proliferation in vivo and reducing RUNX2 expression.

Figure 7. Exosomes carrying miR-205-5p suppress tumor growth in nude mice. (A-D) Tumor size/weight/volume post s.c. implantation with NC-EXO or miR-205-5p-EXO treatment; (E) RUNX2 expression in exosome-treated tumors (IHC). ns: no significant, *p<0.05, **p<0.01, ***p<0.001. EXO: exosomes; miRNA: microRNA; NC: negative control; RUNX2: Runt-related transcription family transcription factor 2.

Figure 7

Discussion

In our study, we transfected cells with miR-205-5p mimics to increase the miR-205-5p content in secreted exosomes. After co-culturing these exosomes with tumor cells, we verified that exosomes could transport miR-205-5p between cells and target RUNX2 to exert antitumor effects, reducing proliferation, migration, and invasion. In animal experiments, through multiple injections of exosomes near the tumor site, we macroscopically confirmed that exosomes could similarly transport miR-205-5p to tumor tissues in nude mice and inhibit xenograft tumor growth. Our study confirms that exosomes can transport anti-cancer miRNAs to inhibit tumor growth, which could be a new intervention strategy for treating NB.

NB is the most common extracranial tumor in children. Prognostic outcomes in NB patients show substantial variability, with cases ranging from spontaneous tumor regression to recurrence.8 9 In advanced stages, NB tumors are highly aggressive and prone to metastasis, leading to a poor prognosis, with a 5-year survival rate of less than 40% even with treatment.10 Early detection and effective treatment remain critical challenges in the clinical management of NB. The aim of this study was to explore the molecular mechanisms underlying the pathogenesis of NB, with the goal of identifying potential therapeutic targets.

miRNAs are small, endogenous non-coding RNAs that regulate gene expression by silencing target genes post-transcriptionally.11 Numerous studies have implicated miRNAs in the pathogenesis of various diseases, including cancer. Specifically, miRNAs such as miR-15a, miR-16, miR-149, miR-195, miR-93, and miR-388 have been identified as tumor suppressors in NB.12,16 miR-205-5p has been shown to function as a tumor suppressor in several cancers, including breast, kidney, prostate, and colorectal.17,20 According to the literature, studies have shown that miR-153-3p and miR-205-5p influence various biological processes in NB cells, including survival, proliferation, and neuroprotection.21 In SH-SY5Y NB cells, modulation of miR-205-5p led to the upregulation of 12 and downregulation of 6 proteins. These proteins are associated with neurobiological processes and include peroxiredoxins 2 and 4, cofilin-1, prefoldin 2, α-enolase, human nucleoside diphosphate kinase B (Nm23), and 14-3-3 epsilon protein. Many of the differentially expressed proteins are involved in metabolism, neurotrophin signaling, actin cytoskeleton regulation, HIF-1 signaling, and the proteasome, suggesting that miR-153-3p and miR-205-5p play a role in regulating multiple biological processes in NB cells.21 Zhou et al.22 found that treatment with a low concentration of Aurora kinase A (AURKA) inhibitor MLN8237 established a cellular senescence model in NB cells. In the senescent cell group, 32 miRNAs, including miR-205-5p, miR-378d, and miR-378f, were significantly downregulated. In the extracellular vesicles secreted by the senescent cells, 48 miRNAs were upregulated, including miR-205-5p. Bioinformatic analysis revealed that these differentially expressed miRNAs might potentially regulate many common target genes involved in metabolic signaling pathways and transcriptional dysregulation in cancer. These findings highlight the important role of miR-205-5p in the initiation and progression of NB. We continue to explore the specific target sites of miR-205-5p in NB in depth and attempt to verify the possibility of using exosomes to deliver miR-205-5p for the treatment of NB by changing the content of miR-205-5p in exosomes co-cultured with cells.

RUNX2 is a major regulator of osteoblast and chondrocyte differentiation, playing a key role in bone and cartilage cell maturation through transcriptional activation and involvement in multiple signaling pathways.23 24 Numerous studies have shown that RUNX2 plays functional roles in coordinating tumor metastasis, tumor proliferation, angiogenesis, and resistance to anticancer agents.25,27 RUNX2 plays a critical role in coordinating cancer progression across various types of tumors and has been shown to promote tumor growth in prostate, pancreatic, and breast cancer. 28,30 To further investigate how miR-205-5p participates in NB cell biology, we used three prediction tools (miRWalk, Targetscan, and miRDB) and identified RUNX2 as the most likely target gene of miR-205-5p. The study of Bao et al. on the YAP-RUNX2-SRSF1 signaling axis demonstrated that it promotes angiogenesis in NB tumors.31 The targeting relationship between miR-205-5p and RUNX2 has also been implicated in the pathogenesis of osteoporosis and osteoblast differentiation. This study found that miR-205-5p inhibits osteogenic differentiation by targeting RUNX2.32 In our study, the dual-luciferase reporter assay confirmed the targeting relationship between miR-205-5p and RUNX2. We found a negative correlation between the expression of RUNX2 and miR-205-5p. qRT-PCR and Western blot analyses confirmed that miR-205-5p downregulates the expression of RUNX2. We increased the expression of RUNX2 in cells using overexpression plasmids and verified the restoration effect of RUNX2 on NB cell function. We found that after the inhibition of RUNX2 expression was reversed, proliferation, migration, and invasion of NB cells inhibited by miR-205-5p was restored. Based on our cell and animal experiments, we conclude that miR-205-5p acts as a tumor suppressor in NB cells and participates in the initiation and progression of NB by targeting and regulating the expression of RUNX2.

Exosomes possess a variety of molecular biological functions and serve as carriers for intercellular material transport and information transmission. They play a crucial role in regulating various physiological and pathological processes. Exosomes are involved in the development, metastasis, and treatment of NB.33 In glioblastoma (GBM), exosomes can promote tumor cell migration.34 However, while exosomes derived from GBM cells exhibit pro-cancer effects, they also contain miR-199a, miR-101-3p, miR-101, and other miRNAs that can inhibit proliferation and migration of GBM cells.35,37 Similarly, exosomes derived from bone marrow mesenchymal stem cells, rich in miR-4461, can inhibit the development of colon cancer,38 and miR-126-3p overexpressed in exosomes can also suppress ovarian cancer progression.39 These studies suggest that exosomes also contain components that inhibit tumor cell proliferation. One hypothesis suggests that to maintain tumorigenesis and metastasis, tumor cells tend to preferentially seclude antitumor miRNAs in vesicles and package them into exosomes for secretion into the extracellular space.40 miR-146 has been shown to be a tumor suppressor in prostate cancer, glioma, breast cancer, and pancreatic cancer. Overexpression of the tumor suppressor miR-146 in stromal cells triggers the release of its exosomes. Additionally, tumor-suppressive miRNA let-7 is highly concentrated in the exosomes of highly metastatic gastric cancer cells compared with those with lower metastatic potential. This suggests a potential link between the decrease of tumor-suppressive miRNAs in tumor cells and their secretion via exosomes. Exosomes have good stability and biocompatibility, making them effective delivery carriers. miRNAs in circulation are unstable, prone to degradation, and have difficulty entering target cells. Delivery via exosomes can prevent miRNAs from being broken down and achieve better therapeutic effects. Since exosomes are non-immunogenic and can protect their encapsulated molecules from degradation by serum proteases and immune system phagocytosis, they are a promising therapeutic tool. Therefore, we considered whether tumor-suppressive miRNAs could be delivered to tumor tissues via exosomes to exert antitumor effects.

In conclusion, we found that miR-205-5p may act as a tumor suppressor in the growth of NB cells. Exosomes carry miR-205-5p and, through targeting RUNX2, participate in the processes of proliferation, migration, and invasion. Our study validates that exosomes can carry miR-205-5p to inhibit proliferation, migration, and invasion. Various technologies derived from exosomes hold tremendous clinical applicability in the diagnosis, treatment, and prognostic assessment of NB. We hope to explore new strategies for tumor diagnosis and treatment using exosomes as a powerful tool.

Supplementary material

online supplemental appendix 1
wjps-8-3-s001.docx (13.9KB, docx)
DOI: 10.1136/wjps-2024-000993

Footnotes

Funding: Shandong Province Medical and Health Science and Technology Development Plan Project (202202080942).

Patient consent for publication: Not applicable.

Ethics approval: Animal experiments were approved by the Ethics Committee for Experimental Animal Welfare of Qingdao University, ethics number 20240401BALB/c2520240506056. The study adhered to the guidelines and regulations set by the committee. In the experimental process, the 3R principles (Replacement, Reduction and Refinement) were strictly followed to minimize the use of animals and alleviate their suffering. The animals were housed in a controlled environment with adequate food, water, and living space and were monitored regularly to ensure their welfare. All procedures were performed by trained personnel in accordance with standard operating procedures to reduce the risk of animal distress.

Provenance and peer review: Not commissioned; externally peer reviewed.

Data availability free text: All data generated or analyzed during this study are included in the published article.

Data availability statement

All data relevant to the study are included in the article or uploaded as supplementary information.

References

  • 1.Su Y, Qin H, Chen C, et al. Treatment and outcomes of 1041 pediatric patients with neuroblastoma who received multidisciplinary care in China. Pediatr Investig . 2020;4:157–67. doi: 10.1002/ped4.12214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Maris JM, Hogarty MD, Bagatell R, et al. Neuroblastoma. Lancet. 2007;369:2106–20. doi: 10.1016/S0140-6736(07)60983-0. [DOI] [PubMed] [Google Scholar]
  • 3.Young JL, Jr, Ries LG, Silverberg E, et al. Cancer incidence, survival, and mortality for children younger than age 15 years. Cancer. 1986;58:598–602. doi: 10.1002/1097-0142(19860715)58:2+&#x0003c;598::aid-cncr2820581332&#x0003e;3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
  • 4.Ren R, Sun H, Ma C, et al. Colon cancer cells secrete exosomes to promote self-proliferation by shortening mitosis duration and activation of STAT3 in a hypoxic environment. Cell Biosci. 2019;9:62.:62. doi: 10.1186/s13578-019-0325-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zamanian M, Fraser LM, Agbedanu PN, et al. Release of Small RNA-containing Exosome-like Vesicles from the Human Filarial Parasite Brugia malayi. PLoS Negl Trop Dis. 2015;9:e0004069. doi: 10.1371/journal.pntd.0004069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977. doi: 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dinkins MB, Dasgupta S, Wang G, et al. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging. 2014;35:1792–800. doi: 10.1016/j.neurobiolaging.2014.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Vanichapol T, Chiangjong W, Panachan J, et al. Secretory High-Mobility Group Box 1 Protein Affects Regulatory T Cell Differentiation in Neuroblastoma Microenvironment In Vitro. J Oncol. 2018;2018:7946021. doi: 10.1155/2018/7946021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Johnsen JI, Dyberg C, Wickström M. Neuroblastoma-A Neural Crest Derived Embryonal Malignancy. Front Mol Neurosci. 2019;12:9. doi: 10.3389/fnmol.2019.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Matthay KK, Maris JM, Schleiermacher G, et al. Neuroblastoma. Nat Rev Dis Primers. 2016;2:16078. doi: 10.1038/nrdp.2016.78. [DOI] [PubMed] [Google Scholar]
  • 11.Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509–24. doi: 10.1038/nrm3838. [DOI] [PubMed] [Google Scholar]
  • 12.Chava S, Reynolds CP, Pathania AS, et al. miR-15a-5p, miR-15b-5p, and miR-16-5p inhibit tumor progression by directly targeting MYCN in neuroblastoma. Mol Oncol. 2020;14:180–96. doi: 10.1002/1878-0261.12588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mao F, Zhang J, Cheng X, et al. miR-149 inhibits cell proliferation and enhances chemosensitivity by targeting CDC42 and BCL2 in neuroblastoma. Cancer Cell Int. 2019;19:357. doi: 10.1186/s12935-019-1082-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhu YJ, Chen X, Han LL, et al. MiR-195 regulates neuroblastoma cell invasion by targeting RET. Chin J Pediatr Surg. 2015;36:775–9. doi: 10.3760/cma.j.issn.0253-3006.2015.10.014. [DOI] [Google Scholar]
  • 15.Chen X, Han L, Jiang Z, et al. Effects and mechanisms of miR-93 on invasion and migration of neuroblastoma cells. Chin J Pediatr Surg. 2015;36:930–5. doi: 10.3760/cma.j.issn.0253-3006.2015.12.012. [DOI] [Google Scholar]
  • 16.Zhou Y, Tang X, Huang Z, et al. KLF5 promotes KIF1A expression through transcriptional repression of microRNA-338 in the development of pediatric neuroblastoma. J Pediatr Surg. 2022;57:192–201. doi: 10.1016/j.jpedsurg.2021.12.020. [DOI] [PubMed] [Google Scholar]
  • 17.Lin LF, Li YT, Han H, et al. MicroRNA-205-5p targets the HOXD9-Snail1 axis to inhibit triple negative breast cancer cell proliferation and chemoresistance. Aging (Albany NY) 2021;13:3945–56. doi: 10.18632/aging.202363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Huang J, Wang X, Wen G, et al. miRNA‑205‑5p functions as a tumor suppressor by negatively regulating VEGFA and PI3K/Akt/mTOR signaling in renal carcinoma cells. Oncol Rep. 2019;42:1677–88. doi: 10.3892/or.2019.7307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Li L, Li S. miR‑205‑5p inhibits cell migration and invasion in prostatic carcinoma by targeting ZEB1. Oncol Lett. 2018;16:1715–21. doi: 10.3892/ol.2018.8862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liu H, Li A, Sun Z, et al. Long non-coding RNA NEAT1 promotes colorectal cancer progression by regulating miR-205-5p/VEGFA axis. Hum Cell. 2020;33:386–96. doi: 10.1007/s13577-019-00301-0. [DOI] [PubMed] [Google Scholar]
  • 21.Patil KS, Basak I, Pal R, et al. A Proteomics Approach to Investigate miR-153-3p and miR-205-5p Targets in Neuroblastoma Cells. PLoS One. 2015;10:e0143969. doi: 10.1371/journal.pone.0143969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhou X, Zhou Q, Zhou D, et al. MicroRNA expression profiles in extracellular vesicles and intracellular of AURKA inhibitor-induced senescent neuroblastoma cells. Transl Cancer Res. 2022;11:2767–82. doi: 10.21037/tcr-21-2438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lian JB, Javed A, Zaidi SK, et al. Regulatory controls for osteoblast growth and differentiation: role of Runx/Cbfa/AML factors. Crit Rev Eukaryot Gene Expr. 2004;14:1–41. [PubMed] [Google Scholar]
  • 24.Komori T. Requisite roles of Runx2 and Cbfb in skeletal development. J Bone Miner Metab. 2003;21:193–7. doi: 10.1007/s00774-002-0408-0. [DOI] [PubMed] [Google Scholar]
  • 25.Wang Y, Pei X, Wang C, et al. LncRNA-gpand promotes gastric cancer progression via runx2 and mmp13. In Review. 2021 doi: 10.21203/rs.3.rs-1061004/v1. Preprint. [DOI] [Google Scholar]
  • 26.Roy A, Prasad S, Chen Y, et al. Protein Kinase D2 and D3 Promote Prostate Cancer Cell Bone Metastasis by Positively Regulating Runx2 in a MEK/ERK1/2-Dependent Manner. Am J Pathol. 2023;193:624–37. doi: 10.1016/j.ajpath.2023.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lin Tsung-Chieh. RUNX2 and Cancer. Int J Mol Sci. 2023;24:7001. doi: 10.3390/ijms24087001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ashe H, Krakowiak P, Hasterok S, et al. Role of the runt-related transcription factor (RUNX) family in prostate cancer. FEBS J. 2021;288:6112–26. doi: 10.1111/febs.15804. [DOI] [PubMed] [Google Scholar]
  • 29.Wu X, Huang J, Yang Z, et al. MicroRNA-221-3p is related to survival and promotes tumour progression in pancreatic cancer: a comprehensive study on functions and clinicopathological value. Cancer Cell Int. 2020;20:443. doi: 10.1186/s12935-020-01529-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yin X, Teng X, Ma T, et al. RUNX2 recruits the NuRD(MTA1)/CRL4B complex to promote breast cancer progression and bone metastasis. Cell Death Differ. 2022;29:2203–17. doi: 10.1038/s41418-022-01010-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bao M, Chen Y, Liu JT, et al. Extracellular matrix stiffness controls VEGF165 secretion and neuroblastoma angiogenesis via the YAP/RUNX2/SRSF1 axis. Angiogenesis. 2022;25:71–86. doi: 10.1007/s10456-021-09804-7. [DOI] [PubMed] [Google Scholar]
  • 32.Huang M, Li X, Zhou C, et al. Noncoding RNA miR‐205‐5p mediates osteoporosis pathogenesis and osteoblast differentiation by regulating RUNX2. J of Cellular Biochemistry. 2020;121:4196–203.:10. doi: 10.1002/jcb.29599. [DOI] [PubMed] [Google Scholar]
  • 33.Chu J. Research progress of extracellular vesicles in the neuroblastoma. Zhonghua Bing Li Xue Za Zhi. 2024;53:317–22. doi: 10.3760/cma.j.cn112151-20230731-00042. [DOI] [PubMed] [Google Scholar]
  • 34.Li N, Luo L, Yang Y, et al. Research progress on the mechanism of action of exosomes in glioblastoma. Chin J Biotechnol. 2023;39:1477–501. doi: 10.13345/j.cjb.220777. [DOI] [PubMed] [Google Scholar]
  • 35.Monfared H, Jahangard Y, Nikkhah M, et al. Potential Therapeutic Effects of Exosomes Packed With a miR-21-Sponge Construct in a Rat Model of Glioblastoma. Front Oncol. 2019;9:782. doi: 10.3389/fonc.2019.00782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xue P, Huang S, Han X, et al. Exosomal miR-101-3p and miR-423-5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ . 2022;29:82–95. doi: 10.1038/s41418-021-00838-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Park J, Cho M, Cho J, et al. MicroRNA-101-3p Suppresses Cancer Cell Growth by Inhibiting the USP47-Induced Deubiquitination of RPL11. Cancers (Basel) 2022;14:964. doi: 10.3390/cancers14040964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chen HL, Li JJ, Jiang F, et al. MicroRNA-4461 derived from bone marrow mesenchymal stem cell exosomes inhibits tumorigenesis by downregulating COPB2 expression in colorectal cancer. Biosci Biotechnol Biochem. 2020;84:338–46. doi: 10.1080/09168451.2019.1677452. [DOI] [PubMed] [Google Scholar]
  • 39.Shimizu A, Sawada K, Kimura T. Pathophysiological Role and Potential Therapeutic Exploitation of Exosomes in Ovarian Cancer. Cells. 2020;9:814. doi: 10.3390/cells9040814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yu X, Odenthal M, Fries J. Exosomes as miRNA Carriers: Formation–Function–Future. IJMS. 2016;17:2028. doi: 10.3390/ijms17122028. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

online supplemental appendix 1
wjps-8-3-s001.docx (13.9KB, docx)
DOI: 10.1136/wjps-2024-000993

Data Availability Statement

All data relevant to the study are included in the article or uploaded as supplementary information.


Articles from World Journal of Pediatric Surgery are provided here courtesy of BMJ Publishing Group

RESOURCES