
Transcriptomic analyses unveil the mechanism of saikosaponin A in inhibiting human neuroblastoma SK‑N‑AS cells
- Authors:
- Published online on: July 2, 2025 https://doi.org/10.3892/ol.2025.15165
- Article Number: 419
-
Copyright: © Gao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Neuroblastoma (NB) is a type of cancer of the sympathetic nervous system characterized by extracranial solid tumors that arise from neural crest cells, and accounts for ~15% of cancer-related deaths in children (1,2). The median age at diagnosis is low (~18 months) with a notable number of cases occurring in infants (<1 year old for 40% of all NB cases) (3,4). Furthermore, over half of patients with NB are classified as high-risk, which results in a low 5-year survival rate (<50%) (3,4). The current major treatments for NB include spontaneous tumor regression, surgery, chemotherapy, radiotherapy and immunotherapy. Although therapy advancements have notably raised survival rates, the prognosis varies by the risk level (3,5–7). Furthermore, relapse, drug resistance and long-term side effects remain significant challenges for the treatment and recovery of these patients (8,9). Therefore, exploring new therapeutic drugs is of great clinical and scientific significance.
Multiple bioactivities have been reported for saikosaponin A (SSa), an oleanane-type saponin derived from Bupleurum chinensis DC. These include antitumor properties, anti-inflammatory effects and liver protection (10,11). Previous studies have shown that the antitumor activities of SSa, including suppressing cell proliferation, inducing apoptosis and inhibiting metastasis and angiogenesis, span various cell types including liver, colon, breast and pancreatic cancer cells (12). Specifically, in human hepatoma cell lines, SSa exhibits an antiproliferative effect (13,14). Such an effect in the HepG2 cell line is correlated with ERK activation and the upregulation of certain genes (15,16). In human colon carcinoma cells, SSa induces apoptosis by activating caspase-4, followed by activating sequential caspase-2 and −8 (17,18). In breast cancer cells, SSa inhibits proliferation and induces apoptosis (19,20). In both in vitro and in vivo conditions, SSa suppresses the activation of Akt and STAT3, decreases the levels of glycolysis and blocks the energy supply (21). In pancreatic cancer, SSa inhibits proliferation and induces apoptosis, which may be related to the inactivation of the EGFR/PI3K/Akt pathway (22).
A study has shown that SSa inhibits the proliferation of NB cells by enhancing apoptosis (23). SSa decreases the expression of Bcl-2 and increases the expression of Bax while activating proteins such as caspase-9, caspase-7 and poly (ADP-ribose) polymerase (23). Furthermore, SSa stops NB cells from invading and migrating by blocking certain signals (such as VEGFR2, Src and Akt) and by regulating proteins connected with epithelial-mesenchymal transition (EMT) (23). However, the molecular mechanism of SSa against NB cell remains unclear. In the present study, human NB SK-N-AS cells were treated with SSa and the transcriptome expression profiles of these cells were analyzed using RNA-sequencing (seq) combined with bioinformatic methods. The differentially expressed genes (DEGs) were then verified by reverse transcription-quantitative PCR (RT-qPCR).
Materials and methods
Cell culture
The human SK-N-AS NB cell line was purchased from Heilongjiang Jiufeng Bioengineering Co., Ltd. The human SH-SY5Y NB cell line was purchased from Haixing Biosciences Co., Ltd. (Suzhou, China) and was authenticated by STR profiling. The human MO3.13 oligodendrocytic cell line (normal cells) was purchased from Cyagen Biosciences, Inc. These cells were initially cultured in Dulbecco's Modified Eagle Medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum [FBS; Cellmax Cell Technology (Beijing) Co., Ltd.] and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) in an incubator maintained at 37°C with a humidified environment of 5% CO2 and 95% air. Afterwards, cells were transferred every 2–3 days depending on their density, and experiments began once they achieved 80–90% confluency.
Cell viability assay
Cell viability was measured using the MTT assay following the protocol by Cheng and Ying (23). In brief, a cell suspension (SK-N-AS/SH-SY5Y/MO13.3) in DMEM containing 10% FBS (106 cells/ml) was inoculated into 96-well plates with 100 µl per well and cultured at 37°C for 24 h with 5% CO2 and 95% air. The cells were then treated with SSa (purity: HPLC ≥98%, Baoji Herbest Bio-Tech Co., Ltd.) at various concentrations [0 µM (control), 5, 10, 15, 20 and 25 µM] for 24 h. Following the treatment, 10 µl of MTT solution (5 mg/ml) was pipetted into each well, allowing the cells to incubate for an additional 4 h at 37°C. The medium was discarded and 100 µl of DMSO was added to each well to dissolve the purple formazan while the absorbance was measured at 490 nm using a Microplate Reader (RT-6000; Rayto Life and Analytical Sciences Co., Ltd.). Cell viability was calculated as follows: Cell viability=[OD490 (SSa)/OD490 (control)] ×100%.
Transwell migration and invasion assay
The methods were performed following the protocol by Cheng and Ying (23). In brief, SK-N-AS cells were digested and suspended in serum-free DMEM containing SSA (0 or 5 µM) at a final concentration of 106 cells/ml. For the invasion assay, 50 µl diluted Matrigel (Matrigel and serum-free DMEM were prepared at a ratio of 1:3 at 4°C) was added to the upper Transwell chamber (24-well; pore size, 8 µm; Corning, Inc.) and incubated at 37°C for 5 h. Subsequently, DMEM containing 20% FBS was added to the lower Transwell chamber. Subsequently, 100 µl of SK-N-AS cell suspension was plated in the upper chamber and incubated at 37°C for 24 h. The cells that invaded into the lower surface of the Transwell membrane were fixed with 100% methanol (room temperature for 30 min) and stained with 0.1% crystal violet (room temperature for 20 min). Stained cells were observed by fluorescence inverted microscopy (Leica Microsystems GmbH). A total of five random fields were imaged to calculate the relative invasion rates based on the ratio of the number of invaded cells in the SSa group to the control group. For the migration assay, all the steps were the same as in the invasion assay but without the addition of Matrigel.
RNA isolation and sequencing
To isolate RNA, ~4 ml of SK-N-AS cells (106 cells/ml) were seeded into a T25 flask. The cells were cultured for ~2 days at standard conditions (37°C for 48 h in humidified atmosphere of 5% CO2 and 95% air) before being divided into two groups and treated for another 24 h: Those receiving SSa treatment (n=3) were incubated in FBS-free DMEM containing 15 µM SSa, and those in the control group did not receive any substance addition (n=3). Post-treatment, total RNA extraction from the SN-K-AS cells was performed using TRIzol Reagent (Invitrogen; Thermo Fisher Scientific, Inc.). NanoDrop spectrophotometer-based measurements (Thermo Fisher Scientific, Inc.) confirmed the concentration and purity levels alongside LabChip GX Touch HT Nucleic Acid analysis (PerkinElmer, Inc.).
The collected RNA samples (a total of 6, 3 for SSa treatment and 3 for control) were then transported to Wuhan Boyue Zhihe Biotechnology Co., Ltd., where library construction and sequencing tasks employing KAPA Stranded RNA-Seq Kit protocols plus multiplexing primer guidance were undertaken. In brief, mRNA was enriched by oligo(dT) beads and sequencing results were obtained using NovaSeq™ 6000 systems (Illumina, Inc.).
Data analysis
Raw data (raw reads) were filtered into clean data (clean reads) using Trimmomatic v0.36 (24). This involved removing reads with adapters, reads containing ploy-N and low-quality reads from the raw data. Clean data were then aligned to the human reference genome (http://asia.ensembl.org/Homo_sapiens/Info/Index) using Hisat 2 (25). The number of perfect clean tags for each gene was calculated and normalized as fragments per kilo bases per million mapped reads using featureCounts v1.5.0 (26). DEGs were identified using the DESeq R package (1.10.1) (27), with the following criteria: Fold-change ≥1.5 or ≤0.667 and adjusted P<0.05 (28).
Gene Ontology (GO) enrichment analysis was conducted by Database for Annotation, Visualization, & Integrated Discovery (DAVID) to reveal the biological function of DEGs (29,30). Kyoto Encyclopedia of Genes and Genomes (KEGG) was utilized to analyze the metabolic and signaling pathways inherent in the DEGs; enrichment and network construction were performed by Metascape 3.5 (31,32). Protein-protein interaction (PPI) networks of putative proteins encoded by DEGs were constructed with STRING 12.0 (33).
Validation by RT-qPCR
Total RNA was isolated from SK-N-AS cells using the SV Total RNA Isolation System (Promega Corporation), then reverse transcribed into cDNA using the GoScript™ kit (Promega Corporation) according to the manufacturer's protocol. qPCR was employed to assess DEG expression using the LineGene 9620 qPCR system (Hangzhou Bioer Co., Ltd.) following the GoTaq® qPCR protocol (Promega Corporation). The primer sequences for amplifying the DEGs are listed in Table I. Relative gene expression level was calculated based on the 2−∆∆Cq method, with GAPDH as the internal control gene (34).
Statistical analysis
Data are presented as mean ± standard deviation. Student's unpaired t-test and one-way analysis of variance with Dunnett's post hoc test were applied for the statistical analyses of the cell viability and qPCR results. Fisher's exact test was used for bioinformatic analysis. P<0.05 was considered to indicate a statistically significant difference. Statistical analysis was performed using SPSS 26.0 (IBM Corp.).
Results
Inhibitory effect of SSa on human NB cells
As depicted in Fig. 1A and B, after 24 h, SSa reduced the viability of human SK-N-AS and SH-SY5Y NB cells in a dose-dependent manner. The half maximal inhibitory concentration values for SSa in SK-N-AS and SH-SY5Y cells after 24 h were 14.5 and 15.8 µM, respectively. SSa had no inhibitory effect on normal MO3.13 cells at doses <25 µM (Fig. 1C). This indicated that SSa was cytotoxic to SK-N-AS and SH-SY5Y cells and was non-toxic to normal cells at the effective doses, which aligned with the results of a previous study (23). In subsequent experiments, 15 µM was selected for transcriptomic analysis.
Inhibitory effect of SSa on the migration and invasion of SK-N-AS cells
Transwell assays were performed to verify the inhibitory effect of SSa on the migration and invasion of SK-N-AS cells. To prevent interference from the inhibitory effect of SSa on SK-N-AS cell viability, a concentration of SSa (5 µM) was used in these assays. As shown in Fig. 2, SSa significantly inhibited the migration and invasion of SK-N-AS cells, consistent with previous literature (23).
DEGs of SK-N-AS cells in the treatment and control groups
RNA-Seq was carried out to analyze the transcriptomic profile of the SK-A-AS cell samples, producing means of 42.5 million and 46.9 million clean reads for the control group and SSa treatment group, respectively. The overall mapped rates of clean reads to the human reference genome in all samples ranged from 92.41 to 94.68% (Table II). Compared with the control group, there were 297 DEGs in the SSa treatment group; among these genes, 23 were upregulated while 274 were downregulated (Table SI). The volcano plot in Fig. 3A illustrates the DEGs between the SSa treatment and control groups; genes were arranged based on their log2 fold change value along the x-axis and their-log10 adjusted P-value along the y-axis. A hierarchical clustered heatmap was created to visualize the gene expression relationship patterns between the samples. The 3 samples from each group were clustered on the same branch, indicating that SSa treatment significantly changed the gene expression patterns in SK-N-AS cells (Fig. 3B).
GO enrichment results of the DEGs
GO annotation enrichment analyses within the DEGs were assessed to determine the significant biological function changes. A total of 717 GO terms were enriched including 610 biological processes, 59 cellular components and 48 molecular functions. In the biological process category, ‘cell adhesion’ (GO: 0007155), ‘biological adhesion’ (GO: 0022610), ‘neurogenesis’ (GO: 0022008), ‘extracellular matrix organization’ (GO: 0030198) and ‘extracellular structure organization’ (GO: 0043062) were the top five significantly enriched processes. Biological processes related to the antitumor activity of SSa on human NB cells were also enriched, such as ‘apoptotic process’ (GO: 0006915), ‘angiogenesis’ (GO: 0001525) and ‘epithelial to mesenchymal transition’ (GO: 0001837). In the molecular function category, ‘extracellular matrix structural component’ (GO: 0005201), ‘integrin binding’ (GO: 0005178), ‘glycosaminoglycan binding’ (GO: 0005539), ‘receptor binding’ (GO: 0005102) and ‘heparin binding’ (GO: 0008201) represented the major functions encompassed by the DEGs. Moreover, in the cellular component category, the DEGs primarily contributed to ‘extracellular matrix’ (GO: 0031012), ‘extracellular region’ (GO: 0005576), ‘extracellular region part’ (GO: 0044421), ‘cell surface’ (GO: 0009986) and ‘extracellular space’ (GO: 0005615). Fig. 4 illustrates the top 20 GO terms within each category.
KEGG pathway enrichment results of the DEGs
The metabolic and signaling pathways contributing to SSa inhibition of SK-N-AS cells were also analyzed using the KEGG database. The results showed that the DEGs were enriched in 55 KEGG pathways (Table SII). Pathways related to the invasion and migration of cancerous cells were significantly enriched, such as ‘ECM-receptor interaction’ (hsa04512), ‘Focal adhesion’ (hsa04510) and ‘Cell adhesion molecules’ (hsa04514). In addition, 14 pathways belonged to signal transduction and 7 pathways were related to cancer. The signaling pathways included ‘PI3K-Akt signaling pathway’ (hsa04151), ‘TNF signaling pathway’ (hsa04668) and ‘Apelin signaling pathway’ (hsa04371). Cancer-related pathways included Metabolism of central carbon in cancer (hsa05230), ‘Proteoglycans in cancer’ (hsa05205) and Non-small cell lung cancer (hsa05223). Other pathways mainly encompassed human disease and metabolic pathways. The top 20 significantly enriched pathways are shown in Fig. 5A, with connections among the enriched pathways shown as a network in Fig. 5B.
PPI networks based on the products of the DEGs
The list of gene symbols was submitted to the STRING database to construct the PPI network of putative proteins encoded by the DEGs, with ‘organisms’ set to Homo sapiens and the ‘minimum required interaction score’ set to high confidence. As shown in Fig. 6, the largest network contained 98 nodes and 201 edges, while 139 isolated nodes without any connections were hidden. Genes such as FN1, COL1A1, DDX58, PTPRC, THBS1, CCL2, IFIH1, IL1B, MX1 and VCAM1 ranked among the 10 most-connected genes and acted as hubs in the network. A total of 29 clusters were obtained based on Markov clustering, with the largest cluster containing 21 genes whose functions mainly referred to cell adhesion (hsa04512, hsa04514, hsa04510, GO:0007155, GO:0030155 and GO:0016477).
Confirmation of transcriptomic sequencing with qPCR
To verify the transcriptomic sequencing results, genes related to the SSa inhibition of SK-N-AS cells were determined by qPCR. These genes included IL24, EGR1, RET, MDK, PDGFRA, HGF, VCAM1, SLIT3, CD34, FN1, COL1A1 and NCAM1. In the group treated with SSa, certain gene expression levels increased (IL24 and EGR1) or decreased (RET, MDK, PDGFRA, HGF, VCAM1, SLIT3, CD34, FN1, COL1A1 and NCAM1) significantly compared with the control group (Fig. 7). Therefore, the RNA-seq and qPCR data were consistent.
Discussion
SSa is one of the main active components of Bupleurum chinensis DC (10,11). In the present study, the antitumor activity of SSa on human NB cells (SK-N-AS and SH-SY5Y) and the absence of SSa inhibition on normal cells (MO3.13) was verified at the effective concentrations, indicating its safety. The inhibitory effects of SSa on SK-N-AS migration and invasion were also confirmed by Transwell assay. Furthermore, RNA-seq was performed in conjunction with bioinformatics analysis, which revealed the molecular mechanism underlying the antitumor effects of SSa against NB at the mRNA level. As shown in the GO enrichment results, DEGs involved in apoptosis were enriched under the GO term GO:0006915 (apoptosis). The RNA-seq results alongside the qPCR verification indicated that the upregulated genes were IL24 and EGR1 and the downregulated genes were RET and MDK, which was consistent with the apoptosis pathway detected following GO enrichment, as indicated in previous studies (35–39). These results suggested that SSa may be an inhibitor of RET and MDK expression, and hence against NB. Notably, RET has been shown to be expressed in numerous NB cells (40), and its activation inhibits apoptosis in NB cells (38). RET inhibition is a promising antitumor therapeutic strategy and drugs targeting RET have shown promising effects in NB (41,42). As to MDK, some therapeutic strategies aimed at inhibiting MDK expression have also shown promising antitumor effects (43–45). In NB, increased MDK levels are correlated with an unfavorable prognosis, and reducing MDK expression may serve as an effective strategy for NB treatment (39).
In the present study, DEGs linked to angiogenesis were enriched under the GO term GO:0001525 (angiogenesis). The RNA-seq results alongside the qPCR verification indicated that the downregulated genes were PDGFRA, HGF, VCAM1, SLIT3 and CD34 after SSa treatment, which was consistent with the angiogenesis pathway detected in the GO enrichment, as suggested in previous studies (46–50). SSa could inhibit angiogenesis in NB by targeting the PDGFRA and HGF genes (and thus HGF/c-MET signaling). Notably, upregulation of PDGFRA has been observed in a number of malignancies and is associated with poor prognosis (51,52). Additionally, HGF promotes angiogenesis in NB and reports suggest that inhibiting the HGF/c-MET signaling pathway may have therapeutic effects against neuroblastoma (47,53). Meanwhile, VCAM1 and SLIT3 may be proangiogenic factors (48,54), and CD34 has been used as a marker in immunohistochemistry to assess angiogenesis in tumors (50,55). However, the roles of VCAM1, SLIT3 and CD34 in the tumorigenesis of NB are currently unknown. In addition, the PI3K-Akt signaling pathway was downregulated according to the KEGG enrichment results obtained in the present study. This pathway influences angiogenesis by activating multiple angiogenic factors and its activation involves both PDGFRA and HGF (56,57). These findings suggest that SSa may suppress angiogenesis by inhibiting the PI3K-Akt signaling pathway, consistent with previous reports (22,23). The results of the present study also showed that after SSa treatment, the ECM-receptor interaction (hsa04512) in NB cells was the most significantly enriched KEGG pathway, which is closely linked to angiogenesis (GO:0001525). This ECM-receptor interaction pathway participates in the regulation of EMT progression, which is critical for metastasis (58). The expression levels of the FN1, COL1A1 and NCAM1 genes are all related to the EMT process (59–61).
In the present study, the PPI network results showed that the proteins in the extracellular matrix (FN1 and COL1A1) were the most connected proteins. Meanwhile, the RNA-seq results alongside the qPCR verification showed that the corresponding FN1 and COL1A1 genes were both downregulated in the treatment group. These results suggest that SSa could inhibit the metastasis-related EMT process by targeting FN1 and COL1A1. The PPI network analysis also demonstrated that the largest protein cluster primarily encompassed cell adhesion. Consistently, the RNA-seq results alongside the qPCR verification showed that after SSa treatment, the cell adhesion molecule gene, NCAM1, was downregulated in NB cells. Therefore, SSa could inhibit the metastasis-related EMT process by suppressing the expression of NCAM1. Notably, the FN1 and COL1A1 proteins (and the corresponding genes) could be potential targets for cancer treatment (62–64), with NCAM1 more specifically for NB (65,66).
There are certain limitations of the present study. The present study only focused on the impact of SSa on SK-N-AS cells. Given the varying genetic makeup of different NB cells (2), further research is needed to determine whether the findings can be generalized to other NB cells. The present study elucidated the mechanism of SSa against NB at the transcriptional level. However, examination at the protein level is equally crucial for the determining the antitumor activity of the drug. Taking the aforementioned PI3K-Akt signaling pathway as an example, in addition to expression levels, the phosphorylation status of core proteins also plays a vital role in pathway activation. Therefore, further investigation is warranted to gain deeper insights into the molecular mechanisms underlying the anti-neuroblastoma effects of SSa. Additionally, subsequent animal studies should be performed to facilitate the clinical translation of SSA for neuroblastoma treatment.
In conclusion, the results of the present study showed that SSa significantly inhibited the viability, migration and invasion of SK-N-AS NB cells within 24 h. SSa likely induced apoptosis in SK-N-AS cells by upregulating IL24 and EGR1 and downregulating RET and MDK expression. SSa may have anti-angiogenesis effects through downregulating PDGFRA, HGF, VCAM1, SLIT3 and CD34. SSa may suppress metastasis by inhibiting EMT through downregulating FN1 (central hub), COL1A1 and NCAM1 expression as well as downregulating the PI3K-Akt signaling pathway. Therefore, the present study provided an in-depth comprehension of the molecular processes and signal transduction pathways driving the effects of SSa against NB through RNA-seq and bioinformatics analyses. However, the specific role of the relevant genes in the anti-NB process of SSa and the direct target of SSa still needs further investigation. These findings suggest that SSa can be a potential therapeutic agent for NB.
Supplementary Material
Supporting Data
Acknowledgements
Not applicable.
Funding
This work was supported by the National Natural Science Foundation of China (grant nos. 81503271 and 81573539), University Nursing Program for Young Scholars with Creative Talents in Heilongjiang Province (grant no. UNPYSCT-2017217) and the Heilongjiang Touyan Innovation Team Program [grant no. (2019) No. 5].
Availability of data and materials
The RNA-seq data generated in the present study may be found in the Sequence Read Archive database under accession no. PRJNA1158969 or at the following URL: https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1158969. All other data generated in the present study may be requested from the corresponding author.
Authors' contributions
NG was responsible for study design and contributed to data interpretation. WZ, LD and HC performed the experiments. JS, BL and YC analyzed the data and modified the manuscript. BL and YC confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Zafar A, Wang W, Liu G, Xian W, McKeon F, Zhou J and Zhang R: Targeting the p53-MDM2 pathway for neuroblastoma therapy: Rays of hope. Cancer Lett. 496:16–29. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bansal M, Gupta A and Ding HF: MYCN and metabolic reprogramming in neuroblastoma. Cancers (Basel). 14:41132022. View Article : Google Scholar : PubMed/NCBI | |
Zafar A, Wang W, Liu G, Wang X, Xian W, McKeon F, Foster J, Zhou J and Zhang R: Molecular targeting therapies for neuroblastoma: Progress and challenges. Med Res Rev. 41:961–1021. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lundberg KI, Treis D and Johnsen JI: Neuroblastoma heterogeneity, plasticity, and emerging therapies. Curr Oncol Rep. 24:1053–1062. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin L, Miao L, Lin H, Cheng J, Li M, Zhuo Z and He J: Targeting RAS in neuroblastoma: Is it possible? Pharmacol Ther. 236:1080542022. View Article : Google Scholar : PubMed/NCBI | |
Qiu B and Matthay KK: Advancing therapy for neuroblastoma. Nat Rev Clin Oncol. 19:515–533. 2022. View Article : Google Scholar : PubMed/NCBI | |
Whittle SB, Smith V, Doherty E, Zhao S, McCarty S and Zage PE: Overview and recent advances in the treatment of neuroblastoma. Expert Rev Anticancer Ther. 17:369–386. 2017. View Article : Google Scholar : PubMed/NCBI | |
Rivera Z, Escutia C, Madonna MB and Gupta KH: Biological insight and recent advancement in the treatment of neuroblastoma. Int J Mol Sci. 24:84702023. View Article : Google Scholar : PubMed/NCBI | |
Gao J, Fosbrook C, Gibson J, Underwood TJ, Gray JC and Walters ZS: Review: Targeting EZH2 in neuroblastoma. Cancer Treat Rev. 119:1026002023. View Article : Google Scholar : PubMed/NCBI | |
Li X, Li X, Huang N, Liu R and Sun R: A comprehensive review and perspectives on pharmacology and toxicology of saikosaponins. Phytomedicine. 50:73–87. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li XQ, Song YN, Wang SJ, Rahman K, Zhu JY and Zhang H: Saikosaponins: A review of pharmacological effects. J Asian Nat Prod Res. 20:399–411. 2018. View Article : Google Scholar : PubMed/NCBI | |
Xiao LX, Zhou HN and Jiao ZY: Present and future prospects of the anti-cancer activities of saikosaponins. Curr Cancer Drug Targets. 23:2–14. 2022.PubMed/NCBI | |
Motoo Y and Sawabu N: Antitumor effects of saikosaponins, baicalin and baicalein on human hepatoma cell lines. Cancer Lett. 86:91–95. 1994. View Article : Google Scholar : PubMed/NCBI | |
Qian L, Murakami T, Kimura Y, Takahashi M and Okita K: Saikosaponin A-induced cell death of a human hepatoma cell line (HuH-7): The significance of the ‘sub-G1 peak’ in a DNA histo. Pathol Int. 45:207–214. 1995. View Article : Google Scholar : PubMed/NCBI | |
Wen-Sheng W: ERK signaling pathway is involved in p15INK4b/p16INK4a expression and HepG2 growth inhibition triggered by TPA and saikosaponin A. Oncogene. 22:955–963. 2003. View Article : Google Scholar : PubMed/NCBI | |
Wu WS and Hsu HY: Involvement of p-15(INK4b) and p-16(INK4a) gene expression in saikosaponin a and TPA-induced growth inhibition of HepG2 cells. Biochem Biophys Res Commun. 285:183–187. 2001. View Article : Google Scholar : PubMed/NCBI | |
Kang SJ, Lee YJ, Kang SG, Cho S, Yoon W, Lim JH, Min SH, Lee TH and Kim BM: Caspase-4 is essential for saikosaponin a-induced apoptosis acting upstream of caspase-2 and γ-H2AX in colon cancer cells. Oncotarget. 8:100433–100448. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kim BM and Hong SH: Sequential caspase-2 and caspase-8 activation is essential for saikosaponin a-induced apoptosis of human colon carcinoma cell lines. Apoptosis. 16:184–197. 2011. View Article : Google Scholar : PubMed/NCBI | |
Chen JC, Chang NW, Chung JG and Chen KC: Saikosaponin-A induces apoptotic mechanism in human breast MDA-MB-231 and MCF-7 cancer cells. Am J Chin Med. 31:363–377. 2003. View Article : Google Scholar : PubMed/NCBI | |
Zhao X, Liu J, Ge S, Chen C, Li S, Wu X, Feng X, Wang Y and Cai D: Saikosaponin A inhibits breast cancer by regulating Th1/Th2 balance. Front Pharmacol. 10:6242019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Dai K, Xu D, Fan H, Ji N, Wang D, Zhao Y and Liu R: Saikosaponin A alleviates glycolysis of breast cancer cells through repression of Akt/STAT3 pathway. Chem Biol Drug Des. 102:115–125. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shi C, Sun L, Fang R, Zheng S, Yu M and Li Q: Saikosaponin-A exhibits antipancreatic cancer activity by targeting the EGFR/PI3K/Akt pathway. Curr Pharm Biotechnol. 24:579–588. 2023. View Article : Google Scholar : PubMed/NCBI | |
Cheng T and Ying M: Antitumor effect of Saikosaponin A on human neuroblastoma cells. Biomed Res Int. 2021:58455542021. View Article : Google Scholar : PubMed/NCBI | |
Bolger AM, Lohse M and Usadel B: Trimmomatic: A flexible trimmer for illumina sequence data. Bioinformatics. 30:2114–2120. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kim D, Paggi JM, Park C, Bennett C and Salzberg SL: Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 37:907–915. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liao Y, Smyth GK and Shi W: featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 30:923–930. 2014. View Article : Google Scholar : PubMed/NCBI | |
Anders S and Huber W: Differential expression analysis for sequence count data. Genome Biol. 11:R1062010. View Article : Google Scholar : PubMed/NCBI | |
Robinson MD, McCarthy DJ and Smyth GK: EdgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI | |
Gene Ontology Consortium, . Aleksander SA, Balhoff J, Carbon S, Cherry JM, Drabkin HJ, Ebert D, Feuermann M, Gaudet P, Harris NL, et al: The gene ontology knowledgebase in 2023. Genetics. 224:iyad0312023. View Article : Google Scholar : PubMed/NCBI | |
Sherman BT, Hao M, Qiu J, Jiao X, Baseler MW, Lane HC, Imamichi T and Chang W: DAVID: A web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 50:W216–W221. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kanehisa M, Furumichi M, Sato Y, Kawashima M and Ishiguro-Watanabe M: KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 51:D587–D592. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C and Chanda SK: Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 10:15232019. View Article : Google Scholar : PubMed/NCBI | |
Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
Pignatelli M, Luna-Medina R, Pérez-Rendón A, Santos A and Perez-Castillo A: The transcription factor early growth response factor-1 (EGR-1) promotes apoptosis of neuroblastoma cells. Biochem J. 373:739–746. 2003. View Article : Google Scholar : PubMed/NCBI | |
Cibelli G, Policastro V, Rössler OG and Thiel G: Nitric oxide-induced programmed cell death in human neuroblastoma cells is accompanied by the synthesis of Egr-1, a zinc finger transcription factor. J Neurosci Res. 67:450–460. 2002. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Zhang H, Zhu X, Feng D, Gong J and Han T: Interleukin-24 induces neuroblastoma SH-SY5Y cell differentiation, growth inhibition, and apoptosis by promoting ROS production. J Interferon Cytokine Res. 33:709–714. 2013. View Article : Google Scholar : PubMed/NCBI | |
Skinner MA, Lackey KE and Freemerman AJ: RET activation inhibits doxorubicin-induced apoptosis in SK-N-MC cells. Anticancer Res. 28:2019–2025. 2008.PubMed/NCBI | |
Kishida S and Kadomatsu K: Involvement of midkine in neuroblastoma tumourigenesis. Br J Pharmacol. 171:896–904. 2014. View Article : Google Scholar : PubMed/NCBI | |
Futami H and Sakai R: RET protein promotes non-adherent growth of NB-39-nu neuroblastoma cell line. Cancer Sci. 100:1034–1039. 2009. View Article : Google Scholar : PubMed/NCBI | |
Steen EA, Basilaia M, Kim W, Getz T, Gustafson JL and Zage PE: Targeting the RET tyrosine kinase in neuroblastoma: A review and application of a novel selective drug design strategy. Biochem Pharmacol. 216:1157512023. View Article : Google Scholar : PubMed/NCBI | |
Ishida M, Ichihara M, Mii S, Jijiwa M, Asai N, Enomoto A, Kato T, Majima A, Ping J, Murakumo Y and Takahashi M: Sprouty2 regulates growth and differentiation of human neuroblastoma cells through RET tyrosine kinase. Cancer Sci. 98:815–821. 2007. View Article : Google Scholar : PubMed/NCBI | |
Erdogan S, Doganlar ZB, Doganlar O, Turkekul K and Serttas R: Inhibition of midkine suppresses prostate cancer CD133+ stem cell growth and migration. Am J Med Sci. 354:299–309. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hao H, Maeda Y, Fukazawa T, Yamatsuji T, Takaoka M, Bao XH, Matsuoka J, Okui T, Shimo T, Takigawa N, et al: Inhibition of the growth factor MDK/midkine by a novel small molecule compound to treat non-small cell lung cancer. PLoS One. 8:e710932013. View Article : Google Scholar : PubMed/NCBI | |
Han X, Li M, Xu J, Fu J, Wang X, Wang J, Xia T, Wang S and Ma G: miR-1275 targets MDK/AKT signaling to inhibit breast cancer chemoresistance by lessening the properties of cancer stem cells. Int J Biol Sci. 19:89–103. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hou Y, Du W, Wu Q, Chai X, Wang Y, Mi Y, Tian Y, Tang M, Li J and Yan D: PDGFRA exhibits potential as an indicator of angiogenesis within the tumor microenvironment and is up-regulated in BLCA. Microvasc Res. 151:1046142024. View Article : Google Scholar : PubMed/NCBI | |
Daudigeos-Dubus E, LeDret L, Bawa O, Opolon P, Vievard A, Villa I, Bosq J, Vassal G and Geoerger B: Dual inhibition using cabozantinib overcomes HGF/MET signaling mediated resistance to pan-VEGFR inhibition in orthotopic and metastatic neuroblastoma tumors. Int J Oncol. 50:203–211. 2017. View Article : Google Scholar : PubMed/NCBI | |
Xiang X, Pathak JL, Wu W, Li J, Huang W, Wu Q, Xin M, Wu Y, Huang Y, Ge L and Zeng S: Human serum-derived exosomes modulate macrophage inflammation to promote VCAM1-mediated angiogenesis and bone regeneration. J Cell Mol Med. 27:1131–1143. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yallowitz AR, Shim JH, Xu R and Greenblatt MB: An angiogenic approach to osteoanabolic therapy targeting the SHN3-SLIT3 pathway. Bone. 172:1167612023. View Article : Google Scholar : PubMed/NCBI | |
Sasano H and Suzuki T: Pathological evaluation of angiogenesis in human tumor. Biomed Pharmacother. 59 (Suppl 2):S334–S336. 2005. View Article : Google Scholar : PubMed/NCBI | |
Lin LH, Lin JS, Yang CC, Cheng HW, Chang KW and Liu CJ: Overexpression of platelet-derived growth factor and its receptor are correlated with oral tumorigenesis and poor prognosis in oral squamous cell carcinoma. Int J Mol Sci. 21:23602020. View Article : Google Scholar : PubMed/NCBI | |
Wei T, Zhang LN, Lv Y, Ma XY, Zhi L, Liu C, Ma F and Zhang XF: Overexpression of platelet-derived growth factor receptor alpha promotes tumor progression and indicates poor prognosis in hepatocellular carcinoma. Oncotarget. 5:10307–10317. 2014. View Article : Google Scholar : PubMed/NCBI | |
Hecht M, Papoutsi M, Tran HD, Wilting J and Schweigerer L: Hepatocyte growth factor/c-Met signaling promotes the progression of experimental human neuroblastomas. Cancer Res. 64:6109–6118. 2004. View Article : Google Scholar : PubMed/NCBI | |
Paul JD, Coulombe KLK, Toth PT, Zhang Y, Marsboom G, Bindokas VP, Smith DW, Murry CE and Rehman J: SLIT3-ROBO4 activation promotes vascular network formation in human engineered tissue and angiogenesis in vivo. J Mol Cell Cardiol. 64:124–131. 2013. View Article : Google Scholar : PubMed/NCBI | |
Liu H and Zhao KY: Application of CD34 expression combined with three-phase dynamic contrast-enhanced computed tomography scanning in preoperative staging of gastric cancer. World J Gastrointest Surg. 15:2513–2524. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mei Y, Wang Z, Zhang L, Zhang Y, Li X, Liu H, Ye J and You H: Regulation of neuroblastoma differentiation by forkhead transcription factors FOXO1/3/4 through the receptor tyrosine kinase PDGFRA. Proc Natl Acad Sci USA. 109:4898–4903. 2012. View Article : Google Scholar : PubMed/NCBI | |
Moosavi F, Giovannetti E, Peters GJ and Firuzi O: Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol. 160:1032342021. View Article : Google Scholar : PubMed/NCBI | |
An Q, Liu T, Wang MY, Yang YJ, Zhang ZD, Lin ZJ and Yang B: CircKRT7-miR-29a-3p-COL1A1 axis promotes ovarian cancer cell progression. Onco Targets Ther. 13:8963–8976. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dehghan MH, Ashrafi MR, Hedayati M, Shivaee S and Rajabi S: Oral contraceptive steroids promote papillary thyroid cancer metastasis by targeting angiogenesis and epithelial-mesenchymal transition. Int J Mol Cell Med. 10:219–226. 2021.PubMed/NCBI | |
Li X, Sun X, Kan C, Chen B, Qu N, Hou N, Liu Y and Han F: COL1A1: A novel oncogenic gene and therapeutic target in malignancies. Pathol Res Pract. 236:1540132022. View Article : Google Scholar : PubMed/NCBI | |
Li J, Yang R, Yang H, Chen S, Wang L, Li M, Yang S, Feng Z and Bi J: NCAM regulates the proliferation, apoptosis, autophagy, EMT, and migration of human melanoma cells via the Src/Akt/mTOR/cofilin signaling pathway. J Cell Biochem. 121:1192–1204. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Cao G, Cai H, Huang H and Zhu X: The effect and clinical significance of FN1 expression on biological functions of gastric cancer cells. Cell Mol Biol (Noisy-le-grand). 66:191–198. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cao M, Xiao D and Ding X: The anti-tumor effect of ursolic acid on papillary thyroid carcinoma via suppressing fibronectin-1. Biosci Biotechnol Biochem. 84:2415–2424. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ding Y, Zhang M, Hu S, Zhang C, Zhou Y, Han M, Li J, Li F, Ni H, Fang S and Chen Q: MiRNA-766-3p inhibits gastric cancer via targeting COL1A1 and regulating PI3K/AKT signaling pathway. J Cancer. 15:990–998. 2024. View Article : Google Scholar : PubMed/NCBI | |
Markovsky E, Eldar-Boock A, Ben-Shushan D, Baabur-Cohen H, Yeini E, Pisarevsky E, Many A, Aviel-Ronen S, Barshack I and Satchi-Fainaro R: Targeting NCAM-expressing neuroblastoma with polymeric precision nanomedicine. J Control Release. 249:162–172. 2017. View Article : Google Scholar : PubMed/NCBI | |
Heinly BE and Grant CN: Cell adhesion molecules in neuroblastoma: Complex roles, therapeutic potential. Front Oncol. 12:7821862022. View Article : Google Scholar : PubMed/NCBI |