Ying-Qiao Wang's research while affiliated with First Affiliated Hospital of China Medical University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (2)


Exosomes derived from sorafenib-resistant Huh-7 cells induce an enhanced resistance to sorafenib in HCC cells. (A) Sorafenib-resistant cell line, Huh-7-SR, was established and confirmed by the IC50 value. (B) Co-culture systems were established to investigate the effects of sorafenib-resistant HCC cells on regular HCC cells. The group with additional GW4869 served as a negative control. (C) Cell viabilities of HCC cells co-cultured with Huh-7 or Huh-7-SR cells under sorafenib stress were evaluated via CCK-8 assays. (D) Assessment of cell proliferation using colony formation assay in Huh-7 and Hep3B cells with the indicated co-culture model. (E) Concentration and size of exosome were measured by NTA. Exosomes were isolated from the supernatant of the culture medium of Huh-7-SR (Exo-SR) and Huh-7 (Exo-Norm) cells by ultracentrifugation. (F) The iconic cup-shaped double-layer closed vesicle structure of isolated exosome was confirmed by TEM. (G) Western blotting analysis of exosomal markers, including CD9, CD63, CD81, LAMP2, TSG101 and Calnexin. An exosome standard from HEK293 cell line was analyzed as a positive control, while Huh-7-SR cell lysate was performed as a negative control. (H) The fluorescent signal of PKH67 labeled Exo-SR was captured in Huh-7 and Hep3B cells. (I) CCK-8 assays were performed to assess the cell viability of Huh-7 and Hep3B cells treated with sorafenib (6 µM) while co-cultured with the indicated exosomes. (J) Colony formation assays were performed to assess the cell proliferation of Huh-7 and Hep3B cells. The data are presented as the mean ± SD of at least three independent experiments. ***P < 0.001
Exo-SR desensitized HCC cells to sorafenib-mediated ferroptosis. (A) The results of Gene Set Enrichment analysis (GSEA) showed that ferroptosis (the green line) was significantly enriched in Huh-7 cells co-cultured with Exo-SR. (B) Assessment of lipid ROS level in HCC cells co-cultured with Exo-SR by flow cytometry using C11-BODIPY. HCC cells were exposed to sorafenib (6 µM) or DMSO for 24 h. Results are quantified as geometric mean (GeoMean) fluorescence intensity. (C), (D) Quantification of Fe²⁺ and GSH levels in HCC cells treated with indicated conditions. (E) Visualization of ferroptotic morphological changes in HCC cells via TEM. (F) Assessment of mitochondrial membrane potential (MMP) using JC-1 staining. (G) The proliferation of HCC cells treated with indicated conditions was analyzed by colony formation assays. The data are presented as the mean ± SD of at least three independent experiments. **P < 0.01, ***P < 0.001
Exo-SR suppresses ferroptosis by promoting the expression of SLC7A11 in HCC cells. (A) Hierarchical cluster analysis showed differentially expressed genes (DEGs) related to ferroptosis in Huh-7 cells co-cultured with Exo-SR (n = 3) compared to those co-cultured with Exo-Norm (n = 3). The columns represent different cell samples. Each row indicates a gene, and the colors represent the abundance of the transcripts. (B) qRT-PCR analysis was applied to confirm the expression of the top 5 upregulated and 5 downregulated genes in HCC cells co-cultured with Exo-SR. (C) Western blotting assays were performed to evaluate the protein expression of SLC7A11, GPX4 and PCBP2 in the indicated groups, suggesting that Exo-SR significantly upregulated SLC7A11 expression in HCC cells. (D) The expression of SLC7A11 was dramatically more abundant in liver hepatocellular carcinoma (LIHC) than paired normal tissues in TCGA database. (E) The prognosis of abnormal SLC7A11 expression in LIHC patients from TCGA data. Results of Kaplan − Meier survival analysis showed a poor Overall survival (OS) and disease-free survival (DFS) in the high SLC7A11 group. (F) Activity of system Xc- in HCC cells was evaluated by cystine uptake assays. (G) Assessment of lipid ROS level in sorafenib-treated HCC cells, while the effect of Exo-SR on reducing lipid ROS level was blocked in cystine free culture medium. (H), (I), (J) Ferroptosis in sorafenib-stressed HCC cells with the indicated treatment was assessed by intracellular Fe²⁺, GSH and lipid ROS levels, and si-SLC7A11 transfection reversed Exo-SR-mediated suppression on ferroptosis. The data are presented as the mean ± SD of at least three independent experiments. *P < 0.05, ***P < 0.001
Exosomal hsa_circ_0017702 (circUPF2) regulated SLC7A11 gene expression in recipient cells. RNA-seq was performed in three pairs of Exo-SR and Exo-Norm to screen differentially expressed circRNAs. (A) Volcano plot illustrated differentially expressed circRNAs in three pairs of Exo-SR and Exo-Norm (fold change ≥ 2 and P < 0.05). Upregulated circRNAs were shown in red, while downregulated circRNAs were shown in green. (B) Heatmap showed clusters of circRNAs differentially expressed in three pairs of Exo-SR and Exo-Norm (fold change ≥ 4 and P < 0.05). (C) Table presented the information of the top 7 upregulated circRNAs in Exo-SR compared to Exo-Norm. (D), (E) qRT-PCR analysis and western blotting analysis were applied to assess the mRNA and protein expression of SLC7A11 in HCC cells co-cultured with exosomes derived from Huh-7-SR cells transfected with corresponding siRNA. (F) Assessment of the activity of system Xc- in HCC cells co-cultured with the indicated exosomes by cystine uptake assays. (G) Detection of lipid ROS level in sorafenib-treated HCC cells indicated that deprivation of exosomal function occurred when hsa_circ_0017702 in Exo-SR was knocked down. (H) qRT-PCR assays were performed to check the content of hsa_circ_0017702 in circulating exosomes isolated from the blood samples. Blood samples were obtained separately from healthy adult volunteers (Healthy), HCC patients receiving sorafenib with no clinical progressive disease (non-PD) and HCC patients who had been treated with sorafenib but had an imaging-confirmed progressive disease (PD). The data are presented as the mean ± SD of at least three independent experiments. **P < 0.01, ***P < 0.001
The characteristics of circUPF2 and the vital role of circUPF2 in suppressing sorafenib-mediated ferroptosis. (A) Schematic diagram of the circularized transcript generated from the UPF2 gene. The back-splicing site of circUPF2 was validated by Sanger sequencing, and the divergent primers were designed based on the spliced junction of circUPF2. (B) Relative expression changes of circUPF2 in the HCC cells after co-culture with Exo-SR were analyzed by qRT-PCR analysis. (C) RNase R was used to remove the linear transcripts from cellular extracts, leaving circRNAs behind. qRT‒PCR assays were applied to assess the resistance of RNAs to RNase R. (D) RNA decay assay evaluated the stability of circUPF2 and linear UPF2 by RT-qPCR after ActD (1 µg/ml) treatment. (E) CircUPF2 was amplified from both cDNA and gDNA samples by divergent or convergent primers; GAPDH was used as a negative control. Agarose gel electrophoresis visualized the products. (F) Huh-7 and Huh-7-SR cell lines transfected with si-circUPF2 (si-ciR) or negative control siRNA (si-NC) were treated with sorafenib and the values of IC50 were calculated via CCK-8 assays. (G) Overexpression vectors for circUPF2 (OE-ciR) were constructed. Agarose gel electrophoresis analysis was performed and confirmed that OE-ciR could accurately express circUPF2 but not UPF2 in HCC cells. (H) Cell proliferation was analyzed by colony formation assays in si-ciR transfected Huh-7-SR and OE-ciR transfected Huh-7 cells. Cells were treated with sorafenib (6 µM). (I) Detection of lipid ROS levels in OE-ciR or Vector transfected HCC cells and Exo-SR co-cultured HCC cells. (J), (K) Quantification of Fe²⁺ and GSH in the indicated group of HCC cells. (L) Activity of system Xc- in the indicated group of HCC cells was assessed by cystine uptake assays. (M) MMP of HCC cells in the indicated group was assessed by JC-1 staining. (N) Correlation analysis of circUPF2 and SLC7A11 expression analyzed by qRT-PCR assays in HCC tissues (n = 36). (O) RNA-FISH assay showed that circUPF2 is colocalized with SLC7A11 mRNA in the cytoplasm. (P), (Q) qRT-PCR and western blotting analysis were applied to assess the expression of SLC7A11 in the indicated group of HCC cells. The data are presented as the mean ± SD of at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001

+3

Exosome-derived circUPF2 enhances resistance to targeted therapy by redeploying ferroptosis sensitivity in hepatocellular carcinoma
  • Article
  • Full-text available

May 2024

·

9 Reads

Journal of Nanobiotechnology

Feng-Lin Dong

·

Zong-Zhen Xu

·

Ying-Qiao Wang

·

[...]

·

Jie Li

Background Advanced hepatocellular carcinoma (HCC) can be treated with sorafenib, which is the primary choice for targeted therapy. Nevertheless, the effectiveness of sorafenib is greatly restricted due to resistance. Research has shown that exosomes and circular RNAs play a vital role in the cancer’s malignant advancement. However, the significance of exosomal circular RNAs in the development of resistance to sorafenib in HCC remains uncertain. Methods Ultracentrifugation was utilized to isolate exosomes (Exo-SR) from the sorafenib-resistant HCC cells’ culture medium. Transcriptome sequencing and differential expression gene analysis were used to identify the targets of Exo-SR action in HCC cells. To identify the targets of Exo-SR action in HCC cells, transcriptome sequencing and analysis of differential expression genes were employed. To evaluate the impact of exosomal circUPF2 on resistance to sorafenib in HCC, experiments involving gain-of-function and loss-of-function were conducted. RNA pull-down assays and mass spectrometry analysis were performed to identify the RNA-binding proteins interacting with circUPF2. RNA immunoprecipitation (RIP), RNA pull-down, electrophoretic mobility shift assay (EMSA), immunofluorescence (IF) -fluorescence in situ hybridization (FISH), and rescue assays were used to validate the interactions among circUPF2, IGF2BP2 and SLC7A11. Finally, a tumor xenograft assay was used to examine the biological functions and underlying mechanisms of Exo-SR and circUPF2 in vivo. Results A novel exosomal circRNA, circUPF2, was identified and revealed to be significantly enriched in Exo-SR. Exosomes with enriched circUPF2 enhanced sorafenib resistance by promoting SLC7A11 expression and suppressing ferroptosis in HCC cells. Mechanistically, circUPF2 acts as a framework to enhance the creation of the circUPF2-IGF2BP2-SLC7A11 ternary complex contributing to the stabilization of SLC7A11 mRNA. Consequently, exosomal circUPF2 promotes SLC7A11 expression and enhances the function of system Xc- in HCC cells, leading to decreased sensitivity to ferroptosis and resistance to sorafenib. Conclusions The resistance to sorafenib in HCC is facilitated by the exosomal circUPF2, which promotes the formation of the circUPF2-IGF2BP2-SLC7A11 ternary complex and increases the stability of SLC7A11 mRNA. Focusing on exosomal circUPF2 could potentially be an innovative approach for HCC treatment. Graphical Abstract

Download
Share

Fig. 4 CircTGFBR2 is the effector cargo in Exo-Ts to enhance protective autophagy in HCC cells. A Schematic diagram of the back-splicing transcript generated from linear TGFBR2. B Relative expression changes of circTGFBR2 in HCC cells cocultured with Exo-Ts were measured by qRT-PCR assays. C RNase R (2.5 U/μg, 37 °C, 15 min) was used to remove the linear transcripts from cellular extracts, leaving circRNAs behind. qRT-PCR assays were applied to assess the resistance of RNAs to RNase R. D RNA decay assay evaluated the stability of circTGFBR2 and TGFBR2 in THLE-2 cells by qRT-PCR after ActD (1 μg/ml) treatment. E Divergent and convergent primers for circTGFBR2 were applied to amplify both cDNA and gDNA; GAPDH was used as a negative control. Agarose gel electrophoresis visualized the products. F Overexpression vectors for circTGFBR2 (oe-ciR) containing front and rear circular frames were constructed. PCR assays were present in HCC cell lines and demonstrated that oe-ciR could accurately express circTGFBR2 but not linear TGFBR2. G Western blotting analysis of LC3B and p62 in HCC cell lines cocultured with the indicated exosomes alone or cotransfected with oe-ciR. H Autophagic flux in Hep3B cells after the indicated treatments was observed under fluorescence microscopy. I Apoptosis was assessed by flow cytometry assay in Hep3B and Huh-7 cells cocultured with the indicated exosomes alone or cotransfected with oe-ciR. J The proliferation of HCC cells in low serum concentrations and cocultured with the indicated exosomes alone or cotransfected with oe-ciR was assessed by colony formation assays. The data are presented as the mean ± SD of at least three independent experiments. ***P < 0.001.
Fig. 5 CircTGFBR2 functions as a sponge for miR-205-5p. A Prediction analysis of the interaction between circTGFBR2 and RBPs through CircInteractome, and the physical interaction between circTGFBR2 and AGO2 in HCC cells was validated by RIP assay. B The Venn diagram shows 5 candidate miRNAs identified from the overlap between the miRanda and RNAhybrid databases. C Relative expression changes of the candidate miRNAs in HCC cells transfected with oe-ciR were measured by qRT-PCR assays. D Interaction between circTGFBR2 and the candidate miRNAs was validated by RNA pulldown assay using an antisense probe spanning the junction site of circTGFBR2 (ciR probe). E A negative correlation between the expression of circTGFBR2 and miR-205-5p was observed in HCC tumors and peritumoral normal tissues (P < 0.001, R 2 = 0.271) F RNA-FISH indicates the location of circTGFBR2 and miR-205-5p in HCC cells. CircTGFBR2 was labeled red with Cy3; miR-205-5p was labeled green with FAM; Nuclei were stained blue with DAPI. G Dual luciferase reporter assay was used to detect the relative luciferase activity (firefly/Renilla) in 293 T cells cotransfected with miR-205-5p mimics and pMIR-circTGFBR2 WT/MUT. H RNA pulldown assays confirmed that the overexpressed circTGFBR2 in HCC cells was dramatically enriched by the biotinylated miR-205-5p. I Results of qRT-PCR assay show the regulatory relationship between circTGFBR2 and miR-205-5p in HCC cells. J Western blotting analysis validated the effect of miR-205-5p on autophagy in HCC cells. The data are presented as the mean ± SD of at least three independent experiments. ***P < 0.001, *P < 0.05.
Fig. 7 CircTGFBR2 promoted HCC progression and tumor cell autophagy in vivo. A Validation of stable circTGFBR2-overexpressing Hep3B cells (Hep3B(oe-ciR)) established by transfecting with pSLenti-EF1-EGFP-F2A-Puro-CMV-S-circTGFBR2-WPRE vectors containing front and rear circular frames. B, C Xenograft tumors of nude mice 26 days after injection of Hep3B(oe-ciR) or Hep3B(vector) cells (n = 6 per group). Tumor growth curves were measured and plotted every 2 days after injection, while tumor weight was measured at the endpoint. D, E IHC staining was implemented to evaluate the expression of Ki-67 and ATG5 in tumor tissues. The results were quantified using Image-Pro Plus 6.0 and are shown as the percentage of positive cells (Ki-67) or the mean density (ATG5). F, G TUNEL staining was implemented to evaluate cell apoptosis in tumor tissues. The results were quantified using Image-Pro Plus 6.0 and are shown as the percentage of positive cells. H The expression of miR-205-5p and ATG5 mRNA in xenograft tumor tissues was measured by qRT-PCR. I Western blotting analysis of ATG5 and the autophagyrelated proteins LC3B and p62 xenograft tumor tissues. The data are presented as the mean ± SD of at least three independent experiments. ***P < 0.001, **P < 0.01.
Circbase ID and information of the candidate circRNAs.
Exosomal circTGFBR2 promotes hepatocellular carcinoma progression via enhancing ATG5 mediated protective autophagy

July 2023

·

42 Reads

·

4 Citations

Cell Death and Disease

Exosomes contribute substantially to the communication between tumor cells and normal cells. Benefiting from the stable structure, circular RNAs (circRNAs) are believed to serve an important function in exosome-mediated intercellular communication. Here, we focused on circRNAs enriched in starvation-stressed hepatocytic exosomes and further investigated their function and mechanism in hepatocellular carcinoma (HCC) progression. Differentially expressed circRNAs in exosomes were identified by RNA sequencing, and circTGFBR2 was identified and chosen for further study. The molecular mechanism of circTGFBR2 in HCC was demonstrated by RNA pulldown, RIP, dual-luciferase reporter assays, rescue experiments and tumor xenograft assay both in vitro and vivo. We confirmed exosomes with enriched circTGFBR2 led to an upregulated resistance of HCC cells to starvation stress. Mechanistically, circTGFBR2 delivered into HCC cells via exosomes serves as a competing endogenous RNA by binding miR-205-5p to facilitate ATG5 expression and enhance autophagy in HCC cells, resulting in resistance to starvation. Thus, we revealed that circTGFBR2 is a novel tumor promoter circRNA in hepatocytic exosomes and promotes HCC progression by enhancing ATG5-mediated protective autophagy via the circTGFBR2/miR-205-5p/ATG5 axis, which may be a potential therapeutic target for HCC.

Citations (1)


... We performed Western blotting using antibodies against CD9 (Ab92726, 1/1000, Abcam), CD63 (Ab216130, 1/1000, Abcam), TSG101 (Ab125011, 1/1000, Abcam), Calnexin (Ab22595,1/1000, Abcam), CD81 (Ab79559, 1/500, Abcam), LAMP2 (Ab199946, 1/2000, Abcam), SLC7A11 (Ab175186, 1/1000, Abcam), GPX4 (67763-1lg, 1/1000, Proteintech), IGF2BP2 (11601-1-AP, 1/1000, Proteintech, ), PCBP2 (Ab184962, 1/1000, Abcam), and GAPDH (10494-1-AP, 1/1000, Proteintech) as described in our previous publications [27]. ...

Reference:

Exosome-derived circUPF2 enhances resistance to targeted therapy by redeploying ferroptosis sensitivity in hepatocellular carcinoma
Exosomal circTGFBR2 promotes hepatocellular carcinoma progression via enhancing ATG5 mediated protective autophagy

Cell Death and Disease