Abstract

BackgroundEmerging evidences suggests that Diosbulbin-B (DB) is effective to improve cisplatin (DDP)-sensitivity in gastric cancer (GC), but its molecular mechanisms were not fully delineated, and this study managed to investigate this issue.MethodsGenes expressions were determined by Real-Time qPCR and Western Blot at transcriptional and translational levels. Cell proliferation and viability were evaluated by cell counting kit-8 (CCK-8) and trypan blue staining assay. Annexin V-FITC/PI double staining assay was used to examine cell apoptosis. The Spheroid formation assay was used to evaluated cell stemness. The xenograft tumor-bearing mice models were established, and the tumors were monitored and the immunohistochemistry (IHC) was employed to examine the expressions and localization of Ki67 protein in mice tumor tissues.ResultsLow-dose DB (12.5 μM) downregulated PD-L1 to activate NLRP3-mediated pyroptosis, and inhibited cancer stem cells (CSCs) properties, to sensitize cisplatin-resistant GC (CR-GC) cells to cisplatin. Mechanistically, the CR-GC cells were obtained, and either low-dose DB or cisplatin alone had little effects on cell viability in CR-GC cells, while low-dose DB significantly induced apoptotic cell death in cisplatin treated CR-GC cells. In addition, low-dose DB triggered cell pyroptosis in CR-GC cells co-treated with cisplatin, which were abrogated by silencing NLRP3. Next, CSCs tended to be enriched in CR-GC cells, instead of their parental cisplatin-sensitive GC (CS-GC) cells, and low-dose DB inhibited spheroid formation and stemness biomarkers (SOX2, OCT4 and Nanog) expressions to eliminate CSCs in CR-GC cells, which were reversed by upregulating programmed death ligand-1 (PD-L1). Furthermore, we proved that PD-L1 negatively regulated NLRP3 in CR-GC cells, and low-dose DB activated NLRP3-mediated pyroptotic cell death in cisplatin treated CR-GC cells by downregulating PD-L1. Also, low-dose DB aggravated the inhibiting effects of cisplatin on tumorigenesis of CR-GC cells in vivo.ConclusionsCollectively, low-dose DB regulated intrinsic PD-L1/NLRP3 pathway to improve cisplatin-sensitivity in CR-GC cells, and this study provided alternative therapy treatments for GC.

Highlights

  • The therapeutic efficacy of the current chemical drugs for gastric cancer (GC) were seriously limited as the results of chemo-resistance [1,2,3], the development of new anti-tumor drugs for GC treatment became urgent and necessary

  • Low‐dose DB sensitized CR‐GC cells to cisplatin treatment DB had been used for cancer treatment [36, 37], and the previous data from our team indicated that low-dose DB (12.5 μM) was advantageous for GC treatment to avoid DB-induced hepatotoxicity [36], which rendered the possibility that low-dose DB (12.5 μM) might be a novel strategy to increase sensitivity of cisplatin-resistant GC (CR-GC) cells to the traditional chemotherapeutic drugs, such as cisplatin

  • The CR-GC cells (SGC7901/CDDP and BGC823/CDDP) were employed to establish xenograft tumor-bearing mice models, and we found that DB and cisplatin co-treatment significantly inhibited tumor weight (P < 0.05, Fig. 1f, Additional file 1: Figure S3) and volume (P < 0.05, Fig. 1g, h) to hamper tumorigenesis of the CR-GC cells in vivo

Read more

Summary

Introduction

The therapeutic efficacy of the current chemical drugs for gastric cancer (GC) were seriously limited as the results of chemo-resistance [1,2,3], the development of new anti-tumor drugs for GC treatment became urgent and necessary. High-dose DB-induced hepatotoxicity [8], and the ineffectiveness of low-dose DB for cancer treatment seriously limited its utilization for GC treatment in clinic [5] To solve this issue, our preliminary work tried to use low-dose DB combined with genes manipulation to treat GC, and the results indicated that this strategy successfully hampered GC development, but had little detrimental effects on human normal hepatocytes (L-02) [5]. Emerging evidences suggests that Diosbulbin-B (DB) is effective to improve cisplatin (DDP)-sensitivity in gastric cancer (GC), but its molecular mechanisms were not fully delineated, and this study managed to investigate this issue

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.