scholarly journals Corrigendum to “LINC01116 targets miR-520a-3p and affects IL6R to promote the proliferation and migration of osteosarcoma cells through the Jak-stat signaling pathway” [Biomed. Pharmacother. 107 (2018) 270–282]

2021 ◽  
Vol 133 ◽  
pp. 110893
Author(s):  
Butian Zhang ◽  
Lili Yu ◽  
Ning Han ◽  
Zhenzhen Hu ◽  
Shuang Wang ◽  
...  
BMC Cancer ◽  
2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Hui Xiao ◽  
Mudan He ◽  
Guogang Xie ◽  
Yanan Liu ◽  
Yuxia Zhao ◽  
...  

Abstract Background Cancer cells release exosomes and can be taken up by mast cells (MCs), but the potential functional effects of MCs on tumor metastasis remain unknown. Method Exosomes were isolated from the lung adenocarcinoma cell line A549, and the uptake of PKH26-labeled exosomes by bone marrow MCs was examined via flow cytometry and fluorescence microscopy. Cytokines and tryptase in MC supernatant were analyzed using an ELISA kit, and the presence of tryptase was evaluated by Western blotting. Cell proliferation and migration were determined through CCK-8 and transwell assays. Proteins in the tryptase-JAK-STAT signaling pathway were detected by Western blotting. Results In this study, we show that exosomes from A549 cells can be taken up by MCs. Moreover, A549 exosomes contain stem cell factor (SCF) to MCs and subsequently induce the activation of MCs through SCF-KIT signal transduction, which leads to MC degranulation and the release of tryptase. Tryptase accelerates the proliferation and migration of human umbilical vein endothelial cells (HUVECs) through the JAK-STAT signaling pathway. Conclusions Our results reveal a mechanism for metastasis in which exosomes can transfer SCF to and activate MCs, which can affect the release of tryptase and the angiogenesis of HUVECs.


2021 ◽  
Author(s):  
ZENG TONG ◽  
Peng Lei ◽  
YIN JUNQIANG ◽  
LIU WEIHAI ◽  
ZHANG DI ◽  
...  

Abstract Background: Osteosarcoma (OS) is the most frequent and high-grade young malignant bone tumor. The prognosis is still poor despite the use of combined therapy involving maximal surgical resection, radiotherapy and chemotherapy. Metabolic reprogramming currently is recognized as one of the hallmarks of cancer. Glutaminase 1(GLS1) has been associated with progression of tumor cell through CDK4 signaling pathway.Methods: In the study, Western blot was used to detect the expression of GLS1 protein in tumor and adjacent normal tissues of osteosarcoma patients, and Western blot was used to detect the expression of GLS1 protein in osteosarcoma cell lines. GLS1 siRNA was transfected into osteosarcoma U2OS cells. Western blot was used to detect the expression of GLS1 protein. MTT and clone formation assay were used to detect cell proliferation. Transwell chamber assay was used to detect migration and invasion. Western blot was used to detect the expression of CDK4 protein in GLS1 knockdown U2OS cells. CB-839 was used to treat U2OS cells. The IC50 value of CB-839 was detected by MTT. The proliferation and migration of CB-839 were detected by clone formation, scratch test, RNA seq sequencing, q-PCR and Western blot.Results: (1) GLS1 was highly expressed in osteosarcoma tissues and cell lines; (2) After transfection, compared with the control group, GLS1 protein expression and CDK4 protein expression of U2OS cells in the knockdown group were significantly down regulated. In vitro experiments showed that the proliferation, migration and invasion of U2OS cells were significantly down regulated; (3) CB-839 promoted apoptosis and inhibited the proliferation and migration of osteosarcoma cells by acting on upstream transcription factors EGR1 and FOXO1. Conclusion: GLS1 can promote the proliferation, migration and invasion of osteosarcoma cells by affecting the cell cycle of CDK4 signaling pathway, and can be used as a potential prognostic indicator and therapeutic target for osteosarcoma patients.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jiong Ma ◽  
Chunxia Zhou ◽  
Xuejun Chen

Abstract Background Hedgehog (Hh) signaling pathway, which is essential for cell proliferation and differentiation, is noted to be aberrantly activated in tumor from increasing studies in recent years. MicroRNAs (miRNAs) as an important non-coding RNA in cells have been proven to possess a regulatory role specific to the Hh signaling pathway. Here, in vitro and in vivo cellular/molecular experiments were adopted to clarify the regulatory mechanism linking miR-636 to the Hh signaling pathway in ovarian cancer (OVC). Methods Protein–protein interaction analysis was performed to identify the hub gene in the Hh pathway. TargetScan database was used to predict the potential upstream regulators for Gli2. qRT-PCR was performed to test the expression of miR-636, while Western blot was conducted to detect the expression of proteins related to the Hh pathway and epithelial-mesenchymal transition (EMT). For cell functional experiments, HO-8910PM OVC cell line was used. MTT assay and wound healing assay were used to measure the effect of miR-636 on cell proliferation and migration. Flow cytometry was carried out to examine the effect of miR-636 on cell cycle, and Western blot was used to identify the change in expression of Hh and EMT-related proteins. Dual-luciferase reporter gene assay was implemented to detect the targeting relationship between miR-636 and Gli2. Xenotransplantation models were established for in vivo examination. Results Gli2 was identified as the hub gene of the Hh pathway and it was validated to be regulated by miR-636 based on the data from TargetScan and GEO databases. In vitro experiments discovered that miR-636 was significantly lowly expressed in OVC cell lines, and overexpressing miR-636 significantly inhibited HO-8910PM cell proliferation, migration and induced cell cycle arrest in G0/G1 phase, while the inhibition of miR-636 caused opposite results. Dual-luciferase reporter gene assay revealed that Gli2 was the target gene of miR-636 in OVC. Besides, overexpressed miR-636 decreased protein expression of Gli2, and affected the expression of proteins related to the Hh signaling pathway and EMT. Rescue experiments verified that overexpression of Gli2 reversed the inhibitory effect of miR-636 on HO-8910PM cell proliferation and migration, and attenuated the blocking effect of miR-636 on cell cycle. The xenotransplantation experiment suggested that miR-636 inhibited cell growth of OVC by decreasing Gli2 expression. Besides, overexpressing Gli2 potentiated the EMT process of OVC cells via decreasing E-cadherin protein expression and increasing Vimentin protein expression, and it reversed the inhibitory effect of miR-636 on OVC cell proliferation in vivo. Conclusion miR-636 mediates the activation of the Hh pathway via binding to Gli2, thus inhibiting EMT, suppressing cell proliferation and migration of OVC. Trial registration: The experimental protocol was established, according to the ethical guidelines of the Helsinki Declaration and was approved by the Human Ethics Committee of The Second Affiliated hospital of Zhejiang University School of Medicine (IR2019001235). Written informed consent was obtained from individual or guardian participants.


2018 ◽  
Vol Volume 11 ◽  
pp. 7019-7029 ◽  
Author(s):  
Sasikamon Khophai ◽  
Malinee Thanee ◽  
Anchalee Techasen ◽  
Nisana Namwat ◽  
Poramate Klanrit ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document