• Title/Summary/Keyword: LN229

Search Result 8, Processing Time 0.029 seconds

Dexamethasone Interferes with Autophagy and Affects Cell Survival in Irradiated Malignant Glioma Cells

  • Komakech, Alfred;Im, Ji-Hye;Gwak, Ho-Shin;Lee, Kyue-Yim;Kim, Jong Heon;Yoo, Byong Chul;Cheong, Heesun;Park, Jong Bae;Kwon, Ji Woong;Shin, Sang Hoon;Yoo, Heon
    • Journal of Korean Neurosurgical Society
    • /
    • v.63 no.5
    • /
    • pp.566-578
    • /
    • 2020
  • Objective : Radiation is known to induce autophagy in malignant glioma cells whether it is cytocidal or cytoprotective. Dexamethasone is frequently used to reduce tumor-associated brain edema, especially during radiation therapy. The purpose of the study was to determine whether and how dexamethasone affects autophagy in irradiated malignant glioma cells and to identify possible intervening molecular pathways. Methods : We prepared p53 mutant U373 and LN229 glioma cell lines, which varied by phosphatase and tensin homolog (PTEN) mutational status and were used to make U373 stable transfected cells expressing GFP-LC3 protein. After performing cell survival assay after irradiation, the IC50 radiation dose was determined. Dexamethasone dose (10 μM) was determined from the literature and added to the glioma cells 24 hours before the irradiation. The effect of adding dexamethasone was evaluated by cell survival assay or clonogenic assay and cell cycle analysis. Measurement of autophagy was visualized by western blot of LC3-I/LC3-II and quantified by the GFP-LC3 punctuated pattern under fluorescence microscopy and acridine orange staining for acidic vesicle organelles by flow cytometry. Results : Dexamethasone increased cell survival in both U373 and LN229 cells after irradiation. It interfered with autophagy after irradiation differently depending on the PTEN mutational status : the autophagy decreased in U373 (PTEN-mutated) cells but increased in LN229 (PTEN wild-type) cells. Inhibition of protein kinase B (AKT) phosphorylation after irradiation by LY294002 reversed the dexamethasone-induced decrease of autophagy and cell death in U373 cells but provoked no effect on both autophagy and cell survival in LN229 cells. After ATG5 knockdown, radiation-induced autophagy decreased and the effect of dexamethasone also diminished in both cell lines. The diminished autophagy resulted in a partial reversal of dexamethasone protection from cell death after irradiation in U373 cells; however, no significant change was observed in surviving fraction LN229 cells. Conclusion : Dexamethasone increased cell survival in p53 mutated malignant glioma cells and increased autophagy in PTEN-mutant malignant glioma cell but not in PTEN-wildtype cell. The difference of autophagy response could be mediated though the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin signaling pathway.

Autophagy Involvement in Olanzapine-Mediated Cytotoxic Effects in Human Glioma Cells

  • Wang, Yi-Xuan;Xu, Shu-Qing;Chen, Xiang-Hui;Liu, Rui-Si;Liang, Zhong-Qin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.19
    • /
    • pp.8107-8113
    • /
    • 2014
  • The aim of this study was to investigate the effects of olanzapine on growth inhibition as well as autophagy in glioma cells in vitro and in vivo. The proliferation of both LN229 and T98 glioma cells, measured by MTT assay, was suppressed in a concentration-dependent and time-dependent manner. Moreover, apoptosis of both cells was significantly increased with the treatment of olanzapine as evidenced by increased Bcl-2 expression, Hoechst 33258 staining and annexinV-FITC/PI staining. Olanzapine treatment also enhanced activation of autophagy with increased expression of LC3-II, expression of protein p62, a substrate of autophagy, being decreased. The growth inhibition by olanzapine in both glioma cell lines could be blocked by co-treatment with 3-MA, an autophagy inhibitor. Furthermore, olanzapine effectively blocked the growth of subcutaneous xenografts of LN229 glioma cells in vivo. The increased level of protein LC3-II and decreased level of p62 followed by a decreased level of Bcl-2, suggesting that autophagy may contribute to apoptosis. In addition, reduced proliferation of glioma cells was shown by a decrease of Ki-67 staining and increased caspase-3 staining indicative of apoptosis in mouse xenografts. These results indicated that olanzapine inhibited the growth of glioma cells accompanied by induction of autophagy and apoptosis both in vitro and in vivo. Olanzapine-induced autophagy plays a tumor-suppressing role in glioma cells.

CXCR4-STAT3 Axis Plays a Role in Tumor Cell Infiltration in an Orthotopic Mouse Glioblastoma Model

  • Han, Ji-hun;Yoon, Jeong Seon;Chang, Da-Young;Cho, Kyung Gi;Lim, Jaejoon;Kim, Sung-Soo;Suh-Kim, Haeyoung
    • Molecules and Cells
    • /
    • v.43 no.6
    • /
    • pp.539-550
    • /
    • 2020
  • Glioblastoma multiforme (GBM) is a fatal malignant tumor that is characterized by diffusive growth of tumor cells into the surrounding brain parenchyma. However, the diffusive nature of GBM and its relationship with the tumor microenvironment (TME) is still unknown. Here, we investigated the interactions of GBM with the surrounding microenvironment in orthotopic xenograft animal models using two human glioma cell lines, U87 and LN229. The GBM cells in our model showed different features on the aspects of cell growth rate during their development, dispersive nature of glioma tumor cells along blood vessels, and invasion into the brain parenchyma. Our results indicated that these differences in the two models are in part due to differences in the expression of CXCR4 and STAT3, both of which play an important role in tumor progression. In addition, the GBM shows considerable accumulation of resident microglia and peripheral macrophages, but polarizes differently into tumor-supporting cells. These results suggest that the intrinsic factors of GBM and their interaction with the TME determine the diffusive nature and probably the responsiveness to non-cancer cells in the TME.

Propranolol Inhibits the Proliferation of Human Glioblastoma Cell Lines through Notch1 and Hes1 Signaling System

  • Kim, Hyun Sik;Park, Young Han;Lee, Heui Seung;Kwon, Mi Jung;Song, Joon Ho;Chang, In Bok
    • Journal of Korean Neurosurgical Society
    • /
    • v.64 no.5
    • /
    • pp.716-725
    • /
    • 2021
  • Objective : The anti-tumor effect of the beta-adrenergic receptor antagonist propranolol in breast cancer is well known; however, its activity in glioblastoma is not well-evaluated. The Notch-Hes pathway is known to regulate cell differentiation, proliferation, and apoptosis. We investigated the effect of propranolol to human glioblastoma cell lines, and the role of Notch and Hes signaling in this process. Methods : We performed immunohistochemical staining on 31 surgically resected primary human glioblastoma tissues. We also used glioblastoma cell lines of U87-MG, LN229, and neuroblastoma cell line of SH-SY5Y in this study. The effect of propranolol and isoproterenol on cell proliferation was evaluated using the MTT assay (absorbance 570 nm). The impact of propranolol on gene expression (Notch and Hes) was evaluated using real-time polymerase chain reaction (RT-PCR, whereas protein levels of Notch1 and Hes1 were measured using Western blotting (WB), simultaneously. Small interfering RNA (siRNA) was used to suppress the Notch gene to investigate its role in the proliferation of glioblastoma. Results : Propranolol and isoproterenol caused a dose-dependent decrease in cell proliferation (MTT assay). RT-PCR showed an increase in Notch1 and Hes1 expression by propranolol, whereas WB demonstrated increase in Notch1 protein, but a decrease in Hes1 by propranolol. The proliferation of U87-MG and LN229 was not significantly suppressed after transfection with Notch siRNA. Conclusion : These results demonstrated that propranolol suppressed the proliferation of glioblastoma cell lines and neuroblastoma cell line, and Hes1 was more closely involved than Notch1 was in glioblastoma proliferation.

Unusual Electrical Transport Characteristic of the SrSnO3/Nb-Doped SrTiO3 Heterostructure

  • De-Peng Wang;Rui-Feng Niu;Li-Qi Cui;Wei-Tian Wang
    • Korean Journal of Materials Research
    • /
    • v.33 no.6
    • /
    • pp.229-235
    • /
    • 2023
  • An all-perovskite oxide heterostructure composed of SrSnO3/Nb-doped SrTiO3 was fabricated using the pulsed laser deposition method. In-plane and out-of-plane structural characterization of the fabricated films were analyzed by x-ray diffraction with θ-2θ scans and φ scans. X-ray photoelectron spectroscopy measurement was performed to check the film's composition. The electrical transport characteristic of the heterostructure was determined by applying a pulsed dc bias across the interface. Unusual transport properties of the interface between the SrSnO3 and Nb-doped SrTiO3 were investigated at temperatures from 100 to 300 K. A diodelike rectifying behavior was observed in the temperature-dependent current-voltage (IV) measurements. The forward current showed the typical IV characteristics of p-n junctions or Schottky diodes, and were perfectly fitted using the thermionic emission model. Two regions with different transport mechanism were detected, and the boundary curve was expressed by ln I = -1.28V - 13. Under reverse bias, however, the temperature- dependent IV curves revealed an unusual increase in the reverse-bias current with decreasing temperature, indicating tunneling effects at the interface. The Poole-Frenkel emission was used to explain this electrical transport mechanism under the reverse voltages.

Fabrication of InP/InGaAs HPT with ITO Transparent Emitter Contact (ITO 투명전극을 갖는 InP/InGaAs HPTs 제작)

  • Kim, Young-Geun;Jang, Eun-Sook;Choi, Byong-Gun;Shin, Ju-Sun;Sung, Kyang-Su;Han, Kyo-Yong
    • Proceedings of the IEEK Conference
    • /
    • 2000.11b
    • /
    • pp.229-232
    • /
    • 2000
  • InP/lnGaAs HPT's were fabricated by employing Indium Tin Oxide(ITO) transparent emitter contact. The device showed the current gaing 70 was obtained but the emitter series resistance was significantly increased. the electrical charateristics of the device were similar to HBT's. However Vceoff was shifted the positive direction. Such a shift ma be resulted from the formation of the shottky barrier rather than the ohmic contact between ITO and n+ InP emitter.

  • PDF

Short-term Outcomes of Pylorus-Preserving Gastrectomy for Early Gastric Cancer: Comparison Between Extracorporeal and Intracorporeal Gastrogastrostomy

  • Alzahrani, Khalid;Park, Ji-Hyeon;Lee, Hyuk-Joon;Park, Shin-Hoo;Choi, Jong-Ho;Wang, Chaojie;Alzahrani, Fadhel;Suh, Yun-Suhk;Kong, Seong-Ho;Park, Do Joong;Yang, Han-Kwang
    • Journal of Gastric Cancer
    • /
    • v.22 no.2
    • /
    • pp.135-144
    • /
    • 2022
  • Purpose: This study aimed to compare the surgical and oncological outcomes between totally laparoscopic pylorus-preserving gastrectomy (TLPPG) with intracorporeal anastomosis and laparoscopy-assisted pylorus-preserving gastrectomy (LAPPG) with extracorporeal anastomosis. Materials and Methods: A retrospective analysis was performed in 258 patients with cT1N0 gastric cancer who underwent laparoscopic pylorus-preserving gastrectomy using two different anastomosis methods: TLPPG with intracorporeal anastomosis (n=88) and LAPPG with extracorporeal anastomosis (n=170). The following variables were compared between the two groups to assess the postoperative surgical and oncological outcomes: proximal and distal margins, number of resected lymph nodes (LNs) in total and in LN station 6, operation time, postoperative hospital stay, and postoperative morbidity including delayed gastric emptying (DGE). Results: The average length of the proximal margin was similar between the TLPPG and LAPPG groups (2.35 vs. 2.73 cm, P=0.070). Although the distal margin was significantly shorter in the TLPPG group than in the LAPPG group (3.15 vs. 4.08 cm, P=0.001), no proximal or distal resection margin-positive cases were reported in either group. The average number of resected LN was similar in both groups (36.0 vs. 33.98, P=0.229; LN station 6, 5.72 vs. 5.33, P=0.399). The operation time was shorter in the TLPPG group than in the LAPPG (200.17 vs. 220.80 minutes, P=0.001). No significant differences were observed between the two groups in terms of postoperative hospital stay (9.38 vs. 10.10 days, P=0.426) and surgical complication rate (19.3% vs. 22.9%), including DGE (8.0% vs. 11.8%, P=0.343). Conclusions: The oncological safety and postoperative complications of TLPPG with intracorporeal anastomosis are similar to those of LAPPG with extracorporeal anastomosis.

MicroRNA-296-5p Promotes Invasiveness through Downregulation of Nerve Growth Factor Receptor and Caspase-8

  • Lee, Hong;Shin, Chang Hoon;Kim, Hye Ree;Choi, Kyung Hee;Kim, Hyeon Ho
    • Molecules and Cells
    • /
    • v.40 no.4
    • /
    • pp.254-261
    • /
    • 2017
  • Glioblastomas (GBM) are very difficult to treat and their aggressiveness is one of the main reasons for this as well as for the frequent recurrences. MicroRNAs post-transcriptionally regulate their target genes through interaction between their seed sequence and 3'UTR of the target mRNAs. We previously reported that miR-296-3p is regulated by neurofibromatosis 2 (NF2) and enhances the invasiveness of GBM cells via SOCS2/STAT3. In this study, we investigated whether miR-296-5p, which originates from the same precursor miRNA as miR-296-3p, can increase the invasiveness of GBM cells. It was observed that miR-296-5p potentiated the invasion of various GBM cells including LN229, T98G, and U87MG. Through bioinformatics approaches, two genes were identified as miR-296-5p targets: caspase-8 (CASP8) and nerve growth factor receptor (NGFR). From results obtained from Ago2 immunoprecipitation and luciferase assays, we found that miR-296-5p downregulates CASP8 and NGFR through direct interaction between seed sequence of the miRNA and 3'UTR of the target mRNA. Knockdown of CASP8 or NGFR also increased the invasive ability of GBM cells, indicating that CASP8 and NGFR are involved in potentiation of invasiveness by miR-296-5p. Consistent with our findings, CASP8 was downregulated in brain metastatic lung cancer cells, which have a high level of miR-296-5p, compared to parental cells, suggesting that miR-296-5p may be generally associated with the acquisition of invasiveness. Collectively, our results implicate miR-296-5p as a potential cause of invasiveness in cancer and suggest it as a promising therapeutic target for GBM.