scholarly journals Xanthohumol-Induced Rat Glioma C6 Cells Death by Triggering Mitochondrial Stress

2021 ◽  
Vol 22 (9) ◽  
pp. 4506
Author(s):  
Shaozhi Hou ◽  
Yang Song ◽  
Di Sun ◽  
Shujun Zhu ◽  
Zhenhua Wang

AIM: To investigate the underlying mechanisms of xanthohumol (XN) on the proliferation inhibition and death of C6 glioma cells. METHODS: To determine the effects of XN on C6 cells, cell proliferation and mortality after XN treatment were assessed by SRB assay and trypan blue assay respectively. Apoptotic rates were evaluated by flowcytometry after Annexin V-FITC/PI double staining. The influence of XN on the activity of caspase-3 was determined by Western blot (WB); and nuclear transposition of apoptosis-inducing factor (AIF) was tested by immunocytochemistry and WB. By MitoSOXTM staining, the mitochondrial ROS were detected. Mitochondrial function was also tested by MTT assay (content of succinic dehydrogenase), flow cytometry (mitochondrial membrane potential (MMP)—JC-1 staining; mitochondrial abundance—mito-Tracker green), immunofluorescence (MMP—JC-1 staining; mitochondrial morphology—mito-Tracker green), WB (mitochondrial fusion-fission protein—OPA1, mfn2, and DRP1; mitophagy-related proteins—Pink1, Parkin, LC3B, and P62), and high-performance liquid chromatography (HPLC) (energy charge). Finally, mitochondrial protein homeostasis of C6 cells after XN treatment with and without LONP1 inhibitor bortezomib was investigated by trypan blue assay (proliferative activity and mortality) and WB (mitochondrial protease LONP1). All cell morphology images were taken by a Leica Microsystems microscope. RESULTS: XN could lead to proliferation inhibition and death of C6 cells in a time- and dose-dependent manner and induce apoptosis of C6 cells through the AIF pathway. After long incubation of XN, mitochondria of C6 cells were seriously impaired, and mitochondria had a diffuse morphology and mitochondrial ROS were increased. The content of succinic dehydrogenase per cell was significantly decreased after XN insults of 24, 48, and 72 h. The energy charge was weakened after XN insult of 24 h. Furthermore, the MMP and mitochondrial abundance were significantly decreased; the protein expression levels of OPA1, mfn2, and DRP1 were down-regulated; and the protein expression levels of Pink1, Parkin, LC3B-II/LC3B-I, and p62 were up-regulated in long XN incubation times (24, 48, and 72 h). XN incubation with bortezomib for 48 h resulted in lower proliferative activity and higher mortality of C6 cells and caused the cell to have visible vacuoles. Moreover, the protein expression levels of LONP1 was up-regulated gradually as XN treatment time increased. CONCLUSION: These data supported that XN could induce AIF pathway apoptosis of the rat glioma C6 cells by affecting the mitochondria.

1988 ◽  
Vol 94 (3) ◽  
pp. 279-284 ◽  
Author(s):  
P.W.L. Tas ◽  
H.G. Kress ◽  
K. Koschel
Keyword(s):  
C6 Cells ◽  

2020 ◽  
Vol 122 (7) ◽  
pp. 151625
Author(s):  
Marta Hałas-Wiśniewska ◽  
Magdalena Izdebska ◽  
Wioletta Zielińska ◽  
Alina Grzanka

2009 ◽  
Vol 2009 (8) ◽  
pp. 511-514 ◽  
Author(s):  
Xiang-Yang Qin ◽  
Gui-Rong Ding ◽  
Xiao-Wu Wang ◽  
Juan Tan ◽  
Guo-Zhen Guo ◽  
...  

Nitroxyl compounds have been previously investigated as potential radioprotection drugs. To develop new radioprotectors, two kinds of novel chiral nitronyl nitroxyl radicals: L- tert-butyl 2-(4, 5-dihydro-4, 4, 5, 5-tetramethyl-3-oxido-1 H-imidazol-3-ium-1-oxyl-2-yl) pyrrolidine-1-carboxylate ( L-NNP) and L- tert-butyl 2-[(4-(4, 5-dihydro-4,4,5,5-tetramethyl-3-oxido-1 H-imidazol-3-ium-1-oxyl-2-yl)-2-methoxyphenoxy)methyl] pyrrolidine-1-carboxylate ( L-NNVP) have been synthesised. The cytotoxic and radioprotective effects of these two compounds were then evaluated in rat glioma C6 cells.


Blood ◽  
2006 ◽  
Vol 109 (5) ◽  
pp. 2205-2209 ◽  
Author(s):  
Ivana De Domenico ◽  
Diane McVey Ward ◽  
Giovanni Musci ◽  
Jerry Kaplan

Abstract Ferroportin (Fpn) (IREG1, SLC40A1, MTP1) is an iron transporter, and mutations in Fpn result in a genetically dominant form of iron overload disease. Previously, we demonstrated that Fpn is a multimer and that mutations in Fpn are dominant negative. Other studies have suggested that Fpn is not a multimer and that overexpression or epitope tags might affect the localization, topology, or multimerization of Fpn. We generated wild-type Fpn with 3 different epitopes, GFP, FLAG, and c-myc, and expressed these constructs in cultured cells. Co-expression of any 2 different epitope-tagged proteins in the same cell resulted in their quantitative coimmunoprecipitation. Treatment of Fpn-GFP/Fpn-FLAG–expressing cells with crosslinking reagents resulted in the crosslinking of Fpn-GFP and Fpn-FLAG. Western analysis of rat glioma C6 cells or mouse bone marrow macrophages exposed to crosslinking reagents showed that endogenous Fpn is a dimer. These results support the hypothesis that the dominant inheritance of Fpn–iron overload disease is due to the dominant-negative effects of mutant Fpn proteins.


Neuropeptides ◽  
1995 ◽  
Vol 29 (5) ◽  
pp. 251-256 ◽  
Author(s):  
R Kaufmann ◽  
T Schöneberg ◽  
C Lindschau ◽  
H Haller ◽  
T Ott
Keyword(s):  
C6 Cells ◽  

1995 ◽  
Vol 81 (2-3) ◽  
pp. 189-195 ◽  
Author(s):  
Klaus Koschel ◽  
Nicole N. Meissner ◽  
Piet W.L. Tas

Neuropeptides ◽  
1998 ◽  
Vol 32 (2) ◽  
pp. 185-189 ◽  
Author(s):  
R Kaufmann ◽  
H Schafberg ◽  
M Zieger ◽  
P Henklein ◽  
G Nowak

2003 ◽  
Vol 2 (3) ◽  
pp. 223-235 ◽  
Author(s):  
Tatyana M. Timiryasova ◽  
Daila S. Gridley ◽  
Bing Chen ◽  
Melba L. Andres ◽  
Radha Dutta-Roy ◽  
...  

The overall goal of this study was to analyze the effect and mechanism of radiation in combination with vaccinia viruses (VV) carrying the p53 gene against glioma. Comparison of two alternative treatments of cultured C6 (p53+) and 9L (p53−) rat glioma cells showed significantly reduced survival for both cell lines, especially 9L, when radiation was applied prior to virus versus radiation alone. High p53 protein expression mediated by VV-TK-p53 was measured in infected cells. Single modality treatment of C6 cells with psoralen and UV (PUV)-inactivated VV-TK-p53 (PUV-VV-TK-53) or radiation significantly decreased survival compared with PUV-inactivated L-15 (PUV-L-15) control virus. However, no difference was observed between radiation and combination treatments of C6 cells. In contrast, radiation followed by PUV-VV-TK-53 resulted in dramatic reduction of 9L cell viability, compared to single modality treatment. Flow cytometry analysis of Annexin-V-stained 9L cells showed that radiation and PUV-VV-TK-53 caused a significant decrease in live cells (17.2%) as compared to other treatments and control (61.6–98.3%). Apoptosis was observed in 37.2% of cells, while the range was 0.7–7.8% in other treatment groups; maximal p53 level was measured on day 7 post-infection. In athymic mice bearing C6 tumors, VV-TK-53 plus radiation in both single and multiple therapies resulted in significantly smaller tumors by day 30 compared to the agents given only once. Immunohistochemical analysis of tumor sections demonstrated p53 protein expression over 20 days after VV-TK-53 treatment. Analysis of blood and spleen cells of mice given multiple combination treatments showed significant splenomegaly, leukocytosis, and increased DNA synthesis and response to mitogen. Multiple combination treatments were also associated with significantly elevated natural killer and B cells in the spleen. There were no overt toxicities, although depression in red blood cell and thrombocyte parameters was noted. Collectively, the data demonstrate that radiation significantly improves the efficacy of VV-mediated tumor suppressor p53 therapy and may be a promising strategy for glioma treatment. Furthermore, the results support the conclusion that the mechanisms underlying the enhanced anti-tumor effect of combination treatment include apoptosis/necrosis and upregulation of innate immune defenses.


Sign in / Sign up

Export Citation Format

Share Document