Modeled microgravity suppressed invasion and migration of human glioblastoma U87 cells through downregulating store-operated calcium entry

2015 ◽  
Vol 457 (3) ◽  
pp. 378-384 ◽  
Author(s):  
Zi-xuan Shi ◽  
Wei Rao ◽  
Huan Wang ◽  
Nan-ding Wang ◽  
Jing-Wen Si ◽  
...  
Author(s):  
Manar Zraikat ◽  
Munir Gharaibeh ◽  
Tasneem Alshelleh

Background: This work studies the effect of different concentrations of soaked ginger on the ability of the U87 glioma cells to invade collagen in a three dimension (3 D) invasion model and compare it with its effect on the ability of the same cell line to migrate in two-dimension (2 D) scratch assay. Methods: The hanging drop spheroids in 3D invasion assay were used to investigate the in invasion of the U87 cells. The 2D scratch assay was used to investigate the migration of the same cell line. Results: Gradual effect of the soaked ginger was noticed on the inhibition of the invasion of U87 in collagen and on the inhibition of the migration of the same cell line in scratch assay. Conclusion: The results in this article are promising and encourage further studies to investigate the effect of ginger active ingredients on tumour progression.


Blood ◽  
2010 ◽  
Vol 115 (3) ◽  
pp. 657-666 ◽  
Author(s):  
Ulrich Y. Schaff ◽  
Neha Dixit ◽  
Emily Procyk ◽  
Itsukyo Yamayoshi ◽  
Tiffany Tse ◽  
...  

Abstract Orai1 was reported to function as a calcium channel subunit that facilitates store operated calcium entry (SOCE) in T cells and is necessary for formation of the immune synapse. We reasoned that SOCE via Orai1 might regulate PMNs activation during recruitment to inflamed endothelium. Orai1 function was assessed by real-time imaging of calcium transients as PMNs were stimulated to roll, arrest, and migrate on E-selectin and ICAM-1 in shear flow. Calcium entry was significantly reduced when Orai1 function was impaired by heterozygous knockout in a mouse model or by siRNA knockdown in HL-60 cells. Reduced Orai-1 expression correlated with the delayed onset of arrest and reduced ability to transition to a polarized migratory phenotype. Inhibition of SOCE by treatment with 2-APB, or blocking phospholipase C (PLC) mediated calcium store release with U73122, abrogated formyl peptide induced calcium elevation, and delayed subsequent cell arrest and polarization. These results suggest that calcium entry via Orai1 is the predominant SOCE that cooperates with cytoplasmic calcium store release in coordinating integrin-dependent PMN arrest and migration in the acute response to inflammation.


2021 ◽  
Author(s):  
Yang Xu ◽  
Jiaguo Chen ◽  
Gao He ◽  
Yuhai Zhang

Abstract Background CTR9 (Cln three requiring 9) has been reported to be implicated in protein modification and oncogenesis of several human cancers. However, the protein expression and mechanism of CTR9 in glioma progression remain unclear. Methods We analyzed mRNA expression of CTR9 and CTR9-related survival curves in the public database. Then we detected CTR9 expression in glioma tissues and constructed U251 and U87 cells with stable silencing or overexpression of CTR9. Cell function tests and Western blot were conducted to explore the effects of CTR9 on glioma proliferation, invasion and migration, as well as the specific mechanism. All the date was presented as means ± SEM. Two-sample t-test and one-way analysis of variance (ANOVA) were used to identify whether there was a significant difference between each group of data. Results We found that CTR9 was overexpressed in glioma and inversely associated with glioma patient survival. The results manifested that knockdown of CTR9 suppressed the proliferation, migration and invasion of glioma cells, while overexpression facilitated them. The underlying molecular mechanism may involve the regulation of JAK2/STAT3 pathway by CTR9. Conclusion Our present study indicates that CTR9 is highly expressed in glioma and related to glioma grading and prognosis. CTR9 regulates malignant behaviors of glioma cells by activating JAK2/STAT3 pathway. Therefore, CTR9 is a promising biomarker for the diagnosis,targeted therapy and prognosis evaluation of glioma.


2012 ◽  
Vol 34 (3) ◽  
pp. 365-371 ◽  
Author(s):  
S. H. Shen ◽  
A. L. Kwan ◽  
Y. Y. Chen ◽  
Z. X. Wang

2019 ◽  
Vol 39 (1) ◽  
Author(s):  
Wei-Li Sun ◽  
Tian Kang ◽  
Yuan-Yu Wang ◽  
Jian-Ping Sun ◽  
Chen Li ◽  
...  

Abstract The present study was undertaken to investigate the underlying mechanisms of long noncoding RNA OIP5-AS1 via regulating miR-410 to modulate Wnt-7b in the progression of glioma. To address this problem, we measured the expression of OIP5-AS1 and miR-410 in glioma tissues by qRT-PCR. Glioma U87 cells were transfected with OIP5-AS1 siRNA or miR-410 inhibitors. The targeting relationships among miR-410, OIP5-AS1 and Wnt-7b were verified by luciferase reporter assays. Western blotting was employed to determine the expression of Wnt-7b/β-catenin pathway-related proteins, while MTT, flow cytometry, Transwell assays and wound-healing assays were used to measure the biological characteristics of glioma cells. The results showed that OIP5-AS1 expression was higher and miR-410 was lower in glioma tissues. Luciferase reporter assays confirmed a targeting relationship between OIP5-AS1 and miR-410, as well as between miR-410 and Wnt-7b. Silencing OIP5-AS1 reduced cell proliferation, invasion and migration of glioma U87 cells and led to depressed expression levels of miR-410, Wnt-7b, p-β-catenin, GSK-3β-pS9, c-Myc and cyclin D1. Furthermore, down-regulation of OIP5-AS1 induced G0/G1 phase cell cycle arrest and apoptosis of glioma cells. Inhibitors of miR-410 abolished the biological effects of OIP5-AS1 siRNA in glioma cells. In vivo, OIP5-AS1 knockdown also inhibited tumor growth. Taken together, this research suggested that silencing OIP5-AS1 may specifically block the Wnt-7b/β-catenin pathway via targeted up-regulating miR-410, thereby inhibiting growth, invasion and migration while promoting apoptosis in glioma cells.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Li Wei ◽  
Ya Peng ◽  
Naiyuan Shao ◽  
Peng Zhou

Abstract Background Glioblastoma remains one of the most lethal brain cancers. T-cell immunoglobulin and mucin domain 1 (Tim-1) is associated with various immune diseases. The molecular mechanism of Tim-1 in regulating glioblastoma cell proliferation, invasion, and migration is still unknown. Moreover, it has shown that miR-133a plays an important role in glioblastoma. However, little is known about the interaction between Tim-1 and miR-133a in glioblastoma. Methods Tim-1 expression in glioblastoma and normal brain tissues was detected by qPCR, Western Blot and IHC. After Tim-1 knockdown in U251 and U87 cells, genes showing significantly differential expression, along with the significant differential miRNAs were analyzed using RNA-seq analysis. The binding sites were verified using dual-luciferase reporter gene assay. U251 and U87 cells were allocated into the small harpin-negative control (sh-NC), sh-Tim-1, sh-Tim-1 + inhibitor NC, and sh-Tim-1 + miR-133a inhibitor group. Cell proliferation, migration, and invasion were determined by CCK-8, flow cytometry, wound-healing and Transwell assays, respectively. Next, U251 and U87 cells were allocated into the mimic NC, miR-133a mimic, miR-133a mimic + pcDNA3.1, and miR-133a mimic + pcDNA3.1-TGFBR1 groups, followed by the detection of cell proliferation, migration, and invasion. Western blot was used to identify the expression of vital kinases in the Wnt/β-catenin pathway. Results Tim-1 was highly expressed in glioblastoma tissues compared with that in normal brain tissues. RNA-seq analysis showed that Tim-1 knockdown could lead to the downregulation of TGFBR1 and the upregulation of miR-133a. The binding sites between TGFBR1 and miR-133a were confirmed. Tim-1 knockdown impaired the invasion, migration, proliferation of U251 and U87 cells, which could be reversed by miR-133a downregulation. miR-133a upregulation inhibited the proliferation, invasion, and migration of U251 and U87 cells, which could be reversed by TGFBR1 upregulation. Tim-1 knockdown and miR-133a upregulation could inhibit the activation of the Wnt/β-catenin pathway, while the elevation of TGFBR1 showed opposite effects. Conclusion Tim-1 knockdown inhibited glioblastoma cell proliferation, invasion, and migration through the miR-133a/TGFBR1 axis and restrained the activation of the Wnt/β-catenin pathway.


MedChemComm ◽  
2015 ◽  
Vol 6 (1) ◽  
pp. 164-173 ◽  
Author(s):  
Shengnan Fu ◽  
Yanqi Xie ◽  
Jue Tuo ◽  
Yalong Wang ◽  
Wenbo Zhu ◽  
...  

This research aims to synthesize lipophilic berberine derivatives and evaluate their antiglioma effects on C6 and U87 cells.


2015 ◽  
Vol 35 (2) ◽  
pp. 419-432 ◽  
Author(s):  
Chi Zhang ◽  
Xian-rui Yuan ◽  
Hao-yu Li ◽  
Zi-jin Zhao ◽  
Yi-wei Liao ◽  
...  

Background: Metabotropic glutamate receptors (mGluRs) are G-protein-coupled receptors that mediate neuronal excitability and synaptic plasticity in the central nervous system, and emerging evidence suggests a role of mGluRs in the biology of cancer. Previous studies showed that mGluR1 was a potential therapeutic target for the treatment of breast cancer and melanoma, but its role in human glioma has not been determined. Methods: In the present study, we investigated the effects of mGluR1 inhibition in human glioma U87 cells using specific targeted small interfering RNA (siRNA) or selective antagonists Riluzole and BAY36-7620. The anti-cancer effects of mGluR1 inhibition were measured by cell viability, lactate dehydrogenase (LDH) release, TUNEL staining, cell cycle assay, cell invasion and migration assays in vitro, and also examined in a U87 xenograft model in vivo. Results: Inhibition of mGluR1 significantly decreased the cell viability but increased the LDH release in a dose-dependent fashion in U87 cells. These effects were accompanied with the induction of caspase-dependent apoptosis and G0/G1 cell cycle arrest. In addition, the results of Matrigel invasion and cell tracking assays showed that inhibition of mGluR1 apparently attenuated cell invasion and migration in U87 cells. All these anti-cancer effects were ablated by the mGluR1 agonist L-quisqualic acid. The results of western blot analysis showed that mGluR1 inhibition overtly decreased the phosphorylation of PI3K, Akt, mTOR and P70S6K, indicating the mitigated activation of PI3K/Akt/mTOR pathway. Moreover, the anti-tumor activity of mGluR1 inhibition in vivo was also demonstrated in a U87 xenograft glioma model in athymic nude mice. Conclusion: The remarkable efficiency of mGluR1 inhibition to induce cell death in U87 cells may find therapeutic application for the treatment of glioma patients.


Tumor Biology ◽  
2015 ◽  
Vol 37 (3) ◽  
pp. 3831-3839 ◽  
Author(s):  
Sonia Aroui ◽  
Feten Najlaoui ◽  
Yassine Chtourou ◽  
Annie-Claire Meunier ◽  
Amel Laajimi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document