Blockage of CGEN-928 Expression Down-Regulates AKT Signaling Pathway and Inhibits Multiple Myeloma (MM) Cell Proliferation

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5102-5102
Author(s):  
Haiming Chen ◽  
Mingjie Li ◽  
Jennifer Li ◽  
Marissa P Dreyer ◽  
Cameryn P Ahles ◽  
...  

Abstract Abstract 5102 We have recently reported that CGEN-928 is highly expressed on the cell membrane of cell lines, human xenografts, and primary tumor cells from MM. Anti-CGEN-928 (anti-TM21) polyclonal antibody blocked the expression of CGEN-928 which decreased MM tumor cell proliferation and increased apoptosis in the MM cell lines MM1s, RPMI8226 and U266 as well as primary MM tumor cells. The mechanism through which blocking CGEN-928 decreases MM tumor cell proliferation and enhances apoptosis has not been elucidated clear. In this study, a CGEN-928 shRNA (lentiviral particles) was used to silence this gene's expression, and determine its impact on the AKT signal transduction pathway which has been shown to play an important role in MM tumor cell metabolism proliferation, and survival. Briefly, MM1s or primary MM tumor cells were cultured in a 12-well plate for 24 hours prior to the viral infection. On the following day, a mixture of 5ug/ml Polybrene and fresh medium were added to the cells. The CGEN-928 shRNA lentiviral particles were then added to the culture. While transducing cells, we treated a portion of the cells with a negative control through introduction of control shRNA lentiviral particles. To ensure we achieved a successful transduction, we also treated another portion of the cells with cop GFP control Lentiviral particles. We confirmed that 75% of MM cells were transduced based on GFP+ cell counts after 24 hours treatment. The day following the transduction, the cultured medium was removed and replaced with fresh medium without polybrene. Two days following transduction, we used fresh 10ug/ml puromycin-containing medium to select stable MM cells. We replaced the medium with fresh puromycin-containing medium every three days until resistant MM tumor cells were stable. Proliferation rate of the MM1s tumor cells transduced with CGEN-928 shRNA (85%) 24 hours was much lower than the tumor cells transduced with control lentiviral particles rate (170%). The proportion of MM cells undergoing apoptosis treated with CGEN-928 shRNA (42%) was higher than MM cells transduced with control lentiviral particles (13%). We next examined several protein phosphorylation sites related to AKT signaling pathway by Western blot. The results showed AKT1 phosphorylation in MM tumor cells transduced with CGEN-928 shRNA or anti- CGEN-928 polyclonal antibody was decreased and phosphorylation of c-Raf, GSK-3β, factors downstream of AKT were also down-regulated. PTEN phosphorylation slightly decreased in MM cell treated with anti-CGEN-928 antibody but did not change in MM cells silenced with CGEN-928 shRNA. We further examined downstream gene expression of the AKT pathway when CGEN-928 was silenced using siRNA or the anti-CGEN-928 TM-21 antibody. We found AKT1 gene expression was reduced in the presence of CGEN-928 siRNA or antibody but it did not impact ATK2 and AKT3. mTOR gene expression in MM tumor cells was decreased with exposure to CGEN-928 siRNA but anti-TM21 showed no effect. Cyclin D1 gene expression in MM tumor cell was not affected by CGEN-928 siRNA and antibody. These studies suggest that blockage of CGEN-928 antigen expression inhibits MM tumor cell proliferation and enhance tumor cell apoptosis through AKT signaling pathway. Currently, a monoclonal anti-CGEN-928 antibody is in development that will be used by our group to evaluate its anti-MM effects both in vitro and in vivo using our SCID-hu models of human MM. Disclosures: Berenson: Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Millennium Pharmaceuticals, Inc.: Consultancy, Honoraria, Research Funding, Speakers Bureau; Onyx Pharmaceuticals: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Medtronic: Consultancy, Honoraria, Research Funding, Speakers Bureau; Merck: Research Funding; Genentech: Research Funding.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1423-1423
Author(s):  
You Hua Yu ◽  
Na Guo ◽  
Yujing Gong ◽  
Baidong Liu ◽  
Hong Liu ◽  
...  

Abstract Abstract 1423 Patients with B cell malignaces initially respond to current treatment modalities, however, such malignances remain incurable. Many new therapeutic options have become available during the past several years but nearly all patients develop resistance to currently available therapeutic options. Ideally, a new treatment should inhibit tumor growth, improve the efficacy of other anti-tumor agents, and improve both the overal survial and the quality of life for patients. Pterostilbene is predominantly found in Rhubarb. We synthesized bipterostilbene (5-(4-(4-(3,5-dihydroxylstyryl)phenoxy)styryl)-benzene-1,3-diol) (C28H22O5) of a molecular weight of 438.48 Kda. In this study, we first examined whether bipterostilbene affects tumor cells proliferation using breast cancer, ovarian cancer, lymphoma and multiple myeloma (MM) cell lines. The results of the MTS assay demonstrated that bipterostilbene significantly inhibited tumor cell proliferation of the lymphoma cell line (Raji) and the MM cell lines (RPMI1640 and MM1s) at 48 hours (IC50: 5μM for Raji, 4μM for RPMI8226, and 2 μM for MM1s). The induction of tumor cell apoptosis was most prominent at 72 hours. The extent of the inhibition of tumor cell proliferation and the induction of apoptosis was concentration-dependent. Bipterostilbene had minimal effects on breast and ovarian cancer cell lines. Noteworthy, bipterostilbene had no detectable cytotoxic effects on normal human peripheral blood mononuclear cells (PBMCs). The molecular mechanism by which bipterostilbene mediates its effects was examined. Both the AKT and the NF-κB signaling transduction pathways have been reported to play key roles in B cell metabolism, proliferation and survival. Using RT-PCR, bipterostilbene specifically inhibited AKT1 and mTOR gene expression when Raji or RPMI8226 tumor cells were treated with the IC50 concentration of bipterostilbene for 24 hours. Analysis of downstream gene products of the AKT pathway revealed that Cyclin D1 expression was slightly reduced and P21Cip and P27 kip expressions were not changed. Bipterostilbene did not alter AKT2 or AKT3 gene expression, demonstrating that this compound is specifically targeting AKT1. We further determined whether bipterostilbene interfered with IGF1-induced AKT/mTOR activation or IL-1β –mediated NF-κB phosphorylation by Western blot. The results showed that bipterostilbene markedly inhibited IGF1-induced phosphorylation of AKT but did not interfere with IL-1β-induced NF-κB activity and IκB phosphorylation. Overall, the results of our in vitro studies demonstrate that bipterostilbene inhibits tumor cell proliferation and enhances apoptosis of B-cell malignancies via inhibition of the AKT/mTOR signaling pathway with no detectable effect on the NF-κB signaling pathway. Importantly, bipterostilbene is not cytotoxic on normal hematopoietic cells at concentrations that were highly toxic to B-cell malignancies. We propose that bipterostilbene may be better tolerated than other anti- cancer drugs that are currently being used for the treatment of B-cell malignancies. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 48 (3) ◽  
pp. 1382-1396 ◽  
Author(s):  
Yu-Xiang Liao ◽  
Zhi-Ping Zhang ◽  
Jie Zhao ◽  
Jing-Ping Liu

Background/Aims: The current study aimed to investigate the role by which fibronectin 1 (FN1) influences the cell cycle, senescence and apoptosis in human glioma cells through the PI3K/ AKT signaling pathway. Methods: Differentially expressed genes (DEGs) were identified based on gene expression data (GSE12657, GSE15824 and GSE45921 datasets) and probe annotation files from Gene Expression Omnibus. The DEGs were identified in connection with gene ontology (GO) enrichment analysis and with the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The positive expression of the FN1 protein was detected by immunohistochemistry. The glioma cell lines U251 and T98G were selected and assigned into blank, negative control (NC) and siRNA-FN1 groups. A dual luciferase reporter gene assay was used to investigate the effects of FN1 on transcriptional activity through the PI3K/AKT signaling pathway. An MTT assay was applied for the detection of cell proliferation, while flow cytometry was employed for cell cycle stage and cellular apoptosis detection. β-galactosidase staining was utilized to detect cellular senescence, a scratch test was applied to evaluate cell migration, and a transwell assay was used to analyze cell invasion. Western blotting and qRT-PCR methods were used to detect the protein and mRNA expression levels, respectively, of the FN1 gene and the related genes in the PI3K/AKT pathway (PI3K, AKT and PTEN), the cell cycle (pRb, CDK4 and Cyclin D1) and cell senescence (p16 and p21) among the collected tissues and cells. Results: GSE12657 profiling revealed FN1 to be the most upregulated gene in glioma. Regarding the GSE12657 and GSE15824 datasets, FN1 gene expression was higher in glioma tissues than in normal tissues. GO enrichment analysis and KEGG pathway enrichment analysis indicated that FN1 is involved in the synthesis of extracellular matrix (ECM) components and the PI3K/AKT signaling pathway. Verification was provided, indicating the role played by the FN1 gene in the regulation of the PI3K/AKT signaling pathway, as silencing the FN1 gene was found to inhibit cell proliferation, promote cell apoptosis and senescence, and reduce migration and invasion through the down-regulation of FN1 gene expression and disruption of the PI3K-AKT signaling pathway. Conclusion: The findings of this study provide evidence highlighting the prominent role played by FN1 in stimulating glioma growth, invasion, and survival through the activation of the PI3K/AKT signaling pathway.


Tumor Biology ◽  
2014 ◽  
Vol 35 (5) ◽  
pp. 4095-4099 ◽  
Author(s):  
Lu Zheng ◽  
Wei Gong ◽  
Ping Liang ◽  
XiaoBing Huang ◽  
Nan You ◽  
...  

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Dongyong Yang ◽  
Yanqing Wang ◽  
Yajing Zheng ◽  
Fangfang Dai ◽  
Shiyi Liu ◽  
...  

Abstract Background Polycystic ovary syndrome (PCOS) is the most common hormonal disorder among reproductive-aged women worldwide, however, the mechanisms and progression of PCOS still unclear due to its heterogeneous nature. Using the human granulosa-like tumor cell line (KGN) and PCOS mice model, we explored the function of lncRNA UCA1 in the pathological progression of PCOS. Results CCK8 assay and Flow cytometry were used to do the cell cycle, apoptosis and proliferation analysis, the results showed that UCA1 knockdown in KGN cells inhibited cell proliferation by blocking cell cycle progression and promoted cell apoptosis. In the in vivo experiment, the ovary of PCOS mice was injected with lentivirus carrying sh-UCA1, the results showed that knockdown of lncRNA UCA1 attenuated the ovary structural damage, increased the number of granular cells, inhibited serum insulin and testosterone release, and reduced the pro-inflammatory cytokine production. Western blot also revealed that UCA1 knockdown in PCOS mice repressed AKT activation, inhibitor experiment demonstrated that suppression of AKT signaling pathway, inhibited the cell proliferation and promoted apoptosis. Conclusions Our study revealed that, in vitro, UCA1 knockdown influenced the apoptosis and proliferation of KGN cells, in vivo, silencing of UCA1 regulated the ovary structural damage, serum insulin release, pro-inflammatory production, and AKT signaling pathway activation, suggesting lncRNA UCA1 plays an important role in the pathological progression of PCOS.


2021 ◽  
Vol 11 (9) ◽  
pp. 1785-1791
Author(s):  
Tangpeng Xu ◽  
Changli Ruan ◽  
Xu Bin ◽  
Mengxue Hu

Hepatocellular carcinoma (HCC) is a serious threat to human health. miR-340 participates in HCC pathogenesis, but its specific mechanism is not completely clear. Therefore, our study assessed the mechanism by how miR-340 involves in HCC. The cancer tissues and paracancerous tissues of HCC patients were collected. miR-340 mimics/NC and Akt siRNA were transfected into HepG2 cells followed by analysis of miR-304 and EMT-related molecules expression by Real-time PCR, cell invasion and migration by Transwell assay, cell proliferation ability by CCK8 assay as well as p-Akt and p-mTOR level by Western blot. miR-340 in HCC tissues was significantly downregulated compared to adjacent tissues (P <0.001). With increased pathological grade, miR-340 expression was decreased gradually. p-Akt and p-mTOR in HCC tissues was significantly upregulated and elevated gradually with increased pathological grade. p-Akt and p-mTOR was negatively associated with miR-340 (P <0.001). After overexpression of miR-340, HepG2 cell proliferation, invasion, migration and epithelialization were significantly inhibited, and p-Akt and p-mTOR was reduced. When Akt expression was interfered with siRNA, cell proliferation and epithelialization was further inhibited. miR-340 inhibits the development of hepatocellular carcinoma through Akt signaling pathway.


2018 ◽  
Vol 17 (7) ◽  
pp. 1243
Author(s):  
Fen Wen ◽  
Yuxian Lu ◽  
Ke Xu ◽  
Yanmei Liu ◽  
Xiaojuan Qian ◽  
...  

2017 ◽  
Vol 4 ◽  
pp. 165-171 ◽  
Author(s):  
Suthat Chottanapund ◽  
M.B.M. Van Duursen ◽  
Anne Zwartsen ◽  
Supatchaya Timtavorn ◽  
Panida Navasumrit ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document