scholarly journals ANTXR1 Facilitates Tumor Growth in Glioma via Deactivating MAPK Signaling Pathway

Author(s):  
Aijun Liang ◽  
Chaoyang Zhou ◽  
Jianzhong Zhang ◽  
Jingxing Leng ◽  
Bin Xi ◽  
...  

Abstract Background: Gliomas are commonly known as primary brain tumors and associated with frequent recurrence and an unsatisfactory prognosis in despite of extensive research in the underlying molecular mechanisms. In this study, we aimed to examine the role of ANTXR1 in glioma tumorigenesis and explore its downstream regulatory mechanism. Methods: We detected overexpression of ANTXR1 in glioma cell lines (SHG-44 and U251) in comparison with those in normal brain tissues.Result: Glioma cell growth and migratory ability were dramatically impaired as a result of silencing ANTXR1 by shANTXR1 lentivirus. ANTXR1 blockade also accelerated cell apoptosis since ANTXR1 held back apoptosis via targeting G2 phrase during cell mitosis. In vivo xenograft models verified in vitro findings above that the solid tumors stripped from mice were much lighter and smaller after depletion of ANTXR1 than controls. We also mechanically probed the downstream pathways and disclosed that overexpression of ANTXR1 abrogated the levels of MAKP9 and apoptisis-related protein HTRA2, but augmenting CCND1 and CDK6 levels in glioma cells. Our findings allow us to demonstrate that ANTXR1 acts as a tumor promoter in glioma induction through attenuating MAPK9-mediated gene transcription and HTRA2-induced apoptisis but intensifying CCND1-mediated proliferation. Conclusion: Together, we declare that ANTXR1 plays an indispensable role in glioma tumorigenesis via deactivating MAPK signaling and apoptosis pathway but activating PI3K/AKT-mediated cell growth. Our study provide a valuable clue to targeting ANTXR1 as a molecular target and a promising anticancer agent in glioma clinical therapeutics.

2021 ◽  
pp. 1-9
Author(s):  
Huan Guo ◽  
Baozhen Zeng ◽  
Liqiong Wang ◽  
Chunlei Ge ◽  
Xianglin Zuo ◽  
...  

BACKGROUND: The incidence of lung cancer in Yunnan area ranks firstly in the world and underlying molecular mechanisms of lung cancer in Yunnan region are still unclear. We screened a novel potential oncogene CYP2S1 used mRNA microassay and bioinformation database. The function of CYP2S1 in lung cancer has not been reported. OBJECTIVE: To investigate the functions of CYP2S1 in lung cancer. METHODS: Immunohistochemistry and Real-time PCR were used to verify the expression of CYP2S1. Colony formation and Transwell assays were used to determine cell proliferation, invasion and migration. Xenograft assays were used to detected cell growth in vivo. RESULTS: CYP2S1 is significantly up-regulated in lung cancer tissues and cells. Knockdown CYP2S1 in lung cancer cells resulted in decrease cell proliferation, invasion and migration in vitro. Animal experiments showed downregulation of CYP2S1 inhibited lung cancer cell growth in vivo. GSEA analysis suggested that CYP2S1 played functions by regulating E2F targets and G2M checkpoint pathway which involved in cell cycle. Kaplan-Meier analysis indicated that patients with high CYP2S1 had markedly shorter event overall survival (OS) time. CONCLUSIONS: Our data demonstrate that CYP2S1 exerts tumor suppressor function in lung cancer. The high expression of CYP2S1 is an unfavorable prognostic marker for patient survival.


2020 ◽  
Vol 52 (2) ◽  
pp. 168-179 ◽  
Author(s):  
Huilin Gong ◽  
Shan Gao ◽  
Chenghuan Yu ◽  
Meihe Li ◽  
Ping Liu ◽  
...  

Abstract Y-box binding protein 1 (YB-1) is manifested as its involvement in cell proliferation and differentiation and malignant cell transformation. Overexpression of YB-1 is associated with glioma progression and patient survival. The aim of this study is to investigate the influence of YB-1 knockdown on glioma cell progression and reveal the mechanisms of YB-1 knockdown on glioma cell growth, migration, and apoptosis. It was found that the knockdown of YB-1 decreased the mRNA and protein levels of YB-1 in U251 glioma cells. The knockdown of YB-1 significantly inhibited cell proliferation, colony formation, and migration in vitro and tumor growth in vivo. Proteome and phosphoproteome data revealed that YB-1 is involved in glioma progression through regulating the expression and phosphorylation of major proteins involved in cell cycle, adhesion, and apoptosis. The main regulated proteins included CCNB1, CCNDBP1, CDK2, CDK3, ADGRG1, CDH-2, MMP14, AIFM1, HO-1, and BAX. Furthermore, it was also found that YB-1 knockdown is associated with the hypo-phosphorylation of ErbB, mTOR, HIF-1, cGMP-PKG, and insulin signaling pathways, and proteoglycans in cancer. Our findings indicated that YB-1 plays a key role in glioma progression in multiple ways, including regulating the expression and phosphorylation of major proteins associated with cell cycle, adhesion, and apoptosis.


2021 ◽  
Author(s):  
Songjia Guo ◽  
Shuhua Shan ◽  
Haili Wu ◽  
huiqiang hao ◽  
Zhuoyu Li

Abstract Nostoc commune Vauch is a nitrogen-fixing blue-green algae, contains a large number of active molecules with medicinal functions. Our previous study found that a water stress protein (WSP1) from Nostoc commune Vauch and its the recombinant protein (Re-WSP1) exhibited significant anti-colon cancer (CRC) activity both in vitro and in vivo. However, the underlying mechanism remains unknown. In this study, the CCK8 and clonogenic assays showed that Re-WSP1 restrained the colon cancer growth in a dose-dependent manner. Mechanistically, Re-WSP1 inhibited the expression of β-catenin, which was partly reversed by LiCl treatment, demonstrating a key role in Re-WSP1-induced inhibition of cell growth. Quantitative PCR analysis showed that the expression of microRNA-539 (miR-539) was significantly up-regulated upon Re-WSP1 treatment. Moreover, miR-539 negatively regulateed the expression of β-catenin through directly binds to the 3’UTR of β-catenin mRNA. Taken together, our data demonstrate that Re-WSP1 suppresses the CRC growth via miR-539/β-catenin axis, which provides new insights into the molecular mechanisms underlying Re-WSP1 against CRC.


2020 ◽  
Author(s):  
Yang Chen ◽  
Huiyan Li ◽  
Chunxun Liu ◽  
Yongmei Han ◽  
Yubao Zhang ◽  
...  

Abstract BACKGROUND: Long non-coding RNAs (lncRNA) have been shown to play important roles in the development and progression of hepatocellular carcinoma (HCC). In this report, we examined the role of lncRNA LINC00645 in HCC. MATERIAL AND METHODS: Based on public databases and integrating bioinformatics analyses, the over-expression of LINC00645 in HCC tissues was detected and further validated in a cohort of liver tissues. A series of in vitro and in vivo functional experiments were executed to investigate the role of LINC00645 in the carcinogenesis and development of HCC. Comprehensive transcriptional analysis, chromatin immunoprecipitation (ChIP) assay, dual-luciferase reporter assay and western blot etc. were performed to explore the molecular mechanisms underlying the functions of LINC00645. RESULTS: LINC00645 was significantly upregulated in HCC cell lines and HCC tissues, which was correlated with poor prognosis in HCC patients. LINC00645 knockdown remarkably suppressed tumor growth in vitro and in vivo. Mechanistically, LINC00645 could competitively bind with miR-141-3p to prevent the degradation of its target gene GP73, which acts as a tumor-promoter in HCC. Furthermore, the ChIP assay showed that the transcription factor MAZ could bind to the LINC00645 promoter and increase its transcription. CONCLUSIONS: Collectively, this study demonstrated that LINC00645 plays a critical regulatory role in hepatocellular carcinoma cells and LINC00645 may serve as a potential diagnostic biomarker and therapeutic target of HCC. Thus, targeting MAZ/LINC00645/miR-141-3p/GP73 signaling axis may prevent the progression of HCC.


2019 ◽  
Author(s):  
Yue-Li Sun ◽  
Wen-Qi Jiang ◽  
Qiu-Yun Luo ◽  
Da-Jun Yang ◽  
Yu-Chen Cai ◽  
...  

Abstract Background: Bcl-2 family members play an important role in the development of malignant lymphoma and can induce drug resistance in anticancer treatment. The development of small molecules targeting Bcl-2 family protein can be new strategy for malignant lymphoma treatment. In this study, we investigate the antitumor effect and the cellular mechanism of a novel Bcl-2/Bcl-xL dual inhibitor BM-1197 in DCBCL and Burkitt lymphoma cells. Methods: CCK-8 assay was used to detect cell viability. Apoptosis was determined by Hoechst 33258 staining and flow cytometry. The activity of caspase-3/caspase-9 was determined using the caspase-3/ caspase-9 activity kit. Western blotting analysis was performed to evaluate the change of protein expression. The functional analysis was evalueated via immunoprecipitation and siRNA interference. Human malignant lymphoma xenograft models in nude mice were established for in vivo efficacy detection. Results: We find that BM-1197 exerts potent growth-inhibitory activity against lymphoma cells which harbor Bcl-2 and Bcl-xL high expression in vitro and has synergistic effect with chemotherapeutic drugs. Mechanistically, we see that the intrinsic apoptosis pathway is activated upon BM-1197 treatment. BM-1197 affects the protein interaction of Bak/Bcl-xl, Bim/Bcl-2, Bim/Bcl-xl, PUMA/Bcl-2 and induced conformational change in the Bax protein.which results in activation of Bax and release cytochrome c, and activated caspase -9, -3, -7 and finally induce cell apoptosis. Furthermore, our data demonstrates that BM-1197 exhibits strong anti-tumor effects against established human malignant lymphoma xenograft models. Conclusions: Our study demonstrated BM-1197 exerts potent antitumor effects both in vitro and in vivo, and provides promising preclinical data for further development of BM-1197 in malignant lymphoma.


Oncogene ◽  
2020 ◽  
Vol 39 (43) ◽  
pp. 6647-6663
Author(s):  
Jinglin Zhang ◽  
Chi Chun Wong ◽  
Kam Tong Leung ◽  
Feng Wu ◽  
Yuhang Zhou ◽  
...  

Abstract Fibroblast growth factor receptor type 2 (FGFR2) has emerged as a key oncogenic factor that regulates gastric cancer (GC) progression, but the underlying mechanism of FGF–FGFR2 signaling pathway remains largely unknown. To identify the potential molecular mechanisms of the oncogenic FGFR2 in gastric carcinogenesis and convey a novel therapeutic strategy, we profiled the FGFR alterations and analyzed their clinical associations in TCGA and Hong Kong GC cohorts. We found that FGFR2 overexpression in GC cell lines and primary tumors predicted poor survival and was associated with advanced stages of GC. Functionally, growth abilities and cell cycle progression of GC were inhibited by inactivation of ERK–MAPK signal transduction after FGFR2 knockdown, while apoptosis was promoted. Meanwhile, the first-line anti-cancer drug sensitivity was enhanced. RNA-seq analysis further revealed that YAP1 signaling serves as a significant downstream modulator and mediates the oncogenic signaling of FGFR2. When stimulating FGFR2 by rhFGF18, we observed intensified F-actin, nuclear accumulation of YAP1, and overexpression of YAP1 targets, but these effects were attenuated by either FGFR2 depletion or AZD4547 administration. Additionally, the FGF18–FGFR2 signaling upregulated YAP1 expression through activating c-Jun, an effector of MAPK signaling. In our cohort, 28.94% of GC cases were characterized as FGFR2, c-Jun, and YAP1 co-positive and demonstrated worse clinical outcomes. Remarkably, we also found that co-targeting FGFR2 and YAP1 by AZD4547 and Verteporfin synergistically enhanced the antitumor effects in vitro and in vivo. In conclusion, we have identified the oncogenic FGF–FGFR2 regulates YAP1 signaling in GC. The findings also highlight the translational potential of FGFR2–c-Jun–YAP1 axis, which may serve as a prognostic biomarker and therapeutic target for GC.


2006 ◽  
Vol 5 (3) ◽  
pp. 271-280 ◽  
Author(s):  
Peiyu Pu ◽  
Chunsheng Kang ◽  
Zhiyong Zhang ◽  
Xiaozhi Liu ◽  
Hao Jiang

Life Sciences ◽  
2007 ◽  
Vol 81 (16) ◽  
pp. 1256-1263 ◽  
Author(s):  
Tao Zhang ◽  
Ming Guan ◽  
Chong Xu ◽  
Yuming Chen ◽  
Yuan Lu

2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii468-iii468
Author(s):  
Kathryn Taylor ◽  
Helena Zhang ◽  
Alexa Hui ◽  
Shawn Gillespie ◽  
Michelle Monje

Abstract Pediatric high-grade gliomas (pHGG) are a devastating group of diseases that urgently require novel therapeutic options. We have previously demonstrated that pHGGs hijack mechanisms of brain development and plasticity to their advantage. Here, we investigated the role of microenvironmental BDNF on pediatric gliomas, independent of the NTRK fusion events commonly identified in infant HGG. Genetic deletion or pharmacological blockade of NTRK2 (TrkB), in patient-derived pediatric glioma increases survival in multiple DIPG and pGBM patient-derived orthotopic xenograft models. Unlike the paracrine BDNF-TrkB signaling observed between subpopulations of adult HGG malignant cells, pediatric glioma express TrkB, but not BDNF ligand. BDNF is secreted by normal brain cells in response to neuronal activity and conditioned medium experiments from cortical slices of mice indicates the brain microenvironment as the chief source of BDNF ligand. Addition of recombinant BDNF protein increases pediatric glioma cell proliferation and activates the canonical downstream MAPK signaling pathway, an effect that is blocked by genetic or pharmacological TrkB inhibition in pHGG. However, the glioma growth-promoting effects of BDNF in vivo cannot be explained by stimulation of MAPK signaling alone. We therefore examined the effects of BDNF signaling on neuron-to-glioma synapse formation, a newly recognized microenvironmental interaction important for pediatric glioma progression. We find that BDNF-TrkB signaling promotes neuron-to-glioma synaptogenesis in neuron-glioma co-culture. We are presently exploring the role for BDNF-TrkB signaling in glioma synaptic plasticity and function. Funding: Abbie’s Army Foundation


Sign in / Sign up

Export Citation Format

Share Document