BF-30 selectively inhibits melanoma cell proliferation via cytoplasmic membrane permeabilization and DNA-binding in vitro and in B16F10-bearing mice

2013 ◽  
Vol 707 (1-3) ◽  
pp. 1-10 ◽  
Author(s):  
Hui Wang ◽  
Mengyun Ke ◽  
Yuwei Tian ◽  
Jing Wang ◽  
Bing Li ◽  
...  
2011 ◽  
Vol 30 (5) ◽  
pp. 1849-1864 ◽  
Author(s):  
Kristopher R. Koch ◽  
Chen-Ou Zhang ◽  
Piotr Kaczmarek ◽  
Joseph Barchi ◽  
Li Guo ◽  
...  

2010 ◽  
Vol 19 (11) ◽  
pp. 958-964 ◽  
Author(s):  
Zhiqi Song ◽  
Chun-Di He ◽  
Changkai Sun ◽  
Yanni Xu ◽  
Xin Jin ◽  
...  

BMC Cancer ◽  
2011 ◽  
Vol 11 (1) ◽  
Author(s):  
Mélissa Labelle-Côté ◽  
Julie Dusseault ◽  
Salma Ismaïl ◽  
Aude Picard-Cloutier ◽  
Peter M Siegel ◽  
...  

Author(s):  
Loreto B. Feril ◽  
Kazuki Yamaguchi ◽  
Yurika Ikeda-Dantsuji ◽  
Yukihiro Furusawa ◽  
Yoshiaki Tabuchi ◽  
...  

Cancers ◽  
2022 ◽  
Vol 14 (2) ◽  
pp. 311
Author(s):  
Mourad Zerfaoui ◽  
Eman Toraih ◽  
Emmanuelle Ruiz ◽  
Youssef Errami ◽  
Abdallah S. Attia ◽  
...  

Background: Previously, we have demonstrated that nuclear BRAFV600E is associated with melanoma aggressiveness and vemurafenib resistance. However, the underlying mechanisms of how nuclear localization of BRAFV600E promotes cell aggressiveness have not yet been investigated. Despite therapeutic advancements targeting cutaneous melanoma, unknown cellular processes prevent effective treatment for this malignancy, prompting an urgent need to identify new biological targets. This study aims to explore the association of inducible heme oxygenase 1 (HMOX-1) with nuclear BRAFV600E in promoting melanoma aggressiveness. Methods: Proteomics analysis was performed to identify the interacting partner(s) of nuclear BRAFV600E. Immunohistochemistry was applied to evaluate the levels of HMOX-1 and nuclear BRAFV600E expression in melanoma and adjacent healthy tissues. Immunofluorescence assessed the nuclear localization of BRAFV600E in vemurafenib-resistant A375R melanoma cells. Further study of HMOX-1 knockdown or BRAFV600E overexpression in melanoma cells suggested a role for HMOX-1 in the regulation of cell proliferation in vivo and in vitro. Finally, Western blot analysis was performed to confirm the pathway by which HMOX-1 mediates Akt signaling. Results: Proteomics results showed that HMOX-1 protein expression was 10-fold higher in resistant A375R cells compared to parental counterpart cells. In vitro and in vivo results illustrate that nuclear BRAFV600E promotes HMOX-1 overexpression, whereas HMOX-1 reduction represses melanoma cell proliferation and tumor growth. Mechanistic studies revealed that HMOX-1 was associated with nuclear BRAFV600E localization, thus promoting melanoma proliferation via a persistent activation of the AKT pathway. Conclusions: Our results highlight a previously unknown mechanism in which the nuclear BRAFV600E/HMOX-1/AKT axis plays an essential role in melanoma cell proliferation. Targeting HMOX-1 could be a novel method for treating melanoma patients who develop BRAF inhibitor resistance.


Oncogenesis ◽  
2018 ◽  
Vol 7 (11) ◽  
Author(s):  
Seung-Shick Shin ◽  
Byeong-Seon Jeong ◽  
Brian A. Wall ◽  
Jiadong Li ◽  
Naing Lin Shan ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yingnan Liu ◽  
Yongqing Zhuang ◽  
Xiaokuan Fu ◽  
Chaofei Li

Abstract Background LncRNA POU3F3 (POU3F3) is overexpressed and plays oncogenic roles in esophageal squamous-cell carcinomas. LncRNA MEG3 (MEG3) has been characterized as a tumor suppressive lncRNA in different types of cancer. Our preliminary deep sequencing analysis revealed the inverse correlation between POU3F3 and MEG2 across melanoma tissues, indicating the interaction between them in melanoma. Therefore, this study was performed to investigate the crosstalk between POU3F3 and MEG3 in melanoma. Methods Tumor and adjacent healthy tissues collected from 60 melanoma patients were subjected to RNA extractions and RT-qPCRs to analyze the differential expression of POU3F3 and MEG2 in melanoma. In melanoma cells, POU3F3 and MEG2 were overexpressed to study the interactions between them. CCK-8 assays were performed to analyze the roles of POU3F3 and MEG2 in regulating melanoma cell proliferation. Results We found that POU3F3 was upregulated, while lncRNA MEG3 was downregulated in melanoma. Expression levels of POU3F3 and MEG3 were inversely correlated across tumor tissues. In vitro experiments showed that POU3F3 overexpression decreased MEG3 expression in melanoma cells, while MEG3 overexpression failed to affect POU3F3. POU3F3 overexpression increased melanoma cell proliferation, while MEG3 overexpression decreased melanoma cell proliferation. In addition, rescue experiments showed that MEG3 overexpression attenuated the enhancing effects of POU3F3 overexpression. Conclusion POU3F3 may promote melanoma cell proliferation by downregulating MEG3.


2007 ◽  
Vol 39 (6) ◽  
pp. 302-307 ◽  
Author(s):  
Chunfang Wang ◽  
Bin Li ◽  
Haiyan Zhang ◽  
Guangpu Shi ◽  
Weihong Li ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document