scholarly journals Inhibition of in vivo breast cancer growth by antisense oligodeoxynucleotides to type I insulin-like growth factor receptor mRNA involves inactivation of ErbBs, PI-3K/Akt and p42/p44 MAPK signaling pathways but not modulation of progesterone receptor activity

Oncogene ◽  
2004 ◽  
Vol 23 (30) ◽  
pp. 5161-5174 ◽  
Author(s):  
Mariana Salatino ◽  
Roxana Schillaci ◽  
Cecilia J Proietti ◽  
Romina Carnevale ◽  
Isabel Frahm ◽  
...  
Oncotarget ◽  
2016 ◽  
Vol 7 (46) ◽  
pp. 75571-75584 ◽  
Author(s):  
Ingunn Holen ◽  
Diane V. Lefley ◽  
Sheila E. Francis ◽  
Sarah Rennicks ◽  
Steven Bradbury ◽  
...  

2007 ◽  
Vol 21 (8) ◽  
pp. 1781-1790 ◽  
Author(s):  
Haibiao Gong ◽  
Ping Guo ◽  
Yonggong Zhai ◽  
Jie Zhou ◽  
Hirdesh Uppal ◽  
...  

Abstract Estrogen plays an important role in normal physiology. It is also a risk factor for breast cancer, and antiestrogen therapies have been shown to be effective in the treatment and prevention of breast cancers. The liver is important for estrogen metabolism, and a compromised liver function has been linked to hyperestrogenism in patients. In this report, we showed that the liver X receptor (LXR) controls estrogen homeostasis by regulating the basal and inducible hepatic expression of estrogen sulfotransferase (Est, or Sult1e1), an enzyme critical for metabolic estrogen deactivation. Genetic or pharmacological activation of LXR resulted in Est induction, which in turn inhibited estrogen-dependent uterine epithelial cell proliferation and gene expression, as well as breast cancer growth in a nude mouse model of tumorigenicity. We further established that Est is a transcriptional target of LXR, and deletion of the Est gene in mice abolished the LXR effect on estrogen deprivation. Interestingly, Est regulation by LXR appeared to be liver specific, further underscoring the role of liver in estrogen metabolism. Activation of LXR failed to induce other major estrogen-metabolizing enzymes, suggesting that the LXR effect on estrogen metabolism is Est specific. In summary, our results have revealed a novel mechanism controlling estrogen homeostasis in vivo and may have implications for drug development in the treatment of breast cancer and other estrogen-related cancerous endocrine disorders.


2020 ◽  
Vol 121 ◽  
pp. 109502 ◽  
Author(s):  
Zhe Liu ◽  
Xianmin Ge ◽  
Yuchen Gu ◽  
Yingying Huang ◽  
Hao Liu ◽  
...  

2016 ◽  
Vol 34 (15_suppl) ◽  
pp. e12574-e12574 ◽  
Author(s):  
Nuha Buchanan Kadri ◽  
Matthew Gdovin ◽  
Nizar Alyassin ◽  
Justin Avila ◽  
Aryana Cruz ◽  
...  

2008 ◽  
Vol 68 (S 01) ◽  
Author(s):  
M Smollich ◽  
M Götte ◽  
J Fischgräbe ◽  
I Radke ◽  
L Macedo ◽  
...  

2018 ◽  
Vol 36 (15_suppl) ◽  
pp. e12581-e12581
Author(s):  
Shristi Bhattarai ◽  
Sergey Klimov ◽  
Mohammed A. Aleskandarany ◽  
Helen Burrell ◽  
Anthony Wormall ◽  
...  

2020 ◽  
Author(s):  
Chengpeng Zhao ◽  
Xiaoling Ling ◽  
Yunxia Xia ◽  
Bingxue Yan ◽  
Quanlin Guan

Abstract Background Breast cancer is a malignant tumor with high incidence in females. Burgeoning studies have analyzed the relationship between long non-coding RNA (lncRNA) and breast cancer. However, the role of LncRNA UAC1 in the pathogenesis of breast cancer remains unclear. Methods LncRNA-UCA1 levels were detected in breast cancer tissues and cells while correlation between LncRNA-UCA1 expression and patient survival was analyzed by the Kaplan-Meier. The proliferation, invasion and apoptosis of cells were measured by CCK-8 assay, Transwell test and flow cytometry, respectively. Protein levels of apoptosis-related factors were assessed by Western blots. RIP test detected the combination of LncRNA-UCA1 and DNA methyltransferases whereas RNA pull-down test showed the interaction of DNA methyltransferases and LncRNA UCA1. Enrichment of DNA transferase of METTL14 promoter in T47D was assessed by ChIP. Interaction between miR-375 and SOX12 was confirmed via dual-luciferase reporter assay. Tumorigenesis was observed in vivo. Results LncRNA-UCA1 levels were increased in breast cancer and a high LncRNA-UCA1 level was a risk factor of the poor breast cancer prognosis. Silencing LncRNA-UCA1 inhibited the proliferation and invasion but promoted apoptosis of breast cancer cells. LncRNA-UCA1 recruited DNA methyltransferase to the METTL14 promoter region to inhibit METTL14 expression in breast cancer. Low METTL14 levels were associated with stronger proliferation and invasion abilities, whereas weaker proliferation and invasion abilities were related to low LncRNA-UCA1 levels. METTL14 mediated the low expression of miR-375 by m6A modification in breast cancer. Conclusion Depleted LncRNA-UCA1 inhibited breast cancer growth by regulating the METTL14-miR-375-SOX12 axis in vitro and in vivo.


Sign in / Sign up

Export Citation Format

Share Document