scholarly journals Interleukin enhancer-binding factor 3 and HOXC8 co-activate cadherin 11 transcription to promote breast cancer cells proliferation and migration

Oncotarget ◽  
2017 ◽  
Vol 8 (64) ◽  
pp. 107477-107491 ◽  
Author(s):  
Yang Zhang ◽  
Chenchen Yang ◽  
Mingsheng Zhang ◽  
Houli Liu ◽  
Chen Gong ◽  
...  
2021 ◽  
Author(s):  
Dale B. Bosco ◽  
Yi Ren ◽  
Karin A. Vallega ◽  
Qing-Xiang Amy Sang

Abstract Background Breast cancer is the most common cancer in women and the leading cause of female cancer deaths worldwide. Obesity causes chronic inflammation and is a risk factor for post-menopausal breast cancer and poor prognosis. Obesity triggers increased infiltration of macrophages into adipose tissue, yet little research has focused on the effects of macrophages in early stages of breast tumor development in obese patients. In this study, the effects of pro-inflammatory macrophages on breast cancer-adipocyte crosstalk were investigated.Methods An innovative human cell co-culture system was used to model the paracrine interactions among adipocytes, macrophages, and breast cancer cells, and how they facilitate tumor progression. The effects on cancer cells were examined using cell counts and migration assays. Quantitative reverse-transcription polymerase chain reaction was used to measure the expression levels of several cytokines and proteases to analyze adipocyte cancer-association.Results Macrophage conditioned media intensified the effects of breast cancer-adipocyte crosstalk. Adipocytes became delipidated and increased production of pro-inflammatory cytokines, even in the absence of cancer cells, although the expression levels were highest with all three cell components. As a result, co-cultured breast cancer cells became more aggressive, with increased proliferation and migration compared to adipocyte-breast cancer co-cultures treated with unconditioned media.Conclusions Macrophage conditioned media promotes adipocyte cancer-association. These macrophage-adipocyte paracrine interactions promote breast cancer cell proliferation and migration. Thus, macrophages may contribute to adipocyte inflammation and cancer-association and promote breast cancer progression.


2017 ◽  
Vol 50 (5) ◽  
pp. 1701-1710 ◽  
Author(s):  
Yanling Ding ◽  
Chunfu Zhang ◽  
Jiahui Zhang ◽  
Nannan Zhang ◽  
Tao Li ◽  
...  

EMBO Reports ◽  
2017 ◽  
Vol 18 (3) ◽  
pp. 420-436 ◽  
Author(s):  
Emad Heidary Arash ◽  
Ahmed Shiban ◽  
Siyuan Song ◽  
Liliana Attisano

2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Jia Cao ◽  
Xi Wang ◽  
Danni Wang ◽  
Rong Ma ◽  
Xiaohan Li ◽  
...  

2016 ◽  
Vol 341 (2) ◽  
pp. 111-122 ◽  
Author(s):  
Marta Guedes ◽  
João R. Araújo ◽  
Ana Correia-Branco ◽  
Inês Gregório ◽  
Fátima Martel ◽  
...  

2020 ◽  
Vol 10 ◽  
Author(s):  
Yuan Huang ◽  
Shi Li ◽  
Zhenhua Jia ◽  
Weiwei Zhao ◽  
Cefan Zhou ◽  
...  

The calcium-permeable cation channel TRPM8 (transient receptor potential melastatin 8) is a member of the TRP superfamily of cation channels that is upregulated in various types of cancer with high levels of autophagy, including prostate, pancreatic, breast, lung, and colon cancers. Autophagy is closely regulated by AMP-activated protein kinase (AMPK) and plays an important role in tumor growth by generating nutrients through degradation of intracellular structures. Additionally, AMPK activity is regulated by intracellular Ca2+ concentration. Considering that TRPM8 is a non-selective Ca2+-permeable cation channel and plays a key role in calcium homoeostasis, we hypothesized that TRPM8 may control AMPK activity thus modulating cellular autophagy to regulate the proliferation and migration of breast cancer cells. In this study, overexpression of TRPM8 enhanced the level of basal autophagy, whereas TRPM8 knockdown reduced the level of basal autophagy in several types of mammalian cancer cells. Moreover, the activity of the TRPM8 channel modulated the level of basal autophagy. The mechanism of regulation of autophagy by TRPM8 involves autophagy-associated signaling pathways for activation of AMPK and ULK1 and phagophore formation. Impaired AMPK abolished TRPM8-dependent regulation of autophagy. TRPM8 interacts with AMPK in a protein complex, and cytoplasmic C-terminus of TRPM8 mediates the TRPM8–AMPK interaction. Finally, basal autophagy mediates the regulatory effects of TRPM8 on the proliferation and migration of breast cancer cells. Thus, this study identifies TRPM8 as a novel regulator of basal autophagy in cancer cells acting by interacting with AMPK, which in turn activates AMPK to activate ULK1 in a coordinated cascade of TRPM8-mediated breast cancer progression.


2020 ◽  
Vol 11 (9) ◽  
pp. 2552-2559 ◽  
Author(s):  
Jae-Yeo Park ◽  
Shi-Eun Kang ◽  
Kwang Seok Ahn ◽  
Jae-Young Um ◽  
Woong Mo Yang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document