Abstract P1-05-05: The effect of microRNA dysregulation on health disparities observed in African American women in triple negative breast cancer

Author(s):  
Ches'que M. Phillips ◽  
Yaguang Xi
2017 ◽  
Author(s):  
Aline S. Fonseca ◽  
Selene Elifio-Esposito ◽  
Marilesia F. Souza ◽  
Akanksha Mahajan ◽  
Yara R. Zabala ◽  
...  

2018 ◽  
Vol Volume 11 ◽  
pp. 1-12 ◽  
Author(s):  
Odalys Torres-Luquis ◽  
Krystal Madden ◽  
N'sanh MR N'dri ◽  
Richard Berg ◽  
Olufunmilayo F. Olopade ◽  
...  

2021 ◽  
Author(s):  
Johnathan Abou-Fadel ◽  
Brian Grajeda ◽  
Xiaoting Jiang ◽  
Alyssa-Marie Cailing-De La O ◽  
Esmeralda Flores ◽  
...  

Breast cancer is the most commonly diagnosed cancer worldwide and remains the second leading cause of cancer death. While breast cancer mortality has steadily declined over the past decades through medical advances, an alarming disparity in breast cancer mortality has emerged between African American women (AAW) and Caucasian American women (CAW); and new evidence suggests more aggressive behavior of triple-negative breast cancer (TNBC) in AAW may contribute to racial differences in tumor biology and mortality. Progesterone (PRG) is capable of exerting its cellular effects through either its classic, non-classic or combined responses through binding to either classic nuclear PRG receptors (nPRs) or non-classic membrane PRG receptors (mPRs), warranting both pathways an equally important status in PRG-mediated signaling. In our previous report, we demonstrated that the CCM signaling complex (CSC) consisting of CCM1, CCM2, and CCM3 proteins can couple both nPRs and mPRs signaling cascades to form a CSC-mPRs-PRG-nPRs (CmPn) signaling network in nPR positive(+) breast cancer cells. In this report, we furthered our research by establishing the CSC-mPRs-PRG (CmP) signaling network in nPR(-) breast cancer cells, demonstrating that a common core mechanism exists, regardless of nPR(+/-) cell type. This is the first report stating that inducible expression patterns exist between CCMs and major mPRs in TNBC cells. Furthermore, we firstly show mPRs in TNBC cells are localized in the nucleus and participate in nucleocytoplasmic shuttling in a coordinately synchronized fashion with CCM proteins under steroid actions, following the same cellular distribution as other well-defined steroid hormone receptors. Finally, for the first time, we deconvoluted the CmP signalosome by using multi-omics approaches, which helped us understand key factors within the CmP network, and identify 21 specific biomarkers with potential clinical applications associated with AAW-TNBC tumorigenesis. These novel biomarkers could have immediate clinical implications to dramatically improve health disparities among AAW-TNBCs.


2016 ◽  
Author(s):  
Julie E. Getz ◽  
Davyd B. Teoh ◽  
Sara Nasser ◽  
Christophe R. Legendre ◽  
Waibhav Tembe ◽  
...  

2008 ◽  
Vol 195 (2) ◽  
pp. 153-158 ◽  
Author(s):  
Chukwuemeka U. Ihemelandu ◽  
Tammey J. Naab ◽  
Haile M. Mezghebe ◽  
Kepher H. Makambi ◽  
Suryanarayana M. Siram ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document