scholarly journals The Nuclear Factor-κB Pathway Controls the Progression of Prostate Cancer to Androgen-Independent Growth

2008 ◽  
Vol 68 (16) ◽  
pp. 6762-6769 ◽  
Author(s):  
Ren Jie Jin ◽  
Yongsoo Lho ◽  
Linda Connelly ◽  
Yongqing Wang ◽  
Xiuping Yu ◽  
...  
2008 ◽  
Vol 22 (2) ◽  
pp. 273-286 ◽  
Author(s):  
Soyoung Ko ◽  
Liheng Shi ◽  
Soyoung Kim ◽  
Chung S. Song ◽  
Bandana Chatterjee

Abstract Increased androgen receptor (AR) levels are associated with prostate cancer progression to androgen independence and therapy resistance. Evidence has suggested that chronic inflammation is closely linked to various cancers including prostate cancer. Herein we show that the proinflammatory cytokine TNFα negatively regulates AR mRNA and protein expression and reduces androgen sensitivity in androgen-dependent LNCaP human prostate cancer cells. Decreased AR expression results from transcription repression involving essential in cis interaction of nuclear factor-κB (NF-κB) with the B-myb transcription factor at a composite genomic element in the 5′-untranslated region of AR. The negative regulation was abrogated when NF-κB activity was inhibited by a superrepressor of the inhibitory κB protein. In contrast, androgen-independent C4-2 (LNCaP-derived) cells fail to show AR down-regulation by TNFα, despite expression of B-myb and TNFα-induced NF-κB activity similar to that in LNCaP cells. The negatively regulated AR gene chromatin region showed TNFα-dependent enrichment of B-myb and the NF-κB proteins p65 and p50. In parallel, the histone deacetylase 1, corepressor silencing mediator of retinoid and thyroid hormone receptor and the corepressor-associated scaffold protein mSin3A were recruited to the inhibitory site. In C4-2 cells, neither NF-κB and B-myb, nor any of the corepressor components, were detected at the negative site in response to TNFα. Apoptosis was induced in TNFα-treated LNCaP cells, likely in part due to the down-regulation of AR. The androgen-independent, AR-expressing C4-2 and C4-2B (derived from C4-2) cells were resistant to TNFα-induced apoptosis. The results linking androgen dependence to the NF-κB and AR pathways may be insightful in identifying novel treatment targets for prostate cancer.


2010 ◽  
Vol 16 (3) ◽  
pp. 286-294 ◽  
Author(s):  
Junxia Min ◽  
Alexander Zaslavsky ◽  
Giuseppe Fedele ◽  
Sara K McLaughlin ◽  
Elizabeth E Reczek ◽  
...  

2012 ◽  
Vol 287 (44) ◽  
pp. 37570-37582 ◽  
Author(s):  
Rachana Garg ◽  
Jorge Blando ◽  
Carlos J. Perez ◽  
HongBin Wang ◽  
Fernando J. Benavides ◽  
...  

Tumor Biology ◽  
2017 ◽  
Vol 39 (5) ◽  
pp. 101042831769838 ◽  
Author(s):  
Chandrashekhar Dasari ◽  
Dattu Prasad Yaghnam ◽  
Reinhard Walther ◽  
Ramesh Ummanni

Our previous study showed that TPD52 overexpression could increase migration and proliferation of LNCaP cells contributing to the development of prostate cancer. However, mechanism of TPD52 in prostate cancer initiation and progression remains elusive. In this study, we investigated the possible underlying mechanism of TPD52 in prostate cancer progression. In LNCaP cells, TPD52 expression was altered by transfecting with either EGFP-TPD52 or specific short hairpin RNA. Overexpression of TPD52 protected LNCaP cells from apoptosis through elevated anti-apoptotic proteins XIAP, Bcl-2, and Cyclin D1, whereas Bax was downregulated. Mechanistically, we found that TPD52 confers transactivation of nuclear factor-κB, thereby enhancing its target gene expression in LNCaP cells. TPD52 promotes LNCaP cell invasion probably via increased matrix metalloproteinase 9 expression and its activity while tissue inhibitor of metalloproteinase expression is significantly downregulated. Notably, TPD52 might be involved in cell adhesion, promoting tumor metastasis by inducing loss of E-cadherin, expression of vimentin and vascular cell adhesion molecule, and additionally activation of focal adhesion kinase. Furthermore, TPD52 directly interacts with nuclear factor-κB p65 (RelA) and promotes accumulation of phosphorylated nuclear factor-κB (p65)S536 that is directly linked with nuclear factor-κB transactivation. Indeed, depletion of TPD52 or inhibition of nuclear factor-κB in TPD52-positive cells inhibited secretion of tumor-related cytokines and contributes to the activation of STAT3, nuclear factor-κB, and Akt. Interestingly, in TPD52 overexpressing LNCaP cells, nuclear factor-κB inhibition prevented the autocrine/paracrine activation of STAT3. TPD52 activates STAT3 through ascertaining a cross talk between the nuclear factor-κB and the STAT3 signaling systems. Collectively, these results reveal mechanism by which TPD52 is associated with prostate cancer progression and highlight the approach for therapeutic targeting of TPD52 in prostate cancer.


Sign in / Sign up

Export Citation Format

Share Document