pituitary tumor cell
Recently Published Documents


TOTAL DOCUMENTS

72
(FIVE YEARS 2)

H-INDEX

20
(FIVE YEARS 1)

Gland Surgery ◽  
2021 ◽  
Vol 0 (0) ◽  
pp. 0-0
Author(s):  
Bowen Sun ◽  
Congxin Dai ◽  
Shangfeng Zhao ◽  
Hao Dong ◽  
Haocheng Liu ◽  
...  

2018 ◽  
Vol 25 (10) ◽  
pp. 837-852 ◽  
Author(s):  
Juan Pablo Petiti ◽  
Liliana del Valle Sosa ◽  
Florencia Picech ◽  
Gabriela Deisi Moyano Crespo ◽  
Jean Zander Arevalo Rojas ◽  
...  

In pituitary adenomas, early recurrences and resistance to conventional pharmacotherapies are common, but the mechanisms involved are still not understood. The high expression of epidermal growth factor receptor 2 (HER2)/extracellular signal-regulated kinase (ERK1/2) signal observed in human pituitary adenomas, together with the low levels of the antimitogenic transforming growth factor beta receptor 2 (TBR2), encouraged us to evaluate the effect of the specific HER2 inhibition with trastuzumab on experimental pituitary tumor cell growth and its effect on the antiproliferative response to TGFB1. Trastuzumab decreased the pituitary tumor growth as well as the expression of ERK1/2 and the cell cycle regulators CCND1 and CDK4. The HER2/ERK1/2 pathway is an attractive therapeutic target, but its intricate relations with other signaling modulators still need to be unraveled. Thus, we investigated possible cross-talk with TGFB signaling, which has not yet been studied in pituitary tumors. In tumoral GH3 cells, co-incubation with trastuzumab and TGFB1 significantly decreased cell proliferation, an effect accompanied by a reduction in ERK1/2 phosphorylation, an increase of SMAD2/3 activation. In addition, through immunoprecipitation assays, a diminution of SMAD2/3-ERK1/2 and an increase SMAD2/3–TGFBR1 interactions were observed when cells were co-incubated with trastuzumab and TGFB1. These findings indicate that blocking HER2 by trastuzumab inhibited pituitary tumor growth and modulated HER2/ERK1/2 signaling and consequently the anti-mitogenic TGFB1/TBRs/SMADs cascade. The imbalance between HER2 and TGFBRs expression observed in human adenomas and the response to trastuzumab on experimental tumor growth may make the HER2/ERK1/2 pathway an attractive target for future pituitary adenoma therapy.


2018 ◽  
Vol 25 (6) ◽  
pp. 665-676 ◽  
Author(s):  
M Fuertes ◽  
M Sapochnik ◽  
L Tedesco ◽  
S Senin ◽  
A Attorresi ◽  
...  

Increased levels of the proto-oncogene pituitary tumor-transforming gene 1 (PTTG) have been repeatedly reported in several human solid tumors, especially in endocrine-related tumors such as pituitary adenomas. Securin PTTG has a critical role in pituitary tumorigenesis. However, the cause of upregulation has not been found yet, despite analyses made at the gene, promoter and mRNA level that show that no mutations, epigenetic modifications or other mechanisms that deregulate its expression may explain its overexpression and action as an oncogene. We describe that high PTTG protein levels are induced by the RWD-containing sumoylation enhancer (RWDD3 or RSUME), a protein originally identified in the same pituitary tumor cell line in which PTTG was also cloned. We demonstrate that PTTG and RSUME have a positive expression correlation in human pituitary adenomas. RSUME increases PTTG protein in pituitary tumor cell lines, prolongs the half-life of PTTG protein and regulates the PTTG induction by estradiol. As a consequence, RSUME enhances PTTG transcription factor and securin activities. PTTG hyperactivity on the cell cycle resulted in recurrent and unequal divisions without cytokinesis, and the consequential appearance of aneuploidies and multinucleated cells in the tumor. RSUME knockdown diminishes securin PTTG and reduces its tumorigenic potential in a xenograft mouse model. Taken together, our findings show that PTTG high protein steady state levels account for PTTG tumor abundance and demonstrate a critical role of RSUME in this process in pituitary tumor cells.


2017 ◽  
Vol 102 (10) ◽  
pp. 3692-3703 ◽  
Author(s):  
Shao Jian Lin ◽  
Ze Rui Wu ◽  
Lei Cao ◽  
Yong Zhang ◽  
Zhi Gen Leng ◽  
...  

Abstract Context The dopamine agonist cabergoline (CAB) has been used widely in the treatment of prolactinomas and other types of pituitary adenomas, but its clinical use is hampered by intolerance in some patients with prolactinoma and lack of effectiveness in other pituitary tumor types. Chloroquine (CQ) is an old drug widely used to treat malaria. Recent studies, including our own, have revealed that CAB and CQ are involved in induction of autophagy and activation of autophagic cell death. Objective To test whether CAB and CQ can function cooperatively to suppress growth of pituitary adenomas as well as other cancers. Results In vitro studies using the rat pituitary tumor cell lines MMQ and GH3, human pituitary tumor cell primary cultures, and several human cancer cell lines showed that CQ enhanced suppression of cell proliferation by CAB. These results were confirmed in in vivo xenograft models in nude mice and estrogen-induced rat prolactinomas. To understand the mechanism of combined CAB and CQ action, we established a low-CAB-dose condition in which CAB was able to induce autophagy but failed to suppress cell growth. Addition of CQ to low-dose CAB blocked normal autophagic cycles and induced apoptosis, evidenced by the further accumulation of p62/caspase-8/LC3-II. Conclusion The data suggest that combined use of CAB and CQ may increase clinical effectiveness in treatment of human pituitary adenomas, as well as other cancers, making it an attractive option in tumor and cancer therapies.


2017 ◽  
Vol 14 (2) ◽  
pp. 1706-1714 ◽  
Author(s):  
Yuanchuan Wang ◽  
Xiaohong Yin ◽  
Long Zhao ◽  
Shun Li ◽  
Jie Duan ◽  
...  

Oncotarget ◽  
2015 ◽  
Vol 6 (30) ◽  
pp. 29413-29427 ◽  
Author(s):  
Hai-qian Liang ◽  
Ren-jie Wang ◽  
Cai-feng Diao ◽  
Jian-wei Li ◽  
Jing-liang Su ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document