scholarly journals Telomerase inhibition, telomere attrition and proliferation arrest of cancer cells induced by phosphorothioate ASO-NLS conjugates targeting hTERC and siRNAs targeting hTERT

2020 ◽  
Vol 39 (1-3) ◽  
pp. 407-425
Author(s):  
Irmina Diala ◽  
Yasuo Shiohama ◽  
Takashi Fujita ◽  
Yojiro Kotake ◽  
Constantinos Demonacos ◽  
...  
Lung Cancer ◽  
2010 ◽  
Vol 68 (3) ◽  
pp. 346-354 ◽  
Author(s):  
Julia Beisner ◽  
Meng Dong ◽  
Sebastian Taetz ◽  
Noha Nafee ◽  
Ernst-Ulrich Griese ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. SCI-40-SCI-40
Author(s):  
Francesca Gazzaniga ◽  
Jue Lin ◽  
Elizabeth Blackburn

Abstract Abstract SCI-40 The ribonucleoprotein enzyme telomerase, which counteracts telomere shortening by adding telomeric DNA repeats to the 3’ ends of chromosomes, contains a core protein reverse transcriptase (called hTERT in humans), the essential template-containing RNA (hTER) and associated factors. Thus, telomerase is capable of circumventing the limit on cell division imposed by telomere attrition. Telomerase enzymatic activity is readily detectable in stem and progenitor cell populations, but is largely downregulated in most adult somatic human cells. In contrast, telomerase is highly active in the large majority (∼80%–90%) of cancer cells. While telomerase is essential for telomere maintenance in cells in vitro and in vivo, several studies now also indicate that telomerase may have telomere-independent functions. We previously showed that lowering the level of the telomerase ribonucleoprotein (RNP) enzyme complex causes rapid changes in cell cycle program in human and mouse cancer cells: partial reduction of telomerase induced by directly lowering telomerase RNP component levels causes altered transcriptional profiles and reduced glucose metabolism in human cancer cells, yet bulk telomere shortening or detectable telomere uncapping are not required for these effects. Work by others has shown that reduction of telomerase core protein component level reduced Wnt signaling and caused Wnt-mutant like developmental defects in Xenopus and mouse and aberrant hematopoiesis in zebrafish embryos. We acutely depleted telomerase RNA components in cultured T cells from healthy human adults using shRNAs targeting both the telomerase protein and the RNA component of telomerase (hTR), delivered immediately after in vitro stimulation. Cell proliferation and telomerase activity levels were quantified through the first and second stimulation cycles. Effects of such telomerase RNP reduction on cell proliferation were observed in time frames too short to be accounted for by bulk telomere attrition. Models to account for these findings will be discussed. Disclosures: Lin: Telome Health, Inc.: Consultancy, Equity Ownership. Blackburn:Telome Health, Inc: Equity Ownership, Membership on an entity’s Board of Directors or advisory committees.


2019 ◽  
Vol 42 (6) ◽  
pp. 906-914 ◽  
Author(s):  
Navakoon Kaewtunjai ◽  
Ratasark Summart ◽  
Ariyaphong Wongnoppavich ◽  
Bannakij Lojanapiwat ◽  
T. Randall Lee ◽  
...  

Lung Cancer ◽  
2003 ◽  
Vol 41 ◽  
pp. S84
Author(s):  
Milena Pantic ◽  
Stefan Zimmermann ◽  
Andreas Schnapp ◽  
Klaus Damm ◽  
Uwe M. Martens

2020 ◽  
Vol 21 (2) ◽  
pp. 653 ◽  
Author(s):  
Luan Wen ◽  
Changzhi Zhao ◽  
Jun Song ◽  
Linyuan Ma ◽  
Jinxue Ruan ◽  
...  

Mammalian telomere lengths are primarily regulated by telomerase, a ribonucleoprotein consisting of a reverse transcriptase (TERT) and an RNA subunit (TERC). TERC is constitutively expressed in all cells, whereas TERT expression is temporally and spatially regulated, such that in most adult somatic cells, TERT is inactivated and telomerase activity is undetectable. Most tumor cells activate TERT as a mechanism for preventing progressive telomere attrition to achieve proliferative immortality. Therefore, inactivating TERT has been considered to be a promising means of cancer therapy. Here we applied the CRISPR/Cas9 gene editing system to target the TERT gene in cancer cells. We report that disruption of TERT severely compromises cancer cell survival in vitro and in vivo. Haploinsufficiency of TERT in tumor cells is sufficient to result in telomere attrition and growth retardation in vitro. In vivo, TERT haploinsufficient tumor cells failed to form xenograft after transplantation to nude mice. Our work demonstrates that gene editing-mediated TERT knockout is a potential therapeutic option for treating cancer.


Sign in / Sign up

Export Citation Format

Share Document