promoter mutant
Recently Published Documents


TOTAL DOCUMENTS

32
(FIVE YEARS 9)

H-INDEX

14
(FIVE YEARS 1)

2021 ◽  
pp. 1-10
Author(s):  
Megan C. Neville ◽  
Alexander Eastwood ◽  
Aaron M. Allen ◽  
Ammerins de Haan ◽  
Tetsuya Nojima ◽  
...  

2021 ◽  
Vol 118 (13) ◽  
pp. e2008772118
Author(s):  
Alexandra M. Amen ◽  
Christof Fellmann ◽  
Katarzyna M. Soczek ◽  
Shawn M. Ren ◽  
Rachel J. Lew ◽  
...  

Most glioblastomas (GBMs) achieve cellular immortality by acquiring a mutation in the telomerase reverse transcriptase (TERT) promoter. TERT promoter mutations create a binding site for a GA binding protein (GABP) transcription factor complex, whose assembly at the promoter is associated with TERT reactivation and telomere maintenance. Here, we demonstrate increased binding of a specific GABPB1L-isoform–containing complex to the mutant TERT promoter. Furthermore, we find that TERT promoter mutant GBM cells, unlike wild-type cells, exhibit a critical near-term dependence on GABPB1L for proliferation, notably also posttumor establishment in vivo. Up-regulation of the protein paralogue GABPB2, which is normally expressed at very low levels, can rescue this dependence. More importantly, when combined with frontline temozolomide (TMZ) chemotherapy, inducible GABPB1L knockdown and the associated TERT reduction led to an impaired DNA damage response that resulted in profoundly reduced growth of intracranial GBM tumors. Together, these findings provide insights into the mechanism of cancer-specific TERT regulation, uncover rapid effects of GABPB1L-mediated TERT suppression in GBM maintenance, and establish GABPB1L inhibition in combination with chemotherapy as a therapeutic strategy for TERT promoter mutant GBM.


2021 ◽  
Author(s):  
Andrew McKinney ◽  
Radhika Mathur ◽  
Nicholas Stevers ◽  
Annette M. Molinaro ◽  
Susan Chang ◽  
...  

Thyroid ◽  
2020 ◽  
Vol 30 (10) ◽  
pp. 1470-1481 ◽  
Author(s):  
Brittany A. McKelvey ◽  
Timothy Gilpatrick ◽  
Yongchun Wang ◽  
Winston Timp ◽  
Christopher B. Umbricht ◽  
...  

Cell Reports ◽  
2020 ◽  
Vol 31 (8) ◽  
pp. 107718
Author(s):  
Josh Lewis Stern ◽  
Richard D. Paucek ◽  
Franklin W. Huang ◽  
Mahmoud Ghandi ◽  
Ronald Nwumeh ◽  
...  

2020 ◽  
Author(s):  
Alexandra M. Amen ◽  
Christof Fellmann ◽  
Katarzyna M. Soczek ◽  
Shawn M. Ren ◽  
Rachel J. Lew ◽  
...  

AbstractMost glioblastomas (GBMs) achieve cellular immortality by acquiring a mutation in the telomerase reverse transcriptase (TERT) promoter. TERT promoter mutations create a binding site for a GA binding protein (GABP) transcription factor complex, whose expression is associated with TERT reactivation and telomere maintenance. Here, using biochemical and cell biology approaches, we show direct evidence that a specific GABP complex containing the subunit protein GABPB1L forms predominantly at the mutant TERT promoter, leading to TERT re-expression. Furthermore, we find that TERT promoter mutant GBM cells, unlike wild-type cells, are immediately dependent on GABPB1L for proliferation in cell culture and post-tumor establishment in vivo. Notably, when combined with frontline temozolomide (TMZ) chemotherapy, GABPB1L knockdown and the associated TERT reduction lead to an impaired DNA damage response that results in profoundly reduced growth of intracranial GBM tumors. Together, these findings provide new insights into the mechanism of cancer-specific TERT regulation, uncover rapid effects of TERT suppression in GBM maintenance, and establish GABPB1L inhibition, alone or in combination with chemotherapy, as a therapeutic strategy for TERTp mutant GBM.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi106-vi107
Author(s):  
Alexandra Amen ◽  
Rachel Lew ◽  
Shawn Ren ◽  
Andrew McKinney ◽  
Andrew Mancini ◽  
...  

Abstract Understanding cancer cell immortality in primary glioblastoma (GBM) is essential for the development of more informed treatments. Multiple cancer types, including >80% of GBMs, undergo immortalization by reactivating Telomerase Reverse Transcriptase (TERT) through acquired mutations in the TERT promoter. TERT, the catalytically active and rate-limiting subunit of telomerase, functions to maintain telomeres, which cap and protect the ends of chromosomes. Our past work has demonstrated that the transcription factor GABP - and specifically its tetramer-forming isoform GABPb1L - binds and activates the mutant TERT promoter. The generation of CRISPR-induced indels in GABPb1L results in a gradual loss of cell viability in TERT promoter mutant but not TERT promoter wild type tumor cells in vitro, but the extent to which GABPb1L function is compromised in this setting is unclear. Thus, the potential for use of GABPb1L as an effective therapeutic target for TERT promoter mutant GBM requires further investigation. Here, we use CRISPR-based strategies to demonstrate that full knockout of GABPb1L is rapidly lethal in TERT promoter mutant cells in vitro, in association with a decrease in both TERT mRNA and telomerase activity. Heterozygous deletion of GABPb1L in the context of TERT promoter mutations leads to slowed growth of orthotopic xenograft tumors in mice, and prolonged survival. Additionally, inducible RNAi-mediated inhibition of GABPb1L in growing tumors is also capable of decreasing tumor burden and increasing survival, further strongly suggesting that targeting GABPb1L in patient tumors could be a viable treatment strategy. Finally, reduced GABPb1L synergizes with temozolomide (TMZ) therapy such that TMZ treatment in the context of low GABPb1L and low TERT leads to a complete ablation of orthotopic GBM xenografts. These results highlight the potential to improve disease outcomes by targeting TERT through inhibition of GABPb1L, particularly in conjunction with TMZ treatment.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi37-vi37
Author(s):  
Elisa Aquilanti ◽  
Duncan Baird ◽  
Jacqueline Watson ◽  
Matthew Meyerson

Abstract TERT promoter mutations are among the most common somatic alterations in cancer and they occur in about 80% of IDH-wildtype glioblastomas. TERT promoter mutations were found to reactivate telomerase by providing a novel binding site for the GABP transcription factor. While the effects of telomerase ablation are well understood in mice and somatic human cells, these effects in cancer are yet to be fully elucidated. In this study, we used a genetic approach with CRISPR-interference to knock down telomerase in TERT promoter-mutant glioblastoma cell lines. We show that this leads to a gradual and significant reduction in proliferation. This phenotype ultimately culminates in telomere crisis, with telomere shortening, activation of the DNA damage response pathway and formation of chromatin bridges. These data suggest that anti-telomerase therapy is a potential effective approach for glioblastoma tumors.


2018 ◽  
Vol 475 (1) ◽  
pp. 355-371 ◽  
Author(s):  
Antara Biswas ◽  
Abhijith Shettar ◽  
Geetashree Mukherjee ◽  
Paturu Kondaiah ◽  
Kartiki V. Desai

Using microarray analysis, we found that HOX transcript antisense intergenic RNA (HOTAIR) is up-regulated by Jumonji domain containing-6 (JMJD6), a bifunctional lysyl hydroxylase and arginine demethylase. In breast cancer, both JMJD6 and HOTAIR RNAs increase tumor growth and associate with poor prognosis but no molecular relationship between them is known. We show that overexpression of JMJD6 increased HOTAIR expression and JMJD6 siRNAs suppressed it in ER+ MCF-7, triple negative MDA-MB-231 and non-breast cancer HEK 293 cells. Therefore, JMJD6 regulates HOTAIR independent of ER status. Using various deletion constructs spanning (−1874 to +50) of the HOTAIR promoter, we identified pHP216 (−216 to +50 bp) as the smallest construct that retained maximal JMJD6 responsiveness. In ChIP assays, JMJD6 bound this region suggesting that JMJD6 may be directly recruited to the HOTAIR promoter. Mutant JMJD6H187A that is devoid of enzymatic activity could bind this site but failed to induce transcription. ChIP and electromobility shift assays identified a JMJD6 interaction region from (−123 to −103 bp) within the HOTAIR promoter. In tumor samples but not normal breast tissue, the expression of JMJD6 linearly correlated with HOTAIR suggesting that JMJD6-mediated up-regulation may occur specifically in tumors. Further, concurrent high expression of both genes correlated with poor survival when individual expression of either gene showed no significant association in TCGA datasets. We propose that high JMJD6 expression may achieve higher levels of HOTAIR in breast tumors. Further, since high levels of HOTAIR promote metastasis and death, blocking JMJD6 may be useful in preventing such events.


Sign in / Sign up

Export Citation Format

Share Document