Discovery of Novel Schizocommunin Derivatives as Telomeric G-Quadruplex Ligands That Trigger Telomere Dysfunction and the Deoxyribonucleic Acid (DNA) Damage Response

2018 ◽  
Vol 61 (8) ◽  
pp. 3436-3453 ◽  
Author(s):  
Tong Che ◽  
Shuo-Bin Chen ◽  
Jia-Li Tu ◽  
Bo Wang ◽  
Yu-Qing Wang ◽  
...  
2019 ◽  
Vol 25 (47) ◽  
pp. 11085-11097 ◽  
Author(s):  
Filippo Doria ◽  
Erica Salvati ◽  
Luca Pompili ◽  
Valentina Pirota ◽  
Carmen D'Angelo ◽  
...  

2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Julio Aguado ◽  
Agustin Sola-Carvajal ◽  
Valeria Cancila ◽  
Gwladys Revêchon ◽  
Peh Fern Ong ◽  
...  

AbstractHutchinson–Gilford progeria syndrome (HGPS) is a genetic disorder characterized by premature aging features. Cells from HGPS patients express progerin, a truncated form of Lamin A, which perturbs cellular homeostasis leading to nuclear shape alterations, genome instability, heterochromatin loss, telomere dysfunction and premature entry into cellular senescence. Recently, we reported that telomere dysfunction induces the transcription of telomeric non-coding RNAs (tncRNAs) which control the DNA damage response (DDR) at dysfunctional telomeres. Here we show that progerin-induced telomere dysfunction induces the transcription of tncRNAs. Their functional inhibition by sequence-specific telomeric antisense oligonucleotides (tASOs) prevents full DDR activation and premature cellular senescence in various HGPS cell systems, including HGPS patient fibroblasts. We also show in vivo that tASO treatment significantly enhances skin homeostasis and lifespan in a transgenic HGPS mouse model. In summary, our results demonstrate an important role for telomeric DDR activation in HGPS progeroid detrimental phenotypes in vitro and in vivo.


Author(s):  
Malissa C. Diehl ◽  
Lynne W. Elmore ◽  
Shawn E. Holt

2008 ◽  
Vol 6 (9) ◽  
pp. 87-88
Author(s):  
A. Biroccio ◽  
A. Rizzo ◽  
E. Salvati ◽  
M. Stevens ◽  
M. Stevens ◽  
...  

2021 ◽  
Author(s):  
Golam Mustafa ◽  
Sajad Shiekh ◽  
Keshav GC ◽  
Sanjaya Abeysirigunawardena ◽  
Hamza Balci

Abstract Single-stranded telomeric overhangs are ∼200 nucleotides long and can form tandem G-quadruplex (GQ) structures, which reduce their accessibility to nucleases and proteins that activate DNA damage response. Whether these tandem GQs further stack to form compact superstructures, which may provide better protection for longer telomeres, is not known. We report single-molecule measurements where the accessibility of 24–144 nucleotide long human telomeric DNA molecules is interrogated by a short PNA molecule that is complementary to a single GGGTTA repeat, as implemented in the FRET-PAINT method. Binding of the PNA strand to available GGGTTA sequences results in discrete FRET bursts which were analyzed in terms of their dwell times, binding frequencies, and topographic distributions. The binding frequencies were greater for binding to intermediate regions of telomeric DNA compared to 3′- or 5′-ends, suggesting these regions are more accessible. Significantly, the binding frequency per telomeric repeat monotonically decreased with increasing telomere length. These results are consistent with telomeres forming more compact structures at longer lengths, reducing accessibility of these critical genomic sites.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2412-2412
Author(s):  
Erik Westin ◽  
Larisa Pereboeva ◽  
Divya Devadasan ◽  
Tim M. Townes ◽  
Frederick D Goldman

Abstract Dyskeratosis Congenita (DC) is a bone marrow failure disorder characterized by a triad of leukoplakia, skin dyspigmentation and nail dystrophy. Pathologies found in these patients arise due to mutations found within a number of genes (DKC1, TERT, TERC, TINF2, TCAB1, CTC1, NOP10, C16orf57, NHP2 and PARN) that limit telomere maintenance/elongation, resulting in severely shortened telomeres. Previous studies in our lab have demonstrated impaired proliferation, limited lifespan and aberrant DNA damage response pathways in DC cells. These studies have also uncovered a significant reactive oxygen species (ROS) increase within every cell type investigated thus far. This ROS increase correlates with telomere dysfunction and the subsequent activation of the p53 DNA damage response pathway, which can be rescued by exogenous TERT or p53-shRNA expression. We have acquired skin punch biopsies from two patients with DC carrying either a TERT or DKC1 mutation. Here, we have investigated a potential candidate pathway largely characterized as a key antioxidant regulator in hematopoietic cells, NRF2 (NFE2L2). NRF2 is a redox-sensitive basic leucine zipper transcription factor that, together with its heterologous partners (small MAF proteins, cJun, ATF, etc), binds to antioxidant response elements (AREs) within gene promoters in a pro-oxidant environment. We compared the RNA expression via QRTPCR of NRF2 in control and DC skin fibroblasts and found a significant reduction in DC cells (TERT mutation: 1.5 fold; DKC1 mutation: 2.6 fold). Protein levels of NRF2 were also decreased in DC fibroblasts compared to controls. TXN is a gene whose expression is increased by NRF2 in a pro-oxidant environment. TXN expression was also significantly reduced (TERT mutation: 2.1 fold; DKC1 mutation: 2.2 fold). To test whether NRF2 suppression in DC cells is due to telomere dysfunction, we exogenously expressed TERT via retrovirus in DC and control fibroblasts. TERT expression led to dramatic increases in NRF2 (TERT mutation: 3.4 fold, DKC1 mutation: 3.7 fold) and TXN (TERT mutation: 3.7 fold, DKC1 mutation: 1.6 fold). In contrast, TERT expression in control cells increased NRF2 only 1.3 fold while TXN decreased 1.4 fold. Finally, we wanted to compare the expression of NRF2/TXN in low and elevated oxidative environments (4% vs 21% O2). Control cells increased the TXN expression in 21% O2 (NRF2: no change, TXN: 2.8 fold) while DC cells suppressed NRF2 (TERT mutation: no change, DKC1 mutation: 3 fold decrease) and TXN expression (TERT mutation: 1.4 fold decrease, DKC mutation: 2.3 fold decrease). Functional studies have found DC cells grown in low oxygen increase their proliferative capacity perhaps due to, in part, the NRF2 pathway. Together, these data support a hypothesis whereby shortened/dysfunctional telomeres suppress NRF2 activity and an antioxidant response to a pro-oxidant environment. Based upon previous research, this pathway is likely dependent on the activation of p53 as an intermediary between dysfunctional telomere signaling and the subsequent suppression of NRF2 activity. An abrogated antioxidant response in shortened telomere cells may promote entry into senescence and pathologies related to aging. Systemic pharmacological intervention that reduces ROS could reverse this process and form the basis to alleviate DC and related symptomology associated with this multi-organ disorder. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document