Telomeres at chromosome ends shorten during cell division and aging in humans. Critically short dysfunctional telomeres trigger cell senescence or apoptosis, which contributes to aging-related diseases and degeneration. Oxidative stress accelerates telomere shortening, and generates reactive oxygen species (ROS), which are particularly damaging to telomeric TTAGGG repeat sequences. The goals of this proposal are to 1) measure the response of base excision repair (BER) proteins to DNA damage at telomeric regions compared with non- telomere regions;and 2) to define the impact of oxidized telomeric DNA on telomere integrity and telomere functions in preventing cell senescence and cell death. A critical barrier to investigating DNA damage and repair at telomeres has been an inability to target damage to the telomeres. To overcome this obstacle we developed a highly innovative system for confining ROS-induced DNA damage to defined regions in the genome. For this we fused KillerRed (KR) protein, a photosensitizer that generates ROS upon light irradiation, to a Tet-repressor (tetR) protein that binds to a single engineered site within condensed chromatin. We established that oxidative damage occurred only at the tetR bound site. To target telomeres we fused KR to the telomere binding protein TRF1 and showed that this restricts ROS-induced damage to the telomeres.
In Aim 1, we will combine the KR system with confocal microscopy to visualize the real time damage response of BER proteins to targeted oxidative damage in telomeric and non-telomeric genomic regions, with 3D resolution in a single cell nucleus. We will measure the mobilization kinetics of various GFP-tagged BER proteins, and the protein domains required for the response, to ROS damage at the TRF1 bound telomeric sites compared with the tetR bound non-telomeric site.
In Aim 2, we will test several endpoints of telomere damage and dysfunction, including cellular senescence and apoptosis, after ROS production by activating KR with light exposure in cells that stably express KR-TRF1, compared with cells expressing HcRed-TRF1 as a control. This study will include both bulk cell population and single cell experiments. Using the microscopy system to target KR activation to defined numbers of KR-TRF1 foci, we will measure the average number of """"""""oxidized"""""""" telomeres required to trigger apoptosis or senescence. Successful completion of the project will provide crucial insights into how oxidative stress accelerates telomere shortening, and how telomeric oxidative base damage impacts telomere function and promotes cell senescence. The results will pave the way for future work examining how telomeric damage and repair change with age and vary with cell type, and should lead to new strategies for preserving telomere function to promote healthy aging. !

Public Health Relevance

Critically short telomeres contribute to aging-related diseases by causing cellular senescence or apoptosis. Oxidative stress accelerates telomere shortening and damages telomeric DNA. This project will advance understanding of how oxidative stress disrupts telomere structure and function, and the mechanisms for repairing damaged telomeres. This study will also establish a new platform for testing factors that influence telomere repair an promote telomere preservation to delay aging-related diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21AG045545-01
Application #
8566953
Study Section
Cellular Mechanisms in Aging and Development Study Section (CMAD)
Program Officer
Guo, Max
Project Start
2013-07-01
Project End
2015-06-30
Budget Start
2013-07-01
Budget End
2014-06-30
Support Year
1
Fiscal Year
2013
Total Cost
$190,625
Indirect Cost
$65,625
Name
University of Pittsburgh
Department
Genetics
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Gao, Ying; Li, Changling; Wei, Leizhen et al. (2017) SSRP1 Cooperates with PARP and XRCC1 to Facilitate Single-Strand DNA Break Repair by Chromatin Priming. Cancer Res 77:2674-2685
Tan, Rong; Nakajima, Satoshi; Wang, Qun et al. (2017) Nek7 Protects Telomeres from Oxidative DNA Damage by Phosphorylation and Stabilization of TRF1. Mol Cell 65:818-831.e5
Tan, Rong; Lan, Li (2017) Induction of Site-Specific Oxidative Damage at Telomeres by Killerred-Fused Shelretin Proteins. Methods Mol Biol 1587:139-146
Yu, Yang; Tan, Rong; Ren, Qian et al. (2017) POT1 inhibits the efficiency but promotes the fidelity of nonhomologous end joining at non-telomeric DNA regions. Aging (Albany NY) 9:2529-2543
Fouquerel, Elise; Parikh, Dhvani; Opresko, Patricia (2016) DNA damage processing at telomeres: The ends justify the means. DNA Repair (Amst) 44:159-168
Fouquerel, Elise; Lormand, Justin; Bose, Arindam et al. (2016) Oxidative guanine base damage regulates human telomerase activity. Nat Struct Mol Biol 23:1092-1100
Sun, Luxi; Tan, Rong; Xu, Jianquan et al. (2015) Targeted DNA damage at individual telomeres disrupts their integrity and triggers cell death. Nucleic Acids Res 43:6334-47
Hwang, Bor-Jang; Jin, Jin; Gao, Ying et al. (2015) SIRT6 protein deacetylase interacts with MYH DNA glycosylase, APE1 endonuclease, and Rad9-Rad1-Hus1 checkpoint clamp. BMC Mol Biol 16:12
Lee, Hong-Jen; Lan, Li; Peng, Guang et al. (2015) Tyrosine 370 phosphorylation of ATM positively regulates DNA damage response. Cell Res 25:225-36
Jiang, Wenxia; Crowe, Jennifer L; Liu, Xiangyu et al. (2015) Differential phosphorylation of DNA-PKcs regulates the interplay between end-processing and end-ligation during nonhomologous end-joining. Mol Cell 58:172-85

Showing the most recent 10 out of 16 publications