A gradual loss of genomic integrity has long been proposed to contribute to the aging process. Consistent with this theory, mutation of genes involved in maintaining genome integrity often leads to premature aging. Identifying the potential genetic predispositions to age-related diseases and, more importantly, understanding how these genetic determinants lead to disease will likely guide our ability to better predict, diagnose and slow the onset of these disorders. The ATR protein kinase maintains genome integrity in mammalian cells and is a central regulator of cell cycle checkpoints. Using a cre/lox system that allows conditional deletion of the ATR gene in 2-3 month old adult mice, our preliminary work has demonstrated that several age-related phenotypes appear 3-6 months after ATR deletion. These aging phenotypes include alopecia, hair graying, kyphosis, osteoporosis, cardiomyopathy, testicular atrophy and acute immune suppression. With this unique mouse model, we propose herein to substantiate these initial findings and further explore the role of ATR in preventing age-related diseases. Experiments are proposed to further explore the effect of ATR loss on several disorders that are components of human aging (hair loss and graying, osteoporosis and reduced hematopoietic regenerative capacity) and, importantly, to address if these phenotypes are caused by a loss of regenerative capacity following ATR deletion and/or a direct effect on differentiated cells. To determine if activation of the p53 transcription factor plays a causative role in age-related phenotypes that result from ATR deletion, the premature aging observed in ATR mice will be compared with that observed in mice that lack both ATR and p53. Finally, experiments are proposed that will determine whether the oxidative DNA base damage that results from normal metabolic processes is exacerbated by ATR loss, leading to highly toxic DNA double strand breaks. Such amplification of DNA damage may accelerate normal aging. To test this hypothesis, oxidative DNA damage will be reduced or increased by treatments that modulate intracellular hydroxyl radical concentrations in ATR knockout cells, and the effect of these treatments will be monitored by chromosome spread analysis. These studies will seek a molecular understanding of how ATR deletion leads to premature aging.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project (R01)
Project #
5R01AG027376-04
Application #
7632093
Study Section
Cellular Mechanisms in Aging and Development Study Section (CMAD)
Program Officer
Velazquez, Jose M
Project Start
2006-07-15
Project End
2011-06-30
Budget Start
2009-07-01
Budget End
2010-06-30
Support Year
4
Fiscal Year
2009
Total Cost
$308,971
Indirect Cost
Name
University of Pennsylvania
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Shastri, Nishita; Tsai, Yu-Chen; Hile, Suzanne et al. (2018) Genome-wide Identification of Structure-Forming Repeats as Principal Sites of Fork Collapse upon ATR Inhibition. Mol Cell 72:222-238.e11
Kim, Hyoung; George, Erin; Ragland, Ryan et al. (2017) Targeting the ATR/CHK1 Axis with PARP Inhibition Results in Tumor Regression in BRCA-Mutant Ovarian Cancer Models. Clin Cancer Res 23:3097-3108
Enriquez-Rios, Vanessa; Dumitrache, Lavinia C; Downing, Susanna M et al. (2017) DNA-PKcs, ATM, and ATR Interplay Maintains Genome Integrity during Neurogenesis. J Neurosci 37:893-905
George, Erin; Kim, Hyoung; Krepler, Clemens et al. (2017) A patient-derived-xenograft platform to study BRCA-deficient ovarian cancers. JCI Insight 2:e89760
Lee, Youngsoo; Brown, Eric J; Chang, Sandy et al. (2014) Pot1a prevents telomere dysfunction and ATM-dependent neuronal loss. J Neurosci 34:7836-44
Brown, Adam D; Sager, Brian W; Gorthi, Aparna et al. (2014) ATR suppresses endogenous DNA damage and allows completion of homologous recombination repair. PLoS One 9:e91222
Anastassiadis, Theonie; Brown, Eric J (2014) Wild-type RAS: keeping mutant RAS in CHK. Cancer Cell 25:137-8
Yamane, Arito; Robbiani, Davide F; Resch, Wolfgang et al. (2013) RPA accumulation during class switch recombination represents 5'-3' DNA-end resection during the S-G2/M phase of the cell cycle. Cell Rep 3:138-47
Royo, Hélène; Prosser, Haydn; Ruzankina, Yaroslava et al. (2013) ATR acts stage specifically to regulate multiple aspects of mammalian meiotic silencing. Genes Dev 27:1484-94
Ragland, Ryan L; Patel, Sima; Rivard, Rebecca S et al. (2013) RNF4 and PLK1 are required for replication fork collapse in ATR-deficient cells. Genes Dev 27:2259-73

Showing the most recent 10 out of 21 publications