- PROJECT 3 DNA damage and its sequelae are now recognized as major drivers of aging. However, many questions remain regarding the nature of age-promoting DNA damage, the role of mutations in non-cancer age-related pathology, and how genomic damage and its sequelae drive age-related phenotypes and pathologies. Project 3 will determine -- in depth, breadth and primarily at the level of cellular responses -- how genes that participate in genome maintenance assure tissue health and ultimately longevity. The primary (but not the only) consequence-generating cell fate responses are cellular senescence and apoptosis. The extent to which cells in aged organisms acquire mutations and/or experience these cellular consequences due to DNA damage are poorly understood. We will explore the relationship between DNA damage-induced cell fates and mutations (with Project 2), molecular consequences of DNA damage (with Project 1), and human gene variants implicated in longevity (with Project 4). Specifically, we will determine cell fate responses of cells carrying genome maintenance genotypes associated with premature and delayed aging identified by Projects 1 and 4, focusing on multifunctional repair genes that participate in repair processes important for relieving replication or transcriptional stress. We will interrogate cellular senescence and apoptosis (and other forms of cell death), and markers of cell function. With Project 2, we will determine whether some genomic changes (e.g., aneuploidy, INDELS) preferentially elicit one cell fate over another. We will use simple and complex culture systems, focusing primarily on the skin, brain and liver and use single cell analyses to determine how different types of DNA damage affect cellular heterogeneity and variability. We will also determine the cellular responses to genotoxic stress in the backgrounds of genome maintenance genotypes and in response to genomic damage in wild-type and mutant mouse models, and naturally aged mice. We will exploit our mouse model in which senescent cells can be eliminated, or use pharmacological means to eliminate senescent cells, to determine how modifying this cell fate affects hallmarks of aging. Finally, once we have a candidate list of human gene variants associated with aging and longevity from Project 4, we will use human embryonic stem cells harboring these variants to assess the cells and their differentiated progeny for their cell fate responses to genomics stressors. Together, this Project will allow us to integrate the genomic and cellular responses to DNA damage during natural, accelerated and delayed aging in mouse and human cells, and provide mechanistic bases for predicting the efficacy of interventions into aging phenotypes and pathologies.

Public Health Relevance

- PROJECT 3 Optimal health and longevity require efficient and accurate maintenance of the genome, which is constantly exposed to damage from endogenous and exogenous sources. When one or more of the several genome maintenance systems fail, or genomic damage overwhelms genome maintenance systems, both life span and health span are compromised -- often as a consequence of how cells respond to failures in genome maintenance. This project will determine precisely how cells respond to different challenges to genome maintenance during aging, using both human cells and mouse models. It will also explore the possibilities of improving health span at least in mice and complex human cells in culture by modulating cell fate responses to genomic damage.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Program Projects (P01)
Project #
5P01AG017242-25
Application #
9964617
Study Section
Special Emphasis Panel (ZAG1)
Project Start
Project End
Budget Start
2020-05-01
Budget End
2021-04-30
Support Year
25
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Albert Einstein College of Medicine
Department
Type
DUNS #
081266487
City
Bronx
State
NY
Country
United States
Zip Code
10461
Hébert, Jean M; Vijg, Jan (2018) Cell Replacement to Reverse Brain Aging: Challenges, Pitfalls, and Opportunities. Trends Neurosci 41:267-279
Lau, Cia-Hin; Suh, Yousin (2018) In vivo epigenome editing and transcriptional modulation using CRISPR technology. Transgenic Res 27:489-509
Wiley, Christopher D; Schaum, Nicholas; Alimirah, Fatouma et al. (2018) Small-molecule MDM2 antagonists attenuate the senescence-associated secretory phenotype. Sci Rep 8:2410
Quispe-Tintaya, Wilber; Lee, Moonsook; Dong, Xiao et al. (2018) Bleomycin-induced genome structural variations in normal, non-tumor cells. Sci Rep 8:16523
Yu, Bo; Dong, Xiao; Gravina, Silvia et al. (2017) Genome-wide, Single-Cell DNA Methylomics Reveals Increased Non-CpG Methylation during Human Oocyte Maturation. Stem Cell Reports 9:397-407
Vijg, Jan; Dong, Xiao; Zhang, Lei (2017) A high-fidelity method for genomic sequencing of single somatic cells reveals a very high mutational burden. Exp Biol Med (Maywood) 242:1318-1324
Ogrodnik, Mikolaj; Miwa, Satomi; Tchkonia, Tamar et al. (2017) Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 8:15691
Dong, Xiao; Zhang, Lei; Milholland, Brandon et al. (2017) Accurate identification of single-nucleotide variants in whole-genome-amplified single cells. Nat Methods 14:491-493
Olivieri, Fabiola; Capri, Miriam; Bonafè, Massimiliano et al. (2017) Circulating miRNAs and miRNA shuttles as biomarkers: Perspective trajectories of healthy and unhealthy aging. Mech Ageing Dev 165:162-170
Perrott, Kevin M; Wiley, Christopher D; Desprez, Pierre-Yves et al. (2017) Apigenin suppresses the senescence-associated secretory phenotype and paracrine effects on breast cancer cells. Geroscience 39:161-173

Showing the most recent 10 out of 253 publications