Abstract

BackgroundDNA methylation changes that accrue in the stem cell pool of an adult tissue in line with the cumulative number of cell divisions may contribute to the observed variation in cancer risk among tissues and individuals. Thus, the construction of epigenetic “mitotic” clocks that can measure the lifetime number of stem cell divisions is of paramount interest.MethodsBuilding upon a dynamic model of DNA methylation gain in unmethylated CpG-rich regions, we here derive a novel mitotic clock (“epiTOC2”) that can directly estimate the cumulative number of stem cell divisions in a tissue. We compare epiTOC2 to a different mitotic model, based on hypomethylation at solo-WCGW sites (“HypoClock”), in terms of their ability to measure mitotic age of normal adult tissues and predict cancer risk.ResultsUsing epiTOC2, we estimate the intrinsic stem cell division rate for different normal tissue types, demonstrating excellent agreement (Pearson correlation = 0.92, R2 = 0.85, P = 3e−6) with those derived from experiment. In contrast, HypoClock’s estimates do not (Pearson correlation = 0.30, R2 = 0.09, P = 0.29). We validate these results in independent datasets profiling normal adult tissue types. While both epiTOC2 and HypoClock correctly predict an increased mitotic rate in cancer, epiTOC2 is more robust and significantly better at discriminating preneoplastic lesions characterized by chronic inflammation, a major driver of tissue turnover and cancer risk. Our data suggest that DNA methylation loss at solo-WCGWs is significant only when cells are under high replicative stress and that epiTOC2 is a better mitotic age and cancer risk prediction model for normal adult tissues.ConclusionsThese results have profound implications for our understanding of epigenetic clocks and for developing cancer risk prediction or early detection assays. We propose that measurement of DNAm at the 163 epiTOC2 CpGs in adult pre-neoplastic lesions, and potentially in serum cell-free DNA, could provide the basis for building feasible pre-diagnostic or cancer risk assays. epiTOC2 is freely available from https://doi.org/10.5281/zenodo.2632938

Highlights

  • DNA methylation changes that accrue in the stem cell pool of an adult tissue in line with the cumulative number of cell divisions may contribute to the observed variation in cancer risk among tissues and individuals

  • There is increasing evidence that the risk of neoplastic transformation of any given tissue in any given individual is a direct function of the mitotic age of the tissue, that is, cancer risk may correlate with the cumulative number of cell divisions within the underlying stem cell pool [1,2,3,4]

  • We first derived a mathematical expression relating the fraction of cells methylated at one of these CpG sites i in a given sample, to the total number of stem cell divisions (TNSC) at cell division time “t”, and to parameters reflecting the site-specific probability of de novo methylation δi and ground-state methylation βi0 (Methods, Fig. 1a)

Read more

Summary

Introduction

DNA methylation changes that accrue in the stem cell pool of an adult tissue in line with the cumulative number of cell divisions may contribute to the observed variation in cancer risk among tissues and individuals. There is increasing evidence that the risk of neoplastic transformation of any given tissue in any given individual is a direct function of the mitotic age of the tissue, that is, cancer risk may correlate with the cumulative number of cell divisions within the underlying (adult) stem cell pool [1,2,3,4]. Given the appeal and importance of such a mitotic stem cell model of oncogenesis, there is increased interest to construct molecular “mitotic-like” clocks that can yield proxies for the cumulative number of stem cell divisions in the tissue of any given individual, which may serve to predict the risk of neoplastic transformation [11,12,13,14,15,16,17,18,19,20,21]. Solo-WCGWs may be subject to substantial confounding by cell type heterogeneity [41, 42], which may preclude a direct interpretation in terms of DNAm changes that accrue because of cell division

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call