Abstract

Advanced donor age is a risk factor for poor survival following lung transplantation. However, recent work identifying epigenetic determinants of aging has shown that biologic age may not always reflect chronologic age and that stressors can accelerate biologic aging. We hypothesized that lung allografts that experienced primary graft dysfunction (PGD), characterized by poor oxygenation in the first three post-transplant days, would have increased biologic age. We cultured airway epithelial cells isolated by transbronchial brush at 1-year bronchoscopies from 13 subjects with severe PGD and 15 controls matched on age and transplant indication. We measured epigenetic age using the Horvath epigenetic clock. Linear models were used to determine the association of airway epigenetic age with chronologic ages and PGD status, adjusted for recipient PGD risk factors. Survival models assessed the association with chronic lung allograft dysfunction (CLAD) or death. Distributions of promoter methylation within pathways were compared between groups. DNA methyltransferase (DNMT) activity was quantified in airway epithelial cells under hypoxic or normoxic conditions. Airway epigenetic age appeared younger but was strongly associated with the age of the allograft (slope 0.38 per year, 95% CI 0.27–0.48). There was no correlation between epigenetic age and recipient age (P = 0.96). Epigenetic age was 6.5 years greater (95% CI 1.7–11.2) in subjects who had experienced PGD, and this effect remained significant after adjusting for donor and recipient characteristics (P = 0.03). Epigenetic age was not associated with CLAD-free survival risk (P = 0.11). Analysis of differential methylation of promoters of key biologic pathways revealed hypomethylation in regions related to hypoxia, inflammation, and metabolism-associated pathways. Accordingly, airway epithelial cells cultured in hypoxic conditions showed suppressed DNMT activity. While airway methylation age was primarily determined by donor chronologic age, early injury in the form of PGD was associated with increased allograft epigenetic age. These data show how PGD might suppress key promoter methylation resulting in long-term impacts on the allograft.

Highlights

  • Advanced donor age increases the risk of allograft failure in most types of solid organ transplantations [1]

  • This epigenetic clock is thought to be driven by the epigenetic maintenance system, in which DNA methyltransferases (DNMTs) maintain CpG methylation and compete with active demethylation driven by ten-eleven translocation (TET) methylcytosine dioxygenases and passive demethylation [6, 7]

  • In lung recipients with severe Primary graft dysfunction (PGD), we found accelerated epigenetic aging of allograft epithelial cells obtained 1 year later

Read more

Summary

Introduction

Advanced donor age increases the risk of allograft failure in most types of solid organ transplantations [1]. Cytosine-phosphate-guanine (CpG) methylation in gene promoters and other regions selectively suppresses transcription, influencing cell phenotypes Chronological age is another major driver of these DNA methylation patterns. Horvath described an epigenetic clock based on 353 representative CpG sites that are hyper- or hypomethylated in association with age across multiple tissue types [6]. This epigenetic clock is thought to be driven by the epigenetic maintenance system, in which DNA methyltransferases (DNMTs) maintain CpG methylation and compete with active demethylation driven by ten-eleven translocation (TET) methylcytosine dioxygenases and passive demethylation [6, 7]. DNMT activity is targetable with existing drugs that could potentially influence aging phenotypes and longevity [11]

Methods
Results
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