Cargando…

Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer

BACKGROUND: Increased epigenetic age acceleration (EAA) in survivors of childhood cancer is associated with specific treatment exposures, unfavorable health behaviors, and presence of certain chronic health conditions. To better understand inter-individual variability, we investigated the genetic ba...

Descripción completa

Detalles Bibliográficos
Autores principales: Dong, Qian, Song, Nan, Qin, Na, Chen, Cheng, Li, Zhenghong, Sun, Xiaojun, Easton, John, Mulder, Heather, Plyler, Emily, Neale, Geoffrey, Walker, Emily, Li, Qian, Ma, Xiaotu, Chen, Xiang, Huang, I-Chan, Yasui, Yutaka, Ness, Kirsten K., Zhang, Jinghui, Hudson, Melissa M., Robison, Leslie L., Wang, Zhaoming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8939156/
https://www.ncbi.nlm.nih.gov/pubmed/35313970
http://dx.doi.org/10.1186/s13073-022-01038-6
_version_ 1784672686205566976
author Dong, Qian
Song, Nan
Qin, Na
Chen, Cheng
Li, Zhenghong
Sun, Xiaojun
Easton, John
Mulder, Heather
Plyler, Emily
Neale, Geoffrey
Walker, Emily
Li, Qian
Ma, Xiaotu
Chen, Xiang
Huang, I-Chan
Yasui, Yutaka
Ness, Kirsten K.
Zhang, Jinghui
Hudson, Melissa M.
Robison, Leslie L.
Wang, Zhaoming
author_facet Dong, Qian
Song, Nan
Qin, Na
Chen, Cheng
Li, Zhenghong
Sun, Xiaojun
Easton, John
Mulder, Heather
Plyler, Emily
Neale, Geoffrey
Walker, Emily
Li, Qian
Ma, Xiaotu
Chen, Xiang
Huang, I-Chan
Yasui, Yutaka
Ness, Kirsten K.
Zhang, Jinghui
Hudson, Melissa M.
Robison, Leslie L.
Wang, Zhaoming
author_sort Dong, Qian
collection PubMed
description BACKGROUND: Increased epigenetic age acceleration (EAA) in survivors of childhood cancer is associated with specific treatment exposures, unfavorable health behaviors, and presence of certain chronic health conditions. To better understand inter-individual variability, we investigated the genetic basis underlying EAA. METHODS: Genome-wide association studies of EAA based on multiple epigenetic clocks (Hannum, Horvath, PhenoAge, and GrimAge) were performed. MethylationEPIC BeadChip array and whole-genome sequencing data were generated with blood-derived DNA from participants in the St. Jude Lifetime Cohort Study (discovery: 2138 pre-existing and 502 newly generated data, all survivors; exploratory: 282 community controls). Linear regression models were fit for each epigenetic age against the allelic dose of each genetic variant, adjusting for age at sampling, sex, and cancer treatment exposures. Fixed-effects meta-analysis was used to combine summary statistics from two discovery data sets. LD (Linkage disequilibrium) score regression was used to estimate single-nucleotide polymorphism (SNP)-based heritability. RESULTS: For EAA-Horvath, a genome-wide significant association was mapped to the SELP gene with the strongest SNP rs732314 (meta-GWAS: β=0.57, P=3.30×10(-11)). Moreover, the stratified analysis of the association between rs732314 and EAA-Horvath showed a substantial heterogeneity between children and adults (meta-GWAS: β=0.97 vs. 0.51, I(2)=73.1%) as well as between survivors with and without chest/abdominal/pelvic-RT exposure (β=0.64 vs. 0.31, I(2)=66.3%). For EAA-Hannum, an association was mapped to the HLA locus with the strongest SNP rs28366133 (meta-GWAS: β=0.78, P=3.78×10(-11)). There was no genome-wide significant hit for EAA-PhenoAge or EAA-GrimAge. Interestingly, among community controls, rs732314 was associated with EAA-Horvath (β=1.09, P=5.43×10(-5)), whereas rs28366133 was not associated with EAA-Hannum (β=0.21, P=0.49). The estimated heritability was 0.33 (SE=0.20) for EAA-Horvath and 0.17 (SE=0.23) for EAA-Hannum, but close to zero for EAA-PhenoAge and EAA-GrimAge. CONCLUSIONS: We identified novel genetic variants in the SELP gene and HLA region associated with EAA-Horvath and EAA-Hannum, respectively, among survivors of childhood cancer. The new genetic variants in combination with other replicated known variants can facilitate the identification of survivors at higher risk in developing accelerated aging and potentially inform drug targets for future intervention strategies among vulnerable survivors. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13073-022-01038-6.
format Online
Article
Text
id pubmed-8939156
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-89391562022-03-23 Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer Dong, Qian Song, Nan Qin, Na Chen, Cheng Li, Zhenghong Sun, Xiaojun Easton, John Mulder, Heather Plyler, Emily Neale, Geoffrey Walker, Emily Li, Qian Ma, Xiaotu Chen, Xiang Huang, I-Chan Yasui, Yutaka Ness, Kirsten K. Zhang, Jinghui Hudson, Melissa M. Robison, Leslie L. Wang, Zhaoming Genome Med Research BACKGROUND: Increased epigenetic age acceleration (EAA) in survivors of childhood cancer is associated with specific treatment exposures, unfavorable health behaviors, and presence of certain chronic health conditions. To better understand inter-individual variability, we investigated the genetic basis underlying EAA. METHODS: Genome-wide association studies of EAA based on multiple epigenetic clocks (Hannum, Horvath, PhenoAge, and GrimAge) were performed. MethylationEPIC BeadChip array and whole-genome sequencing data were generated with blood-derived DNA from participants in the St. Jude Lifetime Cohort Study (discovery: 2138 pre-existing and 502 newly generated data, all survivors; exploratory: 282 community controls). Linear regression models were fit for each epigenetic age against the allelic dose of each genetic variant, adjusting for age at sampling, sex, and cancer treatment exposures. Fixed-effects meta-analysis was used to combine summary statistics from two discovery data sets. LD (Linkage disequilibrium) score regression was used to estimate single-nucleotide polymorphism (SNP)-based heritability. RESULTS: For EAA-Horvath, a genome-wide significant association was mapped to the SELP gene with the strongest SNP rs732314 (meta-GWAS: β=0.57, P=3.30×10(-11)). Moreover, the stratified analysis of the association between rs732314 and EAA-Horvath showed a substantial heterogeneity between children and adults (meta-GWAS: β=0.97 vs. 0.51, I(2)=73.1%) as well as between survivors with and without chest/abdominal/pelvic-RT exposure (β=0.64 vs. 0.31, I(2)=66.3%). For EAA-Hannum, an association was mapped to the HLA locus with the strongest SNP rs28366133 (meta-GWAS: β=0.78, P=3.78×10(-11)). There was no genome-wide significant hit for EAA-PhenoAge or EAA-GrimAge. Interestingly, among community controls, rs732314 was associated with EAA-Horvath (β=1.09, P=5.43×10(-5)), whereas rs28366133 was not associated with EAA-Hannum (β=0.21, P=0.49). The estimated heritability was 0.33 (SE=0.20) for EAA-Horvath and 0.17 (SE=0.23) for EAA-Hannum, but close to zero for EAA-PhenoAge and EAA-GrimAge. CONCLUSIONS: We identified novel genetic variants in the SELP gene and HLA region associated with EAA-Horvath and EAA-Hannum, respectively, among survivors of childhood cancer. The new genetic variants in combination with other replicated known variants can facilitate the identification of survivors at higher risk in developing accelerated aging and potentially inform drug targets for future intervention strategies among vulnerable survivors. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13073-022-01038-6. BioMed Central 2022-03-22 /pmc/articles/PMC8939156/ /pubmed/35313970 http://dx.doi.org/10.1186/s13073-022-01038-6 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Dong, Qian
Song, Nan
Qin, Na
Chen, Cheng
Li, Zhenghong
Sun, Xiaojun
Easton, John
Mulder, Heather
Plyler, Emily
Neale, Geoffrey
Walker, Emily
Li, Qian
Ma, Xiaotu
Chen, Xiang
Huang, I-Chan
Yasui, Yutaka
Ness, Kirsten K.
Zhang, Jinghui
Hudson, Melissa M.
Robison, Leslie L.
Wang, Zhaoming
Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title_full Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title_fullStr Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title_full_unstemmed Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title_short Genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
title_sort genome-wide association studies identify novel genetic loci for epigenetic age acceleration among survivors of childhood cancer
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8939156/
https://www.ncbi.nlm.nih.gov/pubmed/35313970
http://dx.doi.org/10.1186/s13073-022-01038-6
work_keys_str_mv AT dongqian genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT songnan genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT qinna genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT chencheng genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT lizhenghong genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT sunxiaojun genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT eastonjohn genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT mulderheather genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT plyleremily genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT nealegeoffrey genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT walkeremily genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT liqian genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT maxiaotu genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT chenxiang genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT huangichan genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT yasuiyutaka genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT nesskirstenk genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT zhangjinghui genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT hudsonmelissam genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT robisonlesliel genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer
AT wangzhaoming genomewideassociationstudiesidentifynovelgeneticlociforepigeneticageaccelerationamongsurvivorsofchildhoodcancer