ArticlePDF Available

Genetic Evidence for Causal Effects of Socioeconomic, Lifestyle, and Cardiometabolic Factors on Epigenetic-Age Acceleration

Authors:

Abstract and Figures

GrimAge acceleration (GrimAgeAccel) and PhenoAge acceleration (PhenoAgeAccel) are DNA methylation-based markers of accelerated biological aging, standing out in predicting mortality and age-related cardiometabolic morbidities. Causal risk factors for GrimAgeAccel and PhenoAgeAccel are unclear. In this study, we performed two-sample univariable and multivariable Mendelian randomization (MR) to investigate causal associations of 19 modifiable socioeconomic, lifestyle, and cardiometabolic factors with GrimAgeAccel and PhenoAgeAccel. Instrument variants representing 19 modifiable factors were extracted from genome-wide association studies (GWASs) with up to 1 million Europeans. Summary statistics for GrimAgeAccel and PhenoAgeAccel were derived from a GWAS of 34,710 Europeans. We identified 12 and eight factors causally associated with GrimAgeAccel and PhenoAgeAccel, respectively. Smoking was the strongest risk factor (β [SE]: 1.299 [0.107] year) for GrimAgeAccel, followed by higher alcohol intake, higher waist circumference, daytime napping, higher body fat percentage, higher body mass index, higher C-reactive protein, higher triglycerides, childhood obesity, and type 2 diabetes; whereas education was the strongest protective factor (β [SE]: -1.143 [0.121] year), followed by household income. Furthermore, higher waist circumference (β [SE]: 0.850 [0.269] year) and education (β [SE]: -0.718 [0.151] year) were the leading causal risk and protective factors for PhenoAgeAccel, respectively. Sensitivity analyses strengthened the robustness of these causal associations. Multivariable MR analyses further demonstrated independent effects of the strongest risk and protective factors on GrimAgeAccel and PhenoAgeAccel, respectively. In conclusion, our findings provide novel quantitative evidence on modifiable causal risk factors for accelerated epigenetic aging, suggesting promising intervention targets against age-related morbidity and improving healthy longevity.
Content may be subject to copyright.
1083
Journals of Gerontology: Biological Sciences
cite as: J Gerontol A Biol Sci Med Sci, 2023, Vol. 78, No. 7, 1083–1091
https://doi.org/10.1093/gerona/glad078
Advance Access publication March 4, 2023
Editor’s choice
© The Author(s) 2023. Published by Oxford University Press on behalf of The Gerontological Society of America.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.
Original Article
Genetic Evidence for Causal Effects of Socioeconomic,
Lifestyle, and Cardiometabolic Factors on Epigenetic-Age
Acceleration
Lijie Kong, MD,1,2,, Chaojie Ye, MD,1,2, Yiying Wang, MD,1,2 Tianzhichao Hou, MD,1,2
JieZheng, PhD,1,2, ZhiyunZhao, MD, PhD,1,2, MianLi, MD, PhD,1,2 YuXu, MD, PhD,1,2,
JieliLu, MD, PhD,1,2, YuhongChen, MD, PhD,1,2 MinXu, MD, PhD,1,2, WeiqingWang, MD,
PhD,1,2, GuangNing, MD, PhD,1,2, YufangBi, MD, PhD,1,2 and TiangeWang, MD, PhD1,2,*,
1Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai, China. 2Shanghai National Clinical Research Center for Metabolic Diseases, Key
Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for
Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
*Address correspondence to: Tiange Wang, MD, PhD, Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases,
Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rujin 2nd Road,
Shanghai, China. E-mail: tiange.wang@shsmu.edu.cn
These authors contributed equally as first authors.
Received: September 25, 2022; Editorial Decision Date: February 22, 2023
Decision Editor: DavidLeCouteur , MBBS, FRACP, PhD
Abstract
GrimAge acceleration (GrimAgeAccel) and PhenoAge acceleration (PhenoAgeAccel) are DNA methylation-based markers of accelerated
biological aging, standing out in predicting mortality and age-related cardiometabolic morbidities. Causal risk factors for GrimAgeAccel
and PhenoAgeAccel are unclear. In this study, we performed 2-sample univariable and multivariable Mendelian randomization (MR) to
investigate causal associations of 19 modiable socioeconomic, lifestyle, and cardiometabolic factors with GrimAgeAccel and PhenoAgeAccel.
Instrument variants representing 19 modiable factors were extracted from genome-wide association studies (GWASs) with up to 1 million
Europeans. Summary statistics for GrimAgeAccel and PhenoAgeAccel were derived from a GWAS of 34710 Europeans. We identied 12 and
8 factors causally associated with GrimAgeAccel and PhenoAgeAccel, respectively. Smoking was the strongest risk factor (β [standard error
{SE}]: 1.299 [0.107] year) for GrimAgeAccel, followed by higher alcohol intake, higher waist circumference, daytime napping, higher body fat
percentage, higher body mass index, higher C-reactive protein, higher triglycerides, childhood obesity, and type 2 diabetes; whereas education
was the strongest protective factor (β [SE]: −1.143 [0.121] year), followed by household income. Furthermore, higher waist circumference (β
[SE]: 0.850 [0.269] year) and education (β [SE]: −0.718 [0.151] year) were the leading causal risk and protective factors for PhenoAgeAccel,
respectively. Sensitivity analyses strengthened the robustness of these causal associations. Multivariable MR analyses further demonstrated
independent effects of the strongest risk and protective factors on GrimAgeAccel and PhenoAgeAccel, respectively. In conclusion, our ndings
provide novel quantitative evidence on modiable causal risk factors for accelerated epigenetic aging, suggesting promising intervention targets
against age-related morbidity and improving healthy longevity.
Keywords: Causal risk factors, DNA methylation, Epigenetic aging, Mendelian randomization
Biological
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
Aging involves the gradual accumulation of a decline in multiple bio-
logical functions over time, leading to increased risks of developing
age-related diseases and mortality (1,2). Although chronological
aging is uniform and unchangeable, the rate of biological aging is
variable and modiable depending on individual genetics, envir-
onmental exposures, and health-related behaviors (3). Of several
potential types of biological age predictors (eg, epigenetic clock,
leukocyte telomere length, and transcriptomic predictors), the epi-
genetic clock composed of DNA methylation at multiple cytosine–
phosphate–guanine sites is currently the best one, as it correlates
well with age and predicts mortality across populations (4). The
epigenetic-age acceleration is the difference between chronological
age and epigenetic age and represents accelerated biological aging
(5). The second-generation epigenetic-age acceleration indicators,
namely GrimAge acceleration (GrimAgeAccel) and PhenoAge ac-
celeration (PhenoAgeAccel), have evolved to incorporate aging-
related traits, and stand out in terms of predicting mortality and
age-related cardiometabolic morbidities (6–9).
Limited observational evidence has suggested that certain
socioeconomic status (SES), lifestyle behaviors, and cardiometabolic
traits may be related to GrimAgeAccel and PhenoAgeAccel
(6,7,10,11). Whether and to what extent modiable risk factors inu-
ence GrimAgeAccel and PhenoAgeAccel, if causally established, could
shed light on potential contributors to the aging process and elucidate
promising targets for preventing age-related diseases and improving
healthy longevity (12,13). Thus far, such causal evidence isscarce.
To ll this knowledge gap, we applied Mendelian randomiza-
tion (MR) to evaluate the causal associations of 19 common modi-
able socioeconomic, lifestyle, and cardiometabolic factors with
GrimAgeAccel and PhenoAgeAccel. The MR method uses genetic vari-
ants that are robustly associated with exposure as instrumental vari-
ables (IVs) to estimate the causal effect of exposure on an outcome.
Since genetic variants are randomly allocated at conception, the MR
study is less susceptible to confounding and reverse causality than con-
ventional observational studies (14). We performed both univariable
and multivariable MR analyses to discern if modiable factors have
independent causal effects on GrimAgeAccel and PhenoAgeAccel.
Method
StudyDesign
The MR study design is shown in Figure 1. We performed
2-sample univariable and multivariable MR strictly following
the Strengthening the Reporting of Observational Studies in
Epidemiology using Mendelian Randomization (STROBE-MR)
guidelines (Supplementary Table 1) (15). To obtain unbiased esti-
mates of the causal effects, the MR analysis should adhere to three
fundamental assumptions (16): rst, the IVs are truly associated
with the exposures; second, the IVs are independent of confounders
of the relationship between exposures and outcomes (GrimAgeAccel
and PhenoAgeAccel); third, the IVs inuence the outcomes only
through the exposures, but not any direct or indirect pathways. All
data used in this MR study are publicly available. Ethical approval
and informed consent had been obtained in all original studies.
Selection Rationale and Data Sources of Genetic
Instruments
We selected 19 common modiable factors, including SES (educa-
tion and household income), lifestyle behaviors (smoking initiation,
alcohol intake, coffee consumption, daytime napping, sleep dur-
ation, and moderate-to-vigorous physical activity [MVPA]), and
cardiometabolic traits (body mass index [BMI], waist circumfer-
ence, body fat percentage [BF%], childhood obesity, type 2 diabetes,
low-density lipoprotein [LDL] cholesterol, high-density lipopro-
tein [HDL] cholesterol, triglycerides, systolic blood pressure [SBP],
diastolic blood pressure [DBP], and C-reactive protein [CRP]).
Denitions of the 19 modiable factors are shown in Supplementary
Table 2.
We extracted genetic variants for each of the modiable factors
from the most authoritative and appropriate genome-wide asso-
ciation studies (GWASs) of European ancestry (17–29), ensuring
minimum sample overlap with the GWAS of GrimAgeAccel and
PhenoAgeAccel (Supplementary Table 3). We included single nucleo-
tide polymorphisms (SNPs) robustly associated with the 19 modi-
able factors at the genome-wide signicance (p<5×10−8). To select
independent genetic variants, a stringent condition (linkage disequi-
librium threshold of r2 < 0.01) was set to minimize the inuence of
linkage disequilibrium which might bias the results of randomized
allele allocation. Where SNPs for the exposures were not available in
the GWAS summary statistics of GrimAgeAccel or PhenoAgeAccel,
we used proxies of SNPs with r2 > 0.8 as substitutes, by using the
LDproxy search on the online platform LDlink (https://ldlink.nci.
nih.gov/) (30).
Data Source for Epigenetic-Age Acceleration
The genetic associations with GrimAgeAccel and PhenoAgeAccel
were extracted from a recent GWAS meta-analysis (summary stat-
istics available at https://datashare.is.ed.ac.uk/handle/10283/3645),
which included 34710 European participants from 28 cohorts (31).
GrimAgeAccel and PhenoAgeAccel are 2-generation epigenetic-age
acceleration indicators, expressing the biological aging rate in years,
of which GrimAgeAccel is more strongly associated with mortality
than PhenoAgeAccel (11). Detailed denitions of GrimAgeAccel
and PhenoAgeAccel and data preparation in GWAS are shown in
Supplementary Table 2.
Figure 1. Study design and assumptions of the MR analysis. Assumption
1 indicates that the genetic variants proposed as instrumental variables
should be robustly associated with the exposures; assumption 2 indicates
that the used instrumental variables should not be associated with potential
confounders of the relationship between exposures and outcomes; and
assumption 3 indicates that the selected instrumental variables should
influence the outcomes only through the exposures, not via alternative
pathways. GrimAgeAccel = epigenetic-age acceleration obtained using
the GrimAge clock; HDL = high-density lipoprotein; LDL = low-density
lipoprotein; MR = Mendelian randomization; PhenoAgeAccel = epigenetic-
age acceleration obtained using the PhenoAge clock; PRESSO= pleiotropy
residual sum and outlier.
1084 Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
Table 1. MR Results of the Associations Between 19 Modifiable Factors and GrimAgeAccel
Modiable Factor
No. of
SNPs F-Statistic
Main Analysis Sensitivity Analyses
IVW Weighted Median MR-Egger MR-PRESSO
β (SE), year p Value q Value β (SE), years p Value q Value Β (SE), years p Value q Value
No. of
outliers β (SE), years p Value q Value
Socioeconomic status
Education 751 50 1.143 (0.121) 3.31E−21 3.14E20 1.129 (0.172) 5.47E−11 5.20E10 −0.829 (0.452) 6.70E02 2.93E01 4 1.116 (0.116) 6.54E-21 6.21E-20
Household income 51 41 0.774 (0.263) 3.24E−03 8.79E03 −0.710 (0.378) 6.03E02 1.43E01 −1.298 (1.322) 3.31E01 6.29E01 0 0.764 (0.255) 4.14E-03 1.12E-02
Lifestyle behavior
Smoking initiation 299 46 1.299 (0.107) 4.90E−34 9.31E33 1.257 (0.157) 1.42E−15 2.70E14 1.321 (0.446) 3.00E−03 2.85E02 0 1.276 (0.103) 7.63E-29 1.45E-27
Alcohol intake 80 66 0.899 (0.361) 1.30E−02 2.47E02 0.380 (0.567) 5.02E01 5.96E01 0.481 (0.738) 5.16E01 7.56E01 0 0.967 (0.357) 8.00E-03 1.69E-02
Coffee consumption 12 176 0.003 (0.005) 5.89E01 6.22E01 −0.003 (0.006) 6.15E01 6.87E01 −0.008 (0.010) 4.64E01 7.56E01 0 0.003 (0.005) 6.00E-01 6.33E-01
Sleep
Daytime napping 115 47 0.805 (0.355) 2.30E−02 3.64E02 0.524 (0.514) 3.08E01 4.40E01 0.411 (1.235) 7.40E01 8.27E01 0 0.813 (0.346) 2.00E-02 3.17E-02
Sleep duration 77 40 −0.301 (0.266) 2.57E01 3.49E01 −0.403 (0.400) 3.13E01 4.40E01 −0.077 (1.017) 9.40E01 9.40E01 0 −0.275 (0.261) 2.94E-01 3.72E-01
MVPA 6 40 −0.186 (0.903) 8.37E01 8.37E01 −0.036 (1.198) 9.76E01 9.76E01 −9.279 (5.557) 1.70E01 5.38E01 0 −0.186 (0.763) 8.17E-01 8.17E-01
Cardiometabolic trait
Adiposity
BMI 941 59 0.592 (0.079) 9.16E−14 5.80E13 0.490 (0.124) 7.92E−05 3.76E04 0.790 (0.231) 1.00E−03 1.90E02 2 0.595 (0.078) 6.30E-14 3.99E-13
Waist circumference 44 53 0.815 (0.184) 9.26E−06 2.93E05 0.776 (0.272) 4.00E−03 1.52E02 0.913 (0.504) 7.70E02 2.93E01 0 0.815 (0.184) 6.31E-05 2.00E-04
BF% 641 50 0.748 (0.120) 4.12E−10 1.96E09 0.740 (0.182) 4.87E−05 3.08E04 0.843 (0.400) 3.60E−02 2.28E01 3 0.796 (0.115) 1.19E-11 5.65E-11
Childhood obesity 5 43 0.200 (0.075) 7.00E−03 1.48E02 0.142 (0.098) 1.46E01 2.77E01 0.789 (0.554) 2.49E01 5.91E01 0 0.204 (0.057) 1.60E-02 2.76E-02
Type 2 diabetes 232 62 0.095 (0.041) 2.00E−02 3.45E02 0.084 (0.071) 2.36E01 4.08E01 0.043 (0.092) 6.44E01 7.65E01 0 0.100 (0.040) 1.30E-02 2.47E-02
Lipids
LDL cholesterol 145 146 −0.094 (0.096) 3.27E01 3.88E01 −0.064 (0.163) 6.95E01 7.34E01 0.016 (0.142) 9.08E01 9.40E01 1 −0.069 (0.094) 4.63E-01 5.39E-01
HDL cholesterol 223 138 −0.181 (0.095) 5.60E02 8.18E02 −0.267 (0.138) 5.30E02 1.43E01 −0.078 (0.162) 6.32E01 7.65E01 2 −0.175 (0.090) 5.40E-02 7.89E-02
Triglycerides 173 116 0.249 (0.091) 6.00E−03 1.43E02 0.239 (0.144) 9.60E02 2.03E01 0.152 (0.152) 3.19E01 6.29E01 0 0.254 (0.091) 6.00E-03 1.43E-02
Blood pressure
SBP 222 52 0.013 (0.012) 2.79E01 3.53E01 0.015 (0.018) 4.21E01 5.33E01 0.029 (0.049) 5.57E01 7.56E01 0 0.013 (0.012) 2.82E-01 3.72E-01
DBP 264 50 0.011 (0.019) 5.65E01 6.22E01 0.027 (0.027) 3.24E01 4.40E01 0.083 (0.066) 2.13E01 5.78E01 0 0.013 (0.018) 4.82E-01 5.39E-01
CRP 299 170 0.345 (0.073) 2.31E−06 8.78E06 0.245 (0.107) 2.20E−02 6.97E02 0.060 (0.100) 5.53E01 7.56E01 1 0.348 (0.071) 1.64E-06 6.23E-06
Notes: GrimAgeAccel represents epigenetic-age acceleration obtained using the GrimAge clock. The q value represents the false discovery rate (FDR) − adjusted p value. β represents the associations of each modiable
factor with GrimAgeAccel, and the unit for each risk factor is: 1-SD increase in years of schooling; 1-SD increase in household income; Ever smoked regularly compared to never smoked; 1-SD increase in log-transformed
alcoholic drinks per week; 1%-change in coffee consumption; 1-unit increase in napping category (never, sometimes, usually); 1-h/d increase in sleep duration; 1-SD increase in MET-min/wk of MVPA; 1-SD increase in BMI;
1-SD increase in waist circumference; 1-SD increase in BF%; 1-unit increase in log-transformed odds of childhood obesity; 1-unit increase in log-transformed odds of type 2 diabetes; 1-SD increase in LDL cholesterol; 1-SD
increase in HDL cholesterol; 1-SD increase in triglycerides; 1-mmHg increase in SBP; 1-mmHg increase in DBP; 1-SD increase in serum CRP levels. BF%=body fat percentage; BMI=body mass index; CRP=C-reactive
protein; DBP=diastolic blood pressure; HDL=high-density lipoprotein; IVW=Inverse-variance weighted; LDL=low-density lipoprotein; MVPA=moderate-to-vigorous physical activity; MR=Mendelian randomization;
MR-PRESSO=Mendelian randomization pleiotropy residual sum and outlier; No=number; SBP=systolic blood pressure; SD=standard deviation; SNP=single nucleotide polymorphism.
Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7 1085
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
Statistical Analyses
In the main analysis, we used the inverse-variance weighted (IVW)
method to determine MR causal estimates (β coefcients with
standard errors [SEs]) for associations of each modiable factor
with GrimAgeAccel and PhenoAgeAccel. The IVW combines the
Wald ratio estimates of every single SNP in the set of IVs into one
causal estimate using the random-effects meta-analysis approach
(32). To evaluate the robustness of the IVW estimates under different
assumptions and to detect possible pleiotropy, we performed 3 sen-
sitivity analyses, including the MR weighted median, the MR-Egger,
and the MR pleiotropy residual sum and outlier (MR-PRESSO)
methods (Supplementary Table 4). The weighted median method
selects the median MR estimate as the causal estimate and provides
a consistent causal estimate if over 50% of the weight in the analysis
was derived from valid IVs (33). The MR-Egger method which al-
lows the intercept to be freely estimated as an indicator of pleiotropy
was used to identify and adjust for the potential directional pleio-
tropic bias but has limited precision (34). The MR-PRESSO method
detects outlying SNPs that are potentially horizontally pleiotropic
and evaluates whether the exclusion of outlying SNPs inuences the
causal estimates under the assumption that the largest group of can-
didate instruments with similar estimates is the group of valid IVs
(35). We evaluated the heterogeneity for the IVW estimates using
the Cochran’s Q test (36) and identied the horizontal pleiotropy
based on the p value for the intercept in the MR-Egger model (34).
The F-statistics were calculated to measure the strength of IVs,
with larger F-statistics indicating more valid IVs. Afalse discovery
rate (FDR) method was used to correct results for multiple testing,
and FDR q values were provided. In this study, strong causal evi-
dence was dened as an association supported by the main analysis
(FDR q value < 0.05) and at least 1 sensitivity analysis. Suggestive
causal evidence was dened as a suggestive association with p < .05
and FDR q value ≥ 0.05 in the main analysis. Null causal evidence
was dened as no statistically signicant association revealed from
the main analysis (p ≥ .05). Given that the phenotype of GrimAge
is partly determined by smoking, we also conducted leave-one-out
analyses to evaluate the inuence of individual genetic variants of
smoking initiation on the association with GrimAgeAccel.
We further conducted multivariable MR analyses to assess
whether the causal effects of the strongest protective factor and the
strongest risk factor on GrimAgeAccel and PhenoAgeAccel were in-
dependent of other exposure factors (37). Taking into account the ef-
fect size and signicance of causal associations, we selected exposure
factors with β coefcients >0.5 and p < .05 in the main analysis as
covariates in the adjustmentmodels.
Moreover, we performed bidirectional MR analyses to investigate
potential reverse causality from GrimAgeAccel and PhenoAgeAccel to
these modiable socioeconomic, lifestyle, and cardiometabolic factors.
The two-sample MR analyses were conducted with the R pack-
ages “TwoSampleMR” and “MRPRESSO,” and the FDR q values
were estimated using the R package “fdrtool,” in R software (version
4.1.1; R Foundation for Statistical Computing, Vienna, Austria).
Results
Univariable MR Estimates for the Causal Effects of
19 Modifiable Factors on GrimAgeAccel
Ten out of the 19 risk factors showed strong associations with in-
creased GrimAgeAccel after FDR adjustment for multiple com-
parisons (Table 1). Smoking initiation was the strongest risk factor
(β [SE]: 1.299 [0.107] year) for GrimAgeAccel, followed by higher
alcohol intake (β [SE] per 1-standard deviation [SD] increase: 0.899
[0.361] year), higher waist circumference (β [SE] per 1-SD increase:
0.815 [0.184] year), daytime napping (β [SE]: 0.805 [0.355] year),
higher BF% (β [SE] per 1-SD increase: 0.748 [0.120] year), higher
BMI (β [SE] per 1-SD increase: 0.592 [0.079] year), higher CRP (β
[SE] per 1-SD increase: 0.345 [0.073] year), higher triglycerides (β
[SE] per 1-SD increase: 0.249 [0.091] year), childhood obesity (β
[SE]: 0.200 [0.075] year), and type 2 diabetes (β [SE]: 0.095 [0.041]
year; Figure 2A). By contrast, education in years of schooling (β [SE]
per 1-SD increase: −1.143 [0.121] year) and household income (β
[SE] per 1-SD increase: −0.774 [0.263] year) were protective fac-
tors associated with decreased GrimAgeAccel. There was little evi-
dence to support a causal association of other modiable factors
with GrimAgeAccel.
Associations of the above 12 modiable factors with
GrimAgeAccel were robust across sensitivity analyses with con-
sistent effect directions and p < .05 in at least one sensitivity analysis.
The mean F-statistics for the genetic instruments were 40 or greater,
indicating the limited potential for weak instrument bias (Table 1).
The MR-Egger intercept tests indicated potential pleiotropy for CRP
(pintercept < .05; Supplementary Table 5). Cochran’s Q test showed
possible heterogeneity for education, alcohol intake, BMI, BF%,
type 2 diabetes, LDL cholesterol, HDL cholesterol, and CRP (ph <
.05; Supplementary Table 5). With the exclusion of outlying SNPs,
the MR-PRESSO analysis showed consistent results with the IVW
analysis (Table 1). Leave-one-out analysis revealed that no single
SNP drove the MR estimate of smoking initiation on GrimAgeAccel
(Supplementary Figure 1).
For the reverse causality from GrimAgeAccel to modiable fac-
tors, genetically determined each 1-year increased GrimAgeAccel
was associated with lower CRP (β [SE]: −0.042 [0.006] SD), lower
BMI (β [SE]: −0.032 [0.006] SD), and higher alcohol intake (β [SE]:
0.018 [0.007] SD; Supplementary Table 6).
Figure 2. Overview of the findings on associations of 19 modifiable factors
with GrimAgeAccel and PhenoAgeAccel. (A) Causal associations between 19
modifiable factors and GrimAgeAccel; (B) Causal associations between 19
modifiable factors and PhenoAgeAccel. GrimAgeAccel represents epigenetic-
age acceleration obtained using the GrimAge clock; PhenoAgeAccel
represents epigenetic-age acceleration obtained using the PhenoAge
clock; β represents the effect of each modifiable factor on epigenetic-age
acceleration. Red box indicates a strong association with p value of <.05 and
FDR q value < 0.05. Blue box indicates a suggestive association with p value
of <.05 and FDR q value of ≥0.05. Gray box indicates null association with p
value of ≥.05. The IVW method was used for the main analysis. Sensitivity
analyses included the MR-PRESSO, the MR-WM, and the MR-Egger methods.
BF%=body fat percentage; BMI=body mass index; CRP=C-reactive protein;
DBP=diastolic blood pressure; FDR=false discovery rate; HDL=high-density
lipoprotein; IVW=inverse-variance weighted; LDL=low-density lipoprotein;
MR = Mendelian randomization; MVPA = moderate-to-vigorous physical
activity; PRESSO=pleiotropy residual sum and outlier; SBP=systolic blood
pressure; SD=standard deviation; WM=weighted median.
1086 Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
Table 2. MR Results of the Associations Between 19 Modifiable Factors and PhenoAgeAccel
Modiable factor
No. of
SNP F-statistic
Main analysis Sensitivity analyses
IVW Weighted median MR-Egger MR-PRESSO
β (SE), year p Value q Value β (SE), year p Value q Value β (SE), year p Value q Value
No. of
outlier β (SE), year p Value q Value
Socioeconomic factor
Education 751 50 0.718 (0.151) 1.83E-06 1.74E-05 0.633 (0.225) 5.00E-03 3.17E-02 −0.361 (0.565) 5.23E-01 9.03E-01 3 0.689 (0.145) 2.52E-06 2.39E-05
Household income 51 41 −0.565 (0.367) 1.24E-01 2.06E-01 −0.626 (0.466) 1.78E-01 3.64E-01 −1.340 (1.862) 4.75E-01 9.03E-01 0 −0.620 (0.360) 9.12E-02 1.46E-01
Lifestyle behavior
Smoking initiation 299 46 0.519 (0.142) 2.43E-04 9.46E-04 0.404 (0.205) 4.90E-02 2.33E-01 1.222 (0.589) 3.90E-02 2.53E-01 1 0.545 (0.135) 7.20E-05 3.42E-04
Alcohol intake 80 66 0.669 (0.441) 1.30E-01 2.06E-01 1.134 (0.656) 8.40E-02 2.66E-01 1.693 (0.923) 7.00E-02 2.66E-01 0 0.737 (0.432) 9.20E-02 1.46E-01
Coffee consumption 12 176 0.004 (0.008) 6.00E-01 7.30E-01 0.002 (0.007) 7.73E-01 9.79E-01 -0.003 (0.015) 8.61E-01 9.27E-01 0 0.004 (0.008) 6.10E-01 7.03E-01
Sleep
Daytime napping 115 47 0.115 (0.500) 8.18E-01 8.18E-01 0.014 (0.665) 9.84E-01 9.91E-01 0.152 (1.742) 9.31E-01 9.31E-01 0 0.083 (0.488) 8.66E-01 8.66E-01
Sleep duration 77 40 −0.472 (0.357) 1.87E-01 2.73E-01 −0.526 (0.462) 2.55E-01 4.04E-01 2.771 (1.324) 4.00E-02 2.53E-01 0 −0.319 (0.357) 3.75E-01 5.09E-01
MVPA 6 40 0.368 (1.138) 7.47E-01 7.89E-01 0.262 (1.347) 8.46E-01 9.91E-01 −2.565 (6.947) 7.31E-01 9.27E-01 0 0.368 (0.649) 5.96E-01 7.03E-01
Cardiometabolic trait
Adiposity
BMI 941 59 0.586 (0.102) 1.08E-08 2.05E-07 0.680 (0.170) 6.33E-05 1.20E-03 0.510 (0.298) 8.70E-02 2.76E-01 4 0.605 (0.099) 1.53E-09 2.91E-08
Waist circumference 44 53 0.850 (0.269) 2.00E-03 6.33E-03 0.632 (0.339) 6.20E-02 2.36E-01 0.739 (0.740) 3.24E-01 7.43E-01 1 0.771 (0.249) 3.00E-03 9.50E-03
BF% 641 50 0.711 (0.152) 2.88E-06 1.82E-05 0.734 (0.227) 1.00E-03 9.50E-03 1.666 (0.509) 1.00E-03 1.90E-02 4 0.664 (0.145) 5.75E-06 3.64E-05
Childhood obesity 5 43 0.229 (0.095) 1.60E-02 3.80E-02 0.154 (0.123) 2.11E-01 3.64E-01 0.140 (0.724) 8.59E-01 9.27E-01 0 0.223 (0.071) 2.50E-02 5.28E-02
Type 2 diabetes 232 62 0.125 (0.051) 1.40E-02 3.80E-02 0.102 (0.080) 2.05E-01 3.64E-01 0.056 (0.116) 6.28E-01 9.18E-01 0 0.120 (0.049) 1.60E-02 3.80E-02
Lipids
LDL cholesterol 145 146 0.055 (0.123) 6.53E-01 7.30E-01 −0.002 (0.174) 9.91E-01 9.91E-01 0.169 (0.181) 3.52E-01 7.43E-01 1 0.059 (0.123) 6.33E-01 7.03E-01
HDL cholesterol 223 138 −0.127 (0.109) 2.43E-01 3.30E-01 0.068 (0.166) 6.84E-01 9.28E-01 0.103 (0.187) 5.83E-01 9.18E-01 0 −0.121 (0.109) 2.67E-01 3.90E-01
Triglycerides 173 116 −0.058 (0.121) 6.35E-01 7.30E-01 0.009 (0.196) 9.64E-01 9.91E-01 −0.202 (0.203) 3.20E-01 7.43E-01 0 −0.052 (0.121) 6.66E-01 7.03E-01
Blood pressure
SBP 222 52 0.032 (0.016) 5.30E-02 1.01E-01 0.033 (0.023) 1.49E-01 3.56E-01 −0.013 (0.065) 8.40E-01 9.27E-01 1 0.038 (0.016) 1.50E-02 3.80E-02
DBP 264 50 0.049 (0.024) 3.70E-02 7.81E-02 0.048 (0.034) 1.50E-01 3.56E-01 0.161 (0.084) 5.50E-02 2.61E-01 0 0.042 (0.023) 6.90E-02 1.31E-01
CRP 298 170 0.349 (0.095) 2.49E-04 9.46E-04 0.061 (0.150) 6.84E-01 9.28E-01 0.020 (0.132) 8.78E-01 9.27E-01 1 0.362 (0.093) 1.16E-04 4.41E-04
Notes: PhenoAgeAccel represents epigenetic-age acceleration obtained using the PhenoAge clock; The q value represents the false discovery rate (FDR)adjusted p value. β represents the associations of each modiable
factor with PhenoAgeAccel, and the unit for each risk factor is: 1-SD increase in years of educational attainment; 1-SD increase in household income; Ever smoked regularly compared to never smoked; 1-SD increase in
log-transformed alcoholic drinks per week; 1%-change in coffee consumption; 1-unit increase in napping category (never, sometimes, usually); 1-h/d increase in sleep duration; 1-SD increase in MET-min/wk of MVPA; 1-SD
increase in BMI; 1-SD increase in waist circumference; 1-SD increase in BF%; 1-unit increase in log-transformed odds of childhood obesity; 1-unit increase in log-transformed odds of type 2 diabetes; 1-SD increase in LDL
cholesterol; 1-SD increase in HDL cholesterol; 1-SD increase in triglycerides; 1-mmHg increase in SBP; 1-mmHg increase in DBP; 1-SD increase in serum CRP levels. BF%=body fat percentage; BMI=body mass index;
CRP=C-reactive protein; DBP=diastolic blood pressure; HDL=high-density lipoprotein; IVW=inverse-variance weighted; LDL=low-density lipoprotein; MVPA=moderate-to-vigorous physical activity; MR=Mendel-
ian randomization; MR-PRESSO=Mendelian randomization pleiotropy residual sum and outlier; No=number; SBP=systolic blood pressure; SD=standard deviation; SNP=single nucleotide polymorphism.
Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7 1087
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
Univariable MR Estimates for the Causal Effects of
19 Modifiable Factors on PhenoAgeAccel
Eight modiable factors showed strong associations with
PhenoAgeAccel after FDR adjustment for multiple comparisons
(Table 2). Higher waist circumference was the strongest risk factor
(β [SE] per 1-SD increase: 0.850 [0.269] year) for PhenoAgeAccel,
followed by higher BF% (β [SE] per 1-SD increase: 0.711 [0.152]
year), higher BMI (β [SE] per 1-SD increase: 0.586 [0.102] year),
smoking initiation (β [SE]: 0.519 [0.142] year), higher CRP (β [SE]
per 1-SD increase: 0.349 [0.095] year), childhood obesity (β [SE]:
0.229 [0.095] year), and type 2 diabetes (β [SE]: 0.125 [0.051] year;
Figure 2B). Whereas the genetically predicted higher education in
years of schooling was associated with decreased PhenoAgeAccel
(β [SE] per 1-SD increase: −0.718 [0.151] year). Suggestive caus-
ality was identied for the association between genetically predicted
higher DBP and increased PhenoAgeAccel (β [SE] per 1-mmHg in-
crease: 0.049 [0.024] year). No signicant association was observed
with PhenoAgeAccel for the other modiable factors.
Associations for the above eight modiable factors were ro-
bust in sensitivity analyses with consistent effect directions and p
< .05 in at least one sensitivity analysis. The mean F-statistics for
the genetic instruments were above 40, indicating the limited po-
tential for weak instrument bias (Table 2). There was no evidence
of pleiotropy for these risk factors except for CRP (pintercept < .05;
Supplementary Table 7). Potential heterogeneity was observed for
education, smoking initiation, BMI, waist circumference, BF%, type
2 diabetes, and CRP (ph < .05). One to four outliers were detected in
the MR-PRESSO analyses; however, the associations remained con-
sistent after the removal of these outliers (Table 2). For SBP, after
excluding the outlying SNP rs62523863, the MR-PRESSO analysis
revealed a potentially positive association between genetically pre-
dicted higher SBP and PhenoAgeAccel (β [SE] per 1-mmHg increase:
0.038 [0.016]year).
For the reverse causality from PhenoAgeAccel to modiable fac-
tors, genetically determined each 1-year increased PhenoAgeAccel
was associated with decreased napping (β [SE]: −0.004 [0.002]
unit) and sleep duration (β [SE]: −0.004 [0.002] h/d; Supplementary
Table8).
Multivariable MR Estimates for the Independent
Effects of the Strongest Protective and Risk Factors
on GrimAgeAccel and PhenoAgeAccel
In multivariable MR analyses, the inverse association between edu-
cation (the strongest protective factor) and GrimAgeAccel remained
signicant with adjustment for household income, smoking initi-
ation, alcohol intake, waist circumference, daytime napping, BF%,
or BMI (Figure 3A, Supplementary Table 9). Likewise, the positive
association between smoking initiation (the strongest risk factor) and
GrimAgeAccel persisted after adjusting for each of these covariates.
All covariates in multivariable MR analyses were selected based on
their considerable effects on GrimAgeAccel in terms of effect size
(β coefcients >0.5) and signicance (p < .05 in the main analysis).
The associations of education (the strongest protective factor) and
waist circumference (the strongest risk factor) with PhenoAgeAccel
were not substantially changed after adjustment for waist circum-
ference, BF%, BMI, or smoking initiation, which had considerable
causal effects on PhenoAgeAccel (β coefcients > 0.5; Figure 3B).
Discussion
The MR study delineated potential causal relationships of 19
common modiable factors with GrimAgeAccel and PhenoAgeAccel,
the robust second-generation epigenetic-age acceleration indicators.
We identied strong evidence for 12 and 8 factors causally associ-
ated with GrimAgeAccel and PhenoAgeAccel, respectively. Smoking
initiation exhibited the greatest effect on increased GrimAgeAccel
(1.299years), followed by higher alcohol intake, higher waist cir-
cumference, daytime napping, higher BF%, higher BMI, higher CRP,
higher triglycerides, childhood obesity, and type 2 diabetes; whereas
education showed the greatest effect on decreased GrimAgeAccel
(−1.143 years per 1-SD increase in years of schooling), followed
by household income. Higher waist circumference and education
were the leading causal risk and protective factors associated with
PhenoAgeAccel, respectively; BF%, BMI, smoking initiation, CRP,
childhood obesity, and type 2 diabetes were also associated with
increased PhenoAgeAccel. Multiple sensitivity analyses strength-
ened the robustness of these causal relationships. Multivariable
MR analyses further demonstrated the independent causal effects
of the strongest risk and protective factors on GrimAgeAccel and
PhenoAgeAccel, respectively.
In this study, education was the major protective factor for
both GrimAgeAccel and PhenoAgeAccel, and this causal effect was
largely independent of other related factors, such as income, lifestyle,
Figure 3. Multi variable MR assessing the effects of the strongest protective
factor and risk factor on GrimAgeAccel and PhenoAgeAccel. (A) Effect of the
strongest protective factor and risk factor on GrimAgeAccel; (B) Effect of the
strongest protective factor and risk factor on PhenoAgeAccel. GrimAgeAccel
represents epigenetic-age acceleration obtained using the GrimAge clock;
PhenoAgeAccel represents epigenetic-age acceleration obtained using
the PhenoAge clock; Causal estimates are β (95% CI) in years. Exposure
factors with β coefficients >0.5 and p value of <.05 in the main analysis as
shown in Figure 2 were selected as covariates in the adjustment models.
BF%=body fat percentage; BMI=body mass index; CI=confidence intervals;
MR=Mendelian randomization.
1088 Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
and adiposity indicators. By contrast, the protective causal effect of
income on GrimAgeAccel did not persist after adjustment for educa-
tion, suggesting that this effect was largely inuenced by education.
Education is a strong proxy for SES and a more upstream deter-
minant of health, with broad implications for a person’s life-long
lifestyle behaviors and health-promoting resources (38,39). A re-
cent study using UK Biobank data has documented that each 1-year
increase in genetically determined education was associated with
equivalently 4.2 years of age-related increases in telomere length
(40). Telomere length and epigenetic-age acceleration metrics (eg,
GrimAgeAccel and PhenoAgeAccel) point toward distinct mech-
anisms of the aging process that are marked by telomeres and the
DNA methylation-based epigenetic clocks, both of which are inde-
pendently associated with chronological age and mortality risk (41).
Our ndings, together with those of the UK Biobank, highlight the
important impact of education on biological aging rates from two
different aspects of the aging process. Therefore, public health strat-
egies aimed at reducing educational inequalities and improving edu-
cation may slow the biological aging rate and help reduce age-related
health burdens.
This MR study identied several common lifestyle behaviors,
including smoking initiation, alcohol intake, and daytime napping,
causally associated with increased GrimAgeAccel or PhenoAgeAccel
(smoking initiation only), which were consistent with previous
evidence from observational studies (6,7). Smoking methylation
proxy is a component of the GrimAge clock (7), thus it is not sur-
prising that smoking initiation exhibited a large effect on increased
GrimAgeAccel in this study. Our multivariable MR analysis further
conrmed that the effect of smoking on GrimAgeAccel was inde-
pendent of other causal lifestyle behaviors and adiposity indicators.
Various sensitivity analyses also supported the robustness of the
causal association between smoking initiation and GrimAgeAccel.
Similarly, genetically determined smoking has been associated with
shorter telomere length in the UK Biobank (40), and evidence from
the Danish Health Interview Survey suggested that the life expectancy
of a heavy smoker was a little more than seven years shorter than
that of a never smoker (42). Our study also provided strong MR evi-
dence that genetically predicted higher alcohol intake was associated
with increased GrimAgeAccel. Astudy composed of the Hannum
cohort and the Family and Community Health Studies cohort found
that the relationship between alcohol use and the rst-generation
epigenetic clocks seemed to be nonlinear (43). However, given the
dose-dependent relationship of alcohol intake with all-cause mor-
tality and cancers (44), our ndings suggest that reducing alcohol in-
take is necessary to decrease GrimAgeAccel and retard overall health
loss. Previous observational studies also reported positive or inverse
correlations of other lifestyle behaviors such as MVPA, coffee, and
sleep duration with GrimAgeAccel, PhenoAgeAccel, or mortality
(11,45,46). Nevertheless, in this study, there was little evidence
supporting that these associations were causal. The discrepancy be-
tween our ndings and previous observations may result from the
potential confounding or reverse causation in conventional obser-
vational studies, or because of insufcient power of MR analyses
due to the relatively low variance explained by genetic instruments
(eg, MVPA). Moreover, the nonlinear association patterns, as in the
case of sleep duration and mortality, might also partially explain the
inconsistent results (46). Therefore, our null ndings should be cau-
tiously interpreted.
Interestingly, of all cardiometabolic traits (ie, adiposity indica-
tors, type 2 diabetes, triglycerides, and CRP) which causally in-
creased GrimAgeAccel or PhenoAgeAccel, adiposity indicators were
the most dominant traits. Emerging observational studies have pro-
nounced the positive associations of BMI with GrimAgeAccel and
PhenoAgeAccel (11), and a meta-analysis of 87 observational studies
showed each 5kg/m2 higher BMI corresponded to about 1year of
age-related decrease in telomere length (47). Our study further pro-
vided strong evidence for causal associations of various adiposity
indicators, including waist circumference, BF%, BMI, and child-
hood obesity, with increased GrimAgeAccel and PhenoAgeAccel.
In this study, type 2 diabetes, triglycerides, and CRP showed strong
but modest effects on GrimAgeAccel or PhenoAgeAccel, which was
consistent with the ndings of the UK Biobank study on telomere
length (40). Previous cross-sectional studies have found that HDL
cholesterol, LDL cholesterol, and triglycerides were signicantly
related to epigenetic aging (6,7). The prospective Coronary Artery
Risk Development in Young Adults study of 1 118 White and
Black individuals indicated that high triglycerides and low HDL
cholesterol in early adulthood were associated with accelerated
epigenetic aging by midlife (48). In our MR study, only triglycer-
ides were causally associated with GrimAgeAccel, but neither HDL
cholesterol nor LDL cholesterol was identied as a causal factor
for GrimAgeAccel or PhenoAgeAccel. Our ndings suggested that
the signicant associations found in these observational studies
might be partially inuenced by residual confounding. Moreover,
the inammatory phenotype (eg, CRP), which tends to increase
with aging, has been recognized as an important risk factor for
age-related morbidity and mortality (49). Previous cross-sectional
studies have shown that CRP was signicantly related to epigen-
etic aging (6,7). However, one recent longitudinal study found that
although CRP was associated with epigenetic aging measures in
cross-sectional analysis, baseline CRP was not associated with epi-
genetic aging after 11years of follow-up (50). Our study extended
previous observational studies by adding evidence for a causal ef-
fect of CRP on epigeneticaging.
The GWAS for GrimAgeAccel and PhenoAgeAccel also incorp-
orated data-driven MR analyses to investigate causal relationships
between epigenetic aging and 150 UK Biobank traits and found that
adiposity, education, and CRP were causally associated with epigen-
etic aging (31). Based on this study, our hypothesis-driven MR study
further applied GWAS summary statistics from the most authorita-
tive and appropriate consortiums to enhance the precision of causal
estimates. We systematically investigated whether and to what extent
common and clinically signicant modiable factors, including SES,
lifestyle behaviors, and cardiometabolic traits, are likely to inuence
GrimAgeAccel and PhenoAgeAccel, in order to inform clinical prac-
tice and biological understanding. Importantly, we for the rst time
assessed whether these modiable factors had independent causal ef-
fects on GrimAgeAccel and PhenoAgeAccel using multivariable MR
analyses. This study brings us one step closer to understanding the
potential contributors to epigenetic aging and provides promising
intervention targets for healthy aging. Given the enormous burden
induced by age-related morbidity and mortality, strategies to re-
duce educational inequalities, promote healthy lifestyles primarily
through reducing smoking, alcohol intake, and daytime napping,
and improve cardiometabolic traits, specically adiposity, type 2
diabetes, triglycerides, and CRP, to slow epigenetic aging rate are
imminent.
In this study, we included independent and genome-wide signi-
cant SNPs as instruments for each of the modiable factors to en-
sure the rst MR assumption was fullled. Moreover, we applied
strict criteria strengthened by the FDR-corrected signicance and the
cross-validations by main and sensitivity analyses to draw robust
Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7 1089
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
causal conclusions. However, several limitations merited consider-
ation. First, we found potential pleiotropy from the MR-Egger inter-
cept test for CRP. However, we conducted the MR-PRESSO analysis,
and the association remained consistent after the removal of out-
lying SNPs. Second, we could not rule out the possibility that the
associations of certain modiable risk factors such as alcohol intake
and sleep duration with GrimAgeAccel or PhenoAgeAccel may be
nonlinear. Future studies with individual-level data are warranted
to conrm the linear or nonlinear relationships. Third, to ensure the
consistency of genetic background, this MR study was performed
only in European-ancestry participants, thus the generalization of
our results to other ethnic groups should be cautious. Forth, due
to the MR design, this study is not applicable to investigating the
nongenetically determined portion of the trait. However, based on
the MR study design as a natural experiment, the MR estimates
provided in this study are complementary to the previous observa-
tional estimates, providing additional causal information for clinical
practice.
In conclusion, this MR study provided novel quantitative evi-
dence on modiable causal risk factors for accelerated epigen-
etic aging, among which adiposity indicators, low education, and
smoking exhibited the most signicant inuence. Our ndings shed
light on the underlying contributors to biological aging and point
toward promising intervention targets to slow the biological aging
rate and promote healthy longevity.
Supplementary Material
Supplementary data are available at The Journals of Gerontology,
Series A: Biological Sciences and Medical Sciencesonline.
Funding
This work was supported by the grants from the National Natural Science
Foundation of China (82022011, 81970706, 82088102, 81970728), the
“Shanghai Municipal Education Commission–Gaofeng Clinical Medicine
Grant Support” from Shanghai Jiao Tong University School of Medicine
(20171901 Round 2), and the Innovative Research Team of High-level Local
Universities in Shanghai.
Conflict of Interest
None declared.
Acknowledgments
We gratefully acknowledge the investigators and participants of all genome-
wide association studies from which we used data.
Author Contributions
L.K.and T.W.contributed to the conception and design of the study. L.K.and
C.Y. contributed to statistical analysis. L.K. contributed to drafting of the
manuscript. T.W.guaranteed this work and take responsibility for the integrity
of the data. All authors contributed to acquisition or interpretation of data,
critical revision of the manuscript for important intellectual content, and nal
approval of the version to be published.
Ethics Approval
This study is based on publicly available summarized data. Ethical approval
and informed consent had been obtained in all original studies.
Data Availability
The genetic association data of the 19 modiable factors are available in
Supplementary Table 3. The GWAS summary statistics for GrimAgeAccel
and PhenoAgeAccel are available at https://datashare.is.ed.ac.uk/
handle/10283/3645. The analytical script of the MR analyses conducted in
this study is available via the GitHub repository of the “TwoSampleMR” R
package (https://github.com/MRCIEU/TwoSampleMR/).
References
1. Niccoli T, Partridge L. Ageing as a risk factor for disease. Curr Biol.
2012;22(17):R741–R752. doi:10.1016/j.cub.2012.07.024
2. Schumacher B, Pothof J, Vijg J, Hoeijmakers JHJ. The central role of
DNA damage in the ageing process. Nature. 2021;592(7856):695–703.
doi:10.1038/s41586-021-03307-7
3. WarnerHR. Current status of efforts to measure and modulate the bio-
logical rate of aging. J Gerontol ABiol Sci Med Sci. 2004;59(7):692–696.
doi:10.1093/gerona/59.7.b692
4. JylhäväJ, PedersenNL, HäggS. Biological age predictors. EBioMedicine.
2017;21:29–36. doi:10.1016/j.ebiom.2017.03.046
5. HorvathS, RajK. DNA methylation-based biomarkers and the epigenetic
clock theory of ageing. Nat Rev Genet. 2018;19(6):371–384. doi:10.1038/
s41576-018-0004-3
6. Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging
for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573–591.
doi:10.18632/aging.101414
7. Lu AT, Quach A, WilsonJG, etal. DNA methylation GrimAge strongly
predicts lifespan and healthspan. Aging (Albany NY). 2019;11(2):303–
327. doi:10.18632/aging.101684
8. WangC, Ni W, YaoY, etal. DNA methylation-based biomarkers of age
acceleration and all-cause death, myocardial infarction, stroke, and cancer
in two cohorts: the NAS, and KORA F4. EBioMedicine. 2021;63:103151.
doi:10.1016/j.ebiom.2020.103151
9. LiA, Koch Z, IdekerT. Epigenetic aging: biological age prediction and
informing a mechanistic theory of aging. J Intern Med. 2022;292(5):733–
744. doi:10.1111/joim.13533
10. Quach A, Levine ME, Tanaka T, et al. Epigenetic clock analysis of
diet, exercise, education, and lifestyle factors. Aging (Albany NY).
2017;9(2):419–446. doi:10.18632/aging.101168
11. OblakL, vanderZaagJ, Higgins-ChenAT, LevineME, BoksMP. A sys-
tematic review of biological, social and environmental factors associated
with epigenetic clock acceleration. Ageing Res Rev. 2021;69:101348.
doi:10.1016/j.arr.2021.101348
12. SierraF, CaspiA, FortinskyRH, etal. Moving geroscience from the bench
to clinical care and health policy. J Am Geriatr Soc. 2021;69(9):2455–
2463. doi:10.1111/jgs.17301
13. CampisiJ, KapahiP, LithgowGJ, MelovS, NewmanJC, VerdinE. From
discoveries in ageing research to therapeutics for healthy ageing. Nature.
2019;571(7764):183–192. doi:10.1038/s41586-019-1365-2
14. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian ran-
domisation studies: a guide, glossary, and checklist for clinicians. BMJ.
2018;362:k601. doi:10.1136/bmj.k601
15. Skrivankova VW, RichmondRC, WoolfBAR, et al. Strengthening the
Reporting of Observational Studies in Epidemiology using Mendelian
randomisation (STROBE-MR): explanation and elaboration. BMJ.
2021;375:n2233. doi:10.1136/bmj.n2233
16. ZhengJ, BairdD, BorgesMC, etal. Recent developments in Mendelian
randomization studies. Curr Epidemiol Rep. 2017;4(4):330–345.
doi:10.1007/s40471-017-0128-6
17. Lee JJ, WedowR, OkbayA, et al. Gene discovery and polygenic predic-
tion from a genome-wide association study of educational attainment in
1.1 million individuals. Nat Genet. 2018;50(8):1112–1121. doi:10.1038/
s41588-018-0147-3
18. LiuM, JiangY, WedowR, etal. Association studies of up to 1.2 million in-
dividuals yield new insights into the genetic etiology of tobacco and alcohol
use. Nat Genet. 2019;51(2):237–244. doi:10.1038/s41588-018-0307-5
1090 Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
19. Zhong VW, KuangA, Danning RD, etal. A genome-wide association
study of bitter and sweet beverage consumption. Hum Mol Genet.
2019;28(14):2449–2457. doi:10.1093/hmg/ddz061
20. Dashti HS, Daghlas I, Lane JM, et al. Genetic determinants of day-
time napping and effects on cardiometabolic health. Nat Commun.
2021;12(1):900. doi:10.1038/s41467-020-20585-3
21. DashtiHS, Jones SE, WoodAR, et al. Genome-wide association study
identies genetic loci for self-reported habitual sleep duration supported
by accelerometer-derived estimates. Nat Commun. 2019;10(1):1100.
doi:10.1038/s41467-019-08917-4
22. KlimentidisYC, RaichlenDA, BeaJ, etal. Genome-wide association study
of habitual physical activity in over 377,000 UK Biobank participants
identies multiple variants including CADM2 and APOE. Int J Obes
(Lond). 2018;42(6):1161–1176. doi:10.1038/s41366-018-0120-3
23. YengoL, SidorenkoJ, Kemper KE, etal. Meta-analysis of genome-wide
association studies for height and body mass index in <700000 individ-
uals of European ancestry. Hum Mol Genet. 2018;27(20):3641–3649.
doi:10.1093/hmg/ddy271
24. Shungin D, Winkler TW, Croteau-Chonka DC, et al. New genetic
loci link adipose and insulin biology to body fat distribution. Nature.
2015;518(7538):187–196. doi:10.1038/nature14132
25. BradeldJP, TaalHR, TimpsonNJ, etal. A genome-wide association meta-
analysis identies new childhood obesity loci. Nat Genet. 2012;44(5):526–
531. doi:10.1038/ng.2247
26. MahajanA, TaliunD, ThurnerM, etal. Fine-mapping type 2 diabetes loci
to single-variant resolution using high-density imputation and islet-specic
epigenome maps. Nat Genet. 2018;50(11):1505–1513. doi:10.1038/
s41588-018-0241-6
27. Klarin D, DamrauerSM, Cho K, etal. Genetics of blood lipids among
~300,000 multi-ethnic participants of the Million Veteran Program. Nat
Genet. 2018;50(11):1514–1523. doi:10.1038/s41588-018-0222-9
28. EvangelouE, WarrenHR, Mosen-AnsorenaD, etal. Publisher correction:
genetic analysis of over 1 million people identies 535 new loci associated
with blood pressure traits. Nat Genet. 2018;50(12):1755. doi:10.1038/
s41588-018-0297-3
29. HanX, Ong JS, AnJ, HewittAW, GharahkhaniP, MacGregorS. Using
Mendelian randomization to evaluate the causal relationship between
serum C-reactive protein levels and age-related macular degeneration. Eur
J Epidemiol. 2020;35(2):139–146. doi:10.1007/s10654-019-00598-z
30. MachielaMJ, ChanockSJ. LDlink: a web-based application for exploring
population-specic haplotype structure and linking correlated alleles of
possible functional variants. Bioinformatics. 2015;31(21):3555–3557.
doi:10.1093/bioinformatics/btv402
31. McCartneyDL, Min JL, RichmondRC, etal. Genome-wide association
studies identify 137 genetic loci for DNA methylation biomarkers of
aging. Genome Biol. 2021;22(1):194. doi:10.1186/s13059-021-02398-9
32. Lawlor DA, Harbord RM, Sterne JA, Timpson N, Davey Smith G.
Mendelian randomization: using genes as instruments for making
causal inferences in epidemiology. Stat Med. 2008;27(8):1133–1163.
doi:10.1002/sim.3034
33. BowdenJ, DaveySmith G, Haycock PC, Burgess S. Consistent estima-
tion in Mendelian randomization with some invalid instruments using
a weighted median estimator. Genet Epidemiol. 2016;40(4):304–314.
doi:10.1002/gepi.21965
34. BurgessS, BowdenJ, FallT, IngelssonE, ThompsonSG. Sensitivity analyses
for robust causal inference from Mendelian randomization analyses with
multiple genetic variants. Epidemiology. 2017;28(1):30–42. doi:10.1097/
EDE.0000000000000559
35. VerbanckM, ChenCY, NealeB, DoR. Detection of widespread horizontal
pleiotropy in causal relationships inferred from Mendelian randomization
between complex traits and diseases. Nat Genet. 2018;50(5):693–698.
doi:10.1038/s41588-018-0099-7
36. Bowden J, Del Greco MF, Minelli C, Davey Smith G, Sheehan N,
ThompsonJ. A framework for the investigation of pleiotropy in two-sample
summary data Mendelian randomization. Stat Med. 2017;36(11):1783–
1802. doi:10.1002/sim.7221
37. BurgessS, ThompsonSG. Multivariable Mendelian randomization: the use
of pleiotropic genetic variants to estimate causal effects. Am J Epidemiol.
2015;181(4):251–260. doi:10.1093/aje/kwu283
38. FigginsE, Pieruccini-FariaF, SpeechleyM, Montero-OdassoM. Potentially
modiable risk factors for slow gait in community-dwelling older adults:
a systematic review. Ageing Res Rev. 2021;66:101253. doi:10.1016/j.
arr.2020.101253
39. Wang Y, Ye C, Kong L, et al. Independent associations of educa-
tion, intelligence, and cognition with hypertension and the mediating
effects of cardiometabolic risk factors: a Mendelian randomiza-
tion study. Hypertension. 2023;80(1):192–203. doi:10.1161/
HYPERTENSIONAHA.122.20286
40. BountzioukaV, MusichaC, AllaraE, etal. Modiable traits, healthy be-
haviours, and leukocyte telomere length: a population-based study in UK
Biobank. Lancet Healthy Longev. 2022;3(5):e321–e331. doi:10.1016/
S2666-7568(22)00072-1
41. Marioni RE, HarrisSE, ShahS, etal. The epigenetic clock and telomere
length are independently associated with chronological age and mortality.
Int J Epidemiol. 2018;45(2):424–432. doi:10.1093/ije/dyw041
42. Brønnum-Hansen H, Juel K. Abstention from smoking extends life
and compresses morbidity: a population based study of health expect-
ancy among smokers and never smokers in Denmark. Tob Control.
2001;10(3):273–278. doi:10.1136/tc.10.3.273
43. BeachSR, DoganMV, LeiMK, etal. Methylomic aging as a window onto
the inuence of lifestyle: tobacco and alcohol use alter the rate of bio-
logical aging. J Am Geriatr Soc. 2015;63(12):2519–2525. doi:10.1111/
jgs.13830
44. GBD 2016 Alcohol and Drug Use Collaborators. The global burden of
disease attributable to alcohol and drug use in 195 countries and terri-
tories, 1990–2016a systematic analysis for the Global Burden of Disease
Study 2016. Lancet Psychiatry. 2018;5(12):987–1012. doi:10.1016/
s2215-0366(18)30337-7
45. Lofteld E, Cornelis MC, Caporaso N, Yu K, Sinha R, Freedman N.
Association of coffee drinking with mortality by genetic variation in
caffeine metabolism: ndings from the UK biobank. JAMA Intern Med.
2018;178(8):1086–1097. doi:10.1001/jamainternmed.2018.2425
46. WangC, BangdiwalaSI, RangarajanS, etal. Association of estimated sleep
duration and naps with mortality and cardiovascular events: a study of
116 632 people from 21 countries. Eur Heart J. 2019;40(20):1620–1629.
doi:10.1093/eurheartj/ehy695
47. GielenM, HagemanGJ, AntoniouEE, etal. Body mass index is negatively
associated with telomere length: a collaborative cross-sectional meta-
analysis of 87 observational studies. Am J Clin Nutr. 2018;108(3):453–
475. doi:10.1093/ajcn/nqy107
48. GaoT, WilkinsJT, ZhengY, etal. Plasma lipid proles in early adult-
hood are associated with epigenetic aging in the Coronary Artery Risk
Development in Young Adults (CARDIA) study. Clin Epigenetics.
2022;14(1):16. doi:10.1186/s13148-021-01222-2
49. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G.
Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243–278.
doi:10.1016/j.cell.2022.11.001
50. CribbL, HodgeAM, YuC, et al. Inammation and epigenetic aging are
largely independent markers of biological aging and mortality. J Gerontol
ABiol Sci Med Sci. 2022;77(12):2378–2386. doi:10.1093/gerona/glac147
Journals of Gerontology: BIOLOGICAL SCIENCES, 2023, Vol. 78, No. 7 1091
Downloaded from https://academic.oup.com/biomedgerontology/article/78/7/1083/7069451 by guest on 13 September 2023
... Specifically, detrimental dietary habits and low levels of physical activity have been pinpointed as critical accelerators of aging, while moderate alcohol consumption and avoidance of smoking have been associated with a slower aging trajectory (21)(22)(23). Recent studies have further illuminated the associations between sleep, prolonged sitting, physical activity, and PhenoAgeAccel, accentuating the bearing of lifestyle on the aging process (19). ...
Article
Full-text available
Objective To investigate the relationship between Life’s Essential 8 (LE8) and Phenotypic Age Acceleration (PhenoAgeAccel) in United States adults and to explore the impact of LE8 on phenotypic biological aging, thereby providing references for public health policies and health education. Methods Utilizing data from the National Health and Nutrition Examination Survey (NHANES) conducted between 2007 and 2010, this cross-sectional study analyzed 7,339 adults aged 20 and above. Comprehensive assessments of LE8, PhenoAgeAccel, and research covariates were achieved through the integration of Demographics Data, Dietary Data, Laboratory Data, and Questionnaire Data derived from NHANES. Weighted generalized linear regression models and restricted cubic spline plots were employed to analyze the linear and non-linear associations between LE8 and PhenoAgeAccel, along with gender subgroup analysis and interaction effect testing. Results (1) Dividing the 2007–2010 NHANES cohort into quartiles based on LE8 unveiled significant disparities in age, gender, race, body mass index, education level, marital status, poverty-income ratio, smoking and drinking statuses, diabetes, hypertension, hyperlipidemia, phenotypic age, PhenoAgeAccel, and various biological markers (p < 0.05). Mean cell volume demonstrated no intergroup differences (p > 0.05). (2) The generalized linear regression weighted models revealed a more pronounced negative correlation between higher quartiles of LE8 (Q2, Q3, and Q4) and PhenoAgeAccel compared to the lowest LE8 quartile in both crude and fully adjusted models (p < 0.05). This trend was statistically significant (p < 0.001) in the full adjustment model. Gender subgroup analysis within the fully adjusted models exhibited a significant negative relationship between LE8 and PhenoAgeAccel in both male and female participants, with trend tests demonstrating significant results (p < 0.001 for males and p = 0.001 for females). (3) Restricted cubic spline (RCS) plots elucidated no significant non-linear trends between LE8 and PhenoAgeAccel overall and in gender subgroups (p for non-linear > 0.05). (4) Interaction effect tests denoted no interaction effects between the studied stratified variables such as age, gender, race, education level, and marital status on the relationship between LE8 and PhenoAgeAccel (p for interaction > 0.05). However, body mass index and diabetes manifested interaction effects (p for interaction < 0.05), suggesting that the influence of LE8 on PhenoAgeAccel might vary depending on an individual’s BMI and diabetes status. Conclusion This study, based on NHANES data from 2007–2010, has revealed a significant negative correlation between LE8 and PhenoAgeAccel, emphasizing the importance of maintaining a healthy lifestyle in slowing down the biological aging process. Despite the limitations posed by the study’s design and geographical constraints, these findings provide a scientific basis for the development of public health policies focused on healthy lifestyle practices. Future research should further investigate the causal mechanisms underlying the relationship between LE8 and PhenoAgeAccel and consider cross-cultural comparisons to enhance our understanding of healthy aging.
... Notably, there existed a failure to investigate the inverse relationship in that study, with WC and other biological age indicators not being taken into account. A unidirectional Mendelian randomization analysis of BMI and WC with GrimAge and PhenoAge by Kong et al. [29] confirmed a positive facilitating effect, but the reverse relationship has not been explored and the inclusion of biological markers of age is incomplete. Investigating the inherent connection between obesity and biological age markers is instrumental in elucidating the mechanisms through which obesity contributes to the development of other diseases. ...
Article
Full-text available
Background In observational studies, there exists an association between obesity and epigenetic age as well as telomere length. However, varying and partially conflicting outcomes have notably arisen from distinct studies on this topic. In the present study, two-way Mendelian randomization was used to identify potential causal associations between obesity and epigenetic age and telomeres. Methods A genome-wide association study was conducted using data from individuals of European ancestry to investigate bidirectional Mendelian randomization (MR) regarding the causal relationships between obesity, as indicated by three obesity indicators (body mass index or BMI, waist circumference adjusted for BMI or WCadjBMI, and waist-to-hip ratio adjusted for BMI or WHRadjBMI), and four epigenetic age measures (HannumAge, HorvathAge, GrimAge, PhenoAge), as well as telomere length. To assess these causal associations, various statistical methods were employed, including Inverse Variance Weighted (IVW), Weighted Median, MR Egger, Weighted Mode, and Simple Mode. To address the issue of multiple testing, we applied the Bonferroni correction. These methods were used to determine whether there is a causal link between obesity and epigenetic age, as well as telomere length, and to explore potential bidirectional relationships. Forest plots and scatter plots were generated to show causal associations between exposures and outcomes. For a comprehensive visualization of the results, leave-one-out sensitivity analysis plots, individual SNP-based forest plots for MR analysis, and funnel plots were included in the presentation of the results. Results A strong causal association was identified between obesity and accelerated HannumAge, GrimAge, PhenoAge and telomere length shrinkage. The causal relationship between WCadjBMI and PhenoAge acceleration (OR: 2.099, 95%CI: 1.248—3.531, p = 0.005) was the strongest among them. However, only the p-values for the causal associations of obesity with GrimAge, PhenoAge, and telomere length met the criteria after correction using the Bonferroni multiple test. In the reverse MR analysis, there were statistically significant causal associations between HorvathAge, PhenoAge and GrimAge and BMI, but these associations exhibited lower effect sizes, as indicated by their Odds Ratios (ORs). Notably, sensitivity analysis revealed the robustness of the study results. Conclusions The present findings reveal a causal relationship between obesity and the acceleration of epigenetic aging as well as the reduction of telomere length, offering valuable insights for further scientific investigations aimed at developing strategies to mitigate the aging process in humans.
... This MR study was reported following the Strengthening the Reporting of Observational Studies in Epidemiology using Mendelian Randomization (STROBE-MR) guidelines (Additional file 1: Table S1) [22]. We adopted multiple methods to meet the three core assumptions of MR as follows [20,23]. First, the IVs are strongly associated with the exposure (i.e., birthweight) in UVMR analysis or at least one of the multiple exposures in multivariable MR (MVMR) analysis. ...
Article
Full-text available
Background Previous observational studies have documented an inverse association of birthweight with myocardial infarction (MI) but a positive association with atrial fibrillation (AF). However, the causality of these associations and the underlying mediating pathways remain unclear. We aimed to investigate the causal effects of birthweight, incorporating both fetal and maternal genetic effects, on MI and AF, and identify potential mediators in their respective pathways. Methods We performed Mendelian randomization (MR) analyses using genome-wide association study summary statistics for birthweight (N = 297,356 for own birthweight and 210,248 for offspring birthweight), MI (Ncase=61,000, Ncontrol=577,000), AF (Ncase=60,620, Ncontrol=970,216), and 52 candidate mediators (N = 13,848-1,295,946). Two-step MR was employed to identify and assess the mediation proportion of potential mediators in the associations of birthweight with MI and AF, respectively. As a complement, we replicated analyses for fetal-specific birthweight and maternal-specific birthweight. Results Genetically determined each 1-SD lower birthweight was associated with a 40% (95% CI: 1.22–1.60) higher risk of MI, whereas each 1-SD higher birthweight was causally associated with a 29% (95% CI: 1.16–1.44) higher risk of AF. Cardiometabolic factors, including lipids and lipoproteins, glucose and insulin, blood pressure, and fatty acids, each mediated 4.09-23.71% of the total effect of birthweight on MI, followed by body composition and strength traits (i.e., appendicular lean mass, height, and grip strength) and socioeconomic indicators (i.e., education and household income), with the mediation proportion for each factor ranging from 8.08 to 16.80%. By contrast, appendicular lean mass, height, waist circumference, childhood obesity, and body mass index each mediated 15.03-45.12% of the total effect of birthweight on AF. Both fetal-specific birthweight and maternal-specific birthweight were inversely associated with MI, while only fetal-specific birthweight was positively associated with AF. Psychological well-being and lifestyle factors conferred no mediating effect in either association. Conclusions Cardiometabolic factors mainly mediated the association between lower birthweight and MI, while body composition and strength traits mediated the association between higher birthweight and AF. These findings provide novel evidence for the distinct pathogenesis of MI and AF and advocate adopting a life-course approach to improving fetal development and subsequent causal mediators to mitigate the prevalence and burden of cardiovascular diseases.
... According to data from the UK Biobank, the composite indicator, phenotypic age, demonstrated superior predictive effectiveness for chronic lung diseases compared to any of the nine clinical parameters (data not shown). In addition, although results from population-based cohort studies have demonstrated that the derived PhenoAgeAccel is associated with the mortality of cerebrovascular disease, cancer and diabetes [36][37][38][39][40]; however, few efforts have been made to investigate the associations of PhenoAgeAccel with chronic respiratory diseases. ...
Article
Full-text available
Accelerated biological aging has been associated with an increased risk of several chronic respiratory diseases. However, the associations between Phenotypic Age, a new biological age indicator based on clinical chemistry biomarkers, and common chronic respiratory diseases have not been evaluated. We analyzed data from 308 592 participants at baseline in the UK Biobank. The Phenotypic Age was calculated from chronological age and 9 clinical chemistry biomarkers, including albumin, alkaline phosphatase, creatinine, glucose, C-reactive protein, lymphocyte percent, mean cell volume, red cell distribution width, and white blood cell count. Furthermore, Phenotypic Age Acceleration (PhenoAgeAccel) was calculated by regressing Phenotypic Age on chronological age. The associations of PhenoAgeAccel with incident common chronic respiratory diseases and cross-sectional lung function were investigated. Moreover, we constructed polygenic risk scores and evaluated whether PhenoAgeAccel modified the effect of genetic susceptibility on chronic respiratory diseases and lung function. The results showed significant associations of PhenoAgeAccel with increased risk of idiopathic pulmonary fibrosis (IPF) (HR=1.52, 95%CI: 1.45–1.59), chronic obstructive pulmonary disease (COPD) (HR=1.54, 95%CI: 1.51–1.57), and asthma (HR=1.18, 95%CI: 1.15–1.20) per 5-year increase and decreased lung function. There was an additive interaction between PhenoAgeAccel and the genetic risk for IPF and COPD. Participants with high genetic risk and biologically older had the highest risk of incident IPF (HR=5.24, 95%CI: 3.91–7.02), COPD (HR=2.99, 95%CI: 2.66–3.36), and asthma (HR= 2.07, 95%CI: 1.86–2.31). Mediation analysis indicated that PhenoAgeAccel could mediate 10∼20% of the associations between smoking and chronic respiratory diseases, while ∼10% of the associations between PM 2.5 and the disorders were mediated by PhenoAgeAccel. PhenoAgeAccel was significantly associated with incident risk of common chronic respiratory diseases and decreased lung function and could serve as a novel clinical biomarker.
... A study in the United Kingdom Biobank cohort found the acceleration of biological aging would be the consequence of sleep quality (including nighttime sleep duration) (15). A new study has revealed that a higher frequency of napping was causally associated with epigenetic age acceleration based on DNA methylation level (16). Given the potential connections, it seems warranted to evaluate the association of napping duration, BA, with cognitive function, however, the current evidence is limited. ...
Article
Full-text available
Objective The complicated association of daytime napping, biological aging and cognitive function remains inconclusive. We aimed to evaluate the cross-sectional and longitudinal associations of daytime napping and two aging measures with cognition and to examine whether napping affects cognition through a more advanced state of aging. Methods Data was collected from the China Health and Retirement Longitudinal Study. Napping was self-reported. We calculated two published biological aging measures: Klemera and Doubal biological age (KDM-BA) and physiological dysregulation (PD), which derived information from clinical biomarkers. Cognitive z-scores were calculated at each wave. Linear mixed models were used to explore the longitudinal association between napping, two aging measures, and cognitive decline. Mediation analyses were performed to assess the mediating effects of biological age acceleration on the association between napping and cognition. Results Participants aged over 45 years were included in the analyses. Non-nappers had greater KDM-BA and PD [LS means (LSM) = 0.255, p = 0.007; LSM = 0.085, p = 0.011] and faster cognitive decline (LSM = −0.061, p = 0.005)compared to moderate nappers (30–90 min/nap). KDM-BA (β = −0.007, p = 0.018) and PD (β = −0.034, p < 0.001) showed a negative association with overall cognitive z scores. KDM-BA and PD partially mediated the effect of napping on cognition. Conclusion In middle-aged and older Chinese, compared to moderate nappers, non-nappers seem to experience a more advanced state of aging and increased rates of cognitive decline. The aging status possibly mediates the association between napping and cognition. Moderate napping shows promise in promoting healthy aging and reducing the burden of cognitive decline in Chinese middle-aged and older adults.
... Thus, those tests are performed to establish a diagnosis, but no marker is routinely used to measure the patient's healthspan or lifespan potential. However, a few longevity markers do currently exist, such as PhenoAge (algorithms to improve chronological age by adding 9 biomarkers found in routine blood tests) or GrimAge, which is an epigenetic clock that can evaluate the biological age of an individual using DNA methylation-based markers [11,19]. These tests are reliable for determining biological age, but there is little literature linking them to healthspan potential and even less to each of the pillars of lifestyle medicine. ...
Article
Full-text available
Aging is not a disease; it is a natural evolution of human physiology. Medical advances have extended our life expectancy, but chronic diseases and geriatric syndrome continue to affect the increasingly aging population. Yet modern medicine perpetuates an approach based on treatment rather than prevention and education. In order to help solve this ever-growing problem, a new discipline has emerged: lifestyle medicine. Nutrition, physical activity, stress management, restorative sleep, social connection, and avoidance of risky substances are the pillars on which lifestyle medicine is founded. The aim of this discipline is to increase healthspan and reduce the duration of morbidity by making changes to our lifestyle. In this review, we propose the use of klotho protein as a novel biomarker for lifestyle medicine in order to quantify and monitor the health status of individuals, as no integrative tool currently exists.
Article
Epigenetic clocks based on DNA methylation have been known as biomarkers of aging, including principal component (PC) clocks representing the degree of aging and DunedinPACE representing the pace of aging. Prior studies have shown the associations between epigenetic aging and T2DM, but the results vary by epigenetic age metrics and people. This study explored the associations between epigenetic age metrics and T2DM or glycemic traits, based on 1070 twins (535 twin pairs) from the Chinese National Twin Registry. It also explored the temporal relationships of epigenetic age metrics and glycemic traits in 314 twins (157 twin pairs) who participated in baseline and follow‐up visits after a mean of 4.6 years. DNA methylation data were used to calculate epigenetic age metrics, including PCGrimAge acceleration (PCGrimAA), PCPhenoAge acceleration (PCPhenoAA), DunedinPACE, and the longitudinal change rate of PCGrimAge/PCPhenoAge. Mixed‐effects and cross‐lagged modelling assessed the cross‐sectional and temporal relationships between epigenetic age metrics and T2DM or glycemic traits, respectively. In the cross‐sectional analysis, positive associations were identified between DunedinPACE and glycemic traits, as well as between PCPhenoAA and fasting plasma glucose, which may be not confounded by shared genetic factors. Cross‐lagged models revealed that glycemic traits (fasting plasma glucose, HbA1c, and TyG index) preceded DunedinPACE increases, and TyG index preceded PCGrimAA increases. Glycemic traits are positively associated with epigenetic age metrics, especially DunedinPACE. Glycemic traits preceded the increases in DunedinPACE and PCGrimAA. Lowering the levels of glycemic traits may reduce DunedinPACE and PCGrimAA, thereby mitigating age‐related comorbidities.
Preprint
Full-text available
Background Emerging evidence from observational studies suggested that epigenetic age acceleration may result in an increased incidence of stroke and poorer functional outcomes after a stroke. However, the causality of these associations remains controversial and may be confounded by bias. We aimed to investigate the causal effects of epigenetic age on stroke and its functional outcomes. Methods We conducted a two-sample Mendelian randomization (MR) analysis to explore the causal relationships between epigenetic age and stroke and its outcomes. Additionally, a two-step MR analysis was performed to investigate whether lifestyle factors affect stroke via epigenetic age. Datasets of epigenetic age were obtained from a recent meta-analysis (n = 34710), while those of stroke and its outcomes were sourced from the MEGASTROKE (n = 520000) consortium and Genetics of Ischaemic Stroke Functional Outcome network (n = 6165). Results Two-sample MR analysis revealed a causal relationship between PhenoAge and small vessel stroke (OR = 1.07; 95% CI, 1.03–1.12; p = 2.01 × 10− 3). Mediation analysis through two-step MR indicated that the increased risk of small vessel stroke due to smoking initiation was partially mediated by PhenoAge, with a mediation proportion of 9.5% (95% CI, 1.6–20.6%). No causal relationships were identified between epigenetic age and stroke outcomes. Conclusions Our study confirms a causal relationship between epigenetic age acceleration and stroke, indicating that epigenetic age acceleration may mediate the increased stroke risk due to smoking. Interventions specifically aimed at decelerating epigenetic aging, such as specific lifestyle changes, offer effective strategies for reducing stroke risk.
Article
DNA methylation (DNAm) clocks hold promise for measuring biological age, useful for guiding clinical interventions and forensic identification. This study compared the commonly used DNAm clocks, using DNA methylation and SNP data generated from nearly 1000 human blood or buccal swab samples. We evaluated different preprocessing methods for age estimation, investigated the association of epigenetic age acceleration (EAA) with various lifestyle and sociodemographic factors, and undertook a series of novel genome-wide association analyses for different EAA measures to find associated genetic variants. Our results highlighted the Skin&Blood clock with ssNoob normalization as the most accurate predictor of chronological age. We provided novel evidence for an association between the practice of yoga and a reduction in the pace of aging (DunedinPACE). Increased sleep and physical activity were associated with lower mortality risk score (MRS) in our dataset. University degree, vegetable consumption, and coffee intake were associated with reduced levels of epigenetic aging, whereas smoking, higher BMI, meat consumption, and manual occupation correlated well with faster epigenetic aging, with FitAge, GrimAge, and DunedinPACE clocks showing the most robust associations. In addition, we found a novel association signal for SOCS2 rs73218878 (p = 2.87 × 10−8) and accelerated GrimAge. Our study emphasizes the importance of an optimized DNAm analysis workflow for accurate estimation of epigenetic age, which may influence downstream analyses. The results support the influence of genetic background on EAA. The associated SOCS2 is a member of the suppressor of cytokine signaling family known for its role in human longevity. The reported association between various risk factors and EAA has practical implications for the development of health programs to improve quality of life and reduce premature mortality associated with age-related diseases.
Article
Full-text available
BACKGROUND Education, intelligence, and cognition are associated with hypertension, but which one plays the most prominent role in the pathogenesis of hypertension and which modifiable risk factors mediate the causal effects remains unknown. METHODS Using summary statistics of genome-wide association studies of predominantly European ancestry, we conducted 2-sample multivariable Mendelian randomization to estimate the independent effects of education, intelligence, or cognition on hypertension (FinnGen study, 70 651 cases/223 663 controls; UK Biobank, 77 723 cases/330 366 controls) and blood pressure (International Consortium of Blood Pressure, 757 601 participants), and used 2-step Mendelian randomization to evaluate 25 potential mediators of the association and calculate the mediated proportions. RESULTS Meta-analysis of inverse variance weighted Mendelian randomization results from FinnGen and UK Biobank showed that genetically predicted 1-SD (4.2 years) higher education was associated with 44% (95% CI: 0.40–0.79) decreased hypertension risk and 1.682 mm Hg lower systolic and 0.898 mm Hg lower diastolic blood pressure, independently of intelligence and cognition. While the causal effects of intelligence and cognition on hypertension were not independent of education, 6 out of 25 cardiometabolic risk factors were identified as mediators of the association between education and hypertension, ranked by mediated proportions, including body mass index (mediated proportion: 30.1%), waist-to-hip ratio (22.8%), body fat percentage (14.1%), major depression (7.0%), high-density lipoprotein cholesterol (4.7%), and triglycerides (3.4%). These results were robust to sensitivity analyses. CONCLUSIONS Our findings illustrated the causal, independent impact of education on hypertension and blood pressure and outlined cardiometabolic mediators as priority targets for prevention of hypertension attributable to low education.
Article
Full-text available
Limited evidence exists on the link between inflammation and epigenetic ageing. We aimed to 1) assess the cross-sectional and prospective associations of 22 inflammation-related plasma markers and a signature of inflammaging with epigenetic ageing; 2) determine whether epigenetic ageing and inflammaging are independently associated with mortality. Blood samples from 940 participants in the Melbourne Collaborative Cohort Study, collected at baseline (1990-1994) and follow-up (2003-2007) were assayed for DNA methylation and 22 inflammation-related markers, including well-established markers (e.g., interleukins and C-reactive protein) and metabolites of the tryptophan-kynurenine pathway. Four measures of epigenetic ageing (PhenoAge, GrimAge, DunedinPoAm and Zhang) and a signature of inflammaging were considered, adjusted for age, and transformed to Z-scores. Associations were assessed using linear regression, and mortality hazard ratios (HR) and 95% confidence intervals (95%CI) were estimated using Cox regression. Cross-sectionally, most inflammation-related markers were associated with epigenetic ageing measures, although with generally modest effect sizes (regression coefficients per SD≤0.26) and explaining altogether between 1% and 11% of their variation. Prospectively, baseline inflammation-related markers were not, or only weakly, associated with epigenetic ageing after 11 years of follow-up. Epigenetic ageing and inflammaging were strongly and independently associated with mortality, e.g. inflammaging: HR=1.41, 95%CI=1.27-1.56, P=2x10-10; which was only slightly attenuated after adjustment for four epigenetic ageing measures: HR=1.35, 95%CI=1.22-1.51, P=7x10-9). Although cross-sectionally associated with epigenetic ageing, inflammation-related markers accounted for a modest proportion of its variation. Inflammaging and epigenetic ageing are essentially non-overlapping markers of biological ageing and may be used jointly to predict mortality.
Article
Full-text available
Background Telomere length is associated with risk of several age-related diseases and cancers. We aimed to investigate the extent to which telomere length might be modifiable through lifestyle and behaviour, and whether such modification has any clinical consequences. Methods In this population-based study, we included participants from UK Biobank who had leukocyte telomere length (LTL) measurement, ethnicity, and white blood cell count data. We investigated associations of LTL with 117 potentially modifiable traits, as well as two indices of healthy behaviours incorporating between them smoking, physical activity, diet, maintenance of a healthy bodyweight, and alcohol intake, using both available and imputed data. To help interpretation, associations were summarised as the number of equivalent years of age-related change in LTL by dividing the trait β coefficients with the age β coefficient. We used mendelian randomisation to test causality of selected associations. We investigated whether the associations of LTL with 22 diseases were modified by the number of healthy behaviours and the extent to which the associations of more healthy behaviours with greater life expectancy and lower risk of coronary artery disease might be mediated through LTL. Findings 422 797 participants were available for the analysis (227 620 [53·8%] were women and 400 036 [94·6%] were White). 71 traits showed significant (p<4·27 × 10–4) associations with LTL but most were modest, equivalent to less than 1 year of age-related change in LTL. In multivariable analyses of 17 traits with stronger associations (equivalent to ≥2 years of age-related change in LTL), oily fish intake, educational attainment, and general health status retained a significant association of this magnitude, with walking pace and current smoking being additionally significant at this level of association in the imputed models. Mendelian randomisation analysis suggested that educational attainment and smoking behaviour causally affect LTL. Both indices of healthy behaviour were positively and linearly associated with LTL, with those with the most healthy behaviours having longer LTL equivalent to about 3·5 years of age-related change in LTL than those with the least heathy behaviours (p<0·001). However, healthy behaviours explained less than 0·2% of the total variation in LTL and did not significantly modify the association of LTL with risk of any of the diseases studied. Neither the association of more healthy behaviours on greater life expectancy or lower risk of coronary artery disease were substantially mediated through LTL. Interpretation Although several potentially modifiable traits and healthy behaviours have a quantifiable association with LTL, at least some of which are likely to be causal, these effects are not of a sufficient magnitude to substantially alter the association between LTL and various diseases or life expectancy. Attempts to change telomere length through lifestyle or behavioural changes might not confer substantial clinical benefit. Funding UK Medical Research Council, UK Biotechnology and Biological Sciences Research Council, and British Heart Foundation.
Article
Full-text available
Background GrimAge acceleration (GAA), an epigenetic marker that represents physiologic aging, is associated with atherosclerotic cardiovascular disease. However, the associations between early adulthood lipid levels and GAA in midlife are unknown. Also, it is unknown whether GAA mediates the associations between lipid levels in young adults and subclinical atherosclerosis in midlife. Results We estimated measures of epigenetic age acceleration in 1118 White and Black participants from the Coronary Artery Risk Development in Young Adults (CARDIA) Study at examination years (Y) 15 and 20. We used multivariable regression models to examine associations of Y15 and Y20 GAA estimates with plasma lipid levels measured at prior examination years (Y0, Y5, and Y10) and concurrently: triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) levels. Mediation analysis was used to assess the extent to which GAA may mediate associations between plasma lipids and coronary artery calcification (CAC). In our study each 1-SD higher cumulative TG level was associated with an average 0.73 ± 0.12 years older GAA. Each 1-SD higher cumulative HDL-C level was associated with an average 0.57 ± 0.17 years younger GAA. Stratified analyses showed that the associations between TG and GAA were stronger among female and Black participants and the associations between HDL-C and GAA were stronger among female and White participants. GAA statistically mediated 17.4% of the association of cumulative TG with CAC. Conclusions High TG and low HDL-C in early adulthood are associated with accelerated epigenetic aging by midlife. Increased epigenetic age acceleration may partially mediate the associations between high TG levels and the presence of subclinical atherosclerosis.
Article
Full-text available
Mendelian randomisation (MR) studies allow a better understanding of the causal effects of modifiable exposures on health outcomes, but the published evidence is often hampered by inadequate reporting. Reporting guidelines help authors effectively communicate all critical information about what was done and what was found. STROBE-MR (strengthening the reporting of observational studies in epidemiology using mendelian randomisation) assists authors in reporting their MR research clearly and transparently. Adopting STROBE-MR should help readers, reviewers, and journal editors evaluate the quality of published MR studies. This article explains the 20 items of the STROBE-MR checklist, along with their meaning and rationale, using terms defined in a glossary. Examples of transparent reporting are used for each item to illustrate best practices.
Article
Full-text available
Background Biological aging estimators derived from DNA methylation data are heritable and correlate with morbidity and mortality. Consequently, identification of genetic and environmental contributors to the variation in these measures in populations has become a major goal in the field. Results Leveraging DNA methylation and SNP data from more than 40,000 individuals, we identify 137 genome-wide significant loci, of which 113 are novel, from genome-wide association study (GWAS) meta-analyses of four epigenetic clocks and epigenetic surrogate markers for granulocyte proportions and plasminogen activator inhibitor 1 levels, respectively. We find evidence for shared genetic loci associated with the Horvath clock and expression of transcripts encoding genes linked to lipid metabolism and immune function. Notably, these loci are independent of those reported to regulate DNA methylation levels at constituent clock CpGs. A polygenic score for GrimAge acceleration showed strong associations with adiposity-related traits, educational attainment, parental longevity, and C-reactive protein levels. Conclusion This study illuminates the genetic architecture underlying epigenetic aging and its shared genetic contributions with lifestyle factors and longevity.
Article
Full-text available
Aging involves a diverse set of biological changes accumulating over time that leads to increased risk of morbidity and mortality. Epigenetic clocks are now widely used to quantify biological aging, in order to investigate determinants that modify the rate of aging and to predict age-related outcomes. Numerous biological, social and environmental factors have been investigated for their relationship to epigenetic clock acceleration and deceleration. The aim of this review was to synthesize general trends concerning the associations between human epigenetic clocks and these investigated factors. We conducted a systematic review of all available literature and included 156 publications across 4 resource databases. We compiled a list of all presently existing blood-based epigenetic clocks. Subsequently, we created an extensive dataset of over 1300 study findings in which epigenetic clocks were utilized in blood tissue of human subjects to assess the relationship between these clocks and numeral environmental exposures and human traits. Statistical analysis was possible on 57 such relationships, measured across 4 different epigenetic clocks (Hannum, Horvath, Levine and GrimAge). We found that the Horvath, Hannum, Levine and GrimAge epigenetic clocks tend to agree in direction of effects, but vary in size. Body mass index, HIV infection, and male sex were significantly associated with acceleration of one or more epigenetic clocks. Acceleration of epigenetic clocks was also related to mortality, cardiovascular disease, cancer and diabetes. Our findings provide a graphical and numerical synopsis of the past decade of epigenetic age estimation research and indicate areas where further attention could be focused in the coming years.
Article
Aging is driven by hallmarks fulfilling the following three premises: (1) their age-associated manifestation, (2) the acceleration of aging by experimentally accentuating them, and (3) the opportunity to decelerate, stop, or reverse aging by therapeutic interventions on them. We propose the following twelve hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interconnected among each other, as well as to the recently proposed hallmarks of health, which include organizational features of spatial compartmentalization, maintenance of homeostasis, and adequate responses to stress.
Article
Numerous studies have shown that epigenetic age—an individual's degree of aging based on patterns of DNA methylation—can be computed and is associated with an array of factors including diet, lifestyle, genetics, and disease. One can expect that still further associations will emerge with additional aging research, but to what end? Prediction of age was an important first step, but—in our view—the focus must shift from chasing increasingly accurate age computations to understanding the links between the epigenome and the mechanisms and physiological changes of aging. Here, we outline emerging areas of epigenetic aging research that prioritize biological understanding and clinical application. First, we survey recent progress in epigenetic clocks, which are beginning to predict not only chronological age but aging outcomes such as all‐cause mortality and onset of disease, or which integrate aging signals across multiple biological processes. Second, we discuss research that exemplifies how investigation of the epigenome is building a mechanistic theory of aging and informing clinical practice. Such examples include identifying methylation sites and the genes most strongly predictive of aging—a subset of which have shown strong potential as biomarkers of neurodegenerative disease and cancer; relating epigenetic clock predictions to hallmarks of aging; and using longitudinal studies of DNA methylation to characterize human disease, resulting in the discovery of epigenetic indications of type 1 diabetes and the propensity for psychotic experiences.
Article
Geriatricians and others must embrace the emerging field of geroscience. Until recently geroscience research was pursued in laboratory animals, but now this field requires specialized expertise in the care of vulnerable older patients with multiple chronic diseases and geriatric syndromes, the population likely to benefit the most from emerging therapies. While chronological aging measures the inevitable passage of clock time that occurs equally for everyone, biological aging varies among individuals, and importantly, it is modifiable. Advances in our understanding of biological aging, the discovery of strategies for modifying its rate, and an appreciation of aging as a shared risk factor for chronic diseases have jointly led to the Geroscience Hypothesis. This hypothesis states that interventions modifying aging biology can slow its progression—resulting in the delay or prevention of the onset of multiple diseases and disorders. Here we wish to report on the Third Geroscience Summit held at National Institutes of Health on November 4–5, 2019, which highlighted the importance of engaging other disciplines including clinicians. Involvement by scientists with expertise in clinical trials, health outcomes research, behavioral and social sciences, health policy, and economics is urgently needed to translate geroscience discoveries from the bench to clinical care and health policy. Adding to the urgency of broadening this geroscience coalition is the emergence of biological aging as one the most important modifiable factors of COVID‐19, combined with the inability of our society to once again recognize and confront aging as a priority and opportunity when facing these types of public health emergencies.