You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Utility of DNA Methylation as a Biomarker in Aging and Alzheimer’s Disease

Abstract

Epigenetic mechanisms such as DNA methylation have been implicated in a number of diseases including cancer, heart disease, autoimmune disorders, and neurodegenerative diseases. While it is recognized that DNA methylation is tissue-specific, a limitation for many studies is the ability to sample the tissue of interest, which is why there is a need for a proxy tissue such as blood, that is reflective of the methylation state of the target tissue. In the last decade, DNA methylation has been utilized in the design of epigenetic clocks, which aim to predict an individual’s biological age based on an algorithmically defined set of CpGs. A number of studies have found associations between disease and/or disease risk with increased biological age, adding weight to the theory of increased biological age being linked with disease processes. Hence, this review takes a closer look at the utility of DNA methylation as a biomarker in aging and disease, with a particular focus on Alzheimer’s disease.

INTRODUCTION

Dementia is an umbrella term that encompasses a wide variety of neurodegenerative disorders including vascular dementia, Lewy Body dementia, frontotemporal dementia, and Alzheimer’s disease (AD), which is the most common form and accounts for 60-80% of all dementia cases [1]. AD is characterized by cortical atrophy [2] and thinning [3], aggregation of hyperphosphorylated microtubule-associated tau protein (MAPT) resulting in intracellular neurofibrillary tangles, and accumulation of extracellular amyloid-β (Aβ) plaques [4]. Age is regarded as the most important risk factor for neurodegenerative diseases, including AD [5], and as the worldwide population ages, there will be an increase in the prevalence of dementia and AD. In addition to age, there are several accepted modifiable and non-modifiable risk factors for AD [6, 7]. The most common modifiable factors include physical inactivity, smoking, excessive alcohol consumption, air pollution, head injury, infrequent social contact, lower educational attainment, obesity, hypertension, diabetes, depression, and hearing impairment [8]. Non-modifiable risk factors include sex, family history, and genetics, most notable being variation in the Apolipoprotein E (APOE) gene [7]. Currently, several biomarkers for dementia are available that may aid in the diagnosis of possible/probable AD [9]. Cerebrospinal fluid (CSF) obtained via lumbar puncture and brain imaging measures, such as positron emission tomography (PET), computed tomography (CT), and magnetic resonance imaging (MRI), have been the focus in the search for robust and easily accessible biomarkers for AD [10]. These methods focus on the measurement of proteins important in the disease process including tau and Aβ. Notably, while CSF and brain imaging markers have shown good predictive accuracy, particularly in preclinical studies, they are not ideal for use as routine clinical biomarkers due to their cost, level of invasiveness, and need for interpretation of results by specialists [11]. Even though the aforementioned biomarkers are useful in assisting in the diagnosis of AD, a definitive diagnosis of AD can only be made on biopsy postmortem [2]. Currently, there is an urgent need for an easily obtainable, less invasive, cost-effective routine biomarker, that is specific and sensitive to predicting disease progression.

EPIGENETICS AND DNA METHYLATION

Epigenetic modifications are heritable phenotypic changes that do not alter the DNA sequence [12]. These modifications fall into one of three categories; histone modification, non-coding RNAs, DNA hydroxymethylation, and DNA methylation [12]. Epigenetic modifications regulate gene expression patterns by altering DNA accessibility and chromatin structure [13]. Epigenetic processes are a natural occurrence and have critical roles in cellular processes such as gene regulation, chromosome stability, X chromosome inactivation, and genomic imprinting [14]. This review will focus on the most well-studied epigenetic mechanism [15], DNA methylation. Approximately 28 million cytosine-phosphate-guanine (CpG) dinucleotides are distributed unevenly throughout the mammalian genome [16]. These CpG dinucleotides are the target of an epigenetic modification known as DNA methylation, which involves the transfer of a methyl group from S-adenosyl-L-methionine (SAM) onto the 5’ position of a cytosine ring, resulting in the formation of 5-methylcytosine (5mC). The covalent attachment of the methyl group is catalyzed by one of several members belonging to the enzyme family known as DNA methyltransferases (DNMT). DNA methylation is essential in several processes, including silencing retroviral elements, regulation of tissue-specific gene expression, genomic imprinting, and X chromosome inactivation. It has also been found that DNA methylation exerts different influences on gene activities in different genomic regions depending on the underlying genetic sequence. Within intergenic regions, one of the main functions of DNA methylation is to repress the expression of genetic elements that might be potentially unfavorable. These elements, when expressed, may have harmful consequences, with their replication and insertion potentially leading to gene disruption and DNA mutation [17]. Varying degrees of methylation at genes and regulatory sequences can determine the level and integrity of gene expression [16]. Importantly, unmethylated CpGs are not distributed randomly throughout the genome, but are clustered in formations called CpG islands (CGIs), usually located in the promoter regions of genes [18]. CGIs are usually unmethylated in normal cells regardless of their expression levels and gene silencing is promoted post-methylation [16]. Due to their location, they are able to regulate gene expression through silencing of transcription [19]. DNA transcription can be affected in two ways; firstly, methylation of CGIs can impair transcription factors binding and, secondly, methyl-CpG-binding proteins bind to the methylated DNA sequences [20].

Assessment of DNA methylation in neurodegenerative diseases

As stated above, DNA methylation regulates tissue-specific gene expression, complicating its exploration in neurodegenerative diseases, due to the requirement of brain tissue. Brain tissue is only available postmortem making it infeasible for large-scale or longitudinal studies, where repeated measurements are required. Additionally, the heterogeneity of brain tissue poses a barrier in terms of analysis as it is widely known that different cells possess different patterns of methylation [21]. Importantly, postmortem brain tissue only offers an insight into the end stage of processes associated with AD and may not reflect the processes involved in disease development.

To address these concerns, proxy tissues such as blood or saliva have been investigated to determine if their level of DNA methylation is reflective of the methylation state of the tissue of interest, i.e., the brain. Specifically, are the changes observed in the tissue of interest truly reflected in peripheral tissues. Early studies sought to clarify this, with a number demonstrating conservation of DNA methylation across different tissues, using several different methodologies for the assessment of DNA methylation, including the widely used Illumina 450k array, which offered the most comprehensive coverage of the methylome available at the time (now superseded by the Illumina EPIC 850k array). A study by Horvath et al. [22] showed evidence for conservation of DNA methylation patterns across four brain regions and blood tissue, with a correlation of r = 0.85-0.91, using publicly available genome-wide methylation data. This finding was supported in a study assessing 80 matched blood and postmortem brain tissues, which found that subset of probes on the Illumina 450K array were significantly correlated across tissues [21]. Walton et al. [23] have likewise published on the 450K array, reporting that 7.9% of CpGs were strongly correlated between tissues. Further, through the development of the BECon (Blood-Brain Epigenetic Concordance) tool, Edgar and colleagues [24] demonstrated that  9.7% of CpGs from the Illumina 450K array were correlated between tissues. When assessing data from methylated DNA immunoprecipitation sequencing (MeDIP-seq) a high correlation between blood and the cortex and cerebellum (r = 0.82 and 0.77, respectively) was observed in three individuals [25]. More recently, Braun [26] investigated correlations between brain, saliva, blood, and buccal samples obtained from patients undergoing neurosurgical resection for epilepsy, and found that DNA methylation in the brain was most highly correlated with saliva (r = 0.90), followed by blood (r = 0.86) and buccal cells (r = 0.85).

While evidence exists for conservation of DNA methylation between tissues, contradicting studies reporting no significant associations have been published. Specifically, a recent study reported no evidence of a correlation between DNA methylation in peripheral blood CD4 + lymphocytes and prefrontal cortex samples [27]. Currently, the evidence is unclear, and the relationship between DNA methylation in the brain and blood requires additional characterization. However, it has been suggested that a suitable peripheral biomarker does not necessarily need to mirror disease-associated changes observed in the brain and could independently reflect disease responses in the periphery.

DNA METHYLATION AND ALZHEIMER’S DISEASE

Global DNA methylation

DNA methylation can be classified as either global or gene-specific. Global methylation refers to the average percentage methylation across the entire genome, whereas gene-specific DNA methylation refers to the average percentage methylation within a specific gene. Global methylation or overall methylation of the entire genome provides an over-arching picture of methylation status in a sample [28]. However, this measurement should be used with caution as differences in global methylation can result from disease state, tissue collection site, sex, and age [29].

In a 2009 twin study, monozygotic twins discordant for AD were found to have differences in global methylation in the anterior temporal cortex and the superior front gyrus, where the twin with AD had significantly decreased levels of methylation in those two areas of the brain [30]. While both twins had similar levels of education and worked in similar fields, the twin diagnosed with AD worked extensively with pesticides, suggesting this environmental exposure may have contributed to epigenetic changes and the development of AD [30]. Several epidemiological studies have reported phenotypic discordance in monozygotic twins, with the older sibling typically reflecting more discordance for age-related diseases [31, 32]. This is hypothesized to be due to an increased rate of loss of epigenetic architecture, or epigenetic drift, which is described as an increase in DNA methylation errors across the genome during aging [33]. Similarly, several other studies have demonstrated that older monozygotic twins have greater global differences in DNA methylation patterns when compared to younger twin pairs [34–36].

Since it was recognized that DNA methylation could be cell and region-specific, researchers began utilizing immunohistochemical methods to assess brain regions typically affected by AD. In individuals with AD, significant decreases in global DNA methylation have been observed in regions of the brain typically affected by AD, such as the entorhinal cortex [37]. In contrast, within the cerebellum, a region known to be spared in AD, differences in those with AD were not observed [37]. However, in the frontal cortex, a significant site of synaptic loss in AD [38], individuals with AD have been reported to have higher levels of DNA methylation when compared to those classified as cognitively unimpaired. Additional studies have reported increased global DNA methylation levels in the middle frontal gyrus and frontal cortex, in individuals diagnosed with AD compared to those without [39, 40].

The hippocampus is involved in memory formation and is one of the first regions of brain to display atrophy in AD [41]. Similar to the results outlined above, studies assessing DNA methylation in the hippocampus have been inconclusive with higher [42] and lower [43] levels of DNA methylation observed in those diagnosed with AD. Further, DNA methylation is reported to differ depending on the hippocampal subregion and cell type [43]. Similarly, Phipps et al. [44] demonstrated that pyramidal neurons, a particularly vulnerable cell type in AD, but not calretinin interneurons or microglia, have decreased DNA methylation in AD cases compared to controls.

As well as interrogating DNA methylation within brain regions, peripheral blood cells are becoming more common due to the ease of sample access. In 2015, Di Francesco et al. [45] evaluated global DNA methylation in peripheral blood mononuclear cells in AD patients and cognitively unimpaired controls, finding a significant increase in the global methylation levels of AD patients [45]. Additionally, higher global DNA methylation was observed in the presence of the APOE ɛ4 allele, highlighting the importance of combining epigenetic and genetic markers of AD [45]. In a study by Bjornsson et al. [46], peripheral blood was sampled from community cohorts to assess time-dependent changes in global DNA methylation over 11-16 years. Changes in DNA methylation over time varied between individuals, with both increases and decreases observed [46]. These findings are consistent with those published in studies of cancer, where hypomethylation (decreased levels of methylation) and hypermethylation (increased levels of methylation) work in tandem to activate and suppress oncogenes and tumor suppressor genes, respectively and concurrently [47]. It is possible that a similar interplay of gene-specific hypo- and hyper-methylation may be employed during AD progression.

Gene-specific DNA methylation

Initially, studies of gene-specific DNA methylation in AD focused on genes that encode the key proteins implicated in AD pathology, such as Aβ precursor protein (APP), presenilin 1 (PSEN1), microtubule associated protein (MAPT), and apolipoprotein E (APOE). The APOE ɛ4 allele is the strongest genetic risk factor for AD. The ApoE protein functions as the primary cholesterol carrier for the maintenance, growth and repair of neurons and is typically highly expressed in the central nervous system [48]. Wang et al. [49] reported hypermethylation of the APOE promoter region in the prefrontal cortex of AD patients, which is in line with observations of lower levels of circulating ApoE in AD patients [50]. Similarly, Karlsson [51] found an association between dementia and AD, and increased methylation levels at the APOE promoter. However, a number of studies have reported no significant difference in the methylation of APOE as a result of AD [52–55].

The analysis of DNA methylation in genes encoding other important proteins in AD pathogenesis (APP, PSEN1, BACE1, and MAPT), has likewise returned varied results. The level of DNA methylation of these genes within brain [52, 56] and blood [55, 57, 58] samples, was observed in several studies to be associated with AD diagnosis [52, 55, 58], and in other studies not associated [52, 56, 57]. Another commonly targeted gene is BDNF, which encodes the brain-derived neurotrophic factor (BDNF), a protein that is commonly found in decreased levels in AD and is involved in neuronal differentiation, plasticity, and survival [59]. Significantly higher levels of DNA methylation in the BDNF promoter in peripheral samples have been reported in patients with AD [60–62]. However, studies of peripheral blood samples have also found no significant differences in the methylation of BDNF between AD and controls [55, 63]. Neuroinflammation is known to occur during the development of AD [64], and as such the methylation of genes involved in this biological process have been investigated with regard to AD. Nicolia et al. [65] found evidence of hypomethylation of the promoter region of Interleukin-1β (IL-1β) in the early stages of AD, which increased in later stages of the disease, matching the DNA methylation of control participants. Conversely, the same study observed decreasing levels of Interleukin-6 (IL-6) DNA methylation with the progression of the disease [65]. Neuroplasticity and memory formation are also known to be compromised in AD, and a gene thought to be involved in these processes, Sorbin and SH3 Domain Containing 3 (SORBS3), has been found to be hypermethylated in the brains of AD patients [66] and mouse models of AD [67]. Finally, the assessment of genes involved in the process of DNA methylation (DNMT1, DNMT3A, DNMT3B, MTHFR) has revealed associations with AD diagnosis [49]; however, this finding was not replicated in a subsequent study [57].

The development of large-scale DNA methylation arrays has allowed for the unbiased assessment of site-specific DNA methylation across the epigenome. An epigenome-wide association analysis of longitudinal peripheral blood samples identified differentially methylated genetic loci, near known AD risk genes including BDNF, BIN1, and APOC1, to be associated with the diagnosis of mild cognitive impairment and AD in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort [62]. More recently, an additional study in the ADNI cohort identified a CpG site cg00386386, mapped to MED22, as associated with cognitive decline as measured by the Pre-Alzheimer’s Cognitive Composite (PACC) [68].

DNA METHYLATION DYNAMICS DURING AGING

Epigenetic factors, when compared to genetic factors, could be more appropriate in explaining the observed anomalies in LOAD, as DNA methylation patterns are altered during development. In particular, the epigenome is susceptible to deregulation during early embryonal and neonatal development, puberty and most notably, old age [69], the greatest risk factor for AD. Numerous studies have aimed to categorize the cellular and molecular characteristics of aging [70]. Epigenetic alterations represent one of the most crucial mechanisms driving the deterioration of cellular functions, which can be observed during aging and age-related diseases [71]. Specifically, changes in the methylome have been observed to occur over time and are thought to be one of the driving factors for the development of age-related diseases [72]. Epigenetic changes either occur randomly or are driven by internal and external influences [71]. This may explain why aging patterns can be vastly different between genetically identical individuals, such as identical twins, or between animals with similar genetic makeups [34, 73]. Studies in mammals and single cellular models (e.g., Saccharomyces cervevisiae) reveal that the epigenome suffers a progressive loss of architecture during aging, resulting in changes in chromosomal architecture, genomic integrity, and gene expression patterns [71].

Epigenetic drift

DNA methylation changes with age, regardless of the input of maintenance DNA methyltransferases. It has been hypothesized that a complex interplay of genetic, environmental (diet and lifestyle), and stochastic factors (defects in the transmission of epigenetic information through cell division) result in epigenetic drift over time, with the contribution of these factors increasing with age [74]. Epigenetic drift is non-directional, with both hyper- and hypomethylation being observed. Further, it is non-uniform across the genome and variable within individuals of the same age [75]. While epigenetic drift is known to result in unpredictable differences between the methylomes of aging individuals, some changes involve specific genetic regions and are age-associated. This indicates that DNA methylation changes are not purely random but have a role in the aging process.

Age-associated differentially methylated positions/regions

The development of high throughput arrays, such as the Illumina 27k, 450k, and EPIC arrays, has provided an impetus to studying age-specific DNA methylation patterns. These investigations reveal that epigenetic drift appears to be restricted to specific sites in the genome (referred to as age-associated differentially methylated positions/regions; a-DMPs/a-DMRs) and is not purely random [74]. Specific age-related regulatory mechanisms are thought to be involved in age-associated hyper- and hypomethylation of sites detected in a number of epigenome-wide association studies (EWAS) [74]. A study by Bormann et al., in human skin, found that methylation deteriorated with age, with hypermethylated and hypomethylated sites trending towards an intermediate (50%) level of methylation, with a smaller dynamic range [76]. While age-associated, tissue-specific changes in DNA methylation have been identified [77, 78], consistent tissue-type independent a-DMPs have been reported in multiple tissues and cell types [22, 79], and in stem cells [77]. Notably, the causes and functional consequences of age-related changes in DNA methylation, and their relationship to aging, longevity, and disease, are yet undermined [79]. It has been speculated that the identification of age-associated DNA methylation differences between diverse groups of individuals could aid in determining the epigenetic basis of aging and age-related health disparities [80]. Recently, these age-associated changes have been leveraged to aid in the development of DNA methylation clocks.

EPIGENETIC AGE AND DNA METHYLATION CLOCKS

There is great potential for the use of DNA methylation clocks as biomarkers for disease risk and predictors of life expectancy, morbidity, and mortality. Biological aging, like chronological aging, is associated with disease, morbidity and mortality, health, and cognitive and physical decline. Indeed, the varied progression in biological age may allow for the prediction of the aspects of aging outlined above. Measures of biological aging are in development for use in the prediction of disease onset, risk, and progression [81]. Several potential biomarkers of aging, including telomere length, metabolomic, transcriptomic and proteomic markers, and of interest here, DNA methylation, have been proposed [70, 82]. While many biomarkers only capture certain aspects of aging, an increasing body of literature suggests that manifestations of aging are due to epigenetic processes [83]. The “epigenetic clock” is a DNA methylation-based estimate of biological age and has been strongly correlated with chronological age. A measure of age acceleration can be calculated based on the difference between an individual’s biological age (estimated) and chronological age (actual) [84]. Accelerated aging has been linked to an increase in the pathological hallmarks of AD, reduced cognitive and physical fitness and an increase in all-cause mortality.

Approximately 28 million CpG sites exist within the human genome, usually located in clusters of hundreds or thousands, known as CpG islands. Epigenetic clocks have been developed using a select number of these CpG sites, widely distributed throughout the genome. An epigenetic clock, or a DNA methylation clock, is built from DNA methylation markers strongly correlated with chronological age (r ≥ 0.8) [82]. The process of building a DNA methylation clock utilizes supervised machine learning methods such as lasso (least absolute shrinkage and selection operator), elastic net or ridge and training against chronological age. During this process, an informative and predictive set of CpGs is identified [81]. Newer generation epigenetic clocks are specifically trained on age-related biological and health-related measures.

Broadly, epigenetic clocks have been classified as first-generation chronological DNA methylation (DNAm) clocks or second-generation biological DNAm clocks. Chronological DNAm clocks reflect age-related DNAm changes that are common among individuals and reflect the intrinsic aging process. In a pioneering study, Bocklandt et al. [85] demonstrated that DNA methylation levels from saliva could generate an accurate age prediction. Since then, numerous DNAm clocks have been developed that accurately estimate the chronological age of a sample based on DNA methylation levels within the blood and other tissue and cell types (Table 1) [83]. The DNAm clocks developed by Horvath [83] and Hannum [100] were built using similar regression models, are correlated with age and have the ability to predict all-cause mortality. The most well validated of the DNAm clocks is the Horvath clock, which is predictive in all cell types, and encompasses 353 CpGs all weakly correlated with age but when combined are highly predictive of chronological age [83]. The Hannum clock is also well correlated with age, comprising of 71 CpGs sites; however, while works in blood, it requires further calibration for use in other tissues. More recently, the Zhang DNAm clock was developed correcting for cellular composition so it can be accurately used in blood and non-blood samples [86]. Currently there is a movement towards the development of clocks encompassing a smaller number of CpG sites to minimize cost/increase time efficiency [87]. However, these estimations have been demonstrated to be unreliable when used for multiple tissues [82], and have high average errors in their age predictions when compared to clocks with a greater number of CpG sites [74, 88–90].

Table 1

Summary of DNA methylation clocks

ClockGen.No. of CpGsAge RangePlatform/ArrayMethodTissuenModelOutcomeRef.
DNAm-ageFirst318-70Illumina Infinium HumanMethylation 27KBisulphite conversion (EZ DNA Methylation Kit; Zymo)Saliva68Lasso penalized regressionAssociation with chronological age[85]
Epigenetic aging signatureFirst1916-72Illumina Infinium HumanMethylation 27KUsed publicly available data - mixedMixed130Linear regression modelAssociation with chronological age[88]
Passage NumberFirst6-Illumina Infinium HumanMethylation 27KBisulphite conversion (EZ DNA Methylation Kit; Zymo)Dermal fibroblasts &mesenchymal stem cells51Pavlidis Template MatchingTrack passage number in fibroblast cell culture[88]
EVOLV2 epigenetic marker of ageFirst19-99Illumina Infinium HumanMethylation 450KBisulphite conversion (EZ DNA Methylation Gold Kit; Zymo)Blood501Spearman correlationAssociation with chronological age[147]
Florath age predictorFirst1750-75Illumina Infinium HumanMethylation 450KBisulphite conversion (EZ DNA Methylation Kit; Zymo)Blood400Linear regression modelAssociation with chronological age[148]
DNAm-ageFirst3530-101Illumina Infinium HumanMethylation 27K &450KUsed publicly available data - mixedMixed8,000Elastic Net regressionAssociation with chronological age, age-related disease, all-cause mortality, cancer, neurodegenerative phenotypes[83]
DNAm-ageFirst7119-101Illumina Infinium HumanMethylation 450KBisulphite conversionBlood656Elastic Net (combination of Lasso and ridge regression)Association with chronological age and all-cause mortality[101]
DNAm-ageFirst30-78Illumina Infinium HumanMethylation 27K &450KEpiTect Bisulphite Kit; QIAGENBlood575Multivariate linear modelAssociation with chronological age, clinical/lifestyle factors, telomere length, related disease phenotypes[149]
DNAm-ageFirst9919-101Illumina Infinium HumanMethylation 27K &450KEpiTect Bisulphite Kit; QIAGENBlood656Multivariate linear modelAssociation with chronological age, clinical/lifestyle factors, telomere length, related disease phenotypes[149]
DNAm-ageFirst1020-78Illumina Infinium HumanMethylation 27K &450KEpiTect Bisulphite Kit; QIAGENBlood575Multivariate linear modelAssociation with chronological age, clinical/lifestyle factors, telomere length, related disease phenotypes[149]
Huang age predictorFirst59-75PyrosequencingEpiTect Bisulphite Kit; QIAGENBlood89Linear regression modelAssociation with chronological age – for forensic use[90]
Zbiec-Piekarska age predictorFirst52-75PyrosequencingEpiTect Bisulphite Kit; QIAGENBlood420Multivariate linear modelAssociation with chronological age – for forensic use[89]
Buccal Cell SignatureFirst51-85PyrosequencingBisulphite conversion (EZ DNA Methylation Kit; Zymo)Buccal Cells55Multivariate linear modelEpigenetic age predictor[150]
Cho Model 2First520-74PyrosequencingBisulphite Conversion (EZ DNA Methylation - Lightning ™ Kit; Zymo)Blood100Multivariate linear modelAssociation with chronological age[151]
DNAm-age from SalivaFirst718-65Illumina Infinium HumanMethylation 450K &SNaPshotBisulfite ConversionSaliva54Linear regression modelEnabled age prediction in saliva with high accuracy[152]
Skin and BloodFirst3910-94Illumina Infinium HumanMethylation 450K &EPICUsed publicly available data - mixedMixed896Elastic Net regressionPrediction of age[153]
Zhang age predictorFirst5142-104Illumina Infinium HumanMethylation 450K &EPICUsed publicly available data - mixedBlood and Saliva13,661Elastic NetAssociation with chronological age[86]
Zhang age predictorFirst319,6072-104Illumina Infinium HumanMethylation 450K &EPICUsed publicly available data - mixedBlood and Saliva13,661Best Linear Unbiased PredictionAssociation with chronological age[86]
PedBE ClockFirst940-20Illumina Infinium HumanMethylation 450K &EPICUsed publicly available dataBuccal epithelial cells1,721Elastic NetTool for measuring biological age in children[154]
DNAm-ageFirst519-70SNaPShotBisulphite conversion (Imprint™ DNA Modification Kitl Sigma-Aldrich)Blood, saliva, and buccal cells448Linear RegressionAge prediction[155]
DNAm-ageFirst524-86Bisulphite PCRBisulphite conversion (EZ DNA Methylation Gold Kit; Zymo)Blood51Linear RegressionAge prediction for forensic use in deceased individuals[156]
Blood-Bone-Tooth Age Prediction ModelFirst431-94Sanger SequencingBisulphite conversion (EZ DNA Methylation Gold Kit; Zymo)Blood, bone, and tooth185Multivariate linear modelAge prediction for forensic use in alive and deceased individuals[157]
PC Horvath 1  PC Horvath 2 PC Hannum PC PhenoAge PC GrimAge PC DNAmTLSecondPCs trained on 78,464 CpGs0-105-0.3-10119-10121-10024-9224-92Illumina Infinium HumanMethylation 450K &EPIC &Elysium custom arrayUsed publicly available data - mixedMixed4,2978956564,5052,7542,754Principle components and elastic net regressionPC clocks show reduced technical noise allowing for improved detection of clock associations, intervention effects and reliable longitudinal tracking[158]
Cortical DNAm ClockSecond3471–104Illumina Infinium HumanMethylation 450K arrayUsed publicly available data - mixedHuman cortex1,397Elastic Net regressionClock outperforms previously reported clocks[138]
Boroni SkinSecond2,26618–95Illumina Infinium HumanMethylation EPIC arrayUsed publicly available dataHuman skin508Elastic Net regressionSkin specific DNAm age predictor[159]
epiTOCSecond38519–101Illumina Infinium HumanMethylation 450K arrayUsed publicly available dataBlood656Linear regression modelAssociation with mitotic age, cancer[92]
Epigenetic age predictorFirst990–78Illumin Infinium HumanMethylation 27k arrayLothian Birth Cohorts 1921 and 1936Blood575Cox linear regressionAssociation with life expectancy[160]
Biological age predictorFirst8Methylation-sensitive single-nucleotide primer extension (MS-SNuPE)Used publicly available data - mixedBlood390Multiple linear regressionSimplified assay for epigenetic age estimation[161]
All-cause mortalitySecond1031–82Illumina Infinium HumanMethylation 450K arrayBisulphite conversionBlood1,000Lasso penalized regressionAssociation with all-cause CVD, cancer, smoking behavior[97]
MiAgeSecond286Illumina Infinium HumanMethylation 450K arrayUsed publicly available data - mixed8 Cancer cell types4,020Linear regression modelAssociation with mitotic age, cancer outcome and survival prediction[93]
DNAm PhenoAgeSecond513>20Illumina Infinium HumanMethylation 27K, 450K &EPIC arrayUsed publicly available data - mixedBlood9,926Cox penalized regressionAssociation with all cause and cause specific mortality, telomere length, survival, smoking status[94]
DNAm GrimAgeSecond1,030-Illumina Infinium HumanMethylation 450K &EPIC arrayUsed publicly available data - mixedBlood1,731Elastic Net, Cox regressionAssociation with morbidity and mortality, survival, cognitive decline, telomere length[96]
DNAmTLSecond14022-93Illumina Infinium HumanMethylation 450K &EPIC arrayUsed publicly available data - mixedBlood2,256Multiple regression modelsAssociation with chronological age, telomere length, morbidity and mortality, physical function[95]
DunedinPoAmSecond140-Illumina Infinium HumanMethylation 450K &EPIC arrayUsed publicly available data - mixedBlood1,037Elastic NetBlood DNAm measure sensitive to variation in pace of biological aging in individuals of same age[98]
epiTOC2Second16319-101Illumina Inifinium HumanMethylation 450K, Whole genome bisulfite sequencing, RNA-SeqV2Used publicly available data - mixedMixed>2,000Formal dynamic modelPrediction of increased mitotic rate in cancer[162]
HypoclockSecond--Whole genome bisulfite sequencing, Illumina Inifinium HumanMethylation 450KUsed publicly available data - mixedMixed-Gaussian mixture modelDNA methylation loss in late-replicating domains linked to mitotic cell division[163]
DunedinPACESecond173Illumina Infinium HumanMethylation 450K &EPIC arrayThe Dunedin Study, the Understanding Society Study, the Normative Ageing Study, the Framingham Heart Study &the Environmental Risk Longitudinal Twin StudyMixed1,037Elastic Net regressionAssociated with morbidity, mortality, and disability. Novel blood biomarker of the age of pacing for gerontology and geroscience.[164]
GrimAge 2Second1,030Illumina Infinium HumanMethylation 450K &EPIC arrayUsed publicly available data - mixedMixed13,399Elastic Net, Cox regressionGrimAge 2 outperforms GrimAge and is an epigenetic biomarker of mortality and morbidity risk[165]

Biological DNAm clocks, considered “second generation” clocks, are reflective of DNAm changes that vary between individuals and may highlight age-related phenotypes and external factors that may influence age-related DNAm [91]. Specifically, these clocks have been developed against age-related biological processes such as mitotic activity (e.g., epiTOC [92]; MiAge [93]), clinical measures of phenotypic aging (PhenoAge [94]), telomere length (DNAmTL [95]), plasma biomarkers and physiological and stress factors (GrimAge [96]), and all-cause mortality (Zhang [97]). Finally, the Dunedin Pace of Ageing clock, has incorporated the rate of change of 18 blood chemistry and organ system function measurements into the calculation of its score to improve predictive utility [98].

Age acceleration and health and disease

As outlined above, and summarized in Table 1, DNAm clocks have been developed using several different training models and as such reflect different aspects of disease and the aging process. It has been hypothesized that DNA methylation age (DNAm age) measures the cumulative effect of an epigenetic maintenance system [83]. Studies have demonstrated that DNAm age of blood predicts all-cause mortality in later life, which suggests that methylation age is a process that reflects aging [99]. However, it is more probable that the epigenetic clock is a result of the emergent properties of the epigenome [83]. Several studies have proven the usefulness of using age acceleration as a measure for healthy biological aging. For example, semi-supercentenarians and their offspring have a lower biological age compared to chronological age, or age deceleration, when compared to age-matched controls [100]. Similarly, women have shown to have less significant age acceleration compared to men, which is in line with their representative longer lifespans [101, 102].

Moreover, a number of studies have demonstrated the utility of age acceleration as a predictor of disease risk and mortality. A study assessing four longitudinal cohorts identified an association between all-cause mortality and increased epigenetic age in blood [103]. Further, a number of genetic syndromes have been reported to accelerate biological aging, such as Down syndrome [104] and Werner’s syndrome [105]. There is ample evidence of premature biological aging in those with viral infections such as HIV [106, 107], individuals with decreased mental and physical fitness [108], body mass index and metabolic disease [109], non-alcoholic steatohepatitis [110], obesity [111], insomnia [112], and extreme stress [113]. Additionally, individuals with neurodegenerative diseases such as Parkinson’s disease [114], Huntington’s disease [115], and AD [116] show accelerated biological aging.

METHYLATION AGE AND AD PHENOTYPES

The limited research which has been undertaken to explore the relationship between DNAm age and AD related phenotypes has yielded inconsistent findings. The association between DNAm age and AD phenotypes has primarily been characterized by measures of DNA methylation within the blood [108, 117–125], while only one study investigated brain samples [116].

Further, research has focused largely on “first generation” estimates of DNAm age, specifically the Horvath and Hannum clocks, which have been trained to approximate chronological age. Age acceleration, as estimated using the Horvath DNAm age clock in blood, has been associated with lower cognition cross-sectionally [108, 116], longitudinal cognitive decline [122, 123], and reduced white matter integrity. Additionally, Horvath age acceleration in prefrontal cortex samples has been associated with diffuse and neuritic Aβ plaques and higher global Aβ burden [116]. Despite these findings, a number of studies have also reported no significant association between Horvath DNAm age acceleration and cognitive functioning or brain volume, both cross-sectionally [117, 121] and longitudinally [108, 121]. Likewise, inconsistent findings have been reported when assessing DNAm age using the Hannum estimation. Hannum age acceleration has been associated with smaller hippocampal volume [125], but conversely, increased brain connectivity and decreased mean diffusivity (improved axonal integrity) in MRI quantified studies [117]. Additionally, several studies have failed to identify any significant associations between Hannum age acceleration and cognitive functioning [117, 121] or longitudinal cognitive decline [121, 124].

The association between “second generation” biological DNAm age estimates and AD phenotypes has also been explored, although less broadly. GrimAge acceleration has been associated with lower cognitive ability, accelerated cognitive decline, lower intelligence test scores, lower white matter and grey matter volumes, total brain volume, and higher volume of white matter hyperintensities [119]. However, both DNAm telomere length and the Pace of Ageing score were not associated with cognitive decline [120]. Inconsistency in the current literature underscores the need for further investigation of DNA methylation before it can be considered as a robust biomarker for aging-related disease such as AD.

CONSIDERATIONS WHEN UTILIZING DNA METHYLATION

The “tissue issue”

The studies outlined above, and summarized in Table 2, have presented DNA methylation findings from a range of tissue and cell types. As is evident in Table 2, there has been a trend towards large-scale discovery studies (EWAS) using peripheral blood and a shift away from studies exploring global methylation. When exploring these findings, it is important to consider that DNA methylation levels at specific CpG sites typically cannot be compared across different tissue types and DNA methylation at many CpG sites is only weakly correlated across tissue types such as the brain (cortex and cerebellum) and blood [21]. Tissue specificity is one of the most common uncertainties that arise when using peripheral tissues in association studies of DNA methylation. When considering the utility of DNA methylation markers in either the target organ, the brain, or the periphery, the blood, it is important to be aware of the challenges posed by sample types (Table 3).

Table 2

Summary of DNA methylation studies in AD

PhenotypeTissueSample sizeAge range / mean agePlatformGene/sOutcomeRef.
ADFrontal cortex1 case-Southern blotTargetedHypomethylation of APP gene in AD[166]
ADBrain tissue72 (48 case, 24 control)52-93ImmunohistochemistryTargeted genesMethylation levels of PP2AC decreased in AD-affected brain regions[167]
ADBrain tissue12517 weeks-104MethyLight PCRTargeted genesAD-associated differences in SORBS3 and S100A2, DNA methylation dynamically regulated throughout life span[66]
ADPrefrontal cortex and lymphocytes34 brain (24 AD, 10 control); 10 lymphocytes (6 AD, 4 control)AD 81, control 80MALDI-TOF mass spectrometryTargeted genesAge-specific epigenetic drift in AD, significant interindividual epigenetic variability in PSEN1, APOE, MTHFR, DNMT1[49]
ADTemporal neocortex2 (1 case, 1 control)Case 76, control 79ImmunohistochemistryGlobalReduced levels of DNA methylation in the temporal neocortex of the AD twin[30]
ADFrontal cortex &hippocampus66 (43 case, 26 control)46-93MALDI-TOFTargeted genesNo differences in percentage methylation between cases and controls[56]
ADEntorhinal cortex layer II40 (20 case, 20 control)60-97ImmunohistochemistryGlobalDNA methylation status in entorhinal cortex layer II neurons is highly diminished in AD[37]
ADBrain tissue6-Bisulfite sequencingTargeted genesNo associations of APP with AD[168]
ADPeripheral blood83 (43 case, 38 control)55-87Quantitative bisulfite-PCR pyrosequencingTargeted repetitive elementsLINE-1 methylation increased in AD compared to controls[169]
ADFrontal cortex20 (10 case, 10 control)Case 70.60, control 70.20RT-PCRTargeted genes &global methylationHypomethylation of COX-2 and NF-κB promoter in AD, hypermethylation of BDNF and CREB, increased global methylation in AD[40]
ADFrontal cortex50 discovery (12 case, 12 control); 26 validation (13 case, 13 control)Case 78.2, control 78.3Illumina Infinium HumanMethylation 27K arrayEWASHypomethylation of TMEM59 in AD[170]
ADPeripheral blood mononuclear cells66 (33 case, 33 control)Case 79.47, control 79.98Methylation-specific primer RT-PCRTargeted genesDecreased DNA methylation at FAAH gene promoter[171]
ADPeripheral blood mononuclear cells60 (32 case, 28 control)-Methylation-specific primer RT-PCRTargeted genesPromoter methylation of PIN1 in AD increased compared to controls[172]
ADEntorhinal, auditory cortices, hippocampus, and blood99 (44 case, 55 control)Case blood 75.47, case brain 80.1, control blood 72.09, control brain 78.5Mass spectrometryTargeted genesNo AD-associated differences in SNAP25 promotor DNA methylation[173]
ADEntorhinal, auditory cortices, hippocampus, and blood22 brain (12 case, 10 control);84 blood (36 case, 48 control)Case blood 75.4, case brain 81, control blood 72.1, control brain 78.5MALDI-TOF mass spectrometryTargeted genesBrain SORL1 DNA methylation was significantly higher in brain than in blood in both cases and controls[174]
ADBrain and blood57 blood (34 case, 23 control); 22 brain (12 case, 10 control)Blood – case 75.47, control 72.09; Brain – case 81, control 78.50Mass spectrometryTargeted genesNo differences in DNA methylation between cases and controls[175]
ADBrain tissue391 (170 cases, 221 control)Case 78.68, control 76.59PyrosequencingTargeted genesHypermethylation in APP, MAPT and GSK3B in sporadic AD, which is more prominent in APOE ɛ4[52]
ADHippocampus20 (10 case, 10 control)Case 75.36, control 77.91ImmunohistochemistryGlobalDecreased global DNA methylation in the hippocampus of AD[43]
Preclinical AD/ADHippocampus/ parahippocampus and cerebellum17 (12 case, 5 control)Preclinical 89.4, AD 77.4, control 84.6ImmunohistochemistryGlobalAltered DNA methylation patterns in vulnerable brain regions prior to onset of clinical symptoms[42]
ADEntorhinal, auditory cortices, hippocampus22 (12 case, 10 control)Case 81, control 78.5-Mass spectrometryTargeted genesNo AD-associated differences in promoter methylation[176]
ADPeripheral blood55 (27 case, 28 control)Case 79.47, control 79.98Fluorescence-based RT-PCRTargeted genesDecreased DNA methylation at the 5-LOX gene promoter in AD compared to controls[177]
ADGray matter58 (26 case, 26 control)51-98High content analysis methodGlobal methylationIncrease in global 5mC and 5hmC levels in AD[39]
ADBrain tissue and peripheral blood166 (122 brain discovery, 57 blood; 144 brain replication)67-105Illumina Infinium HumanMethylation 450K arrayTargeted genesANK1 differentially methylated and associated with neuropathology in the entorhinal cortex[54]
ADBrain tissue825 (708 discovery, 117 test)>53Illumina Infinium HumanMethylation 450K arrayEWAS71 CpGs associated with AD pathology, 11 validated in independent cohort[53]
ADMiddle frontal gyrus and middle temporal gyrus58 (29 case, 29 control)54-98ImmunohistochemistryGlobalGlobal hypermethylation in AD compared to controls[39]
ADPeripheral blood58 (28 case, 30 control)Case 77.4, control 74.9Methylation-sensitive high-resolution melting quantitative assayGlobalNo differences in LINE-1 methylation between AD and controls[178]
ADHippocampus20 (discovery 15 case, 5 control; validation – 25 case, 25 control)Discovery – case 82.4, control 82.4; Validation case 79.1, control 64.2Illumina Infinium HumanMethylation 27K arrayEWASAD-associated hypermethylation of promoter region of DUSP22[179]
ADDorsolateral pre-frontal cortex740Mean age at death 88Illumina Infinium HumanMethylation 450K arrayTargeted genes16 CpGs associated with neuritic amyloid plaques from 11 AD susceptibility gene regions[180]
ADDorsolateral prefrontal cortex74066-108.3Illumina Infinium HumanMethylation 450K arrayTargeted genesBrain DNA methylation in 5 AD-risk loci is associated with pathological AD[181]
ADEntorhinal cortex and cerebellum26 (14 case, 12 control)38-99Immunohistochemistry &ELISAGlobalNo differences in global methylation between AD and controls[182]
ADHippocampus42 (30 case, 12 control)19-98Bisulfite cloning sequencing further measured by 5hmCTargeted genesTREM2 methylation was significantly higher in AD compared to controls[183]
ADPeripheral blood260 (80 case, 160 control)65-96Melting Curve Analysis-Methylation Assay (MCA-Meth)Targeted genesUQCRC1 was highly methylated in AD, UQCRC1 was significantly associated with gene expression of NRD1, DDT, CTSB and CTSD[184]
ADSuperior temporal gyrus390Cohort 1 – case 85.4, control 77.6; Cohort 2 – case 88.0, control 82.1; Cohort 3 – case 78.4, control 84.1Illumina Infinium HumanMethylation 450K array &pyrosequencingTargeted genesAD-associated hypermethylation in CpG site 289bp upstream of TREM2 in 3 independent cohorts[185]
ADSuperior temporal gyrus68 (34 case, 34 control)66–92Illumina Infinium HumanMethylation 450K arrayEWASIdentified 479 DMRs, with the majority being hypermethylated[186]
ADInferior temporal gyrus20 (15 case, 5 control)58–91ImmunohistochemistryTargetedEpigenetic dysregulation occurs in astrocytes and neurofilament-positive pyramidal neurons in AD[44]
MCIPeripheral blood96 (48 MCI, 48 control)Bisulfite Pyrosequencing, Dual Luciferase AssaysTargeted genesHypermethylation of OPRK1 in females, OPRM1 differential methylation associated with MCI[187]
ADDiscovery - Prefrontal cortex &superior temporal gyrus; test - mixed1004 (147 discovery, 857 test)64–95Illumina Infinium HumanMethylation 450K arrayTargeted genesIncreased DNA methylation associated with AD neuropathology[188]
ADFrontal cortex, temporal cortex, &occipital cortex159 (91 case, 68 control)18–97Illumina Infinium HumanMethylation 450k arrayEWASIdentified numerous genes with cell-type-specific methylation signatures[189]
ADPeripheral blood84 (45 case, 39 control)AD 73.56, control 75.33Illumina Infinium HumanMethylation EPIC arrayEWASHypomethylation of B3GALT4 and ZADH2 associated with the level of Aβ and tau in CSF[190]
ADCortical pyramidal layer32 (18 case, 14 control)AD 77.22, control 70.29Illumina Infinium HumanMethylation 450k arrayEWASDifferential hypermethylation in several genomic regions, including HOXA3, GSTP1, CXXC1-3 & BIN1[191]
ADBrain tissue51 (40 case, 11 control)41–99Illumina Infinium HumanMethylation 450k array &whole genome bisulfite sequencingEWASHypomethylation of KIAA0566 associated with age and presence of NFT pathology[192]
ADPrefrontal cortex10154–105Bisulfite padlock probe techniqueEWAS1224 differentially methylated enhancer regions identified, enhancement in the DCSAML1 gene which targets BACE1[193]
ADHippocampus, entorhinal cortex, dorsolateral pre-frontal cortex and cerebellum73 (24 case, 48 control)53–80Illumina Infinium HumanMethylation 450k arrayEWAS858 differentially methylated sites, annotated to 772 genes, some novel. Sites were overrepresented in AD genetic risk loci[194]
ADBrain tissue60Mean age entorhinal cortex 77.7, superior temporal gyrus 77.6, cerebellum 78.4Bisulfite pyrosequencingTargeted genesDNA hypermethylation in the entorhinal cortex[195]
ADMiddle temporal gyrus and peripheral blood80 brain tissue (45 case, 35 control); 96 peripheral blood (54 AD converters and 42 non-converters)Illumina Infinium HumanMethylation 450K arrayTargeted genesDifferential methylation in OXT promoter in brain of AD, same region in blood associated with converters[196]
ADBrain tissue296 (96 discovery, 104 validation cohort 1, 96 validation 2)Discovery 81.2; Validation 1 84.9; Validation 2 85Illumina Infinium HumanMethylation 450K arrayEWASWNT5B differentially methylated; two differentially methylated regions in ANK1 & ARID5B;[197]
ADBrain Tissue70688 (mean age)Illumina Infinium HumanMethylation 450K arrayTargeted genes249 &115 variably methylated probes associated with Amyloid-β &Neurofibrillary tangles – most not overlapping with DMPs[198]
ADHippocampus38 (26 case, 12 control)19-98Illumina Infinium HumanMethylation 450K arrayTargeted genes118 differentially methylated positions identified in AD hippocampus[199]
ADDorsolateral pre-frontal cortex &122141-104Illumina Infinium HumanMethylation EPIC arrayEWAS334 DMPs associated with AD pathology[200]
MCI/ADPeripheral blood284 (AD 86, MCI 109, control 89)AD 76.8, MCI 75.1, Control 73.8Illumina Infinium HumanMethylation EPIC array and pyrosequencingEWAS12 AD-associated hypermethylated probes identified in HOXB6 and validated by pyrosequencing[201]
ADPeripheral blood653AD 77.19, MCI 72.58, 76.23Illumina Infinium HumanMethylation EPIC arrayEWASIdentified differentially methylated loci were near brain/neurodegeneration related genes[62]
ADBrain tissue134 (72 case, 62 control)AD 81.00, Control 80.50Illumina Infinium HumanMethylation EPIC arrayEWAS22 DMPs and 30 DMRs associated with pathology, novel DMPs and DMRs discovered, replicated in independent cohort[202]
ADPeripheral blood96 test &96 replication (48 case, 48 control), 95 non-AD dementiaTest - AD 72.7, control 71.9;Replication – AD 70.5, control 70.2Sanger SequencingTargeted genesCR1, CLU & PICALM methylation significantly lower in AD[203]
ADPrefrontal cortex, entorhinal cortex, and superior temporal gyrus37 (29 case, 18 control)AD 84.2, control 88.4Illumina Infinium HumanMethylation 450k arrayEWASPsychosis-associated epigenetic changes in a number of loci, genes enriched in schizophrenia-associated genetic and epigenetic variants[204]
ADPeripheral blood and brain tissue50636 (7540 case, 43096 control)Illumina Infinium HumanMethylation 450k arraySummary-based EWAS152 CpGs AD-associated genes corresponding to 113 genes. 10 genes had significant probes in both blood and AD-specific analyses[205]
ADPeripheral blood45256–80Illumina Infinium HumanMethylation 450K arrayEWAS238 gene network identified, including APP and several novel candidate genes[206]
ADBrain tissue2116 (1455 discovery, 661 replication)>65Illumina Infinium HumanMethylation 450K arrayMeta-analysis of six EWAS220 CpGs associated with neuropathology, annotated to 121 genes, 84 novel[207]
ADMiddle temporal gyrus296 (198 case, 98 control)AD 83.8, control 82.2Illumina Infinium HumanMethylation 450K arrayEWAS5246 CpGs and 832 DMRs, some overlap with previous EWAS, some novel[208]
Table 3

Advantages and Disadvantages of commonly used cell types

AdvantagesDisadvantages
Organ of interest (e.g., brain)Desired organ/tissue/cell directly targetedNeed for highly trained professionals,
Relatively expensive compared to other methods,
Difficult/not feasible to obtain samples in some instances, e.g., brain tissue,
Heterogenous in cellular makeup, Cell numbers change in disease
Whole bloodMinimally invasive, cost-effective,Heterogenous in cellular makeup,
Minimally trained professionals required,Some special training required i.e., phlebotomy,
Repeated sampling possible (longitudinal disease/intervention/treatment tracking possible)Potentially peripheral to target of interest
Buccal cellsNon-invasive,Potential to be low in cellular material,
Cost-effective,Contamination from food, beverages, kissing, etc.,
Easily obtained – no specially trained professionals requiredHeterogenous in cellular makeup,
Potentially peripheral to target of interest
MixedAbility to pool multiple publicly available sources of data for increased sample size and analytical powerHeterogenous cell types makes interpretation of results difficult – can only draw general conclusions.
Methods of sample collection/sample handling/bisulfite conversion/data collection may differ between datasets – increased variance due to large number of uncontrollable variables
SalivaNon-invasive and convenient sampling, repeated sampling possible, easy collection without the need for special trainingPotential to be low in cellular material, contamination from food, beverages, etc.,Heterogenous in cellular makeup,Potentially peripheral to target of interest
SkinRelatively non-invasive in some instances, i.e., skin scrapings,Experiments can be replicated in vitro with high fidelityPotentially invasive, i.e., in the instance of skin biopsies,Heterogenous in cellular makeup,Skin is very exposed to environmental stimuli such as the sun (UV exposure) and pollution
Stem CellsEmbryonic stem cells (ESCs) can be cultured indefinitely without compromising pluripotency,Can be differentiated into different cell types in vitro,HomogenousInduced pluripotent stem cell (iPSCs) reprogramming is long, complex and inefficient,Non-CpG methylation needs to be considered when using ESCs,Low passage human iPSCs retain ‘epigenetic memory’, which biases characteristics of tissue of origin,Non-physiological methylation patterns introduced in vitro

When assessing brain tissues in epigenetic clock analysis of neurodegenerative or neuropsychiatric disorders, limited information is available regarding the specific brain regions and cell types present, which has been reflected in differences in epigenetic patterns and gene function [25]. However, with regard to cell-type specific methylation, computational methods, such as the Houseman [126] (cellular proportion estimates in blood) and Guintivano [127] (neuronal and non-neuronal proportion estimates) methods have been developed to address the issues resulting from varying cell-type proportions between individuals [127, 128]. The gold standard for controlling for cell proportions is using sorted cell populations such as laser capture microdissection or fluorescence-activated cell sorting; however, these are relatively expensive options and hard to achieve logistically in large scale studies. Importantly, brain tissue is typically obtained postmortem, which needs to be considered when interpreting data [129]. Specifically, are epigenetic patterns observed post-mortem consequence of the disease process itself, rather than causal? Additionally, detected epigenetic signals could be due to the cause of death, tissue pH and the premortem agonal state [130]. Careful study design via targeted validation employing cell sorting within the tissue of interest can help isolate cell type-specific changes, resulting in a clearer explanation of observed biological effects, such as which epigenetic markers mediate risk for disease or associate with a phenotype. Characterizing and exploring the effects of cellular heterogeneity is a crucial step in the analytical pathway of methylome-wide DNAm data in heterogenous tissues, especially peripheral blood, thus we recommend that it not be overlooked. Moving forward, more research focus needs to be placed on cross-tissue study designs to identify DMPs/DMRs common across two or more cell types such as brain tissue and peripheral blood to uncover common correlations. If the aforementioned limitations can be overcome, the next major issue is sample availability and the capacity to undertake appropriately powered studies to detect associations, which may besubtle.

Conversely, sampling of peripheral tissue offers many advantages over target tissues, such as ease of access, large sample sizes, replication opportunities and longitudinal sampling, both prior to disease onset and during treatments and/or interventions. However, as with brain samples, several limitations need to be considered when performing and interpreting analyses utilizing DNA methylation results from peripheral blood. In addition to tissue specificity, cellular heterogeneity is a significant potential confounder of quantifying DNA methylation levels in the blood and should be considered when interpreting results. Whole blood is composed of many different cell types, including neutrophils, lymphocytes, monocytes, eosinophils and others, and the proportions of these cell types can be influenced by numerous factors such as bacterial or viral infections, inflammation, diet, stress, medication, and environmental exposures [131]. There is an increasing number of studies utilizing peripheral DNA methylation; however, very few take into consideration the proportions of different cell types, and thus do not control for them in analyses, which likely drives some observed associations or masks potential differences between groups [132].

There is currently little evidence to suggest that AD-related epigenetic modifications in the brain are reflected in the periphery [133]. Several studies have measured DNA methylation in specific brain regions and in blood; however, only a few studies reported changes in both tissues, which are speculated to be driven by underlying genetic repeats within the C9orf72 gene [134–136]. While a peripheral biomarker, such as one identifiable in blood, has been proposed as a useful surrogate marker to monitor disease status in major organs or systems, it could independently reflect disease processes in the periphery and be important in disease tracking and prognosis.

Underestimation of age in epigenetic clocks

Several studies have now reported an underestimation of age [137–140], with deviations in biological age from chronological age particularly noticeable in the higher age brackets. This observation is likely due to several contributing factors. One hypothesized factor contributing to an underestimation of age is the saturation of methylation sites, where large proportions of sites are approaching the lower (0%) and upper (100%) limits of methylation, caused by epigenetic drift in aging individuals [141]. Dhingra et al. [139] speculate that differences in results between epigenetic clocks within the same cohort are likely caused by missing probes between arrays, irrespective of probe imputation procedures leading to an underestimation of age. Additionally, survivorship bias, a type of selection bias, may be driving the underestimation of age, particularly in cohorts with relatively high mean age. Individuals with a negative age acceleration are maintained within the sample, and those with a positive age acceleration, are not present within the sample, leading to a skewed cohort. The underlying populations on which the clocks are trained are also an important factor to consider when interpreting results. For example, the Hannum and Horvath clocks were trained on chronological age and given their overall high accuracy in estimating age, they are likely not ideal estimators of biological age. A clock that is capable of predicting chronological age perfectly would contain no information on variation in biological age at an individual level [16]. For this reason, clocks such as the PhenoAge clock, which were trained on age-related and disease phenotypes in combination with chronological age, are preferred when investigating biological age more directly. Further, DNAm age estimates may be susceptible to a ceiling effect, where a plateau is observable. The Horvath clock requires an age transformation past a certain age threshold, indicating that the relationship between chronological age and estimated age is not linear and, past a certain age, estimates need to be treated differently [83]. Recently machine learning and deep learning approaches have been employed in an attempt to mitigate shortfalls such as the ones mentioned here, and have observed more accurate age predictions for older individuals than the original clocks [142].

CONCLUSION

DNA methylation has been implicated in a number of diseases in the last decade including, cancer [143], heart disease [144], autoimmune disorders [145], and neurodegenerative diseases [146]. Many studies, leveraging both global and site specific CpG DNA methylation, have provided evidence for age-associated changes in DNA methylation. However, more recently, clusters of CpG sites whose DNA methylation status provide an accurate measure of chronological age have been identified. Together these sites provide the foundation for epigenetic clocks. Although initially, the clock’s main purpose was to serve as a biomarker of chronological age, there is growing evidence suggesting that epigenetic clocks may have value as biomarkers of biological age. Emerging research supports the idea that age and age-associated diseases or phenotypes are characterized by increased age acceleration in comparison to healthy individuals. Consequently, research focus has been placed on epigenetic clocks as biomarkers to detect aging and aging-associated diseases. The utility of peripheral blood as a biomarker of diseases in specific organs is often questioned, given cells in the periphery may not accurately capture and reflect more central disease processes. However, a good peripheral biomarker does not have to mirror disease-associated changes in the brain. Rather, it could represent a co-occurring peripheral response to central pathology. Despite its limitations, peripheral blood remains the most obvious surrogate tissue for epigenetic studies. DNA methylation is a highly promising biomarker for AD disease-associated pathology. Further investigation of its association with disease trajectories will be essential in identifying individuals at risk of age-related diseases, thus optimizing treatment and intervention strategies.

ACKNOWLEDGMENTS

The authors have no acknowledgments to report.

FUNDING

LM is supported by an Edith Cowan University Higher Degree by Research scholarship and a scholarship top-up from the Australian Alzheimer’s Disease Research Foundation. TP is supported by an Edith Cowan University Strategic Research Fellowship. The authors have no other funding to report that is relevant to this manuscript.

CONFLICT OF INTEREST

The authors have no conflict of interest to report.

REFERENCES

[1] 

Duong S , Patel T , Chang F ((2017) ) Dementia: What pharmacists need to know. Can Pharm J (Ott) 150: , 118–129.

[2] 

Perl DP ((2010) ) Neuropathology of Alzheimer’s disease. Mt Sinai J Med 77: , 32–42.

[3] 

Dickerson BC , Bakkour A , Salat DH , Feczko E , Pacheco J , Greve DN , Grodstein F , Wright CI , Blacker D , Rosas HD , Sperling RA , Atri A , Growdon JH , Hyman BT , Morris JC , Fischl B , Buckner RL ((2009) ) The cortical signature of Alzheimer’s disease: Regionally specific cortical thinning relates to symptom severity in very mild to mild AD dementia and is detectable in asymptomatic amyloid-positive individuals. Cereb Cortex 19: , 497–510.

[4] 

DeTure MA , Dickson DW ((2019) ) The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener 14: , 32.

[5] 

Guerreiro R , Bras J ((2015) ) The age factor in Alzheimer’s disease. Genome Med 7: , 106.

[6] 

Hersi M , Irvine B , Gupta P , Gomes J , Birkett N , Krewski D ((2017) ) Risk factors associated with the onset and progression of Alzheimer’s disease: A systematic review of the evidence. Neurotoxicology 61: , 143–187.

[7] 

Galvin JE ((2017) ) Prevention of Alzheimer’s disease: Lessons learned and applied. J Am Geriatr Soc 65: , 2128–2133.

[8] 

Silva MVF , Loures CdMG , Alves LCV , de Souza LC , Borges KBG , Carvalho MdG ((2019) ) Alzheimer’s disease: Risk factors and potentially protective measures. J Biomed Sci 26: , 33.

[9] 

McKhann GM , Knopman DS , Chertkow H , Hyman BT , Jack CR Jr. , Kawas CH , Klunk WE , Koroshetz WJ , Manly JJ , Mayeux R , Mohs RC , Morris JC , Rossor MN , Scheltens P , Carrillo MC , Thies B , Weintraub S , Phelps CH ((2011) ) The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 7: , 263–269.

[10] 

Olsson B , Lautner R , Andreasson U , Öhrfelt A , Portelius E , Bjerke M , Hölttä M , Roséen C , Olsson C , Strobel G , Wu E , Dakin K , Petzold M , Blennow K , Zetterberg H ((2016) ) CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol 15: , 673–684.

[11] 

Dubois B , Hampel H , Feldman HH , Scheltens P , Aisen P , Andrieu S , Bakardjian H , Benali H , Bertram L , Blennow K , Broich K , Cavedo E , Crutch S , Dartigues JF , Duyckaerts C , Epelbaum S , Frisoni GB , Gauthier S , Genthon R , Gouw AA , Habert MO , Holtzman DM , Kivipelto M , Lista S , Molinuevo JL , O’Bryant SE , Rabinovici GD , Rowe C , Salloway S , Schneider LS , Sperling R , Teichmann M , Carrillo MC , Cummings J , Jack CR Jr. ((2016) ) Preclinical Alzheimer’s disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 12: , 292–323.

[12] 

Qazi TJ , Quan Z , Mir A , Qing H ((2018) ) Epigenetics in Alzheimer’s disease: Perspective of DNA methylation. Mol Neurobiol 55: , 1026–1044.

[13] 

Moore LD , Le T , Fan G ((2013) ) DNA methylation and its basic function. Neuropsychopharmacology 38: , 23–38.

[14] 

Robertson KD ((2005) ) DNA methylation and human disease. Nat Rev Genet 6: , 597–610.

[15] 

Luo C , Hajkova P , Ecker JR ((2018) ) Dynamic DNA methylation: In the right place at the right time. Science 361: , 1336–1340.

[16] 

Field AE , Robertson NA , Wang T , Havas A , Ideker T , Adams PD ((2018) ) DNA methylation clocks in aging: Categories, causes, and consequences. Mol Cell 71: , 882–895.

[17] 

Wu M , Rinchik EM , Wilkinson E , Johnson DK ((1997) ) Inherited somatic mosaicism caused by an intracisternal A particle insertion in the mouse tyrosinase gene. Proc Natl Acad Sci U S A 94: , 890–894.

[18] 

Lim DHK , Maher ER ((2010) ) DNA methylation: A form of epigenetic control of gene expression. Obstet Gynaecol 12: , 37–42.

[19] 

Sant KE , Nahar MS , Dolinoy DC ((2012) ) DNA methylation screening and analysis. Methods Mol Biol 889: , 385–406.

[20] 

Illingworth RS , Bird AP ((2009) ) CpG islands – ‘A rough guide’. FEBS Lett 583: , 1713–1720.

[21] 

Hannon E , Lunnon K , Schalkwyk L , Mill J ((2015) ) Interindividual methylomic variation across blood, cortex, and cerebellum: Implications for epigenetic studies of neurological and neuropsychiatric phenotypes. Epigenetics 10: , 1024–1032.

[22] 

Horvath S , Zhang Y , Langfelder P , Kahn RS , Boks MPM , van Eijk K , van den Berg LH , Ophoff RA ((2012) ) Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol 13: , R97.

[23] 

Walton E , Hass J , Liu J , Roffman JL , Bernardoni F , Roessner V , Kirsch M , Schackert G , Calhoun V , Ehrlich S ((2015) ) Correspondence of DNA methylation between blood and brain tissue and its application to schizophrenia research. Schizophr Bull 42: , 406–414.

[24] 

Edgar RD , Jones MJ , Meaney MJ , Turecki G , Kobor MS ((2017) ) BECon: A tool for interpreting DNA methylation findings from blood in the context of brain. Transl Psychiatry 7: , e1187.

[25] 

Davies MN , Volta M , Pidsley R , Lunnon K , Dixit A , Lovestone S , Coarfa C , Harris RA , Milosavljevic A , Troakes C , Al-Sarraj S , Dobson R , Schalkwyk LC , Mill J ((2012) ) Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood. Genome Biol 13: , R43.

[26] 

Braun PR , Han S , Hing B , Nagahama Y , Gaul LN , Heinzman JT , Grossbach AJ , Close L , Dlouhy BJ , Howard MA , Kawasaki H , Potash JB , Shinozaki G ((2019) ) Genome-wide DNA methylation comparison between live human brain and peripheral tissues within individuals. Transl Psychiatry 9: , 47.

[27] 

Yu L , Chibnik LB , Yang J , McCabe C , Xu J , Schneider JA , De Jager PL , Bennett DA ((2016) ) Methylation profiles in peripheral blood CD4+lymphocytes versus brain: The relation to Alzheimer’s diseaseathology. Alzheimers Dement 12: , 942–951.

[28] 

Yokoyama AS , Rutledge JC , Medici V ((2017) ) DNA methylation alterations in Alzheimer’s disease. Environ Epigenet 3: , dvx008.

[29] 

Vryer R , Saffery R ((2017) ) What’s in a name? Context-dependent significance of ‘global’ methylation measures in human health and disease. Clin Epigenetics 9: , 2.

[30] 

Mastroeni D , McKee A , Grover A , Rogers J , Coleman PD ((2009) ) Epigenetic differences in cortical neurons from a pair of monozygotic twins discordant for Alzheimer’s disease. PLoS One 4: , e6617.

[31] 

Zwijnenburg PJ , Meijers-Heijboer H , Boomsma DI ((2010) ) Identical but not the same: The value of discordant monozygotic twins in genetic research. Am J Med Genet B Neuropsychiatr Genet 153B: , 1134–1149.

[32] 

Castillo-Fernandez JE , Spector TD , Bell JT ((2014) ) Epigenetics of discordant monozygotic twins: Implications for disease. Genome Med 6: , 60.

[33] 

Poulsen P , Esteller M , Vaag A , Fraga MF ((2007) ) The epigenetic basis of twin discordance in age-related diseases. Pediatr Res 61: , 38R–42R.

[34] 

Fraga MF , Ballestar E , Paz MF , Ropero S , Setien F , Ballestar ML , Heine-Suner D , Cigudosa JC , Urioste M , Benitez J , Boix-Chornet M , Sanchez-Aguilera A , Ling C , Carlsson E , Poulsen P , Vaag A , Stephan Z , Spector TD , Wu YZ , Plass C , Esteller M ((2005) ) Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A 102: , 10604–10609.

[35] 

Lévesque ML , Casey KF , Szyf M , Ismaylova E , Ly V , Verner MP , Suderman M , Brendgen M , Vitaro F , Dionne G , Boivin M , Tremblay RE , Booij L ((2014) ) Genome-wide DNA methylation variability in adolescent monozygotic twins followed since birth. Epigenetics 9: , 1410–1421.

[36] 

Wang Y , Karlsson R , Lampa E , Zhang Q , Hedman ÅK , Almgren M , Almqvist C , McRae AF , Marioni RE , Ingelsson E , Visscher PM , Deary IJ , Lind L , Morris T , Beck S , Pedersen NL , Hägg S ((2018) ) Epigenetic influences on aging: A longitudinal genome-wide methylation study in old Swedish twins. Epigenetics 13: , 975–987.

[37] 

Mastroeni D , Grover A , Delvaux E , Whiteside C , Coleman PD , Rogers J ((2010) ) Epigenetic changes in Alzheimer’s disease: Decrements in DNA methylation. Neurobiol Aging 31: , 2025–2037.

[38] 

DeKosky ST , Scheff SW ((1990) ) Synapse loss in frontal cortex biopsies in Alzheimer’s disease: Correlation with cognitive severity. Ann Neurol 27: , 457–464.

[39] 

Coppieters N , Dieriks BV , Lill C , Faull RL , Curtis MA , Dragunow M ((2014) ) Global changes in DNA methylation and hydroxymethylation in Alzheimer’s disease human brain. Neurobiol Aging 35: , 1334–1344.

[40] 

Rao JS , Keleshian VL , Klein S , Rapoport SI ((2012) ) Epigenetic modifications in frontal cortex from Alzheimer’s disease and bipolar disorder patients. Transl Psychiatry 2: , e132.

[41] 

Jahn H ((2013) ) Memory loss in Alzheimer’s disease. Dialogues Clin Neurosci 15: , 445–454.

[42] 

Bradley-Whitman MA , Lovell MA ((2013) ) Epigenetic changes in the progression of Alzheimer’s disease. Mech Ageing Dev 134: , 486–495.

[43] 

Chouliaras L , Mastroeni D , Delvaux E , Grover A , Kenis G , Hof PR , Steinbusch HWM , Coleman PD , Rutten BPF , van den Hove DLA ((2013) ) Consistent decrease in global DNA methylation and hydroxymethylation in the hippocampus of Alzheimer’s disease patients. Neurobiol Aging 34: , 2091–2099.

[44] 

Phipps AJ , Vickers JC , Taberlay PC , Woodhouse A ((2016) ) Neurofilament-labeled pyramidal neurons and astrocytes are deficient in DNA methylation marks in Alzheimer’s disease. Neurobiol Aging 45: , 30–42.

[45] 

Di Francesco A , Arosio B , Falconi A , Micioni Di Bonaventura MV , Karimi M , Mari D , Casati M , Maccarrone M , D’Addario C ((2015) ) Global changes in DNA methylation in Alzheimer’s disease peripheral blood mononuclear cells. Brain Behav Immun 45: , 139–144.

[46] 

Bjornsson HT , Sigurdsson MI , Fallin MD , Irizarry RA , Aspelund T , Cui H , Yu W , Rongione MA , Ekström TJ , Harris TB , Launer LJ , Eiriksdottir G , Leppert MF , Sapienza C , Gudnason V , Feinberg AP ((2008) ) Intra-individual change over time in DNA methylation with familial clustering. JAMA 299: , 2877–2883.

[47] 

Baylin SB , Jones PA ((2011) ) A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer 11: , 726–734.

[48] 

Poirier J , Davignon J , Bouthillier D , Kogan S , Bertrand P , Gauthier S ((1993) ) Apolipoprotein E polymorphism and Alzheimer’s disease. Lancet 342: , 697–699.

[49] 

Wang S-C , Oelze B , Schumacher A ((2008) ) Age-specific epigenetic drift in late-onset Alzheimer’s disease. PLoS One 3: , e2698.

[50] 

Wang C , Yu JT , Wang HF , Jiang T , Tan CC , Meng XF , Soares HD , Tan L ((2014) ) Meta-analysis of peripheral blood apolipoprotein E levels in Alzheimer’s disease. PLoS One 9: , e89041.

[51] 

Karlsson IK , Ploner A , Wang Y , Gatz M , Pedersen NL , Hägg S ((2018) ) Apolipoprotein E DNA methylation and late-life disease. Int J Epidemiol 47: , 899–907.

[52] 

Iwata A , Nagata K , Hatsuta H , Takuma H , Bundo M , Iwamoto K , Tamaoka A , Murayama S , Saido T , Tsuji S ((2014) ) Altered CpG methylation in sporadic Alzheimer’s disease is associated with APP and MAPT dysregulation. Hum Mol Genet 23: , 648–656.

[53] 

De Jager PL , Srivastava G , Lunnon K , Burgess J , Schalkwyk LC , Yu L , Eaton ML , Keenan BT , Ernst J , McCabe C , Tang A , Raj T , Replogle J , Brodeur W , Gabriel S , Chai HS , Younkin C , Younkin SG , Zou F , Szyf M , Epstein CB , Schneider JA , Bernstein BE , Meissner A , Ertekin-Taner N , Chibnik LB , Kellis M , Mill J , Bennett DA ((2014) ) Alzheimer’s disease: Early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat Neurosci 17: , 1156–1163.

[54] 

Lunnon K , Smith R , Hannon E , De Jager PL , Srivastava G , Volta M , Troakes C , Al-Sarraj S , Burrage J , Macdonald R , Condliffe D , Harries LW , Katsel P , Haroutunian V , Kaminsky Z , Joachim C , Powell J , Lovestone S , Bennett DA , Schalkwyk LC , Mill J ((2014) ) Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease. Nat Neurosci 17: , 1164–1170.

[55] 

Fransquet PD , Lacaze P , Saffery R , Phung J , Parker E , Shah RC , Murray A , Woods RL , Ryan J ((2020) ) DNA methylation analysis of candidate genes associated with dementia in peripheral blood. Epigenomics 12: , 2109–2123.

[56] 

Barrachina M , Ferrer I ((2009) ) DNA methylation of Alzheimer disease and tauopathy-related genes in postmortem brain. J Neuropathol Exp Neurol 68: , 880–891.

[57] 

Tannorella P , Stoccoro A , Tognoni G , Petrozzi L , Salluzzo MG , Ragalmuto A , Siciliano G , Haslberger A , Bosco P , Bonuccelli U , Migliore L , Coppedè F ((2015) ) Methylation analysis of multiple genes in blood DNA of Alzheimer’s disease and healthy individuals. Neurosci Lett 600: , 143–147.

[58] 

D’Addario C , Candia SB , Arosio B , Di Bartolomeo M , Abbate C , Casè A , Candeletti S , Romualdi P , Damanti S , Maccarrone M , Bergamaschini L , Mari D ((2017) ) Transcriptional and epigenetic phenomena in peripheral blood cells of monozygotic twins discordant for alzheimer’s disease, a case report. J Neurol Sci 372: , 211–216.

[59] 

Connor B , Young D , Yan Q , Faull RL , Synek B , Dragunow M ((1997) ) Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Brain Res Mol Brain Res 49: , 71–81.

[60] 

Nagata T , Kobayashi N , Ishii J , Shinagawa S , Nakayama R , Shibata N , Kuerban B , Ohnuma T , Kondo K , Arai H , Yamada H , Nakayama K ((2015) ) Association between DNA methylation of the BDNF promoter region and clinical presentation in Alzheimer’s disease. Dement Geriatr Cogn Dis Extra 5: , 64–73.

[61] 

Chang L , Wang Y , Ji H , Dai D , Xu X , Jiang D , Hong Q , Ye H , Zhang X , Zhou X , Liu Y , Li J , Chen Z , Li Y , Zhou D , Zhuo R , Zhang Y , Yin H , Mao C , Duan S , Wang Q ((2014) ) Elevation of peripheral BDNF promoter methylation links to the risk of Alzheimer’s disease. PLoS One 9: , e110773.

[62] 

Vasanthakumar A , Davis JW , Idler K , Waring JF , Asque E , Riley-Gillis B , Grosskurth S , Srivastava G , Kim S , Nho K , Nudelman KNH , Faber K , Sun Y , Foroud TM , Estrada K , Apostolova LG , Li QS , Saykin AJ , Alzheimer’s Disease Neuroimaging Initiative ((2020) ) Harnessing peripheral DNA methylation differences in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) to reveal novel biomarkers of disease. Clin Epigenetics 12: , 84.

[63] 

Carboni L , Lattanzio F , Candeletti S , Porcellini E , Raschi E , Licastro F , Romualdi P ((2015) ) Peripheral leukocyte expression of the potential biomarker proteins Bdnf, Sirt1, and Psen1 is not regulated by promoter methylation in Alzheimer’s disease patients. Neurosci Lett 605: , 44–48.

[64] 

Akiyama H , Barger S , Barnum S , Bradt B , Bauer J , Cole GM , Cooper NR , Eikelenboom P , Emmerling M , Fiebich BL , Finch CE , Frautschy S , Griffin WS , Hampel H , Hull M , Landreth G , Lue L , Mrak R , Mackenzie IR , McGeer PL , O’Banion MK , Pachter J , Pasinetti G , Plata-Salaman C , Rogers J , Rydel R , Shen Y , Streit W , Strohmeyer R , Tooyoma I , Van Muiswinkel FL , Veerhuis R , Walker D , Webster S , Wegrzyniak B , Wenk G , Wyss-Coray T ((2000) ) Inflammation and Alzheimer’s disease. Neurobiol Aging 21: , 383–421.

[65] 

Nicolia V , Cavallaro RA , López-González I , Maccarrone M , Scarpa S , Ferrer I , Fuso A ((2017) ) DNA methylation profiles of selected pro-inflammatory cytokines in Alzheimer disease. J Neuropathol Exp Neurol 76: , 27–31.

[66] 

Siegmund KD , Connor CM , Campan M , Long TI , Weisenberger DJ , Biniszkiewicz D , Jaenisch R , Laird PW , Akbarian S ((2007) ) DNA methylation in the human cerebral cortex is dynamically regulated throughout the life span and involves differentiated neurons. PLoS One 2: , e895.

[67] 

Sanchez-Mut JV , Aso E , Panayotis N , Lott I , Dierssen M , Rabano A , Urdinguio RG , Fernandez AF , Astudillo A , Martin-Subero JI , Balint B , Fraga MF , Gomez A , Gurnot C , Roux JC , Avila J , Hensch TK , Ferrer I , Esteller M ((2013) ) DNA methylation map of mouse and human brain identifies target genes in Alzheimer’s disease. Brain 136: , 3018–3027.

[68] 

Li QS , Vasanthakumar A , Davis JW , Idler KB , Nho K , Waring JF , Saykin AJ , for the Alzheimer’s Disease Neuroimaging Initiative ((2021) ) Association of peripheral blood DNA methylation level with Alzheimer’s disease progression. Clin Epigenetics 13: , 191.

[69] 

Dolinoy DC , Das R , Weidman JR , Jirtle RL ((2007) ) Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr Res 61: , 30r–37r.

[70] 

López-Otín C , Blasco MA , Partridge L , Serrano M , Kroemer G ((2013) ) The hallmarks of aging. Cell 153: , 1194–1217.

[71] 

Pal S , Tyler JK ((2016) ) Epigenetics and aging. Sci Adv 2: , e1600584.

[72] 

Hernandez DG , Nalls MA , Gibbs JR , Arepalli S , van der Brug M , Chong S , Moore M , Longo DL , Cookson MR , Traynor BJ , Singleton AB ((2011) ) Distinct DNA methylation changes highly correlated with chronological age in the human brain. Hum Mol Genet 20: , 1164–1172.

[73] 

Brunet A , Berger SL ((2014) ) Epigenetics of aging and aging-related disease. J Gerontol A Biol Sci Med Sci 69 Suppl 1: , S17–20.

[74] 

Declerck K , Vanden Berghe W ((2018) ) Back to the future: Epigenetic clock plasticity towards healthy aging. Mech Ageing Dev 174: , 18–29.

[75] 

Issa JP ((2014) ) Aging and epigenetic drift: A vicious cycle. J Clin Invest 124: , 24–29.

[76] 

Bormann F , Rodríguez-Paredes M , Hagemann S , Manchanda H , Kristof B , Gutekunst J , Raddatz G , Haas R , Terstegen L , Wenck H ((2016) ) Reduced DNA methylation patterning and transcriptional connectivity define human skin aging. Aging Cell 15: , 563–571.

[77] 

Fernández AF , Bayón GF , Urdinguio RG , Toraño EG , García MG , Carella A , Petrus-Reurer S , Ferrero C , Martinez-Camblor P , Cubillo I ((2015) ) H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Res. 25: , 27–40.

[78] 

Christensen BC , Houseman EA , Marsit CJ , Zheng S , Wrensch MR , Wiemels JL , Nelson HH , Karagas MR , Padbury JF , Bueno R ((2009) ) Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet 5: , e1000602.

[79] 

Bell JT , Tsai P-C , Yang T-P , Pidsley R , Nisbet J , Glass D , Mangino M , Zhai G , Zhang F , Valdes A ((2012) ) Epigenome-wide scans identify differentially methylated regions for age and age-related phenotypes in a healthy ageing population. PLoS Genet 8: , e1002629.

[80] 

Tajuddin SM , Hernandez DG , Chen BH , Noren Hooten N , Mode NA , Nalls MA , Singleton AB , Ejiogu N , Chitrala KN , Zonderman AB , Evans MK ((2019) ) Novel age-associated DNA methylation changes and epigenetic age acceleration in middle-aged African Americans and whites. Clin Epigenetics 11: , 119.

[81] 

Bell CG , Lowe R , Adams PD , Baccarelli AA , Beck S , Bell JT , Christensen BC , Gladyshev VN , Heijmans BT , Horvath S , Ideker T , Issa J-PJ , Kelsey KT , Marioni RE , Reik W , Relton CL , Schalkwyk LC , Teschendorff AE , Wagner W , Zhang K , Rakyan VK ((2019) ) DNA methylation aging clocks: Challenges and recommendations. Genome Biol 20: , 249.

[82] 

Horvath S , Raj K ((2018) ) DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet 19: , 371–384.

[83] 

Horvath S ((2013) ) DNA methylation age of human tissues and cell types. Genome Biol 14: , R115.

[84] 

McCartney DL , Stevenson AJ , Walker RM , Gibson J , Morris SW , Campbell A , Murray AD , Whalley HC , Porteous DJ , McIntosh AM , Evans KL , Deary IJ , Marioni RE ((2018) ) Investigating the relationship between DNA methylation age acceleration and risk factors for Alzheimer’s disease. Alzheimers Dement (Amst) 10: , 429–437.

[85] 

Bocklandt S , Lin W , Sehl ME , Sánchez FJ , Sinsheimer JS , Horvath S , Vilain E ((2011) ) Epigenetic predictor of age. PLoS One 6: , e14821.

[86] 

Zhang Q , Vallerga CL , Walker RM , Lin T , Henders AK , Montgomery GW , He J , Fan D , Fowdar J , Kennedy M , Pitcher T , Pearson J , Halliday G , Kwok JB , Hickie I , Lewis S , Anderson T , Silburn PA , Mellick GD , Harris SE , Redmond P , Murray AD , Porteous DJ , Haley CS , Evans KL , McIntosh AM , Yang J , Gratten J , Marioni RE , Wray NR , Deary IJ , McRae AF , Visscher PM ((2019) ) Improved precision of epigenetic clock estimates across tissues and its implication for biological ageing. Genome Med 11: , 54.

[87] 

Simpson DJ , Chandra T ((2021) ) Epigenetic age prediction. Aging Cell 20: , e13452.

[88] 

Koch CM , Wagner W ((2011) ) Epigenetic-aging-signature to determine age in different tissues. Aging 3: , 1018–1027.

[89] 

Zbieć-Piekarska R , Spólnicka M , Kupiec T , Parys-Proszek A , Makowska Ż , Pałeczka A , Kucharczyk K , Płoski R , Branicki W ((2015) ) Development of a forensically useful age prediction method based on DNA methylation analysis. Forensic Sci Int Genet 17: , 173–179.

[90] 

Huang Y , Yan J , Hou J , Fu X , Li L , Hou Y ((2015) ) Developing a DNA methylation assay for human age prediction in blood and bloodstain. Forensic Sci Int Genet 17: , 129–136.

[91] 

Levine ME ((2020) ) Assessment of epigenetic clocks as biomarkers of aging in basic and population research. J Gerontol A Biol Sci Med Sci 75: , 463–465.

[92] 

Yang Z , Wong A , Kuh D , Paul DS , Rakyan VK , Leslie RD , Zheng SC , Widschwendter M , Beck S , Teschendorff AE ((2016) ) Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol 17: , 205–205.

[93] 

Youn A , Wang S ((2018) ) The MiAge Calculator: A DNA methylation-based mitotic age calculator of human tissue types. Epigenetics 13: , 192–206.

[94] 

Levine ME , Lu AT , Quach A , Chen BH , Assimes TL , Bandinelli S , Hou L , Baccarelli AA , Stewart JD , Li Y , Whitsel EA , Wilson JG , Reiner AP , Aviv A , Lohman K , Liu Y , Ferrucci L , Horvath S ((2018) ) An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY) 10: , 573–591.

[95] 

Lu AT , Seeboth A , Tsai P-C , Sun D , Quach A , Reiner AP , Kooperberg C , Ferrucci L , Hou L , Baccarelli AA , Li Y , Harris SE , Corley J , Taylor A , Deary IJ , Stewart JD , Whitsel EA , Assimes TL , Chen W , Li S , Mangino M , Bell JT , Wilson JG , Aviv A , Marioni RE , Raj K , Horvath S ((2019) ) DNA methylation-based estimator of telomere length. Aging 11: , 5895–5923.

[96] 

Lu AT , Quach A , Wilson JG , Reiner AP , Aviv A , Raj K , Hou L , Baccarelli AA , Li Y , Stewart JD , Whitsel EA , Assimes TL , Ferrucci L , Horvath S ((2019) ) DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11: , 303–327.

[97] 

Zhang Y , Wilson R , Heiss J , Breitling LP , Saum K-U , Schöttker B , Holleczek B , Waldenberger M , Peters A , Brenner H ((2017) ) DNA methylation signatures in peripheral blood strongly predict all-cause mortality. Nat Commun 8: , 14617–14617.

[98] 

Belsky DW , Caspi A , Arseneault L , Baccarelli A , Corcoran DL , Gao X , Hannon E , Harrington HL , Rasmussen LJ , Houts R , Huffman K , Kraus WE , Kwon D , Mill J , Pieper CF , Prinz JA , Poulton R , Schwartz J , Sugden K , Vokonas P , Williams BS , Moffitt TE ((2020) ) Quantification of the pace of biological aging in humans through a blood test, the DunedinPoAm DNA methylation algorithm. eLife 9: , e54870.

[99] 

Chen BH , Marioni RE , Colicino E , Peters MJ , Ward-Caviness CK , Tsai P-C , Roetker NS , Just AC , Demerath EW , Guan W , Bressler J , Fornage M , Studenski S , Vandiver AR , Moore AZ , Tanaka T , Kiel DP , Liang L , Vokonas P , Schwartz J , Lunetta KL , Murabito JM , Bandinelli S , Hernandez DG , Melzer D , Nalls M , Pilling LC , Price TR , Singleton AB , Gieger C , Holle R , Kretschmer A , Kronenberg F , Kunze S , Linseisen J , Meisinger C , Rathmann W , Waldenberger M , Visscher PM , Shah S , Wray NR , McRae AF , Franco OH , Hofman A , Uitterlinden AG , Absher D , Assimes T , Levine ME , Lu AT , Tsao PS , Hou L , Manson JE , Carty CL , LaCroix AZ , Reiner AP , Spector TD , Feinberg AP , Levy D , Baccarelli A , van Meurs J , Bell JT , Peters A , Deary IJ , Pankow JS , Ferrucci L , Horvath S ((2016) ) DNA methylation-based measures of biological age: Meta-analysis predicting time to death. Aging 8: , 1844–1865.

[100] 

Horvath S , Pirazzini C , Bacalini MG , Gentilini D , Di Blasio AM , Delledonne M , Mari D , Arosio B , Monti D , Passarino G , De Rango F , D’Aquila P , Giuliani C , Marasco E , Collino S , Descombes P , Garagnani P , Franceschi C ((2015) ) Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging 7: , 1159–1170.

[101] 

Hannum G , Guinney J , Zhao L , Zhang L , Hughes G , Sadda S , Klotzle B , Bibikova M , Fan J-B , Gao Y , Deconde R , Chen M , Rajapakse I , Friend S , Ideker T , Zhang K ((2013) ) Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell 49: , 359–367.

[102] 

Horvath S , Gurven M , Levine ME , Trumble BC , Kaplan H , Allayee H , Ritz BR , Chen B , Lu AT , Rickabaugh TM , Jamieson BD , Sun D , Li S , Chen W , Quintana-Murci L , Fagny M , Kobor MS , Tsao PS , Reiner AP , Edlefsen KL , Absher D , Assimes TL ((2016) ) An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol 17: , 171.

[103] 

Marioni RE , Shah S , McRae AF , Chen BH , Colicino E , Harris SE , Gibson J , Henders AK , Redmond P , Cox SR , Pattie A , Corley J , Murphy L , Martin NG , Montgomery GW , Feinberg AP , Fallin MD , Multhaup ML , Jaffe AE , Joehanes R , Schwartz J , Just AC , Lunetta KL , Murabito JM , Starr JM , Horvath S , Baccarelli AA , Levy D , Visscher PM , Wray NR , Deary IJ ((2015) ) DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol 16: , 25.

[104] 

Horvath S , Garagnani P , Bacalini MG , Pirazzini C , Salvioli S , Gentilini D , Di Blasio AM , Giuliani C , Tung S , Vinters HV , Franceschi C ((2015) ) Accelerated epigenetic aging in Down syndrome. Aging Cell 14: , 491–495.

[105] 

Maierhofer A , Flunkert J , Oshima J , Martin GM , Haaf T , Horvath S ((2017) ) Accelerated epigenetic aging in Werner syndrome. Aging (Albany NY) 9: , 1143–1152.

[106] 

Gross AM , Jaeger PA , Kreisberg JF , Licon K , Jepsen KL , Khosroheidari M , Morsey BM , Swindells S , Shen H , Ng CT , Flagg K , Chen D , Zhang K , Fox HS , Ideker T ((2016) ) Methylome-wide analysis of chronic HIV infection reveals five-year increase in biological age and epigenetic targeting of HLA. Mol Cell 62: , 157–168.

[107] 

Horvath S , Levine AJ ((2015) ) HIV-1 infection accelerates age according to the epigenetic clock. J Infect Dis 212: , 1563–1573.

[108] 

Marioni , Shah S , McRae AF , Ritchie SJ , Muniz-Terrera G , Harris SE , Gibson J , Redmond P , Cox SR , Pattie A , Corley J , Taylor A , Murphy L , Starr JM , Horvath S , Visscher PM , Wray NR , Deary IJ ((2015) ) The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936. Int J Epidemiol 44: , 1388–1396.

[109] 

Quach A , Levine ME , Tanaka T , Lu AT , Chen BH , Ferrucci L , Ritz B , Bandinelli S , Neuhouser ML , Beasley JM , Snetselaar L , Wallace RB , Tsao PS , Absher D , Assimes TL , Stewart JD , Li Y , Hou L , Baccarelli AA , Whitsel EA , Horvath S ((2017) ) Epigenetic clock analysis of diet, exercise, education, and lifestyle factors. Aging (Albany NY) 9: , 419–446.

[110] 

Loomba R , Gindin Y , Jiang Z , Lawitz E , Caldwell S , Djedjos CS , Xu R , Chung C , Myers RP , Subramanian GM , Goodman Z , Charlton M , Afdhal NH , Diehl AM ((2018) ) DNA methylation signatures reflect aging in patients with nonalcoholic steatohepatitis, JCI Insight 3: , e96685.

[111] 

Horvath S , Erhart W , Brosch M , Ammerpohl O , von Schönfels W , Ahrens M , Heits N , Bell JT , Tsai PC , Spector TD , Deloukas P , Siebert R , Sipos B , Becker T , Röcken C , Schafmayer C , Hampe J ((2014) ) Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci U S A 111: , 15538–15543.

[112] 

Carroll JE , Irwin MR , Levine M , Seeman TE , Absher D , Assimes T , Horvath S ((2017) ) Epigenetic aging and immune senescence in women with insomnia symptoms: Findings from the Women’s Health Initiative Study. Biol Psychiatry 81: , 136–144.

[113] 

Jovanovic T , Vance LA , Cross D , Knight AK , Kilaru V , Michopoulos V , Klengel T , Smith AK ((2017) ) Exposure to violence accelerates epigenetic aging in children. Sci Rep 7: , 8962.

[114] 

Horvath S , Ritz BR ((2015) ) Increased epigenetic age and granulocyte counts in the blood of Parkinson’s disease patients. Aging (Albany NY) 7: , 1130–1142.

[115] 

Horvath S , Langfelder P , Kwak S , Aaronson J , Rosinski J , Vogt TF , Eszes M , Faull RL , Curtis MA , Waldvogel HJ , Choi OW , Tung S , Vinters HV , Coppola G , Yang XW ((2016) ) Huntington’s disease accelerates epigenetic aging of human brain and disrupts DNA methylation levels. Aging (Albany NY) 8: , 1485–1512.

[116] 

Levine ME , Lu AT , Bennett DA , Horvath S ((2015) ) Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging 7: , 1198–1211.

[117] 

Chouliaras L , Pishva E , Haapakoski R , Zsoldos E , Mahmood A , Filippini N , Burrage J , Mill J , Kivimäki M , Lunnon K , Ebmeier KP ((2018) ) Peripheral DNA methylation, cognitive decline and brain aging: Pilot findings from the Whitehall II imaging study. Epigenomics 10: , 585–595.

[118] 

Hodgson K , Carless MA , Kulkarni H , Curran JE , Sprooten E , Knowles EE , Mathias S , Göring HHH , Yao N , Olvera RL , Fox PT , Almasy L , Duggirala R , Blangero J , Glahn DC ((2017) ) Epigenetic age acceleration assessed with human white-matter images. J Neurosci 37: , 4735–4743.

[119] 

Hillary RF , Stevenson AJ , Cox SR , McCartney DL , Harris SE , Seeboth A , Higham J , Sproul D , Taylor AM , Redmond P , Corley J , Pattie A , Hernández M , Muñoz-Maniega S , Bastin ME , Wardlaw JM , Horvath S , Ritchie CW , Spires-Jones TL , McIntosh AM , Evans KL , Deary IJ , Marioni RE ((2021) ) An epigenetic predictor of death captures multi-modal measures of brain health. Mol. Psychiatry 26: , 3806–3816.

[120] 

Belsky DW , Moffitt TE , Cohen AA , Corcoran DL , Levine ME , Prinz JA , Schaefer J , Sugden K , Williams B , Poulton R , Caspi A ((2018) ) Eleven telomere, epigenetic clock, and biomarker-composite quantifications of biological aging: Do they measure the same thing? Am J Epidemiol 187: , 1220–1230.

[121] 

Starnawska A , Tan Q , Lenart A , McGue M , Mors O , Børglum AD , Christensen K , Nyegaard M , Christiansen L ((2017) ) Blood DNA methylation age is not associated with cognitive functioning in middle-aged monozygotic twins. Neurobiol Aging 50: , 60–63.

[122] 

Degerman S , Josefsson M , Nordin Adolfsson A , Wennstedt S , Landfors M , Haider Z , Pudas S , Hultdin M , Nyberg L , Adolfsson R ((2017) ) Maintained memory in aging is associated with young epigenetic age. Neurobiol Aging 55: , 167–171.

[123] 

Vaccarino V , Huang M , Wang Z , Hui Q , Shah AJ , Goldberg J , Smith N , Kaseer B , Murrah N , Levantsevych OM , Shallenberger L , Driggers E , Bremner JD , Sun YV ((2021) ) Epigenetic age acceleration and cognitive decline: A twin study. J Gerontol A Biol Sci Med Sci 76: , 1854–1863.

[124] 

Beydoun MA , Shaked D , Tajuddin SM , Weiss J , Evans MK , Zonderman AB ((2020) ) Accelerated epigenetic age and cognitive decline among urban-dwelling adults. Neurology 94: , e613–e625.

[125] 

Milicic L , Vacher M , Porter T , Doré V , Burnham SC , Bourgeat P , Shishegar R , Doecke J , Armstrong NJ , Tankard R , Maruff P , Masters CL , Rowe CC , Villemagne VL , Laws SM ((2022) ) Comprehensive analysis of epigenetic clocks reveals associations between disproportionate biological ageing and hippocampal volume. Geroscience 44: , 1807–1823.

[126] 

Houseman EA , Accomando WP , Koestler DC , Christensen BC , Marsit CJ , Nelson HH , Wiencke JK , Kelsey KT ((2012) ) DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 13: , 86.

[127] 

Guintivano J , Aryee MJ , Kaminsky ZA ((2013) ) A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics 8: , 290–302.

[128] 

Montaño CM , Irizarry RA , Kaufmann WE , Talbot K , Gur RE , Feinberg AP , Taub MA ((2013) ) Measuring cell-type specific differential methylation in human brain tissue. Genome Biol 14: , R94–R94.

[129] 

Barton AJ , Pearson RC , Najlerahim A , Harrison PJ ((1993) ) Pre- and postmortem influences on brain RNA. J Neurochem 61: , 1–11.

[130] 

Tomita H , Vawter MP , Walsh DM , Evans SJ , Choudary PV , Li J , Overman KM , Atz ME , Myers RM , Jones EG , Watson SJ , Akil H , Bunney WE Jr. ((2004) ) Effect of agonal and postmortem factors on gene expression profile: Quality control in microarray analyses of postmortem human brain. Biol Psychiatry 55: , 346–352.

[131] 

Bakulski KM , Halladay A , Hu VW , Mill J , Fallin MD ((2016) ) Epigenetic research in neuropsychiatric disorders: The “tissue issue”. Curr Behav Neurosci Rep 3: , 264–274.

[132] 

Fransquet PD , Lacaze P , Saffery R , McNeil J , Woods R , Ryan J ((2018) ) Blood DNA methylation as a potential biomarker of dementia: A systematic review. Alzheimers Dement 14: , 81–103.

[133] 

Fransquet PD , Ryan J ((2019) ) The current status of blood epigenetic biomarkers for dementia. Crit Rev Clin Lab Sci 56: , 435–457.

[134] 

Gijselinck I , Van Mossevelde S , van der Zee J , Sieben A , Engelborghs S , De Bleecker J , Ivanoiu A , Deryck O , Edbauer D , Zhang M , Heeman B , Bäumer V , Van den Broeck M , Mattheijssens M , Peeters K , Rogaeva E , De Jonghe P , Cras P , Martin JJ , de Deyn PP , Cruts M , Van Broeckhoven C ((2016) ) The C9orf72 repeat size correlates with onset age of disease, DNA methylation and transcriptional downregulation of the promoter. Mol Psychiatry 21: , 1112–1124.

[135] 

Russ J , Liu EY , Wu K , Neal D , Suh E , Irwin DJ , McMillan CT , Harms MB , Cairns NJ , Wood EM , Xie SX , Elman L , McCluskey L , Grossman M , Van Deerlin VM , Lee EB ((2015) ) Hypermethylation of repeat expanded C9orf72 is a clinical and molecular disease modifier. Acta Neuropathol 129: , 39–52.

[136] 

Xi Z , Zhang M , Bruni AC , Maletta RG , Colao R , Fratta P , Polke JM , Sweeney MG , Mudanohwo E , Nacmias B , Sorbi S , Tartaglia MC , Rainero I , Rubino E , Pinessi L , Galimberti D , Surace EI , McGoldrick P , McKeever P , Moreno D , Sato C , Liang Y , Keith J , Zinman L , Robertson J , Rogaeva E ((2015) ) The C9orf72 repeat expansion itself is methylated in ALS and FTLD patients. Acta Neuropathol 129: , 715–727.

[137] 

El Khoury LY , Gorrie-Stone T , Smart M , Hughes A , Bao Y , Andrayas A , Burrage J , Hannon E , Kumari M , Mill J , Schalkwyk LC ((2019) ) Systematic underestimation of the epigenetic clock and age acceleration in older subjects. Genome Biol 20: , 283.

[138] 

Shireby GL , Davies JP , Francis PT , Burrage J , Walker EM , Neilson GWA , Dahir A , Thomas AJ , Love S , Smith RG , Lunnon K , Kumari M , Schalkwyk LC , Morgan K , Brookes K , Hannon E , Mill J ((2020) ) Recalibrating the epigenetic clock: Implications for assessing biological age in the human cortex. Brain 143: , 3763–3775.

[139] 

Dhingra R , Kwee LC , Diaz-Sanchez D , Devlin RB , Cascio W , Hauser ER , Gregory S , Shah S , Kraus WE , Olden K , Ward-Caviness CK ((2019) ) Evaluating DNA methylation age on the Illumina MethylationEPIC Bead Chip. PLoS One 14: , e0207834.

[140] 

Armstrong NJ , Mather KA , Thalamuthu A , Wright MJ , Trollor JN , Ames D , Brodaty H , Schofield PR , Sachdev PS , Kwok JB ((2017) ) Aging, exceptional longevity and comparisons of the Hannum and Horvath epigenetic clocks. Epigenomics 9: , 689–700.

[141] 

Zampieri M , Ciccarone F , Calabrese R , Franceschi C , Bürkle A , Caiafa P ((2015) ) Reconfiguration of DNA methylation in aging. Mech Ageing Dev 151: , 60–70.

[142] 

Galkin F , Mamoshina P , Kochetov K , Sidorenko D , Zhavoronkov A ((2021) ) DeepMAge: A methylation aging clock developed with deep learning. Aging Dis 12: , 1252–1262.

[143] 

Leygo C , Williams M , Jin HC , Chan MWY , Chu WK , Grusch M , Cheng YY ((2017) ) DNA methylation as a noninvasive epigenetic biomarker for the detection of cancer. Dis Markers 2017: , 3726595.

[144] 

Meder B , Haas J , Sedaghat-Hamedani F , Kayvanpour E , Frese K , Lai A , Nietsch R , Scheiner C , Mester S , Bordalo DM , Amr A , Dietrich C , Pils D , Siede D , Hund H , Bauer A , Holzer DB , Ruhparwar A , Mueller-Hennessen M , Weichenhan D , Plass C , Weis T , Backs J , Wuerstle M , Keller A , Katus HA , Posch AE ((2017) ) Epigenome-wide association study identifies cardiac gene patterning and a novel class of biomarkers for heart failure. Circulation 136: , 1528–1544.

[145] 

Surace AEA , Hedrich CM ((2019) ) The role of epigenetics in autoimmune/inflammatory disease. Front Immunol 10: , 1525.

[146] 

Lu H , Liu X , Deng Y , Qing H ((2013) ) DNA methylation, a hand behind neurodegenerative diseases. Front Aging Neurosci 5: , 85.

[147] 

Garagnani P , Bacalini MG , Pirazzini C , Gori D , Giuliani C , Mari D , Di Blasio AM , Gentilini D , Vitale G , Collino S , Rezzi S , Castellani G , Capri M , Salvioli S , Franceschi C ((2012) ) Methylation of ELOVL2 gene as a new epigenetic marker of age. Aging Cell 11: , 1132–1134.

[148] 

Florath I , Butterbach K , Müller H , Bewerunge-Hudler M , Brenner H ((2014) ) Cross-sectional and longitudinal changes in DNA methylation with age: An epigenome-wide analysis revealing over 60 novel age-associated CpG sites. Hum Mol Genet 23: , 1186–1201.

[149] 

Weidner CI , Lin Q , Koch CM , Eisele L , Beier F , Ziegler P , Bauerschlag DO , Jöckel K-H , Erbel R , Mühleisen TW , Zenke M , Brümmendorf TH , Wagner W ((2014) ) Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol 15: , R24.

[150] 

Eipel M , Mayer F , Arent T , Ferreira MR , Birkhofer C , Gerstenmaier U , Costa IG , Ritz-Timme S , Wagner W ((2016) ) Epigenetic age predictions based on buccal swabs are more precise in combination with cell type-specific DNA methylation signatures. Aging (Albany NY) 8: , 1034–1048.

[151] 

Cho S , Jung SE , Hong SR , Lee EH , Lee JH , Lee SD , Lee HY ((2017) ) Independent validation of DNA-based approaches for age prediction in blood. Forensic Sci Int Genet 29: , 250–256.

[152] 

Hong SR , Jung SE , Lee EH , Shin KJ , Yang WI , Lee HY ((2017) ) DNA methylation-based age prediction from saliva: High age predictability by combination of 7 CpG markers. Forensic Sci Int Genet 29: , 118–125.

[153] 

Horvath S , Oshima J , Martin GM , Lu AT , Quach A , Cohen H , Felton S , Matsuyama M , Lowe D , Kabacik S , Wilson JG , Reiner AP , Maierhofer A , Flunkert J , Aviv A , Hou L , Baccarelli AA , Li Y , Stewart JD , Whitsel EA , Ferrucci L , Matsuyama S , Raj K ((2018) ) Epigenetic clock for skin and blood cells applied to Hutchinson Gilford Progeria Syndrome and ex vivo studies. Aging (Albany NY) 10: , 1758–1775.

[154] 

McEwen LM , O’Donnell KJ , McGill MG , Edgar RD , Jones MJ , MacIsaac JL , Lin DTS , Ramadori K , Morin A , Gladish N , Garg E , Unternaehrer E , Pokhvisneva I , Karnani N , Kee MZL , Klengel T , Adler NE , Barr RG , Letourneau N , Giesbrecht GF , Reynolds JN , Czamara D , Armstrong JM , Essex MJ , de Weerth C , Beijers R , Tollenaar MS , Bradley B , Jovanovic T , Ressler KJ , Steiner M , Entringer S , Wadhwa PD , Buss C , Bush NR , Binder EB , Boyce WT , Meaney MJ , Horvath S , Kobor MS ((2020) ) The PedBE clock accurately estimates DNA methylation age in pediatric buccal cells. Proc Natl Acad Sci U S A 117: , 23329–23335.

[155] 

Jung SE , Lim SM , Hong SR , Lee EH , Shin KJ , Lee HY ((2019) ) DNA methylation of the ELOVL2, FHL2, KLF14, C1orf132/MIR29B2C, and TRIM59 genes for age prediction from blood, saliva, and buccal swab samples. Forensic Sci Int Genet 38: , 1–8.

[156] 

Dias HC , Cordeiro C , Pereira J , Pinto C , Real FC , Cunha E , Manco L ((2020) ) DNA methylation age estimation in blood samples of living and deceased individuals using a multiplex SNaPshot assay. Forensic Sci Int 311: , 110267.

[157] 

Correia Dias H , Manco L , Corte Real F , Cunha E ((2021) ) A blood-bone-tooth model for age prediction in forensic contexts. Biology (Basel) 10: , 1312.

[158] 

Higgins-Chen AT , Thrush KL , Wang Y , Minteer CJ , Kuo P-L , Wang M , Niimi P , Sturm G , Lin J , Moore AZ , Bandinelli S , Vinkers CH , Vermetten E , Rutten BPF , Geuze E , Okhuijsen-Pfeifer C , van der Horst MZ , Schreiter S , Gutwinski S , Luykx JJ , Picard M , Ferrucci L , Crimmins EM , Boks MP , Hägg S , Hu-Seliger TT , Levine ME ((2022) ) A computational solution for bolstering reliability of epigenetic clocks: Implications for clinical trials and longitudinal tracking. Nature Aging 2: , 644–661.

[159] 

Boroni M , Zonari A , Reis de Oliveira C , Alkatib K , Ochoa Cruz EA , Brace LE , Lott de Carvalho J ((2020) ) Highly accurate skin-specific methylome analysis algorithm as a platform to screen and validate therapeutics for healthy aging. Clin Epigenetics 12: , 105.

[160] 

Lin Q , Weidner CI , Costa IG , Marioni RE , Ferreira MR , Deary IJ , Wagner W ((2016) ) DNA methylation levels at individual age-associated CpG sites can be indicative for life expectancy. Aging (Albany N Y) 8: , 394–401.

[161] 

Vidal-Bralo L , Lopez-Golan Y , Gonzalez A ((2016) ) Simplified assay for epigenetic age estimation in whole blood of adults. Front Genet 7: , 126.

[162] 

Teschendorff AE ((2020) ) A comparison of epigenetic mitotic-like clocks for cancer risk prediction. Genome Med 12: , 56.

[163] 

Zhou W , Dinh HQ , Ramjan Z , Weisenberger DJ , Nicolet CM , Shen H , Laird PW , Berman BP ((2018) ) DNA methylation loss in late-replicating domains is linked to mitotic cell division. Nat Genet 50: , 591–602.

[164] 

Belsky DW , Caspi A , Corcoran DL , Sugden K , Poulton R , Arseneault L , Baccarelli A , Chamarti K , Gao X , Hannon E , Harrington HL , Houts R , Kothari M , Kwon D , Mill J , Schwartz J , Vokonas P , Wang C , Williams BS , Moffitt TE ((2022) ) DunedinPACE, a DNA methylation biomarker of the pace of aging. Elife 11: , e73420.

[165] 

Lu AT , Binder AM , Zhang J , Yan Q , Reiner AP , Cox SR , Corley J , Harris SE , Kuo PL , Moore AZ , Bandinelli S , Stewart JD , Wang C , Hamlat EJ , Epel ES , Schwartz JD , Whitsel EA , Correa A , Ferrucci L , Marioni RE , Horvath S ((2022) ) DNA methylation GrimAge version 2. Aging (Albany N Y) 14: , 9484–9549.

[166] 

West RL , Lee JM , Maroun LE ((1995) ) Hypomethylation of the amyloid precursor protein gene in the brain of an Alzheimer’s disease patient. J Mol Neurosci 6: , 141–146.

[167] 

Sontag E , Hladik C , Montgomery L , Luangpirom A , Mudrak I , Ogris E , White CL III ((2004) ) Downregulation of protein phosphatase 2A carboxyl methylation and methyltransferase may contribute to Alzheimer disease pathogenesis. J Neuropathol Exp Neurol 63: , 1080–1091.

[168] 

Brohede J , Rinde M , Winblad B , Graff C ((2010) ) A DNA methylation study of the amyloid precursor protein gene in several brain regions from patients with familial Alzheimer disease. J Neurogenet 24: , 179–181.

[169] 

Bollati V , Galimberti D , Pergoli L , Dalla Valle E , Barretta F , Cortini F , Scarpini E , Bertazzi PA , Baccarelli A ((2011) ) DNA methylation in repetitive elements and Alzheimer disease. Brain Behav Immun 25: , 1078–1083.

[170] 

Bakulski KM , Dolinoy DC , Sartor MA , Paulson HL , Konen JR , Lieberman AP , Albin RL , Hu H , Rozek LS ((2012) ) Genome-wide DNA methylation differences between late-onset Alzheimer’s disease and cognitively normal controls in human frontal cortex. J Alzheimers Dis 29: , 571–588.

[171] 

D’Addario C , Di Francesco A , Arosio B , Gussago C , Dell’Osso B , Bari M , Galimberti D , Scarpini E , Altamura AC , Mari D , Maccarrone M ((2012) ) Epigenetic regulation of fatty acid amide hydrolase in Alzheimer disease. PLoS One 7: , e39186.

[172] 

Arosio B , Bulbarelli A , Bastias Candia S , Lonati E , Mastronardi L , Romualdi P , Candeletti S , Gussago C , Galimberti D , Scarpini E , Dell’Osso B , Altamura C , MacCarrone M , Bergamaschini L , D’Addario C , Mari D ((2012) ) Pin1 contribution to Alzheimer’s disease: Transcriptional and epigenetic mechanisms in patients with late-onset Alzheimer’s disease. Neurodegener Dis 10: , 207–211.

[173] 

Furuya TK , Silva PNO , Payão SLM , Bertolucci PHF , Rasmussen LT , De Labio RW , Braga ILS , Chen ES , Turecki G , Mechawar N , Mill J , Smith MAC ((2012) ) Analysis of SNAP25 mRNA expression and promoter DNA methylation in brain areas of Alzheimer’s disease patients. Neuroscience 220: , 41–46.

[174] 

Furuya TK , da Silva PNO , Payão SLM , Rasmussen LT , de Labio RW , Bertolucci PHF , Braga ILS , Chen ES , Turecki G , Mechawar N , Mill J , de Arruda Cardoso Smith M ((2012) ) SORL1 and SIRT1 mRNA expression and promoter methylation levels in aging and Alzheimer’s disease. Neurochem Int 61: , 973–975.

[175] 

Silva PN , Furuya TK , Braga IL , Rasmussen LT , Labio RW , Bertolucci PH , Chen ES , Turecki G , Mechawar N , Payão SL , Mill J , Smith MC ((2014) ) Analysis of HSPA8 and HSPA9 mRNA expression and promoter methylation in the brain and blood of Alzheimer’s disease patients. J Alzheimers Dis 38: , 165–170.

[176] 

Silva PN , Furuya TK , Sampaio Braga I , Rasmussen LT , de Labio RW , Bertolucci PH , Chen ES , Turecki G , Mechawar N , Payão SL , Mill J , Smith MC ((2013) ) CNP and DPYSL2 mRNA expression and promoter methylation levels in brain of Alzheimer’s disease patients. J Alzheimers Dis 33: , 349–355.

[177] 

Di Francesco A , Arosio B , Gussago C , Dainese E , Mari D , D’Addario C , Maccarrone M ((2013) ) Involvement of 5-lipoxygenase in Alzheimer’s disease: A role for DNA methylation. J Alzheimers Dis 37: , 3–8.

[178] 

Hernández HG , Mahecha MF , Mejía A , Arboleda H , Forero DA ((2014) ) Global long interspersed nuclear element 1 DNA methylation in a Colombian sample of patients with late-onset Alzheimer’s disease. Am J Alzheimers Dis Other Demen 29: , 50–53.

[179] 

Sanchez-Mut JV , Aso E , Heyn H , Matsuda T , Bock C , Ferrer I , Esteller M ((2014) ) Promoter hypermethylation of the phosphatase DUSP22 mediates PKA-dependent TAU phosphorylation and CREB activation in Alzheimer’s disease. Hippocampus 24: , 363–368.

[180] 

Chibnik LB , Yu L , Eaton ML , Srivastava G , Schneider JA , Kellis M , Bennett DA , De Jager PL ((2015) ) Alzheimer’s loci: Epigenetic associations and interaction with genetic factors. Ann Clin Transl Neurol 2: , 636–647.

[181] 

Yu L , Chibnik LB , Srivastava GP , Pochet N , Yang J , Xu J , Kozubek J , Obholzer N , Leurgans SE , Schneider JA , Meissner A , De Jager PL , Bennett DA ((2015) ) Association of brain DNA methylation in SORL1, ABCA7, HLA-DRB5, SLC24A4, and BIN1 with pathological diagnosis of Alzheimer disease. JAMA Neurol 72: , 15–24.

[182] 

Lashley T , Gami P , Valizadeh N , Li A , Revesz T , Balazs R ((2015) ) Alterations in global DNA methylation and hydroxymethylation are not detected in Alzheimer’s disease. Neuropathol Appl Neurobiol 41: , 497–506.

[183] 

Celarain N , Sánchez-Ruiz de Gordoa J , Zelaya MV , Roldán M , Larumbe R , Pulido L , Echavarri C , Mendioroz M ((2016) ) TREM2 upregulation correlates with 5-hydroxymethycytosine enrichment in Alzheimer’s disease hippocampus. Clin Epigenetics 8: , 37.

[184] 

Ma SL , Tang NLS , Lam LCW ((2016) ) Association of gene expression and methylation of UQCRC1 to the predisposition of Alzheimer’s disease in a Chinese population. J Psychiatr Res 76: , 143–147.

[185] 

Smith AR , Smith RG , Condliffe D , Hannon E , Schalkwyk L , Mill J , Lunnon K ((2016) ) Increased DNA methylation near TREM2 is consistently seen in the superior temporal gyrus in Alzheimer’s disease brain. Neurobiol Aging 47: , 35–40.

[186] 

Watson CT , Roussos P , Garg P , Ho DJ , Azam N , Katsel PL , Haroutunian V , Sharp AJ ((2016) ) Genome-wide DNA methylation profiling in the superior temporal gyrus reveals epigenetic signatures associated with Alzheimer’s disease. Genome Med 8: , 5.

[187] 

Liu G , Ji H , Liu J , Xu C , Chang L , Cui W , Ye C , Hu H , Chen Y , Zhou X , Duan S , Wang Q ((2017) ) Association of OPRK1 and OPRM1 methylation with mild cognitive impairment in Xinjiang Han and Uygur populations. Neurosci Lett 636: , 170–176.

[188] 

Smith RG , Hannon E , De Jager PL , Chibnik L , Lott SJ , Condliffe D , Smith AR , Haroutunian V , Troakes C , Al-Sarraj S , Bennett DA , Powell J , Lovestone S , Schalkwyk L , Mill J , Lunnon K ((2018) ) Elevated DNA methylation across a 48-kb region spanning the HOXA gene cluster is associated with Alzheimer’s disease neuropathology. Alzheimers Dement 14: , 1580–1588.

[189] 

Gasparoni G , Bultmann S , Lutsik P , Kraus TFJ , Sordon S , Vlcek J , Dietinger V , Steinmaurer M , Haider M , Mulholland CB , Arzberger T , Roeber S , Riemenschneider M , Kretzschmar HA , Giese A , Leonhardt H , Walter J ((2018) ) DNA methylation analysis on purified neurons and glia dissects age and Alzheimer’s disease-specific changes in the human cortex. Epigenetics Chromatin 11: , 41.

[190] 

Madrid A , Hogan KJ , Papale LA , Clark LR , Asthana S , Johnson SC , Alisch RS ((2018) ) DNA hypomethylation in blood links B3GALT4 and ZADH2 to Alzheimer’s disease. J Alzheimers Dis 66: , 927–934.

[191] 

Hernández HG , Sandoval-Hernández AG , Garrido-Gil P , Labandeira-Garcia JL , Zelaya MV , Bayon GF , Fernández AF , Fraga MF , Arboleda G , Arboleda H ((2018) ) Alzheimer’s disease DNA methylome of pyramidal layers in frontal cortex: Laser-assisted microdissection study. Epigenomics 10: , 1365–1365–1382.

[192] 

Andrés-Benito P , Delgado-Morales R , Ferrer I ((2018) ) Altered regulation of KIAA0566, and katanin signaling expression in the locus coeruleus with neurofibrillary tangle pathology. Front Cell Neurosci 12: , 131.

[193] 

Li P , Marshall L , Oh G , Jakubowski JL , Groot D , He Y , Wang T , Petronis A , Labrie V ((2019) ) Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer’s disease pathology and cognitive symptoms. Nat Commun 10: , 2246.

[194] 

Semick SA , Bharadwaj RA , Collado-Torres L , Tao R , Shin JH , Deep-Soboslay A , Weiss JR , Weinberger DR , Hyde TM , Kleinman JE , Jaffe AE , Mattay VS ((2019) ) Integrated DNA methylation and gene expression profiling across multiple brain regions implicate novel genes in Alzheimer’s disease. Acta Neuropathol 137: , 557–569.

[195] 

Smith AR , Smith RG , Burrage J , Troakes C , Al-Sarraj S , Kalaria RN , Sloan C , Robinson AC , Mill J , Lunnon K ((2019) ) A cross-brain regions study of ANK1 DNA methylation in different neurodegenerative diseases. Neurobiol Aging 74: , 70–76.

[196] 

Lardenoije R , Roubroeks JAY , Pishva E , Leber M , Wagner H , Iatrou A , Smith AR , Smith RG , Eijssen LMT , Kleineidam L , Kawalia A , Hoffmann P , Luck T , Riedel-Heller S , Jessen F , Maier W , Wagner M , Hurlemann R , Kenis G , Ali M , Del Sol A , Mastroeni D , Delvaux E , Coleman PD , Mill J , Rutten BPF , Lunnon K , Ramirez A , van den Hove DLA ((2019) ) Alzheimer’s disease-associated (hydroxy)methylomic changes in the brain and blood. Clin Epigenetics 11: , 164.

[197] 

Smith AR , Smith RG , Pishva E , Hannon E , Roubroeks JAY , Burrage J , Troakes C , Al-Sarraj S , Sloan C , Mill J , van den Hove DL , Lunnon K ((2019) ) Parallel profiling of DNA methylation and hydroxymethylation highlights neuropathology-associated epigenetic variation in Alzheimer’s disease. Clin Epigenetics 11: , 52.

[198] 

Huo Z , Zhu Y , Yu L , Yang J , De Jager P , Bennett DA , Zhao J ((2019) ) DNA methylation variability in Alzheimer’s disease. Neurobiol Aging 76: , 35–44.

[199] 

Altuna M , Urdánoz-Casado A , Sánchez-Ruiz de Gordoa J , Zelaya MV , Labarga A , Lepesant JMJ , Roldán M , Blanco-Luquin I , Perdones Á , Larumbe R , Jericó I , Echavarri C , Méndez-López I , Di Stefano L , Mendioroz M ((2019) ) DNA methylation signature of human hippocampus in Alzheimer’s disease is linked to neurogenesis. Clin Epigenetics 11: , 91.

[200] 

Shireby G , Dempster EL , Policicchio S , Smith RG , Pishva E , Chioza B , Davies JP , Burrage J , Lunnon K , Seiler Vellame D , Love S , Thomas A , Brookes K , Morgan K , Francis P , Hannon E , Mill J ((2022) ) DNA methylation signatures of Alzheimer’s disease neuropathology in the cortex are primarily driven by variation in non-neuronal cell-types. Nat Commun 13: , 5620.

[201] 

Roubroeks JAY , Smith AR , Smith RG , Pishva E , Ibrahim Z , Sattlecker M , Hannon EJ , Kłoszewska I , Mecocci P , Soininen H , Tsolaki M , Vellas B , Wahlund LO , Aarsland D , Proitsi P , Hodges A , Lovestone S , Newhouse SJ , Dobson RJB , Mill J , van den Hove DLA , Lunnon K ((2020) ) An epigenome-wide association study of Alzheimer’s disease blood highlights robust DNA hypermethylation in the HOXB6 gene. Neurobiol Aging 95: , 26–45.

[202] 

Li QS , Sun Y , Wang T ((2020) ) Epigenome-wide association study of Alzheimer’s disease replicates 22 differentially methylated positions and 30 differentially methylated regions. Clin Epigenetics 12: , 149.

[203] 

Mitsumori R , Sakaguchi K , Shigemizu D , Mori T , Akiyama S , Ozaki K , Niida S , Shimoda N ((2020) ) Lower DNA methylation levels in CpG island shores of CR1, CLU, and PICALM in the blood of Japanese Alzheimer’s disease patients. PLoS One 15: , e0239196.

[204] 

Pishva E , Creese B , Smith AR , Viechtbauer W , Proitsi P , van den Hove DLA , Ballard C , Mill J , Lunnon K ((2020) ) Psychosis-associated DNA methylomic variation in Alzheimer’s disease cortex. Neurobiol Aging 89: , 83–88.

[205] 

Nazarian A , Yashin AI , Kulminski AM ((2020) ) Summary-based methylome-wide association analyses suggest potential genetically driven epigenetic heterogeneity of Alzheimer’s disease. J Clin Med 9: , 1489.

[206] 

Soerensen M , Hozakowska-Roszkowska DM , Nygaard M , Larsen MJ , Schwämmle V , Christensen K , Christiansen L , Tan Q ((2020) ) A genome-wide integrative association study of DNA methylation and gene expression data and later life cognitive functioning in monozygotic twins. Front Neurosci 14: , 233.

[207] 

Smith RG , Pishva E , Shireby G , Smith AR , Roubroeks JAY , Hannon E , Wheildon G , Mastroeni D , Gasparoni G , Riemenschneider M , Giese A , Sharp AJ , Schalkwyk L , Haroutunian V , Viechtbauer W , van den Hove DLA , Weedon M , Brokaw D , Francis PT , Thomas AJ , Love S , Morgan K , Walter J , Coleman PD , Bennett DA , De Jager PL , Mill J , Lunnon K ((2021) ) A meta-analysis of epigenome-wide association studies in Alzheimer’s disease highlights novel differentially methylated loci across cortex. Nat Commun 12: , 3517.

[208] 

Piras IS , Brokaw D , Kong Y , Weisenberger DJ , Krate J , Delvaux E , Mahurkar S , Blattler A , Siegmund KD , Sue L , Serrano GE , Beach TG , Laird PW , Huentelman MJ , Coleman PD ((2023) ) Integrated DNA methylation/RNA profiling in middle temporal gyrus of Alzheimer’s disease. Cell Mol Neurobiol, 10.1007/s10571-022-01307-3.