Abstract
A mechanistic understanding of the natural aging process, which is distinct from aging‐related disease mechanisms, is essential for developing interventions to extend lifespan or healthspan. Here, we discuss current trends in aging research and address conceptual and experimental challenges in the field. We examine various molecular markers implicated in aging with an emphasis on the role of heterochromatin and epigenetic changes. Studies in model organisms have been advantageous in elucidating conserved genetic and epigenetic mechanisms and assessing interventions that affect aging. We highlight the use of Drosophila, which allows controlled studies for evaluating genetic and environmental contributors to aging conveniently. Finally, we propose the use of novel methodologies and future strategies using Drosophila in aging research.
Keywords: ageing, aging, aging‐related diseases, Drosophila, epigenetics, geriatrics
1. TOWARD RIGOROUS DEFINITIONS OF THE TERMS USED IN AGING RESEARCH
In the past century, global life expectancy has almost doubled, owing to improved control of infectious diseases and enhanced quality of life. 1 The United Nations predicts that by 2050, the global geriatric population (age of 80 years old and above) will triple that of 2015. 2 Given the expected rise in the proportion of the elderly to the young, studies to elucidate mechanisms that promote healthy aging will be of paramount impact. Indeed, there has been a remarkable upsurge in the number of the articles related to aging in the past decade (Figure 1), reflecting intense interest in aging research. However, a plethora of challenges and questions remain to be addressed.
FIGURE 1.
Overall trend in aging‐related publications. The bar graph shows an exponential increase in the articles in the PubMed database with the keywords, “aging/ageing” in the title or abstract (from a search conducted in January 2020)
1.1. Aging contribution to aging‐related diseases
One of the most challenging questions in aging research is to distinguish the mechanisms and even phenotypes of aging from those of aging‐related diseases. Aging is a risk factor for a multitude of chronic conditions and diseases, including cardiovascular diseases, osteoporosis, dementia, osteoarthritis, type 2 diabetes, cancer, and chronic obstructive pulmonary disease (COPD). 3 The diversity of these diseases suggests that the cellular and physiological changes that determine the onset of aging‐related diseases are highly complex. While some suggest that aging is a distinct syndrome that should be considered separately from aging‐related diseases, others argue that these two overlap in underlying mechanisms to varying degrees. 2 , 4 The lack of consensus may underscore the necessity to characterize the natural aging process. 5 These aging‐related diseases can also manifest in those who are neither old in age nor physiologically aged. For example, cancers could also occur among young people. 6 Another example is Alzheimer's disease (AD), in which early‐ and late‐onset are considered to have separate etiologies, 7 perhaps due to different genetic backgrounds. As such, it is beneficial to have models for evaluating the relative degree of contribution of aging to diseases.
Rothman's 1976 causal pie model 8 was initially employed to model causal inference in the field of epidemiology. We propose that this method can also be applied to describe the causal relationship between aging and aging‐related diseases (Figure 2A), and this application may aid scientists in describing and quantifying the public health relevance to aging. We suggest considering aging or the cellular and physiological consequence of aging as a “component cause” (ie, a slice of a pie representing a factor that is not sufficient on its own to lead to the disease onset) of an aging‐related disease, which explains an increased incidence among older individuals. However, since the disease can also occur in young people, in this model, aging on its own is not considered as a “necessary cause” (ie, a slice present in all the pies, representing a factor that must be present for the disease onset). The “sufficient causes” (ie, complete pies that represent combinations of all the factors that contribute to the disease onset) thus represent different pathways toward the disease onset. The impact of targeting aging on public health can be conceptualized by reducing the proportion of “sufficient causes.” Methods have been developed to apply epidemiological data to causal pie models to quantify the burden of different classes of “sufficient causes” and the effect of interventions on a given “component cause,” 9 , 10 , 11 which may also be used for the aging field.
FIGURE 2.
Possible models to evaluate the contribution of the molecular consequences of aging to the onset of aging‐related diseases. A, Applying Rothman's 1976 causal pie model 8 to understanding the mechanisms of the onset of aging‐related diseases and the public health implications of targeting aging. B, Possible strategy for isolating the effect of aging on the outcome of various aging‐related diseases. It may also be important to elucidate possible multiplicative effects or interactions of aging with other variables, in order to address the degree of contribution of aging to aging‐related diseases. C, The current vs trending approach to tackle aging‐related diseases. A trending approach focuses on preventing aging‐related diseases by targeting aging itself, rather than therapeutic interventions directed at one specific aging‐related disease at a time
Considering the diversity in aging‐related diseases, finding shared molecular mechanisms may aid in isolating the effect of aging (Figure 2B). Such approach is not trivial, as the effect of aging may be additive, or multiplicative, depending on other variables. Previous therapeutic strategies focus on one aging‐related disease at a time and after its onset. In contrast, targeting aging can prevent or delay the onset of an array of aging‐related diseases simultaneously, therefore becoming an important alternative (Figure 2C). Studies using model organisms such as Drosophila can help provide mechanistic insights in the aging process and its interaction with diseases.
1.2. Lifespan and healthspan as distinct aging phenotypes
While “lifespan” refers to the time from birth to death, “healthspan” refers to the length of healthy life. Current goals for aging research emphasize on elucidating the interventions that delay the onset of diseases to extend healthspan. 1 , 5 However, the extension in healthspan is only meaningful when being related to the lifespan. Recently, sub‐definitions of healthspan extension—“chronological” vs “proportional”—were coined. 12 “Chronological” healthspan extension describes a delayed onset of morbidity proportionally with a prolonged lifespan. Therefore, the total morbidity time is shortened. “Proportional” healthspan extension refers to the total morbidity time that remains the same despite a delay in the onset due to extended lifespan.
In human cohort studies, it is challenging to quantify the healthspan extension due to the difficulty in obtaining health records for all different competing morbidities, as well as a lack of a clear consensus of the conditions that constitute morbidity and warrant intervention. For example, geriatric patients often experience circadian rhythm shift, 13 , 14 however, this symptom is often unreported or not considered as a significant morbidity to be included in measuring healthspan. Thus, a recent publication 5 demonstrated a table that indexed healthspan measures based on the World Health Organization's codified patient classification systems, various physiological measurements, other reported measures of frailty such as the Healthy Aging Index, 15 Successful Aging Index, 16 and Healthy Aging Score. 17 It also summarizes the evidence found in model organisms such as mice and Caenorhabditis elegans. Below, we propose to extend this chart by including relevant Drosophila phenotypes and the assays measuring them (part 2b).
2. ASSAYS AND METHODOLOGIES FOR THE AGING RESEARCH IN DROSOPHILA
2.1. Drosophila as a model organism for aging studies
The trend using model organisms in aging research have shifted from ascertaining only longevity (lifespan) to also include physiological assessments (healthspan). 18 Drosophila melanogaster is a genetically tractable model organism with a long history in aging research. The first documented aging experiment was conducted in 1916, which assessed the effect of temperature and food composition. 19 Using Drosophila, the pro‐ or anti‐aging effects of environmental factors and pharmacological interventions can be easily assessed. Since a large number of flies with the same genotype can be investigated under controlled conditions, it allows scientists to conduct statistically powerful experiments at low cost. The fly model is also amenable to largescale screens for new interventions and test the combinations of interventions quickly. Human cohort studies of environmental factors are notoriously susceptible to confounding and reverse causation. The Drosophila model system can help validate the findings in human studies and establish causality. The short lifespan is another major advantage. The typical lifespan of Drosophila in the optimistic temperature of 25°C is approximately 60 days on average. Increasing the temperature to 29°C leads to a reduced lifespan, while decreasing it to 18°C does the opposite. 20
Moreover, Drosophila is a convenient genetic system with numerous mutants and transgenic tools. For example, the well‐established UAS/GAL4 system allows tissue‐specific transgene ectopic expression 21 and temporal control can also be achieved using a temperature or hormone‐inducible variant of the system. 22 , 23 RNAi knockdown studies can be conducted easily using a curated collection of RNAi lines that target most transcripts. 24 In addition, the CRISPR‐Cas9 tools for gene editing in vivo have been developed in Drosophila. 25 Using these methods, scientists have effectively conducted the mechanistic studies, which are challenging in humans, to understand conserved genetic pathways implicated in human aging. Such pathways include sirtuins and other chromatin regulators (Table 2), and those involved in regulating inflammation, 26 oxidative stress detoxification, 27 insulin signaling, 28 and mammalian Target of Rapamycin (mTOR) pathway. 29
TABLE 2.
Histone‐related mechanisms conserved between Drosophila and human aging
Species | Epigenomic modifications | References |
---|---|---|
Histone methylation—transcriptional repression marks | ||
H3K9me3 colocalized with HP1a in heterochromatin | ||
Drosophila |
|
[91] |
|
[32] | |
|
[96] | |
|
[51] | |
|
[93] | |
|
[95] | |
|
[107, 286] | |
|
[123] | |
Human |
|
[287] |
|
[288] | |
|
[289] | |
|
[290] | |
|
[114] | |
|
[291] | |
|
[114, 115, 116, 117, 118] | |
H3K27me3 | ||
Drosophila |
|
[149] |
|
[228] | |
|
[150] | |
|
[151] | |
Human |
|
[152] |
|
[159] | |
Histone methylation—transcriptional activation marks | ||
H3K4me3 | ||
Drosophila |
|
[91] |
|
[168] | |
Human |
|
[292] |
H3K36me3 | ||
Drosophila |
|
[91] |
Human |
|
[169] |
Histone acetylation | ||
Drosophila |
|
[293] |
|
[93] | |
|
[73] | |
|
[71] | |
|
[72] | |
|
[174] | |
|
[176] | |
|
[179] | |
Human |
|
[294] |
|
[295] | |
Chromatin remodeling | ||
NuRD | ||
Drosophila |
|
[179] |
Human |
|
[296] |
2.2. Applying fly‐specific assays to the understanding of human healthspan index
Various assays have been developed in Drosophila for observing aging phenotypes and aging‐related disease models, which may relate to human healthspan ( 5 ; Table 1). For example, survival curve measurements are standard methods for assessing lifespan. The assessment of aging‐related functional decline or functional senescence aids the quantification of the healthspan extension resulting from specific interventions. 30 Investigators may combine both measurements to evaluate how an intervention affects lifespan and healthspan.
TABLE 1.
Suggested assays in Drosophila to investigate traits that contribute to healthspan in humans
Healthspan traits conserved between Drosophila and humans | Drosophila phenotype concept in the Gene Ontology (GO) | Relevant assays to investigate healthspan traits in Drosophila |
---|---|---|
Physiological function | ||
Stress resistance | GO:0006950—response to stress | Survival kinetics in the presence of environmental stressors |
Thermotolerance heat shock tolerance | GO:0009408—response to heat | Heat shock survival kinetics 41 |
Hypoxic stress tolerance | GO:0001666—response to hypoxia | Survival in hypoxic environment. 54 |
Oxidative stress tolerance | GO:0006979—response to oxidative stress | Survival kinetics fed with paraquat 55 and other oxidative stress‐inducers |
Metabolic status Metabolic homeostasis Metabolic pathology |
GO:0019222—regulation of metabolic process | Assays to quantify metabolites, mitochondrial activity, and other markers of metabolic state 273 , 274 , 275 |
Redox status Redox homeostasis Redox pathology |
GO:0051775—response to redox state | Assays to quantify reactive oxygen species 276 , 277 |
Immune status Immune homeostasis Immune pathology |
GO:0006955—immune response | Assays to quantify innate immunity and phagocytic response |
Both physical strength and cognitive function | ||
Motivated locomotion Stimulated locomotion |
GO:0007626—locomotion in response to stimulus |
Geotaxis assay 31 , 32 , 33 with shaking Response to light, olfaction, and taste 44 Variations of olfactory maze tests 278 , 279 , 280 Taste preference assay 281 Real‐time imaging of response to gentle pricking and rotation, odor‐stimulated movement, and neuroactivity imaging 282 |
Motor balance dexterity | GO:0050885—regulation of balance | High‐speed real‐time machine vision robot developed for Drosophila to capture behavioral response to gentle pricking and rotation, odor‐stimulated movement, and neuroactivity imaging 282 |
Cellular integrity: Muscle Neuronal Intestinal Pathological loss of cell integrity |
GO:0046716—muscle fiber maintenance GO:0070050—neuron maintenance GO:0060730—epithelial structure maintenance |
Muscle fiber integrity by actin filament staining 32 Brain degeneration vacuoles 31 , 41 , 42 Retinal degeneration and loss of ommatidium photoreceptor integrity 41 , 42 Neuromuscular junction (NMJ) marker abnormality quantification 42 , 43 |
Physical strength | ||
Locomotion Unmotivated Unstimulated |
GO:0040011—locomotion (self‐propelled) |
Geotaxis assay 31 , 32 , 33 with no shaking Variety of flight duration 33 , 34 and exploratory activity tests 33 , 35 , 36 |
Grip strength | Measured in mice, but not a conserved trait in Drosophila | |
Gait speed, chair rising | Similar traits measured in mice, but not a conserved trait in Drosophila | |
Muscle integrity pathological loss of muscle integrity | GO:0046716—muscle fiber maintenance | Muscle fiber integrity by actin filament staining 32 |
Cognitive function | ||
Sensory perception | GO:0007600—sensory perception |
Response to light, olfaction and taste 44 Variations of olfactory maze tests 278 , 279 , 280 Taste preference assay 281 Odor‐stimulated movement real‐time imaging 282 |
Short‐term memory processing speed | GO:0007614—short‐term memory |
Response to light, olfaction, and taste 44 |
Sleep defective | GO:0030431—sleep |
Drosophila Activity Monitor (DAM) device and accompanying analysis software called ShinyR‐DAM 283 Locomotor activity monitoring and phase‐response curves 284 Sleep behavior assay 285 |
Cardiac rhythm | GO:0042749—regulation of circadian sleep/wake cycle | Heart rate measurement 52 |
Executive function verbal function | Drosophila is not verbal | |
Neuronal integrity neuronal pathology | GO:0070050—neuron maintenance |
Brain degeneration vacuoles 31 , 41 , 42 Retinal degeneration and loss of ommatidium photoreceptor integrity 41 , 42 Neuromuscular junction (NMJ) marker abnormality quantification 42 , 43 Neuroactivity real‐time imaging 282 |
Reproductive function | ||
Number of offspring Offspring health Surviving offspring |
GO:0019953—sexual reproduction |
Eggs laid, number of progeny relative to copulation 48 , 49 Quantify germ line stem cell number to evaluate reproductive function 50 , 51 |
Among commonly used assays for assessing healthspan are those that measure movement. These include the negative geotaxis assay for evaluating climbing ability, 31 , 32 , 33 and a variety of flight duration, 33 , 34 and exploratory activity tests. 33 , 35 , 36 The movement weakening during aging may be due to skeletal muscle deterioration or cognitive decline and can be further examined using histological methods. For example, staining of actin filaments help visualize the loss of muscle fiber integrity, 32 and various neuronal markers allow the detection of the loss of specific neurons. 37 , 38 , 39 , 40 Other aging‐related neurological phenotypes can be also quantified, including brain degeneration vacuoles, 31 , 41 , 42 retinal degeneration and loss of ommatidium photoreceptor integrity, 41 , 42 and neuromuscular junction abnormalities. 42 , 43 Aging‐related cognitive decline can be assessed by evaluating the fly response to light, olfaction and taste, 44 and memory and learning. 45 , 46 , 47
Reproductive decline also occurs during aging and can easily be investigated by quantifying egg‐laying or the number of progeny relative to copula copulation. 48 , 49 Additionally, germ line stem cells can be stained and quantified. 50 , 51 Cardiac function assays include monitoring heart rate 52 and cardiac tube wall movements. 53 Heat shock survival kinetics 41 or survival in hypoxic environment 54 can be used to assess the resistance to stress. Another commonly used assay is to evaluate oxidative stress resistance through the survival on paraquat. 55 Assays were also developed for assessing the impact of social interaction on lifespan. 56
2.3. Using Drosophila for the mechanistic studies of gene‐environment interactions
A large number of aging studies have been conducted in Drosophila to characterize the effect of exposure to various environmental agents (notably, diet, microbial interactions, pharmacological interventions, and pollutants) on aging. Such strength is best exemplified by the studies investigating the aging impact of dietary restriction (DR), pharmacological and genetic manipulation of Sirtuin family of histone deacetylases (HDACs), mammalian target of rapamycin (mTOR) signaling, and insulin signaling. Here we highlight the aging studies in Drosophila that helped the mechanistic understanding in mammals.
2.3.1. Dietary Restriction
It is well‐established that DR enhances lifespan, and this appears to be conserved across species ranging from yeast to invertebrates to rodents. However, whether this conservation is simply due to reduced caloric intake remains elusive. Several Drosophila studies have shown that the effect was determined by the relative composition of yeast, sugar and essential amino acids (EAAs), rather than a mere reduction in calories. 57 , 58 , 59 , 60 , 61 One study found that supplementing with EAA negated the lifespan extending effect of DR, thus isolating the relevant dietary component, 60 and interestingly, this effect was accompanied by reduced fecundity. 60 Another study found that a short‐term DR was also effective, suggesting that the intervention later in life in humans might be a valuable method. 62 Recently, a randomized control trial of DR, the Comprehensive Assessment of Long‐term Effects of Reducing Intake of Energy (CALERIE) study was conducted, in which healthy and young subjects were randomly assigned to 2 years of caloric restriction or control diet. 63 , 64 , 65 , 66 , 67 , 68 , 69 , 70 Ongoing analyses of physiological measurements and molecular profiling of biobank tissue sample are expected to yield invaluable information about the effect of DR on humans. Nevertheless, since controlled experiments and isogenic models are not feasible in human studies, using Drosophila is still expected to be advantageous for elucidating mechanistic understanding.
2.3.2. Sirtuins and Dietary Restriction
The link between aging and the sirtuin family of histone deacetylases (HDACs) have been controversial. Studies in Drosophila helped draw the conclusion that the expression level of Sirtuin 2 (Sirt2) is important for the effect (described in detail in Section 3.1). For instance, the lifespan extension by DR was abrogated in Sirt2 mutant flies, and different dietary conditions altered the magnitude of the effect. 71 Such findings again highlight the advantage of using the Drosophila model for meticulous and controlled studies of gene‐environment interactions, which are challenging in humans. Additionally, the Drosophila model was advantageous in ascertaining the lifespan extension effects of the Sirtuin‐activating compounds including resveratrol, fisetin, and alkylresorcinols. 72 , 73
2.3.3. mTOR signaling and dietary restriction
Drosophila has proven to be advantageous for understanding the impact of the mTOR signaling pathway on aging genetically, pharmacologically, and under different dietary conditions. It is well‐established that mTOR is a master regulator of protein synthesis and cell growth, and that an augmented mTOR signaling is negatively correlated with lifespan in multiple species. 74 It has been demonstrated that supplementing with EAA negates the lifespan extending effect of DR by upregulating mTOR, while rapamycin, a mTOR inhibitor abolished this effect and extend the lifespan. 60 Further characterization of tissue‐specific gene expression changes identified GATA transcription factors, the zinc finger DNA binding proteins, as the downstream mediator of mTOR signaling 59 ; GATA transcription factors have been shown to regulate lifespan in multiple organisms. These studies exemplify the strength of combining genetic tools with pharmacological treatments to dissect molecular mechanism of aging.
2.3.4. Insulin/IGF and JNK/foxo signaling, metabolism, and dietary composition
It is well‐established that reducing insulin/insulin‐like growth factor (IGF) signaling extends lifespan across species, including humans and Drosophila, and that one major signaling pathway mediating the effect is the c‐Jun N‐terminal Kinase (JNK)/Forkhead box and sub‐group O (foxo) pathway. 75 , 76 , 77 The JNK/foxo pathway is a conserved mechanism that confers protection against stress, as well as counters the activity of Insulin/IGF signaling. 28 Consistent with this notion, a study found that in the fly head, the number of genes regulated by the Foxo transcription factor significantly decreased with age, and many Foxo‐targeted genes were also altered in their expression profile (up‐ or downregulation) during aging. 78 Moreover, a high sugar diet that reduces lifespan was found to augment the insulin/IGF signaling and reduce the Foxo signaling. 79
Studies using the Drosophila model organism underscore the importance of considering various environmental exposures that interact with this pathway. For instance, while overexpressing foxo in the head's fat body leads to lifespan extension, it was found that this only occurred under a high level of dietary yeast. 80 Moreover, this study showed that foxo heteroallelic null mutants on DR had the lifespan comparable to wildtype controls, suggesting that the lifespan extension effect of DR may not be mediated by Foxo. 80 DR or JNK/Foxo signaling could also regulate separate pathways to extend lifespan; DR was found to reduce the expression of insulin‐like peptide‐5 (ilp5), while foxo overexpression lead to the reduction of insulin‐like peptide‐2 (ilp2). Another study showed that different food concentrations are required for wildtype and the insulin receptor substrate, chico mutants to achieve a maximum median of lifespan. 81 This study suggests that the genetic effects of longevity may depend on food composition.
Another gene that was found to prolong lifespan in Drosophila is I'm not dead yet (Indy). Indy encodes a plasma membrane transporter of Krebs cycle metabolites. 82 Given the well‐established role of metabolism in aging across species, this finding may be relevant widely. By simultaneously assessing whether there is a trade‐off between lifespan extension and healthspan measures of fecundity, locomotion and metabolism, the study found that increased lifespan with Indy mutation did not alter healthspan measures under normal food conditions, however, the reduced fecundity occurred under the DR conditions. This suggests that the trade‐off between reproduction and lifespan depend on both environmental conditions and genetic susceptibility. Together, these studies highlight the advantage of using Drosophila to gain mechanistic insights in the conditions that affect both lifespan and healthspan.
3. EPIGENETIC MECHANISMS IMPLICATED IN DROSOPHILA AND HUMAN AGING
The mechanisms involving epigenetic regulation (histone modifications, DNA methylation, noncoding RNAs, RNA modifications) are among the major models of aging, presented as the “hallmarks” of aging 83 and the “seven pillars” of aging by the NIH steering conference on aging. 84 The genetic heritability of longevity in humans is estimated to be only 15% to 40%, 85 suggesting the likely significant contribution of epigenetic and environmental factors. In this section, we present major studies describing different epigenetic mechanisms in aging, particularly those conserved between Drosophila and humans. These studies are summarized in Tables 2, 3, 4.
TABLE 3.
Conserved mechanisms in noncoding RNAs, RNA modification, and RNA editing implicated in aging in Drosophila and human
Species | Molecular mechanism and aging phenotypes | Species‐Gene | Concepts | References |
---|---|---|---|---|
RNAi pathway | ||||
Drosophila |
Loss‐of‐function mutations Argonaute 2 (AGO2) decreased lifespan Also increased TE expression in the brain, and memory impairment |
Dme‐AGO2 | Determination of adult lifespan; transposable element; gene expression; learning; or memory | [140] |
Dcr‐2 (Dicer‐2) overexpression increased lifespan and repressed TE elevation in the head and fatbody seen during aging Dicer‐2 loss‐of‐function mutation increased DNA double‐strand breaks Reverse transcriptase inhibitor 3TC treatment attenuated TE activation and increased life span in Dicer‐2 mutants |
Dme‐Dcr‐2 | Determination of adult lifespan; DNA double strand breaks; transposable element retrotransposon; gene expression | [93] | |
Mt2 (Dnmt2 ortholog) overexpression increased lifespan | Dme‐Mt2 | Determination of adult lifespan; gene expression | [141] | |
tRNA methylation by Dnmt2 and fragmentation during heatshock was shown to be required for efficient Dcr‐2 function in Drosophila and thus the lifespan effect may be relevant to RNAi pathway |
Dme‐Mt2 Dme‐Dcr‐2 |
Determination of adult lifespan; gene expression; RNAi; heat shock | [297] | |
PIWI pathway | ||||
Drosophila |
piwi attenuated intestinal stem cell activity reduction during aging Loss‐of‐function piwi in intestinal stem cells induced Gypsy retrotransposon transcriptional activation and insertion, DNA damage and apoptosis |
Dme‐piwi | Intestinal stem cells; DNA damage response; apoptotic process | [146] |
Human | Multiple studies have found many different PIWI‐interacting RNAs (piRNAs) to be relevant to human cancers | Many different piRNAs | Gene expression; cancer | Reviewed in [147] |
Micro‐RNAs (miRNAs) | ||||
mir‐34 | ||||
Drosophila |
Loss of mir‐34 resulted in gene profile alterations associated with accelerated brain aging and reduced lifespan Upregulation of mir‐34 increases lifespan and alleviates neurodegeneration induced by human pathogenic polyglutamine disease protein |
Dme‐mir‐34 | Determination of adult lifespan; polyQ models neurodegenerative disease | [41] |
Age‐associated mir‐34 reduction and its effect on brain aging was mediated by translational repression of Eip74EF and putatively via developmental gene silencing |
Dme‐mir‐34 Dme‐Eip74EF |
miRNA mediated inhibition of translation | [227] | |
miR‐34 RNA inhibits polycomb repressive complex 2 to modulate chaperone expression and promote healthy brain aging H3K27me3 levels increase with age and alleviated in mir‐34 mutants |
Dme‐mir‐34 |
ESC‐E(Z) complex H3K27me3 |
[228] | |
Human |
MIR34A overexpressed with aging SIRT1 gene is downregulated in PBMCs |
Hsa‐MIR34A Hsa‐SIRT1 |
Gene expression PBMC |
[229] |
GWAS association |
Hsa‐SIRT1 rs7096385‐T |
GWAS on atrial fibrillation | [298] | |
Type 2 diabetes is associated with increased expression of MIR34A in the heart | Hsa‐MIR34A |
Type 2 diabetes mellitus gene expression Cardiac muscle tissue |
[230] | |
Overexpressed MIR34A in the lungs of patients with idiopathic pulmonary fibrosis, and functional studies found it promotes senescence and reduces cell proliferation | Hsa‐MIR34A |
Gene expression Idiopathic pulmonary fibrosis Lung Cell senescence Cell proliferation |
[231] | |
Increased MIR34A expression was associated with advanced hepatocellular carcinoma | Hsa‐MIR34A |
Gene expression Hepatocellular carcinoma |
[232] | |
miR‐125 | ||||
Drosophila | Loss of miR‐125 results in reduced lifespan, climbing activity, and increased vacuoles in the brain | Dme‐miR‐125 | Determination of adult lifespan | [31] |
Human | Several studies have shown dysregulation of MIR125b (mature sequence of the MIR125B1 and MIR125B2 stem loop transcripts) in various cancer, and it appears to have tumor suppressive and oncogenic activity depending on tumor type |
Hsa‐MIR125B1 Hsa‐MIR125B2 |
Tumor suppressor gene oncogene | Reviewed in [234] |
miR‐9 | ||||
Drosophila |
The level of Dme‐miR‐9a expression is elevated in testes germline stem cells miR‐9a targets transcripts encoding N‐cadherin, adhesion molecule miR‐9a‐null mutants can maintain larger number of stem cells despite aging, however, have decreased tissue regeneration and reduced fertility |
Dme‐miR‐9a Dme‐CadN Dme‐CadN2 |
Gene expression male germ‐line stem cell population maintenance tissue regeneration fertility | [233] |
Human |
MIR9 (mature sequence of the MIR9‐1, MIR9‐2, and MIR9‐3 stem loop transcripts) expression increases with aging and SIRT1 gene is downregulated in PBMCs Functional assay found that miR‐9 interacts with the 3′UTR of SIRT1 mRNA |
Hsa‐MIR9‐1 Hsa‐MIR9‐2 Hsa‐MIR9‐3 Hsa‐SIRT1 |
Gene expression PBMC miRNA mediated inhibition of translation |
[229] |
MIR9 expression increases and IGF‐1R and FOXO1 are downregulated, in PBMCs |
Hsa‐MIR9‐1 Hsa‐MIR9‐2 Hsa‐MIR9‐3 Hsa‐IGF1R Hsa‐FOXO1 |
Gene expression PBMC |
[235] | |
Long non‐coding RNAs (lncRNAs) | ||||
Drosophila | Relevance of lncRNA in aging is suggested by a study that profiled lncRNAs in the head, gut and fat body of flies reared in dietary restrictive condition that was found to increase lifespan | 102 differentially expressed lncRNAs |
Gene expression Response to caloric restriction Determination of adult lifespan |
[224] |
Human |
lncRNA involved in aging lncRNA involved in aging‐related diseases |
15 lncRNAs, mainly associated with senescence and cell cycle regulation | Reviewed in [213, 217] | |
Circular RNAs (circRNAs) | ||||
Drosophila | Accumulation of circRNAs in neural tissues to occur with aging |
circRNA Drosophila neuron |
[240] | |
Strong increase of circRNAs during aging in photoreceptors |
circRNA Drosophila photoreceptor cell |
[241] | ||
Human | Several lines of evidence collectively suggest a key role for circRNAs in aging | circRNA | Reviewed in [242] | |
RNA methylation—tRNA m 5 C | ||||
Drosophila | Mt2 (Dnmt2) overexpression increased lifespan | Dme‐Mt2 |
Gene expression Determination of adult lifespan |
[141] |
Mt2 (Dnmt2) was found to be an tRNA m5C methyltransferase and thus the lifespan effect may be relevant to tRNA methylation | Dme‐Mt2 | tRNA methyltransferase | [204, 205, 206, 207] | |
Human | Knocking down TRDMT1 (Dnmt2) in human fibroblasts result in increased oxidative stress, DNA damage, upregulation of miRNAs related to proliferation, and results in senescence | Hsa‐TRDMT1 |
tRNA methyltransferase Fibroblast Oxidative stress‐induced premature senescence |
[258] |
RNA editing—Adenosine‐to‐inosine editing | ||||
Drosophila | Adenosine deaminase acting on RNA (ADAR) extended lifespan, with increased levels of histone modifications associated with heterochromatic silencing in brain neuronal cells | Dme‐ADAR | Determination of adult lifespan | [259] |
Human | 18 SNPs in ADARB1 (5 SNPs) and ADARB2 (13 SNPs) are associated with extreme longevity in a cohort combining the New England Centenarian Study and Southern Italian Centenarian Study. They were replicated in the Ashkenazi Jewish Centenarian Study and Japanese Centenarian Study |
Hsa‐ADARB1 Hsa‐ADARB2 |
Bayesian approach GWAS Determination of adult lifespan |
[260] |
GWAS SNP interaction association to age related hearing impairment |
Hsa‐ADARB2 rs787274‐? x rs11250795‐? |
Age‐related hearing impairment | [299] | |
GWAS association to Alzheimer diagnosis accelerated after diagnosis of cognitive impairment |
Hsa‐ADARB2 rs10903488‐? |
GWAS on Alzheimer's disease, cognitive decline measurement, and cognitive impairment | [300] | |
GWAS association to age at menarche |
Hsa‐ADARB2 rs1874984‐C |
GWAS on age at menarche | [301] | |
GWAS association to age at menarche |
Hsa‐ADARB2 rs1874984‐? |
GWAS on age at menarche | [302] |
TABLE 4.
Conserved mechanisms involving nuclear and chromosomal architecture implicated in aging in Drosophila, mouse, and human
Species | Nuclear and chromosomal architecture | References |
---|---|---|
Maintenance of meiotic sister chromatid cohesion | ||
Drosophila |
|
[303] |
|
[304] | |
Mouse |
|
[305] |
Human |
|
[306] |
|
Reviewed in [307, 308] | |
Integrity of the nuclear lamina | ||
Drosophila |
|
[132, 133, 134] |
Human |
|
[129, 130] |
|
[131] | |
|
[309] |
3.1. Heterochromatin redistribution and epigenetic changes during aging
3.1.1. Overview
The eukaryotic genome is highly organized with nucleosomes (DNA with histone proteins, H2A, H2B, H3, and H4) as a fundamental unit of chromatin. The histone proteins with various types of chemical modifications are highly conserved across species. 86 Chromatin consists of heterochromatin and euchromatin regions. 86 , 87 , 88 Heterochromatin are gene‐poor, transcriptionally silent, and highly condensed regions, which are generally characterized by histone hypoacetylation and the enrichment of repressive histone marks, H3K9me2 and H3K9me3, and HP1α proteins. 86 , 88 In contrast, euchromatin regions are relatively gene‐rich, transcriptionally active, less condensed, and enriched with histone hyperacetylation and active histone marks such as H3K4me2 and H3K4me3. 86 , 88
Heterochromatin redistribution in the aging process has been widely reported. While an aberrant gain of heterochromatin at specific gene promoters induces transcriptional repression, in other promoters, the loss of heterochromatin results in abnormal transcriptional activity. It was speculated that the loss of protective heterochromatin at constitutive heterochromatin loci during the aging process might increase the rate of chromosomal aberrations and the susceptibility to double‐stranded DNA breaks in the genome. 89 , 90 Another notable consequence is the activation of transposable elements (TEs) that can cause mutagenesis events such as insertion and deletion. 89 , 90 Identifying the role of heterochromatin in specific regions has therefore become one of the major goals in the aging field.
3.1.2. Repressive hallmark of heterochromatin—H3K9 methylation and HP1a
Generally, aging‐related disruptions in the repressive marks, H3K9me3 and HP1a, especially in constitutive heterochromatin, are correlated with reduced lifespan and compromised healthspan measures in Drosophila. 32 , 51 , 91 , 92 , 93 , 94 , 95 Specifically, a study 91 found that these marks were most dramatically reduced at pericentric heterochromatin and the fourth chromosome in aged flies. Another study in Drosophila demonstrated that the loss‐of‐function HP1a heterozygous mutants with loss of heterochromatin resulted in the disruption of ribosomal RNA (rRNA) gene loci and an increase in ribosomal RNA (rRNA) transcription, and these flies exhibited significantly shortened lifespan and the loss of muscle integrity, thus demonstrating a link between the loss of heterochromatin and metabolism. 32 It was also shown in this study that overexpressing HP1a had the opposite effects, resulting in increased lifespan and the maintenance of muscle integrity. 32 Additionally, knocking down HP1a or Su(var)3‐9 (H3K9 methyltransferase) in the male germ cells caused an early reduction of stem cells 51 ; stem cell exausting is widely regarded as one of aging phenotypes. Together, these studies strongly support that H3K9 methylation and HP1a play an important role in maintaining the heterochromatin, and the disruption of heterochromatin can negatively impact both lifespan and healthspan.
Other pathways were found to regulate heterochromatin through their interaction with HP1 and Su(var)3‐9. A genetic screen in Drosophila that intended to identify novel components in the Janus kinase (JAK) signaling pathway, found that HP1 and Su(var)3‐9 mutations enhanced a hyperactive JAK allele, which globally disrupts heterochromatic gene silencing. 92 It was demonstrated that a JAK downstream transcription factor, the signal‐transducer and activator of transcription protein at 92E (Stat92E), in its unphosphorylated form, interacts with HP1a, promoted heterochromatin formation, 92 , 96 , 97 and enhanced genome stability 98 ; moreover, this interaction was also found to protect against leukemia‐like cancer formation. 92 A similar mechanism was also demonstrated in mammals to suppress cancer development, 99 , 100 highlighting the advantage of using Drosophila for discovering conserved mechanisms.
These findings in Drosophila is consistent with an aging model in mammals, 101 , 102 , 103 in which constitutive heterochromatin regions including TEs and rDNA also lose the major heterochromatin markers, HP1a and H3K9me3, while aberrant heterochromatin forms in other regions. The loss of heterochromatin during aging is conserved in budding yeast, mice, and human cells, underscoring the importance of this mechanism. 104 Mobilization of TEs during the aging process has become increasingly appreciated. 105 For example, TE activation and genomic instability were found in the neurons of a Drosophila model of AD as well as the postmortem brain tissue from AD patients. 106 Recently, an in vivo drug screen in Drosophila identified methotrexate, a compound for treating arthritis, to augment H3K9me3 and HP1a foci, thus promoting heterochromatin formation and genome stability. 107 This may lead to new interventions targeting aging. These findings highlight the relevance of the Drosophila as a model for understanding the aging process in humans.
During mammalian aging, prominent genomic regions acquire the aberrant heterochromatin including the Senescence‐associated heterochromatin foci (SAFH). This can be triggered by the upregulation of the p16(INK4a) tumor suppressor and the consequent repression of E2F‐target genes mediated by the Retinoblastoma (Rb) tumor suppressor. 108 , 109 , 110 , 111 , 112 , 113 The SAFH formation involves the recruitment of H3K9me2/3 and HP1α, the enrichment of a histone variant, macroH2A and a DNA damage marker, phosphorylated H2AX (γH2AX). 114 , 115 , 116 , 117 , 118 Other pathways are also involved. It was speculated that senescent cells drive aging phenotypes by secreting signaling molecules, including those that promote chronic inflammation 119 ; inflammation is widely accepted as an aging hallmark Replicative senescence, an irreversible arrest of cell proliferation perhaps due to DNA repairing, can also be triggered by p53 115 ; DNA damages generally accumulate with aging. Studies of cellular senescence in the mammalian system are complicated because senescence occurs in progressive steps, 120 and different types of senescent states appear to exist, which are caused by different triggers 121 and involve different molecular pathways. 115 , 122
However, the Drosophila genome lacks the conservation of p16, which is one of the well‐established triggers of senescence and plays an important role in regulating aging in mammalian cells. Instead, overexpression of a hyperactive allele of the Ras oncogene, along with mitochondrial dysfunction, could induce senescence. 123 The downstream events are more conserved, including the activation of p53 and JNK pathways and the inhibition of the Hippo pathway; and in turn, these events lead to the upregulation of the Unpaired (Upd) inflammatory cytokine, an aging hallmark Clonal analyses also found an increase in H3K9me3 in the cells in which senescence was induced. Therefore, the Drosophila model appears to have a mammalian‐like mechanism of cellular senescence. Future studies that address the regulation of specific loci subject to heterochromatin redistribution are important in both Drosophila and humans.
3.1.3. Nuclear lamina and heterochromatin
Nuclear lamina (NL), a lining of the inner nuclear envelope, consists of filaments, lamins, and lamin‐associated proteins, and is directly tethered with heterochromatin. 124 Heterochromatin domains of the genome are anchored to the NL, allowing them to reside in the nuclear periphery. It has been suggested that the spatial positioning of the heterochromatin is mechanistically important for their repressiveness of transcriptional activity. 125 The lamin‐B receptor (LBR) forms a complex with HP1α, demonstrating a direct link. 126 Moreover, Proline Rich 14 (PRR14), a protein that binds to lamin‐A (LMNA), can also interact with HP1α. In human cells, the PRR14 depletion results in the redistribution of H3K9me2 and H3K9me3, releasing chromatin from nuclear periphery to the interior area. 126 The recent development of chromosome conformation‐capture technologies allows the assessment of 3D chromatin structure. Using this technique, it was found that aging‐associated heterochromatin redistribution and chromatin alterations also occurred at higher orders of the structure, suggesting a complex role played by chromosomal organization in senescence and aging. 127 , 128 Further understanding of chromatin organization and sub‐nuclear localization is expected to lead to important findings in the aging mechanism and aging‐related diseases.
The Hutchinson‐Gilford progeria syndrome (HGPS, https://www.omim.org/entry/176670), which is characterized by an extreme premature aging phenotype, is caused by various germline mutations in LMNA, suggesting a mechanistic link between nuclear architecture, heterochromatin, and aging. 129 , 130 In senescent human dermal fibroblasts and keratinocytes, lamin‐B1 (LMNB1) and lamina‐associated polypeptide 2 (LAP2) protein levels are reduced, and this reduction was also recapitulated in skin harvested from young vs old individuals. 131
In Drosophila, studies have also provided evidences for the relevance of the NL in natural aging. 132 , 133 , 134 A progressive reduction in LMNB expression in the fat body was observed with aging, and moreover, found to contribute to chronic inflammation and gut hyperplasia. 134 Intriguingly, knocking down LMNB in young adult or larval fat body resulted in reduced heterochromatin, and an increase in retrotransposon expression and DNA damage. 132 , 133 Collectively, these studies provide evidence of the importance of NL in controlling nuclear architecture, heterochromatin, TE activation, genome instability and chronic inflammation in aging. This again suggests the conservation between Drosophila and human aging.
3.1.4. RNAi pathway related to heterochromatin and TE silencing
One of the major roles of the RNAi pathway is in regulating heterochromatin‐induced silencing of repetitive DNA loci, such as TEs, pericentric satellite repeats, and telomeric repeats. 135 , 136 , 137 (Its other major role in post‐transcriptional gene silencing is discussed in Section 3.3). The RNAi pathway's regulation of heterochromatin formation involves the interaction between RNA and H3K9me3 methyltransferases and promoting HP1α recruitment. 138 , 139 In Drosophila, RNAi pathway components, Dicer‐2 (Dcr‐2) and Agonaute (Ago2), have been shown to regulate lifespan and other aging‐related phenotypes by suppressing TEs. 93 , 140 , 141 Overexpressing Dcr‐2 in Drosophila led to increased lifespan, with concomitant repression of TEs, while the loss‐of‐function of Dcr‐2 resulted in an increase in DNA double‐strand breaks. 93 Lamivudine (3TC), a rreverse transcriptase inhibitor, prolonged lifespan and attenuated TE mobilization caused by Dcr‐2 loss‐of‐function mutants. 93 An Ago2 loss‐of‐function mutation reduced lifespan and caused neuronal phenotypes such as memory impairment and TE activation in the brain, 140 , 142 and neurodegeneration and increased brain vacuoles ( 142 ; described in detail in Section 3.4). In humans, knocking down DICER 1 in HEK293T and HepG2 cells increased double‐stranded DNA damage, 143 however, the mechanistic studies in this area have not been conducted in Drosophila.
3.1.5. PIWI/piRNA related to heterochromatin and TE silencing
It is well‐established that the PIWI/piRNA pathway plays a crucial role in silencing TEs in the germline of Drosophila 144 by regulating heterochromatin formation. 145 More recently, a Drosophila study has found that Piwi is crucial for suppressing age‐related TE expression in intestinal stem cells and the maintenance of epithelial homeostasis, thus demonstrating its new role in somatic stem cell maintenance. 146 The PIWI pathway has recently been appreciated as a key player in cancer as well. 147
3.1.6. Silencing histone methylation mark, H3K27me3
It is well‐established that H3K27me3 promotes gene silencing and is associated with polycomb repressive complex (PRC) 1 and 2. 148 These complexes and sub‐complexes share some components and have distinct and overlapping target loci. Evidence in Drosophila supports the presence of specific genomic areas with aging‐associated augmentation of H3K27me3, 149 , 150 , 151 and this increase is particularly high in the head. 149 Loss‐of‐function mutations in the genes encoding specific PRC1 and PRC2 components suppressed the elevation of H3K27me3 during aging and improved lifespan and healthspan. Among PRC components, Enhancer‐of‐zeste (E[z]) mutants have been particularly well‐characterized. 150 , 151 Observed healthspan effects include improved stress resistance to hyperthermia, oxidative stress and endoplasmic reticulum stress, as well as enhanced fecundity. 150 , 151 The stress resistance was accompanied by the upregulation of many target genes including the stress resistance gene, Ornithine Decarboxylase 1 (Odc1). 150 Further genome‐wide transcriptome profiling of these animals found altered expressions of 239 genes that were mainly involved in metabolism, immune response, cell cycle, and ribosome biogenesis. 150 Further characterization of the H3K27me3 regulation during aging may find potential targets for slowing down the aging in the epigenome.
In human studies, a bulk increase in H3K27me3 was also detected with aging, when assessing hematopoietic stem cells (HSCs) and progenitor cells. 152 Moreover, DNA methylations at the CpG sites of the epigenetic clock model of human aging (described in detail in Section 3.2) are significantly correlated with Polycomb‐group (PcG) target loci. 153 These observations are in line with the findings in Drosophila where H3K27me3 increases with natural aging. Cross‐talks between DNA methylation and H3K27me3 also occur in other contexts. 154 , 155 , 156 As opposed to the Drosophila genome where PRCs are mainly recruited at Polycomb repressive elements (PREs), far fewer PREs have been described in the mammalian genome. 157 One possibility could be that due to the lack of DNA methylation, PREs are increasingly important to guide PRCs in the Drosophila genome.
There are other loci that can be affected by augmented levels of H3K27me3. 158 For example, a study in cell culture demonstrated that replicative senescence and SAFH formation resulted from a reduced level of H3K27me3 at the CDKN2A (INK4/ARF) locus. 159 Metformin, an anti‐diabetic drug, was found to be a specific inhibitor of the H3K27me3 demethylase, Kdm6A, 158 and the cells treated with Metformin had a global increase in H3K27me3. Therefore, metformin may be applicable as an anti‐aging intervention by inhibiting the H3K27me3 demethylation at the locus such as CDKN2A. However, it is critically important to note that inactivating this locus may drive oncogenesis, therefore, a balanced activity or expression is crucial. 160 , 161
3.1.7. Gene activation marks, H3K4me3, and H3K36me3
In the study that found an overall decrease in the repressive H3K9me3 mark in aging Drosophila, the active marks, H3K4me3 and H3K36me3 showed an overall increase. 91 H3K4me3 that located at the promoter regions are associated with transcriptionally active genes. 162 H3K36me3 is enriched in the gene bodies, regulating transcriptional fidelity and alternative splicing events. 163 , 164 Studies demonstrated that both activating marks can antagonize H3K27me3, 165 , 166 , 167 and the interplay between these histone marks might be important during aging. For instance, a mutation in the trithorax (trx) encoding a H3K4 methyltransferase was found to negate the lifespan increasing effect of E(z) mutation. 150 Elucidating the loci related to these marks during aging, including the identification of the regulatory enzymes and other factors can help find the interventions to suppress aging‐related phenotypes. It is easy to conduct genetic studies in this area using the Drosophila model.
H3K4me3 and H3K36me3 appear to be also relevant to human aging brain. A genome‐wide H3K4me3 profiling found that in neurons isolated from infant prefrontal cortex, H3K4me3 peaks in several hundred loci, compared to approximately a hundred in samples from old people (>60 years). 168 The H3K4me3 peak sites in infants consisted mainly of the genes involved in neurogenesis, neuronal growth, and differentiation. The link between H3K36 methylation and human aging was also observed in Sotos syndrome patients who had a mutation in a gene encoding H3K36 methyltransferase. 169 Future studies of H3K4me3 in other tissues and H3K36me3 in natural aging are expected to yield informative results. Concomitantly, assessing transcript levels and alternative splicing isoforms would be useful.
3.1.8. Sirtuin as an anti‐aging regulator
The Sirtuin family of NAD‐dependent histone deacetylases (HDACs) has been in the limelight in aging studies because its stimulation or overexpression has been shown as a promising avenue for anti‐aging. 170 , 171 Although it is well‐established that Sirtuins mediate the longevity effect of caloric restriction (diet condition studies are described in detail in Section 2.3), its role played in Drosophila lifespan has been controversial. Whereas one study reported no effect, 172 some others observed lifespan extension, but only when the expression level of Sirtuin 2 (Sirt2) was two to fivefold upregulated. 71 , 173 , 174 Above this range, Sirt2 was found to be detrimental, as evidenced by the triggering of the JNK negative feedback loop kinase, puckered (puc) and the heat‐shock stress response gene, DnaJ homolog (dnaJ‐H), as well as caspase‐mediated apoptosis. 174 , 175
Consistent with the idea that Sirt2 overexpression can increase lifespan, Sirtuin‐activating compounds, resveratrol and fisetin were found to improve longevity and prolong fecundity when functional Sirt2 was present. 73 A later Drosophila study confirmed the effect of resveratrol, and further demonstrated that a mixture of Sirtuin‐activating compounds, alkylresorcinols, similarly extended lifespan in a Sirt2‐dependent manner. 72 Taken together, these studies support that the lifespan enhancement effect of Sirt2 is conserved across species including Drosophila, and that the level of its expression is functionally important. In Drosophila, Sirt2 overexpression can repress the age‐related TE activation in the head and fat body, 93 suggesting its potential link to H3K9me3‐HP1a and RNAi, suggesting its relevance to the lifespan extension involving Sirtuins in humans.
Different classes of histone deacetylases have been shown to work together while retaining distinct function. Drosophila mutants of the histone deacetylase, HDAC1 are long‐lived. 176 HDAC1 is highly conserved across species and belongs to Class I—different from sirtuins that belong to Class III HDACs. 177 Sirt2 upregulation was found in HDAC1 mutants, 176 and the lifespan effect was similar to caloric restriction or other interventions that enhanced the level of Sirt2. Interestingly, caloric restriction did not extend the lifespan of the HDAC1 mutants. 176 How these two histone deacetylases interact requires further investigation.
Sirturins also regulate nonhistone targets. In both Drosophila and mammalian studies, p53 protein has been identified to be a major target of deacetylation by SIRT2. 178
3.1.9. Nucleosome remodeling regulators in aging
The interaction between the NuRD—HDAC complex and PRC might be important in regulation of nucleosome remodeling during aging across species. Increased lifespan was found in Drosophila with Mi2 knockdown. 179 Mi2 protein is a component of the nucleosome remodeling and deacetylase (NuRD) complex that also includes HDACs and conserved across species. 180 This protein was initially found to participate in the PcG‐mediated repression at Hox clusters. 181 The deacetylation of H3K27ac facilitates the methylation at the H3K27 residue, and this antagonistic mechanism was previously observed in both mammalian cells 182 and in Drosophila. 183 Ascertaining the age‐dependent effects of nucleosome remodeling vs HDAC‐dependent gene expression at NuRD target sites can help elucidate the interactions between these mechanisms. It can also aid in understanding their relevance to the heterochromatin marks such as H3K27me3.
3.2. . DNA methylation alteration during aging
3.2.1. 5‐methylcytosine
A recent advance in aging research, which has received extensive attention, is the development of the “epigenetic clock” for measuring the biological age of tissues or cells. 153 , 184 , 185 , 186 , 187 , 188 , 189 The clock is based on a linear regression model that correlates the changes of 5‐methylcytosine (5mC) marks at specific CpGs with the age. The age‐dependent alteration of DNA methylation in the clock appears to be conserved in mice, chimpanzees and other organisms. Using the clock, liver samples obtained from obesity patients were found to have accelerated epigenetic age. 190 It has been informative to evaluate the effect of different lifestyle factors and other interventions on aging, 191 as well as applying the model longitudinally. 192 , 193
In Drosophila, C elegans, and yeast species, the presence of 5mC in the genome has been long debated. 194 , 195 , 196 , 197 In Drosophila, 5mC was found relatively more abundantly in early embryos, 195 , 198 but with a low level in adult tissues. 195 , 197 , 198 , 199 , 200 , 201 , 202 , 203 In its genome, only one methyltransferase, Methyltransferase 2 (Mt2, previously named as DNA methyltransferase [Dnmt2]) is present with a mammalian orthologue. It lacks the conservation with the human de novo DNA methyltransferases (DNMTs), Dnmt3A/B, and the maintenance DNMT, Dnmt1. Mt2/Dnmt2 was initially named as a DNMT, but later found to act predominantly as a tRNA m5C methyltransferase, 204 , 205 , 206 , 207 although in the context of tRNA‐DNA hybrids, it can also act on DNA. 208 In Drosophila, the overexpression of Mt2/Dnmt2 increases lifespan, however the mechanism remains elusive. 141 The minimal presence of 5mC in adult flies suggest that it likely does not mimic methylation changes in 5mC in human aging, thus not a suitable model for the aging clock.
3.2.2. 6‐methyldeoxyadenosine
It was found recently that N6 ‐6‐methyldeoxyadenosine (6mA) is another DNA methylation mark that is abundantly present in both the human 209 and Drosophila genome. 210 In Drosophila, 6mA was demonstrated to be regulated by DNA 6mA demethylase (DMAD), and its presence was correlated with TE expression in the ovary. 210 A different study showed that 6mA transcriptionally regulated zelda via the recruitment of Jumu, a 6mA reader, to facilitate the maternal‐to‐zygotic transition. 211 It was also demonstrated that 6mA might dynamically regulate the genes involved in neurodevelopment and neuronal functions and TE activity in the brain. 212 However, the role of 6mA in aging is still lacking in any organism. Given the relevance of TE in aging, 105 6mA and its regulators warrant further investigation for its role as a functional substitute for 5mC and a potential aging clock in Drosophila. It is also of great interest to delineate the similarities and differences between the mechanistic roles of 6mA and 5mC.
3.3. Noncoding RNAs
The transcriptional dysregulation due to chromatin alterations has been shown to affect both protein‐coding gene transcripts and noncoding RNAs. Studies of different noncoding RNAs involved in Drosophila aging, and their relevance to human aging are summarized in Table 2. The RNAi and Piwi/piRNA pathways, relevant to small noncoding RNAs, has been discussed in relation to heterochromatin in Section 3.1.
3.3.1. Long noncoding RNAs (lncRNAs)
LncRNAs can act as an enhancer to facilitate transcription, as well as a decoy to prevent or guide the recruitment of transcription factors and chromatin modifying factors, and the interactions between DNA methylation or histone modification mechanisms and lncRNAs are evident. 213 For instance, MALAT1 lncRNA, which was found to be upregulated in various cancer types in humans, 214 can regulate the recruitment of histone‐lysine N‐methyltransferase enzyme, EZH2 to promote H3K27 methylation at specific genomic loci. 215 LncRNAs can also act as a scaffold to promote the formation of protein complexes, which regulate various processes such as transcription, alternative splicing, translation, rRNA maturation, microRNAs binding to their target mRNAs, and signaling molecule phosphorylation. 213 Moreover, they can serve as a precursor to small RNAs. 213 A plethora of other studies have linked noncoding RNAs to human aging 216 , 217 and aging‐related diseases 213 including cancer, 218 , 219 , 220 cardiovascular, 221 and neuronal aging 222 and metabolism and cellular senescence. 223
LncRNAs have not been investigated extensively in the context of aging in Drosophila. A study profiled lncRNAs in the head, gut and fat body of flies reared in the lifespan‐extension dietary restrictive condition and identified 102 differentially expressed lncRNAs. 224 Bioinformatic analyses indicated that these lncRNAs mainly regulated the metabolism pathways such as mTOR, foxo, and Wnt signaling pathways, which have been implicated in aging across species including humans. Such profiling studies can be highly informative guiding the investigation on the targets of identified lncRNAs. Given that a high level of conservation of major lncRNAs have been noted, 225 , 226 studies in Drosophila are expected to lead to mechanistic insights.
3.3.2. Small noncoding RNAs
In a Drosophila Hungtington's disease model that overexpressed the human pathogenic polyglutamine disease protein, a knockout in miR‐34 resulted in reduced lifespan with accelerated brain aging, whereas its upregulation extended lifespan and alleviated neurodegeneration. 41 The Ecdysone‐induced protein 74EF gene, Eip74EF was found to be a target of miR‐34 and upregulated in the miR‐34 deletion strain. 41 Another study confirmed the disruption of the Ecdysone signaling in miR‐34 knockouts and reported a defect in innate immune response. 227 These observations suggest a new role of the Ecdysone pathway in aging. Another study showed that miR‐34 targeted PRC2 components, Polycomblike (Pcl) and Su(z)12, and that in miR‐34 deletion animals, the H3K27me3 accumulated in the brain and the gene expression profile was associated with advanced aging. 228 Also, in this study, miR‐34 upregulation was shown to lead to the alleviation of neurodegeneration induced by pathogenic polyglutamine protein overexpression. 228
The miR‐34 family is conserved across species including humans. The human miR‐34a (member of the miR‐34 family) was found to be upregulated with aging in peripheral blood mononuclear cells (PBMCs) and target SIRT genes. 229 Given the importance of maintaining the level of Sirtuins (described in part 3b.iv. above), targeting miR‐34a may be a feasible anti‐aging intervention. Other studies have also shown the relevance of miR‐34a to cancer and diabetes. 230 , 231 , 232 These observations underscore the importance of miR‐34a in aging across species.
Other miRNAs relevant to aging include miR‐125 and miR‐9a. The loss of miR‐125 in Drosophila was found to reduce lifespan, impact climbing activity, and increase neurodegeneration in the brain. 31 miR‐9a was found to regulate the maintenance of male germline stem cell. 233 The human homologs of these miRNAs have been implicated in aging and cancer in humans. 229 , 234 , 235 Identifying other miRNAs involved in aging may yield new putative targets to counter aging phenotypes.
Age‐related chromatin alterations can affect the expression of various miRNAs, as shown by some examples above. MiRNAs have also been shown to also downregulate DNMTs to promote aging‐related cancer progression. 236 , 237 , 238 , 239
3.3.3. Circular RNAs
Circular RNAs (circRNAs), a new species of small RNAs have emerged as being important in regulating aging In Drosophila, the age‐related accumulation of circRNAs was evident in the brain and photoreceptors. 240 , 241 Further mechanistic studies in Drosophila are expected to be informative, especially since circRNAS have been increasingly appreciated in human aging. 242
3.4. Emerging roles of RNA methylation and RNA editing
RNA methylation and other types of chemical modifications such as hydroxylmethylation and pseudouridylation exist abundantly. 243 , 244 Although the first RNA methylation mark, N 6 ‐Methyladenosine (m6A) was found in 1970's, the regulators (writers and erasers) and the effectors (readers) have begun to be understood recently. 243 , 245 The abbreviation, m6A, is used for RNA modification, distinguishing from 6mA for DNA modification. 246 RNA methylation (m6A and other types) has been shown to regulate the stability of various RNA species, mRNA alternative splicing, translation, nuclear‐cytoplasmic shuttling, and interactions between different RNA species. The writers, erasers, and readers of m6A have been shown to be conserved across species including Drosophila and humans. 247
Although m6A has not been directly implicated in aging in Drosophila, phenotypes that are loosely relevant have been found. For example, the loss‐of‐function mutations in m6A regulators result in the aging‐like defects in neuronal activity and oogenesis. 248 , 249 The high expression of the m6A methyltransferase complex during early embryogenesis suggest that these aging phenotypes likely arise during early development rather thanthe aging process. Thus, evaluating m6A profile changes, or a temporal knockdown (presented in Section 2.1) of m6A regulators later in life may help discover the links between m6A and aging. In a human study profiling m6A‐containing transcripts in peripheral blood mononuclear cell (PBMCs), it was found that the overall enrichment level of m6A decreased in old people, compared to young people. 250 Specifically, for AGO2 mRNA, both its abundance and the m6A enrichment were reduced. m6A was also found to regulate other biological processes relevant to aging in humans, including self‐renewal in stem cells, 251 the circadian rhythm, 252 promoting cancer stem cells, 253 and enhancing tumorigenicity. 254
Other significant RNA modifications including 5‐methylcytosine (m5C), N 6 ,2′‐O‐dimethyladenosine (m6Am), 2′‐O‐methylation (2′Ome), and pseudouridylation might be also involved in aging. 255 , 256 , 257 As mentioned above in Section 3.2, in Drosophila, the overexpression of Mt2/Dnmt2, which acts mainly as a tRNA m5C methyltransferase, 204 , 205 , 206 increases lifespan. 141 Conversely, knocking down TRDMT1/Dnmt2 in human fibroblasts increased oxidative stress, DNA damage, miRNAs targeting transcripts related to proliferation, resulting in senescence. 258 In Drosophila, 2′Ome becomes enriched in specific miRNAs with age. 142 The study also reported that more miRNAs become associated with Ago2, but not with Ago1 during aging. 142 Indeed, Ago2 mutation led to decreased lifespan and neurodegeneration accompanied with increased brain vacuoles. 142 Thus, further studies of RNA modification mechanisms and their relation to the Ago2 loading in the context of aging may be important.
In addition to RNA base modifications, RNA editing has been recently described to play a role in aging. An interplay between RNA m6A modification and RNA adenosine‐to‐inosine (A‐to‐I) base editing was found; the presence of the two alterations appeared to be negatively correlated. 243 In Drosophila, a hypomorphism of the adenosine deaminase acting on RNA (ADAR) gene, which encodes an A‐to‐I editor, was found to cause the extended lifespan. This was accompanied in neurons with increased levels of histone modifications that are associated with heterochromatic silencing. 259 Human cohort studies found single nucleotide polymorphisms (SNPs) in the orthologues, ADARB1 and ADARB2 to be associated with longevity, 260 suggesting the clinical relevance of the findings in Drosophila. In addition, various recent studies have also linked the misregulation of RNA editing to the development of cancers and metabolic disorders. 261
4. CONSIDERATIONS FOR FUTURE STUDIES OF AGING
4.1. Tissue specificity, single‐cell methods, ‐omics, and integrative approaches
Aging‐related epigenetic alterations and gene expression changes are distinctive in different tissues and cell types, demonstrated by epigenetic aging clocks. 153 , 262 , 263 Additional studies to identify other biomarkers through transcriptomic, proteomic and metabolomic profiling with multi‐tissue approach may lead to the identification of molecular changes across different tissues and cells and further improve the epigenetic clocks. Recent development for single‐cell DNA methylation analysis 264 is expected to yield deep insights in epigenetic drift mechanisms and human aging‐related heterogeneity with high resolution. 191 The trending approach, not just in the aging field, but widely, involves emerging technologies that allow molecular signature assessment of single cells, in addition to bulk tissues. The obvious limitation for the human studies is that not all tissues and cells can be harvested for investigation.
Moreover, an integrative ‐omics obtaining information from a wide variety of types of molecular signatures (genomics, epigenomics, transcriptomics, proteomics, metabolomics, metagenomics, and other ‐omics) is an emerging approach that provides comprehensive and unbiased information. 265 , 266 , 267 It would invariably increase the accuracy of predicting the aging development and improve our understanding of the underlying molecular mechanisms. It is particularly powerful, if combined with rich genetics in Drosophila. It is clear from animal models that it is important to consider genetic backgrounds for aging research, which presents a challenge in human studies in which heterogeneity is a given.
Applying machine learning algorithms to select panels of predictors and construct regression models for estimating biological age, or for risk profiling of a specific aging‐related outcome, could potentially be conducted using samples from geriatric patients as well as model organisms including Drosophila. Moreover, such algorithms would provide additional clinical research tools for standardizing populations or measuring the success of therapeutic interventions. Another type of analyses is to identify differentially expressed biomarkers and monitoring them longitudinally. Such information could be used for elucidating molecular mechanisms underlying the aging process. Network analyses of the relevant factors and identifying central nodes would aid the identification of effective targets and development of pharmacological interventions, and this is particularly useful for Drosophila and other model organisms that have rich network information available. The contribution of the microbiome and microvirome to aging remains to be largely elusive and the efforts including metagenomics is highly in demand. Developing computational and visualization pipelines for integrative‐omics approaches are challenging because it requires the consideration of various molecular types with different scales and formats to high dimension and is an on‐going effort. 268 , 269 , 270 , 271 , 272
4.2. Other uses of the Drosophila system in aging research
As indicated in Table 1, Drosophila, with the plethora of healthspan phenotypes that are relevant to human aging, has been shown to contribute significantly to the aging studies in vivo. It would also be advantageous for meticulous quantification of “proportional” vs “chronological” healthspan extension, which is not easy in human studies.
Drosophila have been used extensively as a model to elucidate fundamental mechanisms of all aspects of biology and there are still endless discoveries that could be made in this highly characterized genetic system. As we discussed, the limited presence of 5mC in the adult Drosophila, points to the skepticism over its being a major epigenetic mark for measuring aging and the importance of developing aging models based on other epigenetic marks. The abundance of 6mA in the Drosophila genome and the lack of studies on its possible role in aging in any organism represents an exciting opportunity in aging research. Limited studies on lncRNAs in aging in Drosophila, and the emerging importance of circRNAs may suggest the need for identifying new noncoding RNAs and mechanisms in aging. The possible involvement of various RNA modifications, such as m6A and m5C, as well as RNA editing in aging remains elusive and warrant further investigation.
CONFLICT OF INTEREST
The authors declare that they have no conflict of interest.
AUTHOR CONTRIBUTIONS
Amy Tsurumi: Supervision; writing‐original draft; writing‐review and editing. Willis Li: Supervision; writing‐original draft; writing‐review and editing.
PEER REVIEW
The peer review history for this article is available at https://publons.com/publon/10.1002/ggn2.10026.
Supporting information
Transparent‐Peer‐Review‐Record
ACKNOWLEDGMENT
Willis X. Li is supported by a grant from the National Institute of General Medical Sciences of the National Institutes of Health (R01GM131044). Willis X. Li was supported by the Shriners Hospitals Research Fellowship #84293. We would like to acknowledge Ankita Banerjee for her input and aid editing.
Tsurumi A, Li WX. Aging mechanisms—A perspective mostly from Drosophila . Advanced Genetics. 2020;1:e10026. 10.1002/ggn2.10026
Funding information National Institute of General Medical Sciences, Grant/Award Number: R01GM131044; Shriners Hospitals Research Fellowship, Grant/Award Number: #84293
REFERENCES
- 1. Crimmins EM. Lifespan and Healthspan: past, present, and promise. Gerontologist. 2015;55(6):901‐911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Jaul E, Barron J. Age‐related diseases and clinical and public health implications for the 85 years old and over population. Front Public Health. 2017;5:335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. MacNee W, Rabinovich RA, Choudhury G. Ageing and the border between health and disease. Eur Respir J. 2014;44(5):1332‐1352. [DOI] [PubMed] [Google Scholar]
- 4. Franceschi C, Garagnani P, Morsiani C, et al. The continuum of aging and age‐related diseases: Common mechanisms but different rates. Front Med (Lausanne). 2018;5:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Fuellen G, Jansen L, Cohen AA, et al. Health and aging: unifying concepts, scores. Biomarkers Pathways Aging Dis. 2019;10(4):883‐900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Aunan JR, Cho WC, Soreide K. The biology of aging and cancer: a brief overview of shared and divergent molecular hallmarks. Aging Dis. 2017;8(5):628‐642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Alzheimer's Association . 2016 Alzheimer's disease facts and figures. Alzheimers Dement. 2016;12(4):459‐509. [DOI] [PubMed] [Google Scholar]
- 8. Rothman KJ. Causes. Am J Epidemiol. 1976;104(6):587‐592. [DOI] [PubMed] [Google Scholar]
- 9. Liao SF, Lee WC. Weighing the causal pies in case‐control studies. Ann Epidemiol. 2010;20(7):568‐573. [DOI] [PubMed] [Google Scholar]
- 10. Lee WC. Completion potentials of sufficient component causes. Epidemiology. 2012;23(3):446‐453. [DOI] [PubMed] [Google Scholar]
- 11. Hoffmann K, Heidemann C, Weikert C, Schulze MB, Boeing H. Estimating the proportion of disease due to classes of sufficient causes. Am J Epidemiol. 2006;163(1):76‐83. [DOI] [PubMed] [Google Scholar]
- 12. Hansen M, Kennedy BK. Does longer lifespan mean longer Healthspan? Trends Cell Biol. 2016;26(8):565‐568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Duffy JF, Zitting KM, Chinoy ED. Aging and circadian rhythms. Sleep Med Clin. 2015;10(4):423‐434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Martinez‐Nicolas A, Madrid JA, Garcia FJ, et al. Circadian monitoring as an aging predictor. Sci Rep. 2018;8(1):15027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Sanders JL, Minster RL, Barmada MM, et al. Heritability of and mortality prediction with a longevity phenotype: the healthy aging index. J Gerontol A Biol Sci Med Sci. 2014;69(4):479‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Tyrovolas S, Haro JM, Mariolis A, et al. Successful aging, dietary habits and health status of elderly individuals: a k‐dimensional approach within the multi‐national MEDIS study. Exp Gerontol. 2014;60:57‐63. [DOI] [PubMed] [Google Scholar]
- 17. Jaspers L, Schoufour JD, Erler NS, et al. Development of a healthy aging score in the population‐based Rotterdam study: evaluating age and sex differences. J Am Med Dir Assoc. 2017;18(3):276‐276. [DOI] [PubMed] [Google Scholar]
- 18. Bansal A, Zhu LJ, Yen K, Tissenbaum HA. Uncoupling lifespan and healthspan in Caenorhabditis elegans longevity mutants. Proc Natl Acad Sci U S A. 2015;112(3):E277‐E286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Loeb J, Northrop JH. Is there a temperature coefficient for the duration of life? Proc Natl Acad Sci U S A. 1916;2(8):456‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Linford NJ, Bilgir C, Ro J, Pletcher SD. Measurement of lifespan in Drosophila melanogaster . J Vis Exp. 2013;71:50068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Brand AH, Perrimon N. Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development. 1993;118(2):401‐415. [DOI] [PubMed] [Google Scholar]
- 22. Osterwalder T, Yoon KS, White BH, Keshishian H. A conditional tissue‐specific transgene expression system using inducible GAL4. Proc Natl Acad Sci U S A. 2001;98(22):12596‐12601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. McGuire SE, Mao Z, Davis RL. Spatiotemporal gene expression targeting with the TARGET and gene‐switch systems in Drosophila. Sci STKE. 2004;2004(220):pl6. [DOI] [PubMed] [Google Scholar]
- 24. Perkins LA, Holderbaum L, Tao R, et al. The transgenic RNAi project at Harvard Medical School: resources and validation. Genetics. 2015;201(3):843‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bier E, Harrison MM, O'Connor‐Giles KM, Wildonger J. Advances in engineering the fly genome with the CRISPR‐Cas system. Genetics. 2018;208(1):1‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Garschall K, Flatt T. The interplay between immunity and aging in Drosophila. F1000Res. 2018;7:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Orr WC, Radyuk SN, Sohal RS. Involvement of redox state in the aging of Drosophila melanogaster . Antioxid Redox Signal. 2013;19(8):788‐803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Biteau B, Karpac J, Hwangbo D, Jasper H. Regulation of Drosophila lifespan by JNK signaling. Exp Gerontol. 2011;46(5):349‐354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Papadopoli D, Boulay K, Kazak L, et al. mTOR as a central regulator of lifespan and aging. F1000Res. 2019;8:F1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Grotewiel MS, Martin I, Bhandari P, Cook‐Wiens E. Functional senescence in Drosophila melanogaster . Ageing Res Rev. 2005;4(3):372‐397. [DOI] [PubMed] [Google Scholar]
- 31. Chawla G, Deosthale P, Childress S, Wu YC, Sokol NS. A let‐7‐to‐miR‐125 MicroRNA switch regulates neuronal integrity and lifespan in Drosophila. PLoS Genet. 2016;12(8):e1006247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Larson K, Yan SJ, Tsurumi A, et al. Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Genet. 2012;8(1):e1002473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sun Y, Yolitz J, Wang C, Spangler E, Zhan M, Zou S. Aging studies in Drosophila melanogaster . Methods Mol Biol. 2013;1048:77‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Petrosyan A, Hsieh IH, Phillips JP, Saberi K. Enhanced tethered‐flight duration and locomotor activity by overexpression of the human gene SOD1 in Drosophila motorneurons. Genet Mol Biol. 2015;38(1):107‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Liu L, Davis RL, Roman G. Exploratory activity in Drosophila requires the kurtz nonvisual arrestin. Genetics. 2007;175(3):1197‐1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Soibam B, Shah S, Gunaratne GH, Roman GW. Modeling novelty habituation during exploratory activity in Drosophila. Behav Processes. 2013;97:63‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Barone MC, Bohmann D. Assessing neurodegenerative phenotypes in Drosophila dopaminergic neurons by climbing assays and whole brain immunostaining. J Vis Exp. 2013;74:e50339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kasture AS, Hummel T, Sucic S, Freissmuth M. Big lessons from tiny flies: Drosophila melanogaster as a model to explore dysfunction of dopaminergic and serotonergic neurotransmitter systems. Int J Mol Sci. 2018;19(6):1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Niens J, Reh F, Coban B, et al. Dopamine modulates serotonin innervation in the Drosophila brain. Front Syst Neurosci. 2017;11:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. White KE, Humphrey DM, Hirth F. The dopaminergic system in the aging brain of Drosophila. Front Neurosci. 2010;4:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Liu N, Landreh M, Cao K, et al. The microRNA miR‐34 modulates ageing and neurodegeneration in Drosophila. Nature. 2012;482(7386):519‐523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. McGurk L, Berson A, Bonini NM. Drosophila as an in vivo model for human neurodegenerative disease. Genetics. 2015;201(2):377‐402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Kim NC, Tresse E, Kolaitis RM, et al. VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations. Neuron. 2013;78(1):65‐80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Vang LL, Medvedev AV, Adler J. Simple ways to measure behavioral responses of Drosophila to stimuli and use of these methods to characterize a novel mutant. PLoS One. 2012;7(5):e37495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Pacifico R, MacMullen CM, Walkinshaw E, Zhang X, Davis RL. Brain transcriptome changes in the aging Drosophila melanogaster accompany olfactory memory performance deficits. PLoS One. 2018;13(12):e0209405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Pletcher SD. The modulation of lifespan by perceptual systems. Ann N Y Acad Sci. 2009;1170:693‐697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Masek P, Keene AC. Gustatory processing and taste memory in Drosophila. J Neurogenet. 2016;30(2):112‐121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Rogina B, Wolverton T, Bross TG, Chen K, Muller HG, Carey JR. Distinct biological epochs in the reproductive life of female Drosophila melanogaster . Mech Ageing Dev. 2007;128(9):477‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Somashekar K, Krishna MS, Hegde SN, Jayaramu SC. Effects of age on female reproductive success in Drosophila bipectinata. J Insect Sci. 2011;11:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Mair W, McLeod CJ, Wang L, Jones DL. Dietary restriction enhances germline stem cell maintenance. Aging Cell. 2010;9(5):916‐918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Xing Y, Li WX. Heterochromatin components in germline stem cell maintenance. Sci Rep. 2015;5:17463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Wessells RJ, Fitzgerald E, Cypser JR, Tatar M, Bodmer R. Insulin regulation of heart function in aging fruit flies. Nat Genet. 2004;36(12):1275‐1281. [DOI] [PubMed] [Google Scholar]
- 53. Piazza N, Wessells RJ. Drosophila models of cardiac disease. Prog Mol Biol Transl Sci. 2011;100:155‐210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Zhou D, Visk DW, Haddad GG. Drosophila, a golden bug, for the dissection of the genetic basis of tolerance and susceptibility to hypoxia. Pediatr Res. 2009;66(3):239‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Lee KS, Iijima‐Ando K, Iijima K, et al. JNK/FOXO‐mediated neuronal expression of fly homologue of peroxiredoxin II reduces oxidative stress and extends life span. J Biol Chem. 2009;284(43):29454‐29461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Ruan H, Wu CF. Social interaction‐mediated lifespan extension of Drosophila cu/Zn superoxide dismutase mutants. Proc Natl Acad Sci U S A. 2008;105(21):7506‐7510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Mair W, Piper MD, Partridge L. Calories do not explain extension of life span by dietary restriction in Drosophila. PLoS Biol. 2005;3(7):e223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Bass TM, Grandison RC, Wong R, Martinez P, Partridge L, Piper MD. Optimization of dietary restriction protocols in Drosophila. J Gerontol A Biol Sci Med Sci. 2007;62(10):1071‐1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Dobson AJ, He X, Blanc E, et al. Tissue‐specific transcriptome profiling of Drosophila reveals roles for GATA transcription factors in longevity by dietary restriction. NPJ Aging Mech Dis. 2018;4:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Emran S, Yang M, He X, Zandveld J, Piper MD. Target of rapamycin signalling mediates the lifespan‐extending effects of dietary restriction by essential amino acid alteration. Aging (Albany NY). 2014;6(5):390‐398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Grandison RC, Piper MD, Partridge L. Amino‐acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature. 2009;462(7276):1061‐1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Mair W, Goymer P, Pletcher SD, Partridge L. Demography of dietary restriction and death in Drosophila. Science. 2003;301(5640):1731‐1733. [DOI] [PubMed] [Google Scholar]
- 63. Rochon J, Bales CW, Ravussin E, et al. Design and conduct of the CALERIE study: comprehensive assessment of the long‐term effects of reducing intake of energy. J Gerontol A Biol Sci Med Sci. 2011;66(1):97‐108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Das SK, Roberts SB, Bhapkar MV, et al. Body‐composition changes in the comprehensive assessment of long‐term effects of reducing intake of energy (CALERIE)‐2 study: a 2‐y randomized controlled trial of calorie restriction in nonobese humans. Am J Clin Nutr. 2017;105(4):913‐927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Racette SB, Das SK, Bhapkar M, et al. Approaches for quantifying energy intake and %calorie restriction during calorie restriction interventions in humans: the multicenter CALERIE study. Am J Physiol Endocrinol Metab. 2012;302(4):E441‐E448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Redman LM, Smith SR, Burton JH, Martin CK, Il'yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab. 2018;27(4):805‐815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Belsky DW, Huffman KM, Pieper CF, Shalev I, Kraus WE. Change in the rate of biological aging in response to caloric restriction: CALERIE biobank analysis. J Gerontol A Biol Sci Med Sci. 2017;73(1):4‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Stewart TM, Bhapkar M, Das S, et al. Comprehensive assessment of long‐term effects of reducing intake of energy phase 2 (CALERIE phase 2) screening and recruitment: methods and results. Contemp Clin Trials. 2013;34(1):10‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Ravussin E, Redman LM, Rochon J, et al. A 2‐year randomized controlled trial of human caloric restriction: feasibility and effects on predictors of health span and longevity. J Gerontol A Biol Sci Med Sci. 2015;70(9):1097‐1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Il'yasova D, Fontana L, Bhapkar M, et al. Effects of 2years of caloric restriction on oxidative status assessed by urinary F2‐isoprostanes: the CALERIE 2 randomized clinical trial. Aging Cell. 2018;17(2): e12719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Rogina B, Helfand SL. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc Natl Acad Sci U S A. 2004;101(45):15998‐16003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Kayashima Y, Katayanagi Y, Tanaka K, Fukutomi R, Hiramoto S, Imai S. Alkylresorcinols activate SIRT1 and delay ageing in Drosophila melanogaster . Sci Rep. 2017;7:43679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Wood JG, Rogina B, Lavu S, et al. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature. 2004;430(7000):686‐689. [DOI] [PubMed] [Google Scholar]
- 74. Weichhart T. mTOR as regulator of lifespan, aging, and cellular senescence: a mini‐review. Gerontology. 2018;64(2):127‐134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Yamamoto R, Tatar M. Insulin receptor substrate Chico acts with the transcription factor FOXO to extend Drosophila lifespan. Aging Cell. 2011;10(4):729‐732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Slack C, Giannakou ME, Foley A, Goss M, Partridge L. dFOXO‐independent effects of reduced insulin‐like signaling in Drosophila. Aging Cell. 2011;10(5):735‐748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Martins R, Lithgow GJ, Link W. Long live FOXO: unraveling the role of FOXO proteins in aging and longevity. Aging Cell. 2016;15(2):196‐207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Birnbaum A, Wu X, Tatar M, Liu N, Bai H. Age‐dependent changes in transcription factor FOXO targeting in female Drosophila. Front Genet. 2019;10:312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Dobson AJ, Ezcurra M, Flanagan CE, et al. Nutritional programming of lifespan by FOXO inhibition on sugar‐rich diets. Cell Rep. 2017;18(2):299‐306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Min KJ, Yamamoto R, Buch S, Pankratz M, Tatar M. Drosophila lifespan control by dietary restriction independent of insulin‐like signaling. Aging Cell. 2008;7(2):199‐206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Clancy DJ, Gems D, Hafen E, Leevers SJ, Partridge L. Dietary restriction in long‐lived dwarf flies. Science. 2002;296(5566):319. [DOI] [PubMed] [Google Scholar]
- 82. Marden JH, Rogina B, Montooth KL, Helfand SL. Conditional tradeoffs between aging and organismal performance of Indy long‐lived mutant flies. Proc Natl Acad Sci U S A. 2003;100(6):3369‐3373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Lopez‐Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194‐1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kennedy BK, Berger SL, Brunet A, et al. Geroscience: linking aging to chronic disease. Cell. 2014;159(4):709‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Brooks‐Wilson AR. Genetics of healthy aging and longevity. Hum Genet. 2013;132(12):1323‐1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074‐1080. [DOI] [PubMed] [Google Scholar]
- 87. Richards EJ, Elgin SC. Epigenetic codes for heterochromatin formation and silencing: rounding up the usual suspects. Cell. 2002;108(4):489‐500. [DOI] [PubMed] [Google Scholar]
- 88. Wang J, Jia ST, Jia S. New insights into the regulation of heterochromatin. Trends Genet. 2016;32(5):284‐294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Benayoun BA, Pollina EA, Brunet A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat Rev Mol Cell Biol. 2015;16(10):593‐610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Munoz‐Najar U, Sedivy JM. Epigenetic control of aging. Antioxid Redox Signal. 2011;14(2):241‐259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Wood JG, Hillenmeyer S, Lawrence C, et al. Chromatin remodeling in the aging genome of Drosophila. Aging Cell. 2010;9(6):971‐978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Shi S, Calhoun HC, Xia F, Li J, Le L, Li WX. JAK signaling globally counteracts heterochromatic gene silencing. Nat Genet. 2006;38(9):1071‐1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Wood JG, Jones BC, Jiang N, et al. Chromatin‐modifying genetic interventions suppress age‐associated transposable element activation and extend life span in Drosophila. Proc Natl Acad Sci U S A. 2016;113(40):11277‐11282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Peng JC, Karpen GH. H3K9 methylation and RNA interference regulate nucleolar organization and repeated DNA stability. Nat Cell Biol. 2007;9(1):25‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Peng JC, Karpen GH. Heterochromatic genome stability requires regulators of histone H3 K9 methylation. PLoS Genet. 2009;5(3):e1000435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Shi S, Larson K, Guo D, et al. Drosophila STAT is required for directly maintaining HP1 localization and heterochromatin stability. Nat Cell Biol. 2008;10(4):489‐496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Li WX. Canonical and non‐canonical JAK‐STAT signaling. Trends Cell Biol. 2008;18(11):545‐551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Yan SJ, Lim SJ, Shi S, Dutta P, Li WX. Unphosphorylated STAT and heterochromatin protect genome stability. FASEB J. 2011;25(1):232‐241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Dutta P, Zhang L, Zhang H, et al. Unphosphorylated STAT3 in heterochromatin formation and tumor suppression in lung cancer. BMC Cancer. 2020;20(1):145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Hu X, Dutta P, Tsurumi A, et al. Unphosphorylated STAT5A stabilizes heterochromatin and suppresses tumor growth. Proc Natl Acad Sci U S A. 2013;110(25):10213‐10218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Villeponteau B. The heterochromatin loss model of aging. Exp Gerontol. 1997;32(4–5):383‐394. [DOI] [PubMed] [Google Scholar]
- 102. Sidler C, Kovalchuk O, Kovalchuk I. Epigenetic regulation of cellular senescence and aging. Front Genet. 2017;8:138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Tsurumi A, Li WX. Global heterochromatin loss: a unifying theory of aging? Epigenetics. 2012;7(7):680‐688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Pal S, Tyler JK. Epigenetics and aging. Sci Adv. 2016;2(7):e1600584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Orr WC. Tightening the connection between transposable element mobilization and aging. Proc Natl Acad Sci U S A. 2016;113(40):11069‐11070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Guo C, Jeong HH, Hsieh YC, et al. Tau activates transposable elements in Alzheimer's disease. Cell Rep. 2018;23(10):2874‐2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Loyola AC, Zhang L, Shang R, Dutta P, Li J, Li WX. Identification of methotrexate as a heterochromatin‐promoting drug. Sci Rep. 2019;9(1):11673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16Ink4a‐positive senescent cells delays ageing‐associated disorders. Nature. 2011;479(7372):232‐236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Janzen V, Forkert R, Fleming HE, et al. Stem‐cell ageing modified by the cyclin‐dependent kinase inhibitor p16INK4a. Nature. 2006;443(7110):421‐426. [DOI] [PubMed] [Google Scholar]
- 110. Krishnamurthy J, Ramsey MR, Ligon KL, et al. p16INK4a induces an age‐dependent decline in islet regenerative potential. Nature. 2006;443(7110):453‐457. [DOI] [PubMed] [Google Scholar]
- 111. Liu Y, Johnson SM, Fedoriw Y, et al. Expression of p16(INK4a) prevents cancer and promotes aging in lymphocytes. Blood. 2011;117(12):3257‐3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Molofsky AV, Slutsky SG, Joseph NM, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443(7110):448‐452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Sato S, Kawamata Y, Takahashi A, et al. Ablation of the p16(INK4a) tumour suppressor reverses ageing phenotypes of klotho mice. Nat Commun. 2015;6:7035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Narita M, Nunez S, Heard E, et al. Rb‐mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113(6):703‐716. [DOI] [PubMed] [Google Scholar]
- 115. Ye X, Zerlanko B, Zhang R, et al. Definition of pRB‐ and p53‐dependent and ‐independent steps in HIRA/ASF1a‐mediated formation of senescence‐associated heterochromatin foci. Mol Cell Biol. 2007;27(7):2452‐2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. O'Sullivan RJ, Karlseder J. The great unravelling: chromatin as a modulator of the aging process. Trends Biochem Sci. 2012;37(11):466‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Kennedy AL, Morton JP, Manoharan I, et al. Activation of the PIK3CA/AKT pathway suppresses senescence induced by an activated RAS oncogene to promote tumorigenesis. Mol Cell. 2011;42(1):36‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Aird KM, Zhang R. Detection of senescence‐associated heterochromatin foci (SAHF). Methods Mol Biol. 2013;965:185‐196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Sanada F, Taniyama Y, Muratsu J, et al. Source of chronic inflammation in aging. Front Cardiovasc Med. 2018;5:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Crowe EP, Nacarelli T, Bitto A, Lerner C, Sell C, Torres C. Detecting senescence: methods and approaches. Methods Mol Biol. 2014;1170:425‐445. [DOI] [PubMed] [Google Scholar]
- 121. Kosar M, Bartkova J, Hubackova S, Hodny Z, Lukas J, Bartek J. Senescence‐associated heterochromatin foci are dispensable for cellular senescence, occur in a cell type‐ and insult‐dependent manner and follow expression of p16(ink4a). Cell Cycle. 2011;10(3):457‐468. [DOI] [PubMed] [Google Scholar]
- 122. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010;24(22):2463‐2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Nakamura M, Ohsawa S, Igaki T. Mitochondrial defects trigger proliferation of neighbouring cells via a senescence‐associated secretory phenotype in Drosophila. Nat Commun. 2014;5:5264. [DOI] [PubMed] [Google Scholar]
- 124. Lochs SJA, Kefalopoulou S, Kind J. Lamina associated domains and gene regulation in development and cancer. Cell. 2019;8(3):E271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Bank EM , Gruenbaum Y. The nuclear lamina and heterochromatin: a complex relationship. Biochem Soc Trans. 2011;39(6):1705‐1709. [DOI] [PubMed] [Google Scholar]
- 126. Shevelyov YY, Ulianov SV. The nuclear lamina as an organizer of chromosome architecture. Cell. 2019;8(2):136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Parry AJ, Narita M. Old cells, new tricks: chromatin structure in senescence. Mamm Genome. 2016;27(7–8):320‐331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Sun L, Yu R, Dang W. Chromatin architectural changes during cellular senescence and aging. Genes (Basel). 2018;9(4):211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Eriksson M, Brown WT, Gordon LB, et al. Recurrent de novo point mutations in lamin A cause Hutchinson‐Gilford progeria syndrome. Nature. 2003;423(6937):293‐298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Taimen P, Pfleghaar K, Shimi T, et al. A progeria mutation reveals functions for lamin A in nuclear assembly, architecture, and chromosome organization. Proc Natl Acad Sci U S A. 2009;106(49):20788‐20793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Dreesen O, Chojnowski A, Ong PF, et al. Lamin B1 fluctuations have differential effects on cellular proliferation and senescence. J Cell Biol. 2013;200(5):605‐617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Chen H, Zheng X, Zheng Y. Lamin‐B in systemic inflammation, tissue homeostasis, and aging. Nucleus. 2015;6(3):183‐186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Chen H, Zheng X, Xiao D, Zheng Y. Age‐associated de‐repression of retrotransposons in the Drosophila fat body, its potential cause and consequence. Aging Cell. 2016;15(3):542‐552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Chen H, Zheng X, Zheng Y. Age‐associated loss of Lamin‐B leads to systemic inflammation and gut hyperplasia. Cell. 2014;159(4):829‐843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Slotkin RK, Martienssen R. Transposable elements and the epigenetic regulation of the genome. Nat Rev Genet. 2007;8(4):272‐285. [DOI] [PubMed] [Google Scholar]
- 136. Volpe T, Martienssen RA. RNA interference and heterochromatin assembly. Cold Spring Harb Perspect Biol. 2011;3(9):a003731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Martienssen R, Moazed D. RNAi and heterochromatin assembly. Cold Spring Harb Perspect Biol. 2015;7(8):a019323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Iglesias N, Moazed D. Silencing repetitive DNA. Elife. 2017;6:e29503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Djupedal I, Ekwall K. Epigenetics: heterochromatin meets RNAi. Cell Res. 2009;19(3):282‐295. [DOI] [PubMed] [Google Scholar]
- 140. Li W, Prazak L, Chatterjee N, et al. Activation of transposable elements during aging and neuronal decline in Drosophila. Nat Neurosci. 2013;16(5):529‐531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Lin MJ, Tang LY, Reddy MN, Shen CK. DNA methyltransferase gene dDnmt2 and longevity of Drosophila. J Biol Chem. 2005;280(2):861‐864. [DOI] [PubMed] [Google Scholar]
- 142. Abe M, Naqvi A, Hendriks GJ, et al. Impact of age‐associated increase in 2′‐O‐methylation of miRNAs on aging and neurodegeneration in Drosophila. Genes Dev. 2014;28(1):44‐57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Tang KF, Ren H, Cao J, et al. Decreased Dicer expression elicits DNA damage and up‐regulation of MICA and MICB. J Cell Biol. 2008;182(2):233‐239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Saito K. The epigenetic regulation of transposable elements by PIWI‐interacting RNAs in Drosophila. Genes Genet Syst. 2013;88(1):9‐17. [DOI] [PubMed] [Google Scholar]
- 145. Brower‐Toland B, Findley SD, Jiang L, et al. Drosophila PIWI associates with chromatin and interacts directly with HP1a. Genes Dev. 2007;21(18):2300‐2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Sousa‐Victor P, Ayyaz A, Hayashi R, et al. Piwi is required to limit exhaustion of aging somatic stem cells. Cell Rep. 2017;20(11):2527‐2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Yu Y, Xiao J, Hann SS. The emerging roles of PIWI‐interacting RNA in human cancers. Cancer Manag Res. 2019;11:5895‐5909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Wiles ET, Selker EU. H3K27 methylation: a promiscuous repressive chromatin mark. Curr Opin Genet Dev. 2017;43:31‐37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Ma Z, Wang H, Cai Y, et al. Epigenetic drift of H3K27me3 in aging links glycolysis to healthy longevity in Drosophila. Elife. 2018;7:e35368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Siebold AP, Banerjee R, Tie F, Kiss DL, Moskowitz J, Harte PJ. Polycomb repressive complex 2 and Trithorax modulate Drosophila longevity and stress resistance. Proc Natl Acad Sci U S A. 2010;107(1):169‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Moskalev AA, Shaposhnikov MV, Zemskaya NV, et al. Transcriptome analysis of long‐lived Drosophila melanogaster E(z) mutants sheds light on the molecular mechanisms of longevity. Sci Rep. 2019;9(1):9151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Graffmann N, Brands J, Gorgens A. Vitoriano da Conceicao Castro S, Santourlidis S, Reckert a, et al. age‐related increase of EED expression in early hematopoietic progenitor cells is associated with global increase of the histone modification H3K27me3. Stem Cells Dev. 2015;24(17):2018‐2031. [DOI] [PubMed] [Google Scholar]
- 153. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Brinkman AB, Gu H, Bartels SJ, et al. Sequential ChIP‐bisulfite sequencing enables direct genome‐scale investigation of chromatin and DNA methylation cross‐talk. Genome Res. 2012;22(6):1128‐1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Kondo Y. Shall we crosstalk? ‐ the relationship between DNA methylation and histone H3 lysine 27 trimethylation. Bioessays. 2014;36(2):128. [DOI] [PubMed] [Google Scholar]
- 156. Reddington JP, Sproul D, Meehan RR. DNA methylation reprogramming in cancer: does it act by re‐configuring the binding landscape of Polycomb repressive complexes? Bioessays. 2014;36(2):134‐140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Simon JA, Kingston RE. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol. 2009;10(10):697‐708. [DOI] [PubMed] [Google Scholar]
- 158. Cuyas E, Verdura S, Llorach‐Pares L, et al. Metformin directly targets the H3K27me3 demethylase KDM6A/UTX. Aging Cell. 2018;17(4):e12772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Bracken AP, Kleine‐Kohlbrecher D, Dietrich N, et al. The Polycomb group proteins bind throughout the INK4A‐ARF locus and are disassociated in senescent cells. Genes Dev. 2007;21(5):525‐530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Sherr CJ. Ink4‐Arf locus in cancer and aging. Wiley Interdiscip Rev Dev Biol. 2012;1(5):731‐741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. LaPak KM, Burd CE. The molecular balancing act of p16(INK4a) in cancer and aging. Mol Cancer Res. 2014;12(2):167‐183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Howe FS, Fischl H, Murray SC, Mellor J. Is H3K4me3 instructive for transcription activation? Bioessays. 2017;39(1):1‐12. [DOI] [PubMed] [Google Scholar]
- 163. Zhou HL, Luo G, Wise JA, Lou H. Regulation of alternative splicing by local histone modifications: potential roles for RNA‐guided mechanisms. Nucleic Acids Res. 2014;42(2):701‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Huang C, Zhu B. Roles of H3K36‐specific histone methyltransferases in transcription: antagonizing silencing and safeguarding transcription fidelity. Biophys Rep. 2018;4(4):170‐177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Yuan W, Xu M, Huang C, Liu N, Chen S, Zhu B. H3K36 methylation antagonizes PRC2‐mediated H3K27 methylation. J Biol Chem. 2011;286(10):7983‐7989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Kim DH, Tang Z, Shimada M, et al. Histone H3K27 trimethylation inhibits H3 binding and function of SET1‐like H3K4 methyltransferase complexes. Mol Cell Biol. 2013;33(24):4936‐4946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Schmitges FW, Prusty AB, Faty M, et al. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell. 2011;42(3):330‐341. [DOI] [PubMed] [Google Scholar]
- 168. Li L, Greer C, Eisenman RN, Secombe J. Essential functions of the histone demethylase lid. PLoS Genet. 2010;6(11):e1001221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Martin‐Herranz DE, Aref‐Eshghi E, Bonder MJ, et al. Screening for genes that accelerate the epigenetic aging clock in humans reveals a role for the H3K36 methyltransferase NSD1. Genome Biol. 2019;20(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Grabowska W, Sikora E, Bielak‐Zmijewska A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology. 2017;18(4):447‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: promises, achievements, and challenges. Pharmacol Ther. 2018;188:140‐154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Burnett C, Valentini S, Cabreiro F, et al. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature. 2011;477(7365):482‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Woods JK, Rogina B. The effects of Rpd3 on fly metabolism, health, and longevity. Exp Gerontol. 2016;86:124‐128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Whitaker R, Faulkner S, Miyokawa R, et al. Increased expression of Drosophila Sir2 extends life span in a dose‐dependent manner. Aging (Albany NY). 2013;5(9):682‐691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Griswold AJ, Chang KT, Runko AP, Knight MA, Min KT. Sir2 mediates apoptosis through JNK‐dependent pathways in Drosophila. Proc Natl Acad Sci U S A. 2008;105(25):8673‐8678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Rogina B, Helfand SL, Frankel S. Longevity regulation by Drosophila Rpd3 deacetylase and caloric restriction. Science. 2002;298(5599):1745. [DOI] [PubMed] [Google Scholar]
- 177. Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 2014;6(4):a018713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Frankel S, Ziafazeli T, Rogina B. dSir2 and longevity in Drosophila. Exp Gerontol. 2011;46(5):391‐396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. De Vaux V, Pfefferli C, Passannante M, et al. The Caenorhabditis elegans LET‐418/Mi2 plays a conserved role in lifespan regulation. Aging Cell. 2013;12(6):1012‐1020. [DOI] [PubMed] [Google Scholar]
- 180. Basta J, Rauchman M. The nucleosome remodeling and deacetylase complex in development and disease. Transl Res. 2015;165(1):36‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Kehle J, Beuchle D, Treuheit S, et al. dMi‐2, a hunchback‐interacting protein that functions in polycomb repression. Science. 1998;282(5395):1897‐1900. [DOI] [PubMed] [Google Scholar]
- 182. Reynolds N, Salmon‐Divon M, Dvinge H, et al. NuRD‐mediated deacetylation of H3K27 facilitates recruitment of Polycomb repressive complex 2 to direct gene repression. EMBO J. 2012;31(3):593‐605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Tie F, Banerjee R, Stratton CA, et al. CBP‐mediated acetylation of histone H3 lysine 27 antagonizes Drosophila Polycomb silencing. Development. 2009;136(18):3131‐3141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Wagner W. Epigenetic aging clocks in mice and men. Genome Biol. 2017;18(1):107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Petkovich DA, Podolskiy DI, Lobanov AV, Lee SG, Miller RA, Gladyshev VN. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 2017;25(4):954‐960 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Cole JJ, Robertson NA, Rather MI, et al. Diverse interventions that extend mouse lifespan suppress shared age‐associated epigenetic changes at critical gene regulatory regions. Genome Biol. 2017;18(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Wang T, Tsui B, Kreisberg JF, et al. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol. 2017;18(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Stubbs TM, Bonder MJ, Stark AK, et al. Multi‐tissue DNA methylation age predictor in mouse. Genome Biol. 2017;18(1):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Thompson MJ, Chwialkowska K, Rubbi L, et al. A multi‐tissue full lifespan epigenetic clock for mice. Aging (Albany NY). 2018;10(10):2832‐2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Horvath S, Erhart W, Brosch M, et al. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci U S A. 2014;111(43):15538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Horvath S, Raj K. DNA methylation‐based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet. 2018;19(6):371‐384. [DOI] [PubMed] [Google Scholar]
- 192. Grant CD, Jafari N, Hou L, et al. A longitudinal study of DNA methylation as a potential mediator of age‐related diabetes risk. Geroscience. 2017;39(5–6):475‐489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Li X, Ploner A, Wang Y, et al. Longitudinal trajectories, correlations and mortality associations of nine biological ages across 20‐years follow‐up. Elife. 2020;9:e51507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Yi S. Birds do it, bees do it, worms and ciliates do it too: DNA methylation from unexpected corners of the tree of life. Genome Biol. 2012;13(10):174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Dunwell TL, Pfeifer GP. Drosophila genomic methylation: new evidence and new questions. Epigenomics. 2014;6(5):459‐461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Wenzel D, Palladino F, Jedrusik‐Bode M. Epigenetics in C. elegans: facts and challenges. Genesis. 2011;49(8):647‐661. [DOI] [PubMed] [Google Scholar]
- 197. Capuano F, Mulleder M, Kok R, Blom HJ, Ralser M. Cytosine DNA methylation is found in Drosophila melanogaster but absent in Saccharomyces cerevisiae, Schizosaccharomyces pombe, and other yeast species. Anal Chem. 2014;86(8):3697‐3702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Takayama S, Dhahbi J, Roberts A, et al. Genome methylation in D. melanogaster is found at specific short motifs and is independent of DNMT2 activity. Genome Res. 2014;24(5):821‐830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Lyko F, Ramsahoye BH, Jaenisch R. DNA methylation in Drosophila melanogaster . Nature. 2000;408(6812):538‐540. [DOI] [PubMed] [Google Scholar]
- 200. Kunert N, Marhold J, Stanke J, Stach D, Lyko F. A Dnmt2‐like protein mediates DNA methylation in Drosophila. Development. 2003;130(21):5083‐5090. [DOI] [PubMed] [Google Scholar]
- 201. Rasmussen EMK, Vagbo CB, Munch D, et al. DNA base modifications in honey bee and fruit fly genomes suggest an active demethylation machinery with species‐ and tissue‐specific turnover rates. Biochem Biophys Rep. 2016;6:9‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Deshmukh S, Ponnaluri VC, Dai N, Pradhan S, Deobagkar D. Levels of DNA cytosine methylation in the Drosophila genome. PeerJ. 2018;6:e5119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Lian T, Gaur U, Wu QI, et al. DNA methylation is not involved in dietary restriction induced lifespan extension in adult Drosophila. Genet Res (Camb). 2018;100:e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Goll MG, Kirpekar F, Maggert KA, et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science. 2006;311(5759):395‐398. [DOI] [PubMed] [Google Scholar]
- 205. Jeltsch A, Ehrenhofer‐Murray A, Jurkowski TP, et al. Mechanism and biological role of Dnmt2 in nucleic acid methylation. RNA Biol. 2017;14(9):1108‐1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Jurkowski TP, Meusburger M, Phalke S, et al. Human DNMT2 methylates tRNA(asp) molecules using a DNA methyltransferase‐like catalytic mechanism. RNA. 2008;14(8):1663‐1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Tuorto F, Liebers R, Musch T, et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat Struct Mol Biol. 2012;19(9):900‐905. [DOI] [PubMed] [Google Scholar]
- 208. Kaiser S, Jurkowski TP, Kellner S, Schneider D, Jeltsch A, Helm M. The RNA methyltransferase Dnmt2 methylates DNA in the structural context of a tRNA. RNA Biol. 2017;14(9):1241‐1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Xiao CL, Zhu S, He M, et al. N(6)‐methyladenine DNA modification in the human genome. Mol Cell. 2018;71(2):306‐318 e7. [DOI] [PubMed] [Google Scholar]
- 210. Zhang G, Huang H, Liu D, et al. N6‐methyladenine DNA modification in Drosophila. Cell. 2015;161(4):893‐906. [DOI] [PubMed] [Google Scholar]
- 211. He S, Zhang G, Wang J, et al. 6mA‐DNA‐binding factor Jumu controls maternal‐to‐zygotic transition upstream of Zelda. Nat Commun. 2019;10(1):2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Yao B, Li Y, Wang Z, et al. Active N(6)‐methyladenine demethylation by DMAD regulates gene expression by coordinating with Polycomb protein in neurons. Mol Cell. 2018;71(5):848‐857. e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. He J, Tu C, Liu Y. Role of lncRNAs in aging and age‐related diseases. Aging Med (Milton). 2018;1(2):158‐175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Li ZX, Zhu QN, Zhang HB, Hu Y, Wang G, Zhu YS. MALAT1: a potential biomarker in cancer. Cancer Manag Res. 2018;10:6757‐6768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Wang X, Sehgal L, Jain N, Khashab T, Mathur R, Samaniego F. LncRNA MALAT1 promotes development of mantle cell lymphoma by associating with EZH2. J Transl Med. 2016;14(1):346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Marttila S, Chatsirisupachai K, Palmer D, de Magalhaes JP. Ageing‐associated changes in the expression of lncRNAs in human tissues reflect a transcriptional modulation in ageing pathways. Mech Ageing Dev. 2020;185:111177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Degirmenci U, Lei S. Role of lncRNAs in cellular aging. Front Endocrinol (Lausanne). 2016;7:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Hanly DJ, Esteller M, Berdasco M. Interplay between long non‐coding RNAs and epigenetic machinery: emerging targets in cancer? Philos Trans R Soc Lond B Biol Sci. 2018;373(1748):20170074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Bennett RL, Licht JD. Targeting epigenetics in Cancer. Annu Rev Pharmacol Toxicol. 2018;58:187‐207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Anastasiadou E, Jacob LS, Slack FJ. Non‐coding RNA networks in cancer. Nat Rev Cancer. 2018;18(1):5‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Bink DI, Lozano‐Vidal N, Boon RA. Long non‐coding RNA in vascular disease and aging. Noncoding RNA. 2019;5(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Fernandes JCR, Acuna SM, Aoki JI, Floeter‐Winter LM, Muxel SM. Long non‐coding RNAs in the regulation of gene expression: physiology and disease. Noncoding RNA. 2019;5(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Mori MA. Editorial: non‐coding RNAs: entwining metabolism and aging. Front Endocrinol (Lausanne). 2018;9:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Yang D, Lian T, Tu J, et al. LncRNA mediated regulation of aging pathways in Drosophila melanogaster during dietary restriction. Aging (Albany NY). 2016;8(9):2182‐2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Rogoyski OM, Pueyo JI, Couso JP, Newbury SF. Functions of long non‐coding RNAs in human disease and their conservation in Drosophila development. Biochem Soc Trans. 2017;45(4):895‐904. [DOI] [PubMed] [Google Scholar]
- 226. Li K, Tian Y, Yuan Y, et al. Insights into the functions of LncRNAs in Drosophila. Int J Mol Sci. 2019;20(18):4646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Xiong XP, Kurthkoti K, Chang KY, et al. miR‐34 modulates innate immunity and Ecdysone signaling in Drosophila. PLoS Pathog. 2016;12(11):e1006034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Kennerdell JR, Liu N, Bonini NM. MiR‐34 inhibits polycomb repressive complex 2 to modulate chaperone expression and promote healthy brain aging. Nat Commun. 2018;9(1):4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Owczarz M, Budzinska M, Domaszewska‐Szostek A, et al. miR‐34a and miR‐9 are overexpressed and SIRT genes are downregulated in peripheral blood mononuclear cells of aging humans. Exp Biol Med (Maywood). 2017;242(14):1453‐1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Fomison‐Nurse I, Saw EEL, Gandhi S, et al. Diabetes induces the activation of pro‐ageing miR‐34a in the heart, but has differential effects on cardiomyocytes and cardiac progenitor cells. Cell Death Differ. 2018;25(7):1336‐1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Cui H, Ge J, Xie N, et al. miR‐34a inhibits lung fibrosis by inducing lung fibroblast senescence. Am J Respir Cell Mol Biol. 2017;56(2):168‐178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Xu X, Chen W, Miao R, et al. miR‐34a induces cellular senescence via modulation of telomerase activity in human hepatocellular carcinoma by targeting FoxM1/c‐Myc pathway. Oncotarget. 2015;6(6):3988‐4004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Epstein Y, Perry N, Volin M, et al. miR‐9a modulates maintenance and ageing of Drosophila germline stem cells by limiting N‐cadherin expression. Nat Commun. 2017;8(1):600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Banzhaf‐Strathmann J, Edbauer D. Good guy or bad guy: the opposing roles of microRNA 125b in cancer. Cell Commun Signal. 2014;12:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Budzinska M, Owczarz M, Pawlik‐Pachucka E, Roszkowska‐Gancarz M, Slusarczyk P, Puzianowska‐Kuznicka M. miR‐96, miR‐145 and miR‐9 expression increases, and IGF‐1R and FOXO1 expression decreases in peripheral blood mononuclear cells of aging humans. BMC Geriatr. 2016;16(1):200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Braconi C, Huang N, Patel T. MicroRNA‐dependent regulation of DNA methyltransferase‐1 and tumor suppressor gene expression by interleukin‐6 in human malignant cholangiocytes. Hepatology. 2010;51(3):881‐890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Wang YS, Chou WW, Chen KC, Cheng HY, Lin RT, Juo SH. MicroRNA‐152 mediates DNMT1‐regulated DNA methylation in the estrogen receptor alpha gene. PLoS One. 2012;7(1):e30635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Wang L, Yao J, Sun H, et al. MicroRNA‐101 suppresses progression of lung cancer through the PTEN/AKT signaling pathway by targeting DNA methyltransferase 3A. Oncol Lett. 2017;13(1):329‐338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Lee YS, Dutta A. MicroRNAs in cancer. Annu Rev Pathol. 2009;4:199‐227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Westholm JO, Miura P, Olson S, et al. Genome‐wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age‐dependent neural accumulation. Cell Rep. 2014;9(5):1966‐1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Hall H, Medina P, Cooper DA, et al. Transcriptome profiling of aging Drosophila photoreceptors reveals gene expression trends that correlate with visual senescence. BMC Genomics. 2017;18(1):894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Cai H, Li Y, Niringiyumukiza JD, Su P, Xiang W. Circular RNA involvement in aging: An emerging player with great potential. Mech Ageing Dev. 2019;178:16‐24. [DOI] [PubMed] [Google Scholar]
- 243. Yue Y, Liu J, He C. RNA N6‐methyladenosine methylation in post‐transcriptional gene expression regulation. Genes Dev. 2015;29(13):1343‐1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Li X, Xiong X, Yi C. Epitranscriptome sequencing technologies: decoding RNA modifications. Nat Methods. 2016;14(1):23‐31. [DOI] [PubMed] [Google Scholar]
- 245. Batista PJ. The RNA modification N(6)‐methyladenosine and its implications in human disease. Genomics Proteomics Bioinformatics. 2017;15(3):154‐163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Li Z, Zhao P, Xia Q. Epigenetic methylations on N6‐adenine and N6‐adenosine with the same input but different output. Int J Mol Sci. 2019;20(12):E2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Lence T, Soller M, Roignant JY. A fly view on the roles and mechanisms of the m(6)A mRNA modification and its players. RNA Biol. 2017;14(9):1232‐1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Lence T, Akhtar J, Bayer M, et al. M(6)a modulates neuronal functions and sex determination in Drosophila. Nature. 2016;540(7632):242‐247. [DOI] [PubMed] [Google Scholar]
- 249. Hongay CF, Orr‐Weaver TL. Drosophila inducer of MEiosis 4 (IME4) is required for notch signaling during oogenesis. Proc Natl Acad Sci U S A. 2011;108(36):14855‐14860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Min KW, Zealy RW, Davila S, et al. Profiling of m6A RNA modifications identified an age‐associated regulation of AGO2 mRNA stability. Aging Cell. 2018;17(3):e12753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Cui Q, Shi H, Ye P, et al. m(6)A RNA methylation regulates the self‐renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 2017;18(11):2622‐2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Fustin JM, Doi M, Yamaguchi Y, et al. RNA‐methylation‐dependent RNA processing controls the speed of the circadian clock. Cell. 2013;155(4):793‐806. [DOI] [PubMed] [Google Scholar]
- 253. Barbieri I, Tzelepis K, Pandolfini L, et al. Promoter‐bound METTL3 maintains myeloid leukaemia by m(6)A‐dependent translation control. Nature. 2017;552(7683):126‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Liu J, Eckert MA, Harada BT, et al. M(6)a mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat Cell Biol. 2018;20(9):1074‐1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Zaccara S, Ries RJ, Jaffrey SR. Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 2019;20(10):608‐624. [DOI] [PubMed] [Google Scholar]
- 256. Trixl L, Lusser A. The dynamic RNA modification 5‐methylcytosine and its emerging role as an epitranscriptomic mark. Wiley Interdiscip Rev RNA. 2019;10(1):e1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Mauer J, Jaffrey SR. FTO, m(6) Am, and the hypothesis of reversible epitranscriptomic mRNA modifications. FEBS Lett. 2018;592(12):2012‐2022. [DOI] [PubMed] [Google Scholar]
- 258. Lewinska A, Adamczyk‐Grochala J, Kwasniewicz E, et al. Reduced levels of methyltransferase DNMT2 sensitize human fibroblasts to oxidative stress and DNA damage that is accompanied by changes in proliferation‐related miRNA expression. Redox Biol. 2018;14:20‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Savva YA, Jepson JE, Chang YJ, et al. RNA editing regulates transposon‐mediated heterochromatic gene silencing. Nat Commun. 2013;4:2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Sebastiani P, Montano M, Puca A, et al. RNA editing genes associated with extreme old age in humans and with lifespan in C. elegans . PLoS One. 2009;4(12):e8210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Kung CP, Maggi LB Jr, Weber JD. The role of RNA editing in cancer development and metabolic disorders. Front Endocrinol (Lausanne). 2018;9:762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Hannum G, Guinney J, Zhao L, et al. Genome‐wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013;49(2):359‐367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Marioni RE, Shah S, McRae AF, et al. DNA methylation age of blood predicts all‐cause mortality in later life. Genome Biol. 2015;16:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Karemaker ID, Vermeulen M. Single‐cell DNA methylation profiling: technologies and biological applications. Trends Biotechnol. 2018;36(9):952‐965. [DOI] [PubMed] [Google Scholar]
- 265. Song Y, Xu X, Wang W, Tian T, Zhu Z, Yang C. Single cell transcriptomics: moving towards multi‐omics. Analyst. 2019;144(10):3172‐3189. [DOI] [PubMed] [Google Scholar]
- 266. Yan J, Risacher SL, Shen L, Saykin AJ. Network approaches to systems biology analysis of complex disease: integrative methods for multi‐omics data. Brief Bioinform. 2018;19(6):1370‐1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Hasin Y, Seldin M, Lusis A. Multi‐omics approaches to disease. Genome Biol. 2017;18(1):83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Pinu FR, Beale DJ, Paten AM, et al. Systems biology and multi‐Omics integration: viewpoints from the metabolomics research community. Metabolites. 2019;9(4):E76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Canzler S, Schor J, Busch W, et al. Prospects and challenges of multi‐omics data integration in toxicology. Arch Toxicol. 2020;94(2):371‐388. [DOI] [PubMed] [Google Scholar]
- 270. Misra BB, Langefeld CD, Olivier M, Cox LA. Integrated Omics: tools, advances, and future approaches. J Mol Endocrinol. 2018;62(1):R21‐R45. [DOI] [PubMed] [Google Scholar]
- 271. Chappell L, Russell AJC, Voet T. Single‐cell (multi)omics technologies. Annu Rev Genomics Hum Genet. 2018;19:15‐41. [DOI] [PubMed] [Google Scholar]
- 272. Hu Y, An Q, Sheu K, Trejo B, Fan S, Guo Y. Single cell multi‐Omics technology: methodology and application. Front Cell Dev Biol. 2018;6:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Burman JL, Itsara LS, Kayser EB, et al. A Drosophila model of mitochondrial disease caused by a complex I mutation that uncouples proton pumping from electron transfer. Dis Model Mech. 2014;7(10):1165‐1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Chen J, Shi X, Padmanabhan R, et al. Identification of novel modulators of mitochondrial function by a genome‐wide RNAi screen in Drosophila melanogaster . Genome Res. 2008;18(1):123‐136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Yoon W, Hwang SH, Lee SH, Chung J. Drosophila ADCK1 is critical for maintaining mitochondrial structures and functions in the muscle. PLoS Genet. 2019;15(5):e1008184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Mast JD, Tomalty KM, Vogel H, Clandinin TR. Reactive oxygen species act remotely to cause synapse loss in a Drosophila model of developmental mitochondrial encephalopathy. Development. 2008;135(15):2669‐2679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Oswald MC, Brooks PS, Zwart MF, et al. Reactive oxygen species regulate activity‐dependent neuronal plasticity in Drosophila. Elife. 2018;7:e39393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Ichinose T, Tanimoto H. Dynamics of memory‐guided choice behavior in Drosophila. Proc Jpn Acad Ser B Phys Biol Sci. 2016;92(8):346‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Simonnet MM, Berthelot‐Grosjean M, Grosjean Y. Testing Drosophila olfaction with a Y‐maze assay. J Vis Exp. 2014;88:51241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. Ali YO, Escala W, Ruan K, Zhai RG. Assaying locomotor, learning, and memory deficits in Drosophila models of neurodegeneration. J Vis Exp. 2011;49:2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Bantel AP, Tessier CR. Taste preference assay for adult Drosophila. J Vis Exp. 2016;115:54403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Savall J, Ho ETW, Huang C, Maxey JR, Schnitzer MJ. Dexterous robotic manipulation of alert adult Drosophila for high‐content experimentation. Nat Methods. 2015;12(7):657‐660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Cichewicz K, Hirsh J. ShinyR‐DAM: a program analyzing Drosophila activity, sleep and circadian rhythms. Commun Biol. 2018;1:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Tataroglu O, Emery P. Studying circadian rhythms in Drosophila melanogaster . Methods. 2014;68(1):140‐150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Linford NJ, Ro J, Chung BY, Pletcher SD. Gustatory and metabolic perception of nutrient stress in Drosophila. Proc Natl Acad Sci U S A. 2015;112(8):2587‐2592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Bach EA, Vincent S, Zeidler MP, Perrimon N. A sensitized genetic screen to identify novel regulators and components of the Drosophila janus kinase/signal transducer and activator of transcription pathway. Genetics. 2003;165(3):1149‐1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Djeghloul D, Kuranda K, Kuzniak I, et al. Age‐associated decrease of the histone Methyltransferase SUV39H1 in HSC perturbs heterochromatin and B lymphoid differentiation. Stem Cell Reports. 2016;6(6):970‐984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Scaffidi P, Misteli T. Reversal of the cellular phenotype in the premature aging disease Hutchinson‐Gilford progeria syndrome. Nat Med. 2005;11(4):440‐445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Scaffidi P, Misteli T. Lamin A‐dependent nuclear defects in human aging. Science. 2006;312(5776):1059‐1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Zhang W, Li J, Suzuki K, et al. Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science. 2015;348(6239):1160‐1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. O'Sullivan RJ, Kubicek S, Schreiber SL, Karlseder J. Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nat Struct Mol Biol. 2010;17(10):1218‐1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Cheung I, Shulha HP, Jiang Y, et al. Developmental regulation and individual differences of neuronal H3K4me3 epigenomes in the prefrontal cortex. Proc Natl Acad Sci U S A. 2010;107(19):8824‐8829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Peleg S, Feller C, Forne I, et al. Life span extension by targeting a link between metabolism and histone acetylation in Drosophila. EMBO Rep. 2016;17(3):455‐469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Bellizzi D, Rose G, Cavalcante P, et al. A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics. 2005;85(2):258‐263. [DOI] [PubMed] [Google Scholar]
- 295. Rose G, Dato S, Altomare K, et al. Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. Exp Gerontol. 2003;38(10):1065‐1070. [DOI] [PubMed] [Google Scholar]
- 296. Pegoraro G, Kubben N, Wickert U, Gohler H, Hoffmann K, Misteli T. Ageing‐related chromatin defects through loss of the NURD complex. Nat Cell Biol. 2009;11(10):1261‐1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Durdevic Z, Mobin MB, Hanna K, Lyko F, Schaefer M. The RNA methyltransferase Dnmt2 is required for efficient Dicer‐2‐dependent siRNA pathway activity in Drosophila. Cell Rep. 2013;4(5):931‐937. [DOI] [PubMed] [Google Scholar]
- 298. Nielsen JB, Thorolfsdottir RB, Fritsche LG, et al. Biobank‐driven genomic discovery yields new insight into atrial fibrillation biology. Nat Genet. 2018;50(9):1234‐1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Fransen E, Bonneux S, Corneveaux JJ, et al. Genome‐wide association analysis demonstrates the highly polygenic character of age‐related hearing impairment. Eur J Hum Genet. 2015;23(1):110‐115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Lee E, Giovanello KS, Saykin AJ, et al. Single‐nucleotide polymorphisms are associated with cognitive decline at Alzheimer's disease conversion within mild cognitive impairment patients. Alzheimers Dement (Amst). 2017;8:86‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Perry JR, Day F, Elks CE, et al. Parent‐of‐origin‐specific allelic associations among 106 genomic loci for age at menarche. Nature. 2014;514(7520):92‐97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Kichaev G, Bhatia G, Loh PR, et al. Leveraging polygenic functional enrichment to improve GWAS power. Am J Hum Genet. 2019;104(1):65‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Subramanian VV, Bickel SE. Aging predisposes oocytes to meiotic nondisjunction when the cohesin subunit SMC1 is reduced. PLoS Genet. 2008;4(11):e1000263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. Subramanian VV, Bickel SE. Heterochromatin‐mediated association of achiasmate homologs declines with age when cohesion is compromised. Genetics. 2009;181(4):1207‐1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Hodges CA, Revenkova E, Jessberger R, Hassold TJ, Hunt PA. SMC1beta‐deficient female mice provide evidence that cohesins are a missing link in age‐related nondisjunction. Nat Genet. 2005;37(12):1351‐1355. [DOI] [PubMed] [Google Scholar]
- 306. Tsutsumi M, Fujiwara R, Nishizawa H, et al. Age‐related decrease of meiotic cohesins in human oocytes. PLoS One. 2014;9(5):e96710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Cheng JM, Liu YX. Age‐related loss of cohesion: causes and effects. Int J Mol Sci. 2017;18(7):1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Gilliland WD, Hawley RS. Cohesin and the maternal age effect. Cell. 2005;123(3):371‐373. [DOI] [PubMed] [Google Scholar]
- 309. van Steensel B, Belmont AS. Lamina‐associated domains: links with chromosome architecture, heterochromatin, and gene repression. Cell. 2017;169(5):780‐791. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Transparent‐Peer‐Review‐Record