Skip to main content
Briefings in Functional Genomics logoLink to Briefings in Functional Genomics
. 2021 Mar 19;21(1):35–42. doi: 10.1093/bfgp/elab010

Epigenetic alterations in stem cell ageing—a promising target for age-reversing interventions?

Andromachi Pouikli, Peter Tessarz
PMCID: PMC8789308  PMID: 33738480

Abstract

Ageing is accompanied by loss of tissue integrity and organismal homeostasis partly due to decline in stem cell function. The age-associated decrease in stem cell abundance and activity is often referred to as stem cell exhaustion and is considered one major hallmark of ageing. Importantly, stem cell proliferation and differentiation potential are tightly coupled to the cellular epigenetic state. Thus, research during the last years has started to investigate how the epigenome regulates stem cell function upon ageing. Here, we summarize the role of epigenetic regulation in stem cell fate decisions and we review the impact of age-related changes of the epigenome on stem cell activity. Finally, we discuss how targeted interventions on the epigenetic landscape might delay ageing and extend health-span.

Keywords: epigenetics, chromatin, ageing, stem cells

Introduction

Chromatin describes the macromolecular complex of proteins and DNA that can be found in the nucleus of every eukaryotic cell. It provides the scaffold for packaging the entire genetic material, facilitating its compaction and protecting DNA. The core of chromatin consists of the nucleosome. Research during the last decades has revealed that post-translational modifications on histones, which are the protein components of nucleosomes, and on DNA itself regulate gene expression and allow time- and tissue-controlled read-out of the genetic information [1, 2]. These studies have highlighted the contribution of chromatin architecture and dynamics during physiological development as well as upon tumorigenesis [3, 4]. Recent data also underscore the central role of epigenetics in the development and progression of ageing and age-related diseases, such as cardiovascular, neurodegenerative and metabolic disorders [5].

Adult stem cells play a vital role in tissue repair and regeneration; thus, they are essential for the maintenance of tissue homeostasis. Stem cell exhaustion, which describes the decline in the stem cell number and/or function, is one well-established hallmark of ageing. During the last years, the role of the epigenome in the regulation of adult stem cell activity, particularly in the context of ageing, has been the subject of increased scientific interest. Given the major contribution of stem cells in the maintenance of tissue integrity and homeostasis, investigating the mechanisms that govern stem cell ageing is of exceptional importance for interventions aiming at delaying or even preventing ageing and age-associated pathologies. However, stem cell ageing research is extremely challenging, mostly due to technical limitations, such as the isolation of pure stem cell populations in sufficient numbers. As a result, we still have only limited insight into the age-related changes of the stem cell epigenome. Nevertheless, the increasing progress in identifying suitable markers for purification of homogenous stem cell populations, the constant improvement of flow cytometers allowing cell sorting based on several markers simultaneously and, more importantly, the development of sensitive (epi-)genomic assays to study the chromatin landscape even at single-cell resolution has now made it possible to address these intriguing questions during the course of ageing. Here, we summarize the current knowledge on the role of epigenetics in the ageing process focusing on the impact of age-dependent alterations of the epigenome on stem cell physiology. We conclude our review discussing how careful interventions to the epigenetics-ageing axis might represent a potential way to alter stem cell fate decisions, favouring rejuvenation of aged stem cells and enhancing tissue homeostasis.

Age-associated alterations of the epigenome

DNA methylation was discovered almost simultaneously with the identification of DNA as the genetic material [6, 7]. We know now that ~70% of cytosines are methylated genome-wide, as part of the dinucleotide motif CpG [8] and that DNA methylation at gene promoters correlates with transcriptional repression. The responsible mechanisms for the establishment and maintenance of DNA methylation as well as its role in transcriptional regulation are extensively discussed elsewhere [1, 9].

Given the early discovery of DNA methylation, it is not surprising that the first data on age-associated epigenetic modifications focused on the analysis of the DNA methylation profile in young and old individuals. Early studies showed that DNA methylation decreases with age in several organs in salmons, rats and mice [10–12]. Around ~25% of the CpG sites in mice exhibit age-related methylation changes across tissues, with the most prominent alterations observed in highly proliferative organs, such as the gut and the spleen [13]. Interestingly, the alterations of the DNA methylation profile occur progressively and linearly upon ageing, with similar rates between hyper- and hypomethylation; a phenomenon that has been described as epigenetic drift. The progressive modulation of the DNA methylation status with age explains the use of methylation patterns as a biomarker of ageing. Indeed, there are now available several global and tissue-specific epigenetic ageing clocks that are built on statistical models, taking into consideration the methylation level at specific genomic loci [14].

Apart from the changes in the DNA methylation profile, the chromatin landscape undergoes dramatic alterations upon ageing, including re-organization and loss of heterochromatic regions. In particular, reduction or redistribution of the transcriptional repressing H3K9me2/3 marks disrupts HP1 localization [12] and affects heterochromatin organization. Such heterochromatin rearrangements have been correlated with ageing in C. elegans, D. melanogaster and humans [15–17]. Similarly, epigenetic modifications in euchromatin have also been reported to change with age. Although this subject has been covered in detail elsewhere [18, 19], for the purpose of this review, we would like to discuss two important observations that highlight the fundamental role of an altered epigenetic landscape in the ageing process. Firstly, genetic manipulation of chromatin modifiers has been shown to elicit a profound effect on longevity in different model organisms; deletion of the chromatin-associated proteins ASH-2, WDR-5, and SET-2 leads to decreased H3K4me3 levels and subsequently, lifespan extension in C. elegans [20]. In contrast, mutations in set1 and met-1 genes, which encode for the enzymes that deposit H3K36me3, reduce lifespan of S. cerevisiae and C. elegans, respectively [21, 22]. Furthermore, in a landmark publication, Ocampo et al. [23] demonstrated that a transient and subtle induction of Oct4, Sox2, Klf4 and Myc genes ameliorates progeria and improves regeneration of various murine tissues, via epigenetic remodelling. These examples indicate that epigenetic reprogramming has a beneficial effect on organismal function and is associated with extension of the health-span and lifespan.

Changes in epigenetic modifications represent one extensively studied hallmark of ageing. However, one important issue to consider when interpreting results from epigenetic studies in aged organisms is that so far most of them have been carried out in complex biological systems or tissues composed of multiple cell types. Thus, these data likely reflect the average of all the cell types found in each tissue. Therefore, the possibility that the epigenetic drift is also—at least in part—caused by a change in tissue composition with age cannot be excluded. To this end, development of methods to analyse DNA methylation and chromatin architecture at single-cell resolution and establishment of ChIP-like approaches which require low cell numbers, such as the CUT&RUN [24] and the CUT&Tag [25] technics, will considerably facilitate research on the role of epigenetics in ageing.

Epigenetic regulation of fate decisions in adult stem cells

Adult stem cells have the dual capacity to proliferate and differentiate into specific lineages in response to various internal and external stimuli [26, 27]. Given that stem cells share the same genetic information with somatic cells, their epigenome and the associated transcriptional signature distinguish them from their differentiated counterparts. Importantly, the characteristic epigenetic profile of stem cells reflects their wide developmental potential. During the last years, the molecular mechanisms via which changes in the chromatin landscape control stem cell fate decisions have been the subject of intense scientific research, both in embryonic stem cells (ESCs) as well as in various adult stem cell populations.

As discussed above, DNA methylation at regulatory regions, such as gene promoters and enhancers, impacts stem cell fate decisions via altering the transcriptional output (Figure 1). For instance, differentiating ESCs display increased DNA methylation and high levels of H3K9me3 and H3K27me3, but low levels of H3K4me3 [28–30]. This increase in the transcriptional repressive marks promotes silencing of self-renewal genes and enhances lineage commitment. In contrast, regulatory regions of differentiation-associated genes are usually methylated in quiescent hair follicle stem cells (HFSCs), whereas they undergo a progressive, but profound de-methylation upon differentiation [31]. Likewise, neural stem cells (NSCs) display high DNA methylation, which is lost upon induction of differentiation, favouring efficient development of mature neurons [32]. The critical role of DNA and histone methylation in the regulation of stem cell function has also been investigated using various genetic models. For example, loss of the DNA methyltransferase 3A and/or 3B (DNMT3A/B) impairs the differentiation capacity of murine haematopoietic stem cells (HSCs) [33, 34]. Similarly, deletion of KMT5B, the enzyme that deposits H4K20me2, activates muscle stem cells (MuSCs) and forces them to differentiate. This leads to depletion of the MuSC population and impairs muscle integrity [36]. Furthermore, deletion of Tet1, the enzyme involved in DNA hydroxymethylation, regulates the expression of Wnt target genes in mouse intestinal stem cells (ISCs), altering their self-renewal capacity and influencing the integrity of the intestinal epithelium [35].

Figure 1 .


Figure 1

Epigenetic changes during stem cell differentiation. Overview of the changes that occur on the epigenome during differentiation of stem cells. DNA methylation increases in the differentiating HSCs, while it decreases upon NSCs differentiation. H3acetylation (H3ac) is higher in the differentiating NSCs, whereas it is reduced in MSCs during lineage commitment. Differentiating MuSCs exhibit high levels of H4K16ac and low levels of H4K20me2. Importantly, these changes occur at specific genomic loci, favouring the expression of lineage-specific genes and preventing expression of self-renewal and stemness-related genes. In blue boxes are indicated small molecules and metabolites that influence the activity of the respective epigenetic enzymes, affecting stem cell fate decisions.

Histone acetylation is associated with activation of gene transcription and requires acetyl-CoA as the donor of the acetyl-group. Several studies have investigated the role of histone acetylation in different stem cell populations both in proliferating cells as well as during the course of differentiation. In mesenchymal stem cells (MSCs), elevated levels of global H3 acetylation (H3ac) correlate with enhanced stemness; indeed, in proliferating human MSCs increased H3ac promotes expression of core pluripotency genes, whereas osteogenic differentiation is accompanied by loss of global H3ac, leading to repression of self-renewal genes [37]. Consistently, histone deacetylation by SIRT3 is essential for efficient adipogenesis. In contrast, in mouse MuSCs elevated H4K16ac drives myogenic differentiation [38], whereas inhibition of histone deacetylases in NSCs is crucial for neurogenesis in mice and rats, favouring the development of both neuronal and glial lineages [39, 40]. In ISCs, nutritional interventions in the form of fasting or caloric restriction leads to elevated b-hydroxybutyrate levels and inhibition of histone deacetylases [41, 42]. These alterations influence ISC function via changing the H3K27ac abundance on promoters of genes involved in the Notch pathway [41]. Therefore, it is evident that the effect of histone acetylation on the regulation of the proliferation/differentiation balance depends heavily on the cell type and the modified residue.

Findings from epigenetic studies in adult stem cell populations have also led to the discovery of drugs, usually small molecules and metabolites, which can redirect—and even reprogram—stem cells towards specific lineages. For instance, vorinostat and sodium butyrate, two inhibitors of histone deacetylases that are commonly used in cancer treatment, arrest murine NSCs in the G1 phase and control cell cycle progression and differentiation [36]. Furthermore, Kohyama et al. [43] showed that treatment of mature osteoblasts with 5-azacytidine (5-AzaC), an inhibitor of DNA methylation, converts them to NSCs, via alterations in their epigenome and their gene expression profile. Similarly, osteocytes treated with valproic acid, a histone deacetylase inhibitor, are able to re-differentiate to functional NSCs [44]. In this context, it is worth mentioning that metabolism might represent a potential means to alter the epigenome and thus the stem cell identity. More specifically, a growing body of evidence suggests that chromatin interacts with metabolism in a direct and dynamic manner. Similar to the central role of acetyl-CoA in the establishment of histone acetylation marks, the metabolism-chromatin interaction is primarily mediated via intermediate metabolites, including α-ketoglutarate (α-KG) and S-adenosylmethionine (SAM). These metabolites serve as cofactors and substrates for epigenetic writers and erasers, modulating their activity and thus affecting stem cell fate decisions [45–48].

These are just a few examples—and by no means an exhaustive overview—that illustrate the tight control of stem cell function by epigenetics.

Epigenetic changes in ageing stem cells

Age-dependent decline in stem cell number and function is observed virtually in all tissues and organs and is an important hallmark of ageing [49]. As discussed above, stem cells play a fundamental role in maintaining tissue homeostasis throughout the lifespan of an organism [50] and their potency is tightly linked with their epigenome. Therefore, it is not surprising that chromatin-mediated changes in stem cell function upon ageing have a profound impact not only on their resident tissue but ultimately on the physiology of the whole organism. The contribution of epigenetic modifications on stem cell ageing is underscored by genetic models. For instance, Sirt7 knock out in mice is shown to drive the exit of HSCs from quiescence and to promote aberrant HSCs proliferation. This phenotype is reminiscent—at least in part—to that of aged HSCs. Mechanistically, this is achieved by loss of H3K56 deacetylation at the promoter of Wnt target genes [51]. Furthermore, in HFSCs, SIRT7 controls hair growth by regulating the transition from telogen to anagen during the hair-follicle life cycle. Sirt7 is downregulated in aged HFSCs, which correlates with the well-known age-related loss and thinning of hair. Importantly, overexpression of Sirt7 in HFSCs of older animals restores hair-growth. Mechanistically, this is mediated via deacetylation of the transcription factor NFATc1, which leads to its destabilization and the subsequent initiation of the hair follicle cycle [52]. For an extensive review of similar examples that highlight the impact of epigenetics on stem cell function during ageing, we refer the interested readers to recent reviews on this topic [53–55] and to Table 1.

Table 1.

Summary of age-related epigenetic alterations in several stem cell types and their functional consequences on stem cell activity

Stem cell type Modification Functional consequence Ref.
Enzyme HSC DNMT1 Lineage bias & self-renewal defects [56–58]
DNMT3A/B Arrest of HSC differentiation [33]
TET1 Enhanced HSC self-renewal & myeloid lineage skewing [59]
MSCs HDAC Senescence [60]
SIRT6 Redox imbalance through H3K56ac on Nrf2 promoter [61]
MuSCs DNMT1 Lineage bias & self-renewal defects [62]
NSCs DNMT3A/B Arrest of NSC differentiation [63]
SIRT1 Abnormal expansion of oligodendrocyte progenitors [64]
ISCs DNMT1 Lineage bias & self-renewal defects [36]
TET1 Enhanced ISC proliferation
HFSCs SIRT7 Arrest of the hair follicle life-cycle transition from telogen to anagen [52]
Histone modification HSCs H3K4me3 local broadening of peaks Broadening at genes involved in self-renewal & loss of differentiation [65]
H3K27ac Altered expression of tumour-suppressor genes [66]
H4K16ac diffuse pattern Myeloid lineage skewing & misformed nuclei [67, 68]
MSCs H3ac & H4ac Impaired osteogenesis [69]
H3K9me3 [70]
MuSCs H3K4me3 Impaired stem cell function & chromatin remodelling [71]
H3K27me3
NSCs H3K27me3 Inhibition of senescence-associated genes [64]

But why does the chromatin landscape and the DNA and histone modification profile change upon ageing? Several studies in a wide range of organisms and cell types suggest that age-dependent DNA damage elicits permanent changes on the epigenome, redistributing chromatin-associated proteins and resetting the chromatin landscape. These alterations are not restored after repair of the DNA damage and have a profound impact on both stem cell and organismal function. For instance, in HFSCs, SIRT7, apart from regulating cell cycle, plays a significant role in the repair of DNA double-strand breaks (DSBs), via H3K18 deacetylation at the break site. This triggers the recruitment of the damage response factor 53BP1 to the DSB site. Thus, Sirt7 deletion has dramatic consequences on the organismal function, resulting in shorter lifespan in mice [72]. In addition to DNA damage, other mechanisms contribute to the age-dependent changes in the epigenome, e.g. metabolic alterations that impact chromatin modifications [73], as metabolism and epigenome are tightly linked via the abundance of central metabolites [53]. Recent data also point towards an impact of age-related changes in cellular polarity on the epigenome [68].

While data from genetic approaches, like the examples described above, are essential to provide functional insights into the role of epigenetics during stem cell ageing, it is also vital to start incorporating age-associated changes in the DNA methylation profile, histone modification patterns and chromatin architecture in order to understand the general trends and identify commonly affected genes. Of note, these integrated studies should be performed in homogenous cell populations, ideally over the course of the lifetime and in both sexes to fully characterize the age-related changes as well as the complex underlying mechanisms. Although technically challenging, such studies will definitely shed light on the role of epigenetics in stem cell ageing.

One of the first studies analysing global changes in the DNA methylation profile of purified HSCs from young and old mice used reduced representation bisulfite sequencing (RRBS) [74]. The authors found that the DNA methylation pattern is fairly stable upon ageing, with only a slight increase in residue-specific DNA methylation levels, particularly in sites associated with the stem cell haematopoietic capacity. This study also revealed that ageing in HSCs is linked to DNA hypermethylation at genes that are regulated by the polycomb repressive complex 2 (PRC2). Importantly, these findings were confirmed later by an independent study [65]. Recently, also human HSCs were profiled using RRBS [66]. Of the ~3 million identified CpGs sites, around ~2200 sites changed their DNA methylation status upon ageing. Interestingly, in the same study the authors investigated age-related changes in histone modifications and integrated these results to differentially methylated DNA regions at functional sites of the genome, such as enhancers. Data analysis revealed a strong change in promoter and enhancer DNA methylation of genes that are involved in developmental processes and cancer. Remarkably, these genes are also affected in a similar manner in acute myeloid leukaemia (AML) [66], indicating that the ageing DNA methylation pattern resembles a tumour-like state. These studies on HSCs propose that the DNA methylation profile changes with age. However, these changes are subtle and affect only a subset of sites, suggesting that overall DNA methylation is fairly stable. Likewise, single-cell DNA methylation analysis of MuSCs revealed that the DNA methylation profile remains relatively unchanged upon ageing with only modest increase of DNA methylation, particularly over SINE elements and regions marked by H3K36me3 [75]. However, it is important to highlight that the examples discussed above refer to studies that were carried out in quiescent stem cells. Results of genomic experiments might be different in other compartments, in which stem cells divide more frequently, such as the intestine, in which DNA methylation changes are sufficient to predict the donor’s age using epigenetic clocks [76].

In contrast to DNA methylation, histone modifications undergo profound alterations during ageing in various types of somatic stem cells. In human HSCs, ChIP-seq analysis revealed a strong reduction of H3K4me1, H3K27ac and H3K4me3 levels upon ageing, while H3K27me3 levels are only mildly affected [66]. In contrast, aged murine HSCs display a significant increase in H3K4me3 and H3K27me3 levels in comparison to the young HSCs. In addition to its altered levels, H3K4me3 mark in aged HSCs exhibits a broader distribution over regions associated with HSC identity and self-renewal [65]—a phenomenon that is thought to enhance transcriptional consistency and increase transcriptional output [77]. On the other hand, MuSCs are characterized by a global loss of H3K4me3 and a slight increase of H3K27me3 levels [71]. Importantly, in all of the described cases, the epigenetic changes were corroborated by transcriptional alterations. We recently performed ATAC-sequencing on MSCs isolated from young and old mice and observed a strong decrease in chromatin accessibility, which is in agreement to a global histone hypoacetylation in aged MSCs [69], contrary to the belief that generally, chromatin becomes more accessible with age.

Together, these data point towards a cell-type-specific change in the chromatin modification pattern with age, and they highlight the necessity for a deeper understanding of the contribution of histone modifications to the ageing process of stem cells. Integration of the histone modification profile with chromatin accessibility patterns, the DNA methylation distribution and the transcriptional output will hopefully help us generate a more complete view regarding the precise mechanisms via which the epigenome regulates ageing in somatic stem cells.

Outlook: interfering with the epigenome to positively impact longevity

Although we have only recently started to study the changes that occur in the epigenome of ageing stem cells, it is clear that chromatin structure plays a fundamental role in regulating stem cell fate and function. Interestingly, since modulation of the chromatin architecture enables targeted alteration of the transcriptional output and ultimately of the stem cell identity, the epigenome represents an attractive target for development of anti-ageing strategies (Figure 2). Of note, such approaches to direct stem cell fate decisions towards specific lineages could be combined with the currently used approaches during autologous stem cell therapy. For instance, ex vivo manipulation of the chromatin landscape to enhance stem cell activity prior to HSC and MSC transplantation could potentially increase the efficiency of stem cell therapies. Proof-of-concept studies have been published in the last years using model organisms to explore the potentially beneficial effects of such approaches. For example, ex vivo treatment of aged MuSCs using a combination of biophysical (rigidity of culture matrix) and biochemical (p38a/b inhibitor) approaches resulted in MuSC rejuvenation. Indeed, after transplantation, treated MuSCs contributed to extensive myofiber repair and restored strength to injured muscles in aged mice [78]. While this study did not use any epigenetic intervention, it clearly illustrates the potential of an ex vivo therapy. On the other hand, inhibition of DNMTs by 5-azacytidine has been used in various studies to enhance stem cell differentiation capacity of MSCs purified from elderly human donors [79, 80]. Furthermore, two recent studies demonstrated that supplementation of MSCs with the epigenetic-related metabolites a-KG and acetate improves osteogenesis in aged MSCs. Mechanistically, both metabolites elicit changes on chromatin architecture; in particular, a-KG leads to decreased repressive marks on the promoters of osteogenic genes [81], whereas acetate restores histone acetylation levels and thus, chromatin accessibility on osteogenesis-involved genes [69]. In line to this, manipulating the intracellular metabolism by regulation of nutrient-sensing pathways via caloric restriction and/or pharmacological interventions, such as rapamycin and resveratrol treatment, influences stem cell fate decisions, via altering the epigenome. Therefore, ex vivo treatments to manipulate the chromatin landscape are emerging as a potential tool for efficient stem cell rejuvenation.

Figure 2 .


Figure 2

The epigenome and cell fate decisions. (A) In stem cells, the epigenome plays an important role in the regulation of fate decisions, which are heavily affected by age-associated epigenetic alterations. Targeting the ageing epigenome represents a potential tool to reverse these changes and re-establish the full potency of the stem cell population. (B) MSCs are used as an example to illustrate the effect of epigenetic interventions in stem cell ageing. Upon physiological ageing, MSCs exhibit skewed differentiation at the expense of osteoblasts, which leads to accumulation of adipocytes (upper panel). Treatment of aged MSCs with acetate, α-KG or 5-azacytidine (5-AzaC) enhances stemness and re-establishes a balanced differentiation potential, via altering the stem cell epigenome.

Although there has been significant progress in the use of epigenetic interventions in various cancer types [82], before we start following such approaches to delay age-related decline in stem cell function, we should address several key-issues, including (i) a more complete understanding of the profile of epigenetic modifications in ageing stem cells and the underlying molecular mechanisms and (ii) the development of safe and efficient protocols for stem cell purification and transplantation.

Key Points

  • The epigenetic state of a stem cell is tightly linked with cellular fate.

  • Ageing leads to changes on the epigenetic level and thus, alters stem cell potential and fate.

  • Targeting the ageing epigenome might represent a potent strategy to re-establish stem cell potency in the elderly and to extend health-span.

Acknowledgements

We thank members of the Tessarz lab for discussions around this topic. Figures were drawn using Biorender.

Andromachi Pouikli is a graduate student at the MPI for Biology of Ageing. She is interested in understanding the connection between metabolism and epigenetics during stem cell ageing.

Peter Tessarz is a group leader at the MPI for Biology of Ageing. His main interest lies in understanding how chromatin-mediated regulation of transcription is impacted by ageing.

Funding

This work was funded by the Max Planck Society and the Onassis Foundation–Scholarship ID: F ZP 047-1/2019-2020 (to A.P).

References

  • 1. Greenberg  MVC, Bourc’his  D. The diverse roles of DNA methylation in mammalian development and disease. Nat Rev Mol Cell Biol  2019;20:590–607. [DOI] [PubMed] [Google Scholar]
  • 2. Tessarz  P, Kouzarides  T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol  2014;15:703–708. [DOI] [PubMed] [Google Scholar]
  • 3. Perino  M, Veenstra  GJC. Chromatin control of developmental dynamics and plasticity. Dev Cell  2016;38:610–620. [DOI] [PubMed] [Google Scholar]
  • 4. Zhao  Z, Shilatifard  A. Epigenetic modifications of histones in cancer. Genome Biol  2019;20:245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Booth  LN, Brunet  A. The aging epigenome. Mol Cell  2016;62:728–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Avery  OT, MacLeod  CM, McCarty  M. Studies on the chemical nature of the substance inducing transformation of pneumococcal types: induction of transformation by a desoxyribonucleic acid fraction isolated from pneumococcus type III. J Exp Med  1944;79:137–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. McCarty  M, Avery  OT. Studies on the chemical nature of the substance inducing transformation of pneumococcal types: II. Effect of desoxyribonuclease on the biological activity of the transforming substance. J Exp Med  1946;83:89–96. [PubMed] [Google Scholar]
  • 8. Jabbari  K, Bernardi  G. Cytosine methylation and CpG, TpG (CpA) and TpA frequencies. Gene  2004;333:143–149. [DOI] [PubMed] [Google Scholar]
  • 9. Parry  A, Rulands  S, Reik  W. Active turnover of DNA methylation during cell fate decisions. Nat Rev Genet  2020. doi: 10.1038/s41576-020-00287-8. [DOI] [PubMed] [Google Scholar]
  • 10. Berdyshev  GD, Korotaev  GK, Boiarskikh  GV, et al.  Nucleotide composition of DNA and RNA from somatic tissues of humpback and its changes during spawning. Biokhimiia  1967;32:988–993. [PubMed] [Google Scholar]
  • 11. Vanyushin  BF, Nemirovsky  LE, Klimenko  VV, et al.  The 5-methylcytosine in DNA of rats. Gerontology  1973;19:138–152. [PubMed] [Google Scholar]
  • 12. Wilson  VL, Smith  RA, Ma  S, et al.  Genomic 5-methyldeoxycytidine decreases with age. J Biol Chem  1987;262:9948–9951. [PubMed] [Google Scholar]
  • 13. Maegawa  S, Hinkal  G, Kim  HS, et al.  Widespread and tissue specific age-related DNA methylation changes in mice. Genome Res  2010;20:332–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Horvath  S. DNA methylation age of human tissues and cell types. Genome Biol  2013;14:R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Haithcock  E, Dayani  Y, Neufeld  E, et al.  Age-related changes of nuclear architecture in Caenorhabditis elegans. Proc Natl Acad Sci U S A  2005;102:16690–16695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Larson  K, Yan  S-J, Tsurumi  A, et al.  Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Genet  2012;8:e1002473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. 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;348:1160–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Benayoun  BA, Pollina  EA, Brunet  A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat Rev Mol Cell Biol  2015;16:593–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Maleszewska  M, Mawer  JSP, Tessarz  P. Histone modifications in ageing and lifespan regulation. Current Molecular Biology  2016;2:26–35. [Google Scholar]
  • 20. Greer  EL, Maures  TJ, Hauswirth  AG, et al.  Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature  2010;466:383–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sen  P, Dang  W, Donahue  G, et al.  H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes Dev  2015;29:1362–1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pu  M, Ni  Z, Wang  M, et al.  Trimethylation of Lys36 on H3 restricts gene expression change during aging and impacts life span. Genes Dev  2015;29:718–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ocampo  A, Reddy  P, Martinez-Redondo  P, et al.  In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell  2016;167:1719–1733.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Hainer  SJ, Bošković  A, McCannell  KN, et al.  Profiling of pluripotency factors in single cells and early embryos. Cell  2019;177:1319–1329.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kaya-Okur  HS, Wu  SJ, Codomo  CA, et al.  CUT&tag for efficient epigenomic profiling of small samples and single cells. Nat Commun  2019;10:1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Weissman  IL, Anderson  DJ, Gage  F. Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiations. Annu Rev Cell Dev Biol  2001;17:387–403. [DOI] [PubMed] [Google Scholar]
  • 27. Seita  J, Weissman  IL. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip Rev Syst Biol Med  2010;2:640–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Bernstein  BE, Mikkelsen  TS, Xie  X, et al.  A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell  2006;125:315–326. [DOI] [PubMed] [Google Scholar]
  • 29. Shiraki  N, Shiraki  Y, Tsuyama  T, et al.  Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab  2014;19:780–794. [DOI] [PubMed] [Google Scholar]
  • 30. Carey  BW, Finley  LWS, Cross  JR, et al.  Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature  2015;518:413–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Lee  J, Kang  S, Lilja  KC, et al.  Signalling couples hair follicle stem cell quiescence with reduced histone H3 K4/K9/K27me3 for proper tissue homeostasis. Nat Commun  2016;7:11278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Singh  RP, Shiue  K, Schomberg  D, et al.  Cellular epigenetic modifications of neural stem cell differentiation. Cell Transplant  2009;18:1197–1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Challen  GA, Sun  D, Mayle  A, et al.  Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells. Cell Stem Cell  2014;15:350–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Mayle  A, Yang  L, Rodriguez  B, et al.  Dnmt3a loss predisposes murine hematopoietic stem cells to malignant transformation. Blood  2015;125:629–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Boonsanay  V, Zhang  T, Georgieva  A, et al.  Regulation of skeletal muscle stem cell quiescence by Suv4-20h1-dependent facultative heterochromatin formation. Cell Stem Cell  2016;18:229–242. [DOI] [PubMed] [Google Scholar]
  • 36. Kim  R, Sheaffer  KL, Choi  I, et al.  Epigenetic regulation of intestinal stem cells by Tet1-mediated DNA hydroxymethylation. Genes Dev  2016;30:2433–2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Li  Z, Liu  C, Xie  Z, et al.  Epigenetic dysregulation in mesenchymal stem cell aging and spontaneous differentiation. PLoS One  2011;6:e20526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Ryall  JG, Dell’Orso  S, Derfoul  A, et al.  The NAD(+)-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell  2015;16:171–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Zhou  Q, Dalgard  CL, Wynder  C, et al.  Histone deacetylase inhibitors SAHA and sodium butyrate block G1-to-S cell cycle progression in neurosphere formation by adult subventricular cells. BMC Neurosci  2011;12:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Hsieh  J, Nakashima  K, Kuwabara  T, et al.  Histone deacetylase inhibition-mediated neuronal differentiation of multipotent adult neural progenitor cells. Proc Natl Acad Sci U S A  2004;101:16659–16664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Cheng  C-W, Biton  M, Haber  AL, et al.  Ketone body Signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell  2019;178:1115–1131.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Gebert  N, Cheng  C-W, Kirkpatrick  JM, et al.  Region-specific proteome changes of the intestinal epithelium during aging and dietary restriction. Cell Rep  2020;31:107565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Kohyama  J, Abe  H, Shimazaki  T, et al.  Brain from bone: efficient “meta-differentiation” of marrow stroma-derived mature osteoblasts to neurons with noggin or a demethylating agent. Differentiation  2001;68:235–244. [DOI] [PubMed] [Google Scholar]
  • 44. Woodbury  D, Reynolds  K, Black  IB. Adult bone marrow stromal stem cells express germline, ectodermal, endodermal, and mesodermal genes prior to neurogenesis. J Neurosci Res  2002;69:908–917. [DOI] [PubMed] [Google Scholar]
  • 45. Lu  C, Thompson  CB. Metabolic regulation of epigenetics. Cell Metab  2012;16:9–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Kaelin  WG, Jr, McKnight  SL. Influence of metabolism on epigenetics and disease. Cell  2013;153:56–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Etchegaray  J-P, Mostoslavsky  R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes. Mol Cell  2016;62:695–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Reid  MA, Dai  Z, Locasale  JW. The impact of cellular metabolism on chromatin dynamics and epigenetics. Nat Cell Biol  2017;19:1298–1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. López-Otín  C, Blasco  MA, Partridge  L, et al.  The hallmarks of aging. Cell  2013;153:1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Goodell  MA, Rando  TA. Stem cells and healthy aging. Science  2015;350:1199–1204. [DOI] [PubMed] [Google Scholar]
  • 51. Wang  H, Diao  D, Shi  Z, et al.  SIRT6 controls hematopoietic stem cell homeostasis through epigenetic regulation of Wnt Signaling. Cell Stem Cell  2016;18:495–507. [DOI] [PubMed] [Google Scholar]
  • 52. Li  G, Tang  X, Zhang  S, et al.  SIRT7 activates quiescent hair follicle stem cells to ensure hair growth in mice. EMBO J  2020;39:e104365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Ren  R, Ocampo  A, Liu  G-H, et al.  Regulation of stem cell aging by metabolism and epigenetics. Cell Metab  2017;26:460–474. [DOI] [PubMed] [Google Scholar]
  • 54. Buisman  SC, de  Haan  G. Epigenetic changes as a target in aging haematopoietic stem cells and age-related malignancies. Cell  2019;8. doi: 10.3390/cells8080868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Cakouros  D, Gronthos  S. Epigenetic regulation of bone marrow stem cell aging: revealing epigenetic signatures associated with hematopoietic and mesenchymal stem cell aging. Aging Dis  2019;10:174–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Bröske  A-M, Vockentanz  L, Kharazi  S, et al.  DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet  2009;41:1207–1215. [DOI] [PubMed] [Google Scholar]
  • 57. Trowbridge  JJ, Snow  JW, Kim  J, et al.  DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell  2009;5:442–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Liu  X, Jia  X, Yuan  H, et al.  DNA methyltransferase 1 functions through C/ebpa to maintain hematopoietic stem and progenitor cells in zebrafish. J Hematol Oncol  2015;8:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Cimmino  L, Dawlaty  MM, Ndiaye-Lobry  D, et al.  Erratum: TET1 is a tumor suppressor of hematopoietic malignancy. Nat Immunol  2015;16:889. [DOI] [PubMed] [Google Scholar]
  • 60. Jung  J-W, Lee  S, Seo  M-S, et al.  Histone deacetylase controls adult stem cell aging by balancing the expression of polycomb genes and jumonji domain containing 3. Cell Mol Life Sci  2010;67:1165–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Pan  H, Guan  D, Liu  X, et al.  SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res  2016;26:190–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Bigot  A, Duddy  WJ, Ouandaogo  ZG, et al.  Age-associated methylation suppresses SPRY1, leading to a failure of re-quiescence and loss of the reserve stem cell pool in elderly muscle. Cell Rep  2015;13:1172–1182. [DOI] [PubMed] [Google Scholar]
  • 63. Wu  H, Coskun  V, Tao  J, et al.  Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science  2010;329:444–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Rafalski  VA, Ho  PP, Brett  JO, et al.  Expansion of oligodendrocyte progenitor cells following SIRT1 inactivation in the adult brain. Nat Cell Biol  2013;15:614–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Sun  D, Luo  M, Jeong  M, et al.  Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell  2014;14:673–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Adelman  ER, Huang  H-T, Roisman  A, et al.  Aging human hematopoietic stem cells manifest profound epigenetic reprogramming of enhancers that may predispose to Leukemia. Cancer Discov  2019;9:1080–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Florian  MC, Dörr  K, Niebel  A, et al.  Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell  2012;10:520–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Grigoryan  A, Guidi  N, Senger  K, et al.  LaminA/C regulates epigenetic and chromatin architecture changes upon aging of hematopoietic stem cells. Genome Biol  2018;19:189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Pouikli  A, Parekh  S, Maleszewska  M, et al.  Deregulated Mito-nuclear communication alters chromatin plasticity and differentiation 2 potential of mesenchymal stem cells upon ageing. bioRxiv  2020. doi: 10.1101/2020.04.02.022293. [DOI] [Google Scholar]
  • 70. Morganti  C, Bonora  M, Marchi  S, et al.  Citrate mediates crosstalk between mitochondria and the nucleus to promote human mesenchymal stem cell in vitro osteogenesis. Cell  2020;9. doi: 10.3390/cells9041034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Liu  L, Cheung  TH, Charville  GW, et al.  Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep  2013;4:189–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Vazquez  BN, Thackray  JK, Simonet  NG, et al.  SIRT7 promotes genome integrity and modulates non-homologous end joining DNA repair. EMBO J  2016;35:1488–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Dai  Z, Ramesh  V, Locasale  JW. The evolving metabolic landscape of chromatin biology and epigenetics. Nat Rev Genet  2020;21:737–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Beerman  I, Bock  C, Garrison  BS, et al.  Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell  2013;12:413–425. [DOI] [PubMed] [Google Scholar]
  • 75. Hernando-Herraez  I, Evano  B, Stubbs  T, et al.  Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat Commun  2019;10:4361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Lewis  S, Nachun  D, Martin  MG, et al.  DNA methylation analysis validates organoids as a viable model for studying human intestinal aging. Cell Mol Gastroenterol Hepatol  2019. doi: 10.1016/j.jcmgh.2019.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Benayoun  BA, Pollina  EA, Ucar  D, et al.  H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell  2014;158:673–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Cosgrove  BD, Gilbert  PM, Porpiglia  E, et al.  Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med  2014;20:255–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Yan  X, Ehnert  S, Culmes  M, et al.  5-azacytidine improves the osteogenic differentiation potential of aged human adipose-derived mesenchymal stem cells by DNA demethylation. PLoS One  2014;9:e90846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Kornicka  K, Marycz  K, Marędziak  M, et al.  The effects of the DNA methyltranfserases inhibitor 5-Azacitidine on ageing, oxidative stress and DNA methylation of adipose derived stem cells. J Cell Mol Med  2017;21:387–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Wang  Y, Deng  P, Liu  Y, et al.  Alpha-ketoglutarate ameliorates age-related osteoporosis via regulating histone methylations. Nat Commun  2020;11:5596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Cheng  Y, He  C, Wang  M, et al.  Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther  2019;4:62. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Briefings in Functional Genomics are provided here courtesy of Oxford University Press

RESOURCES