Elsevier

Experimental Cell Research

Volume 314, Issue 9, 15 May 2008, Pages 1909-1917
Experimental Cell Research

Review
Aging by epigenetics—A consequence of chromatin damage?

https://doi.org/10.1016/j.yexcr.2008.02.023Get rights and content

Abstract

Chromatin structure is not fixed. Instead, chromatin is dynamic and is subject to extensive developmental and age-associated remodeling. In some cases, this remodeling appears to counter the aging and age-associated diseases, such as cancer, and extend organismal lifespan. However, stochastic non-deterministic changes in chromatin structure might, over time, also contribute to the break down of nuclear, cell and tissue function, and consequently aging and age-associated diseases.

Introduction

As reviewed in this volume and elsewhere, organismal longevity and aging may be influenced by many complex interacting factors [1]. These include accumulation of nuclear and mitochondrial genome mutations; shortened and dysfunctional chromosome telomeres; oxidative damage of DNA and other macromolecules; systemic hormonal factors, such as insulin/IGF signaling; and senescence, apoptosis and altered differentiation in self-renewing tissues that depend on stem cells. In many cases, these factors are, or are likely to be, overlapping and/or interacting. In this chapter, we discuss another possible determinant of aging, “epigenetics”.

Strictly speaking, “epigenetics” refers to chromatin and DNA modifications that are heritable through cell division, but do not involve changes in the underlying DNA sequence [2]. However, since the aging process is not restricted to proliferating tissues and may, in fact, be influenced by irreversible proliferation arrest, we will use the term more loosely to refer to effects mediated by chromatin structure and DNA methylation, but not DNA sequence.

Section snippets

The dynamic and stochastic properties of chromatin

The basic repeating unit of chromatin structure is the nucleosome, comprised of about 146 bp of DNA wrapped around an octamer of histones [3]. Chromatin can be broadly divided into two types, heterochromatin and euchromatin [4]. Euchromatin is decondensed during interphase, relatively transcriptionally active and replicates early in S-phase. Conversely, heterochromatin remains condensed during interphase, is more transcriptionally silent and is late replicating. Heterochromatin is itself

Aging is associated with epigenetic changes, from yeast to humans

The enzymes that add and remove acetyl groups, the histone acetyltransferases (HATs) and histone deacetylases (HDACs) respectively, determine the steady-state level of histone acetylation [10]. In Saccharomyces cerevisiae, inactivation of the histone deacetylase, Sir2, shortens replicative lifespan [20], [21], [22]. Conversely, activation of Sir2 extends lifespan. The anti-aging effects of Sir2 in yeast are, at least in part, due to translocation of a Sir2-containing protein complex from

Cellular senescence is associated with redistribution of heterochromatin

Cellular senescence is characterized by an irreversible proliferation arrest [60]. Cellular senescence can be initiated by various triggers, including an excessive number of cell divisions, in part due to shortened chromosome telomeres (so-called replicative senescence). Because of senescence, most primary human cells have a finite proliferative lifespan, and evidence has been presented that senescence contributes to tissue aging in vivo, in part by limiting the self-renewal of tissues [61],

Progeria cells exhibit accelerated changes in chromatin

Hutchinson–Gilford Progeria Syndrome (HGPS) is a rare syndrome that is characterized by rapid onset of some debilitating phenotypes that are typically indicative of aging [95]. These include severe growth retardation, loss of subcutaneous fat, alopecia, reduced bone density and poor muscle development. The average age of death in HGPS is 12–15 years, usually by myocardial infarction or stroke. However, HGPS patients do not accelerate all aspects of aging. For example, cancer incidence is rare

The consequences of age-associated epigenetic changes

Not surprisingly, the consequence of these age-associated epigenetic changes is best understood in yeast. As discussed above, in this organism redistribution of Sir2 and heterochromatic proteins counteracts the aging process [21], [22]. The consequence of the various modes of age-associated epigenetic regulation in mammals remains somewhat speculative. However, aging is accompanied by various altered phenotypes that might be linked to age-associated epigenetic changes. Studies going back to the

Summary, outstanding questions and therapeutic implications

From the preceding discussion, it is apparent that chromatin structure does change with aging, in organisms as diverse as yeast and mammals. However, with the exception of Sir2 in yeast, the extent to which this impacts the aging process has not yet been defined. Although it is early days for this field, we propose that some age-associated epigenetic alterations in mammals, such as formation of SAHF, might extend lifespan by suppressing age-associated diseases, such as cancer. However, as

Acknowledgments

PDA is supported by the NIH (RO1 GM062281, P01 AG031862 and R01 CA129334-01) and is a scholar of the Leukemia and Lymphoma Society. JMS is supported by the NIH (R01 GM041690 and R01 AG016694), and a Senior Scholar Award from the Ellison Medical Foundation. PDA thanks members of the Adams lab as well as Shelley Berger, Ronen Marmorstein and Brad Johnson for discussions.

References (118)

  • B.K. Kennedy et al.

    Redistribution of silencing proteins from telomeres to the nucleolus is associated with extension of life span in S. cerevisiae

    Cell

    (1997)
  • V.L. Wilson et al.

    Genomic 5-methyldeoxycytidine decreases with age

    J. Biol. Chem.

    (1987)
  • R.P. Singhal et al.

    DNA methylation in aging of mice

    Mech. Ageing Dev.

    (1987)
  • G.A. Romanov et al.

    Methylation of reiterated sequences in mammalian DNAs. Effects of the tissue type, age, malignancy and hormonal induction

    Biochim. Biophys. Acta

    (1981)
  • J.Y. Kim et al.

    Age-related human small intestine methylation: evidence for stem cell niches

    BMC Med.

    (2005)
  • B. Sarg et al.

    Postsynthetic trimethylation of histone H4 at lysine 20 in mammalian tissues is associated with aging

    J. Biol. Chem.

    (2002)
  • S.E. Polo et al.

    Chromatin assembly: a basic recipe with various flavours

    Curr. Opin. Genet. Dev.

    (2006)
  • J.C. Jeyapalan et al.

    Accumulation of senescent cells in mitotic tissue of aging primates

    Mech. Ageing Dev.

    (2007)
  • H. Tagami et al.

    Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis

    Cell

    (2004)
  • D. Dimova et al.

    A role for transcriptional repressors in targeting the yeast Swi/Snf complex

    Mol. Cell

    (1999)
  • J.A. Sharp et al.

    Yeast histone deposition protein Asf1p requires Hir proteins and PCNA for heterochromatic silencing

    Curr. Biol.

    (2001)
  • B. Villeponteau

    The heterochromatin loss model of aging

    Exp. Geronto.

    (1997)
  • M. Collado et al.

    Cellular senescence in cancer and aging

    Cell

    (2007)
  • M.K. Francis et al.

    Loss of EPC-1/PEDF expression during skin aging in vivo

    J. Invest. Dermatol.

    (2004)
  • M.A. Blasco et al.

    Telomere shortening and tumor formation by mouse cells lacking telomerase RNA

    Cell

    (1997)
  • K.L. Rudolph et al.

    Longevity, stress response, and cancer in aging telomerase-deficient mice

    Cell

    (1999)
  • D.M. Feldser et al.

    Short telomeres limit tumor progression in vivo by inducing senescence

    Cancer Cell

    (2007)
  • P. Sun et al.

    PRAK is essential for ras-induced senescence and tumor suppression

    Cell

    (2007)
  • M. Narita et al.

    Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence

    Cell

    (2003)
  • R. Zhang et al.

    Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA

    Dev. Cell

    (2005)
  • X. Ye et al.

    Downregulation of Wnt signaling is a trigger for formation of facultative heterochromatin and onset of cell senescence in primary human cells

    Mol. Cell

    (2007)
  • K. Luger et al.

    Crystal structure of the nucleosome core particle at 2.8 A resolution

    Nature

    (1997)
  • S.I. Grewal et al.

    Heterochromatin revisited

    Nat. Rev. Genet.

    (2007)
  • H.J. Muller

    Types of visible variations induced by X-rays in Drosophila

    J. Genet.

    (1930)
  • S.L. Berger

    The complex language of chromatin regulation during transcription

    Nature

    (2007)
  • R.D. Phair et al.

    High mobility of proteins in the mammalian cell nucleus

    Nature

    (2000)
  • T. Cheutin et al.

    Maintenance of stable heterochromatin domains by dynamic HP1 binding

    Science

    (2003)
  • T.S. Mikkelsen et al.

    Genome-wide maps of chromatin state in pluripotent and lineage-committed cells

    Nature

    (2007)
  • E. Birney et al.

    Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project

    Nature

    (2007)
  • S. Schoeftner et al.

    Developmentally regulated transcription of mammalian telomeres by DNA-dependent RNA polymerase II

    Nat. Cell Bio.

    (2008)
  • C.M. Azzalin et al.

    Telomeric repeat containing RNA and RNA surveillance factors at mammalian chromosome ends

    Science

    (2007)
  • M. Kaern et al.

    Stochasticity in gene expression: from theories to phenotypes

    Nat. Rev. Genet.

    (2005)
  • M.C. Haigis et al.

    Mammalian sirtuins — emerging roles in physiology, aging, and calorie restriction

    Genes Dev.

    (2006)
  • M. Sommer et al.

    DeltaNp63alpha overexpression induces downregulation of Sirt1 and an accelerated aging phenotype in the mouse

    Cell Cycle

    (2006)
  • T. Sasaki et al.

    Progressive loss of SIRT1 with cell cycle withdrawal

    Aging Cell

    (2006)
  • J.Y. Kim et al.

    Counting human somatic cell replications: methylation mirrors endometrial stem cell divisions

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • Y. Yatabe et al.

    Investigating stem cells in human colon by using methylation patterns

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • N. Ahuja et al.

    Aging and DNA methylation in colorectal mucosa and cancer

    Cancer Res.

    (1998)
  • J.P. Issa et al.

    Switch from monoallelic to biallelic human IGF2 promoter methylation during aging and carcinogenesis

    Proc. Natl. Acad. Sci. U. S. A.

    (1996)
  • J.P. Issa et al.

    Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon

    Nat. Genet.

    (1994)
  • Cited by (0)

    View full text