Elsevier

Gene

Volume 527, Issue 1, 15 September 2013, Pages 10-25
Gene

Review
SIR–nucleosome interactions: Structure–function relationships in yeast silent chromatin

https://doi.org/10.1016/j.gene.2013.05.088Get rights and content

Highlights

  • Histone PTMs regulate chromatin structure and assembly of yeast silent chromatin

  • Sir2-dependent turnover of H4K16ac actively reinforces assembly of silent chromatin

  • Protein contacts within and between adjacent SIR complexes are crucial to silencing

  • Variegation and regulation of subtelomeric silencing respond to environmental changes

Abstract

Discrete regions of the eukaryotic genome assume a heritable chromatin structure that is refractory to gene expression, referred to as heterochromatin or “silent” chromatin. Constitutively silent chromatin is found in subtelomeric domains in a number of species, ranging from yeast to man. In addition, chromatin-dependent repression of mating type loci occurs in both budding and fission yeasts, to enable sexual reproduction. The silencing of chromatin in budding yeast is characterized by an assembly of Silent Information Regulatory (SIR) proteins—Sir2, Sir3 and Sir4—with unmodified nucleosomes. Silencing requires the lysine deacetylase activity of Sir2, extensive contacts between Sir3 and the nucleosome, as well as interactions among the SIR proteins, to generate the Sir2–3–4 or SIR complex. Results from recent structural and reconstitution studies suggest an updated model for the ordered assembly and organization of SIR-dependent silent chromatin in yeast. Moreover, studies of subtelomeric gene expression reveal the importance of subtelomeric silent chromatin in the regulation of genes other than the silent mating type loci. This review covers recent advances in this field.

Introduction

The packaging of DNA into nucleosomes and the interaction of the nucleosomal fiber with transcription modulators, regulate promoter accessibility and gene expression. Recent structure–function studies have yielded insights into the molecular details of gene silencing, and in no organism is this more advanced than in Saccharomyces cerevisiae. Here we focus in particular on the yeast-specific Silent Information Regulator (SIR) complex and its contacts with nucleosomes, which control transcription both at silent mating type loci and in subtelomeric domains.

Section snippets

Structural aspects of the chromatin fiber

Silent chromatin reflects an integrated assembly of non-histone and histone proteins into a chromatin structure that represses promoter activity independent of the specific promoter sequence. To understand gene silencing it is first necessary to describe the structure of the nucleosomal core particle (NCP), which lies at the core of silent chromatin. A canonical NCP is composed of 147 base pairs (bp) of DNA wrapped around a histone octamer in 1.65 superhelical turns. The highly basic core

Chromatin regulates gene expression

Gene expression results from both the binding of transcription factors to regulatory sequences and changes in nucleosomal organization in these control regions. This latter involves the post-translational modification (PTM) of histone tails and core residues, the exchange of histone variants, and the shifting and eviction of nucleosomal themselves. Here we summarize evidence showing that nucleosomes and their positioning information alone can influence transcriptional efficiency, before we

Silent chromatin in S. cerevisiae

Silent chromatin in yeast occurs at the subtelomeric regions (Gottschling et al., 1990) and at silent homothallic mating-type loci (HM), HML and HMR (Rine and Herskowitz, 1987). It requires the loading onto the chromatin fiber of the Silent Information Regulator (SIR) proteins Sir2, Sir3 and Sir4 (Fig. 2). These three proteins form a heterotrimeric Sir2–3–4 complex—the SIR complex—which together with histones, constitutes the essential silencing machinery in budding yeast (Cubizolles et al.,

Outlook

It is clear that the combination of genetics, biochemistry and structural work has put the SIR system of gene repression at the forefront of model systems for the study of chromatin-mediated gene repression. It remains to determine the structure of the SIR-nucleosomal complex and to define the transition from “SIR binding” to “silencing” in molecular terms. Indeed, the precise step in the transcription process that is impaired by the SIR complex is still debated. Nonetheless, a wealth of

Conflict of interest

The authors declare that they have no conflict of interest.

Acknowledgments

The Gasser laboratory is funded by the Swiss National Science Foundation, the NCCR Frontiers in Genetics, the SystemsX RTD CINA, various EU FP7 programs, and the Novartis Research Foundation. We thank colleagues in the laboratory for critical reading of this review and for a stimulating environment. We thank David Shore, and Nicolas Thomae for comments on earlier versions of the text and Laurent Rueff for artistic help. SK also thanks EMBO and FWF for support.

References (285)

  • J.F. Chang et al.

    Structure of the coiled-coil dimerization motif of Sir4 and its interaction with Sir3

    Structure

    (2003)
  • L. Chen et al.

    Mechanism of transcriptional silencing in yeast

    Cell

    (2005)
  • K. Chen

    Stabilization of the promoter nucleosomes in nucleosome-free regions by the yeast Cyc8-Tup1 corepressor

    Genome Res.

    (2013)
  • C.C. Chou et al.

    Bypassing Sir2 and O-acetyl-ADP-ribose in transcriptional silencing

    Mol. Cell

    (2008)
  • M.N. Conrad et al.

    RAP1 protein interacts with yeast telomeres in vivo: overproduction alters telomere structure and decreases chromosome stability

    Cell

    (1990)
  • F. Cubizolles et al.

    A homotrimer–heterotrimer switch in Sir2 structure differentiates rDNA and telomeric silencing

    Mol. Cell

    (2006)
  • C.A. Davey et al.

    Solvent mediated interactions in the structure of the nucleosome core particle at 1.9 A resolution

    J. Mol. Biol.

    (2002)
  • H. Dehghani et al.

    Organization of chromatin in the interphase mammalian cell

    Micron

    (2005)
  • B. Dorigo et al.

    Chromatin fiber folding: requirement for the histone H4 N-terminal tail

    J. Mol. Biol.

    (2003)
  • E.A. Feeser et al.

    Structural and functional studies of the Rap1 C-terminus reveal novel separation-of-function mutants

    J. Mol. Biol.

    (2008)
  • Q. Feng

    Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain

    Curr. Biol.

    (2002)
  • E. Fussner et al.

    Living without 30 nm chromatin fibers

    Trends Biochem. Sci.

    (2011)
  • M.R. Gartenberg et al.

    Sir-mediated repression can occur independently of chromosomal and subnuclear contexts

    Cell

    (2004)
  • S.M. Gasser et al.

    The molecular biology of the SIR proteins

    Gene

    (2001)
  • S.M. Gasser et al.

    A glimpse at chromosomal order

    Trends Genet.

    (1987)
  • E. Gilson et al.

    Distortion of the DNA double helix by RAP1 at silencers and multiple telomeric binding sites

    J. Mol. Biol.

    (1993)
  • J.M. Gottesfeld et al.

    Nonrandom alignment of nucleosomes on 5S RNA genes of X. laevis

    Cell

    (1980)
  • D.E. Gottschling et al.

    Position effect at S. cerevisiae telomeres: reversible repression of Pol II transcription

    Cell

    (1990)
  • S.A. Grigoryev et al.

    Chromatin organization—the 30 nm fiber

    Exp. Cell Res.

    (2012)
  • A. Halme et al.

    Genetic and epigenetic regulation of the FLO gene family generates cell-surface variation in yeast

    Cell

    (2004)
  • J. Han et al.

    Acetylation of lysine 56 of histone H3 catalyzed by RTT109 and regulated by ASF1 is required for replisome integrity

    J. Biol. Chem.

    (2007)
  • N. Abramova et al.

    Reciprocal regulation of anaerobic and aerobic cell wall mannoprotein gene expression in Saccharomyces cerevisiae

    J. Bacteriol.

    (2001)
  • V.G. Allfrey et al.

    Acetylation and methylation of histones and their possible role in the regulation of rna synthesis

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

    (1964)
  • A.J. Andrews et al.

    Nucleosome structure(s) and stability: variations on a theme

    Annu. Rev. Biophys.

    (2011)
  • E.D. Andrulis et al.

    Perinuclear localization of chromatin facilitates transcriptional silencing

    Nature

    (1998)
  • E.D. Andrulis

    Esc1, a nuclear periphery protein required for Sir4-based plasmid anchoring and partitioning

    Mol. Cell. Biol.

    (2002)
  • K.J. Armache et al.

    Structural basis of silencing: Sir3 BAH domain in complex with a nucleosome at 3.0 A resolution

    Science

    (2011)
  • A.J. Bannister et al.

    Regulation of chromatin by histone modifications

    Cell Res.

    (2011)
  • C. Beisel et al.

    Silencing chromatin: comparing modes and mechanisms

    Nat. Rev. Genet.

    (2011)
  • S.P. Bell et al.

    Yeast origin recognition complex functions in transcription silencing and DNA replication

    Science

    (1993)
  • O. Bell et al.

    Determinants and dynamics of genome accessibility

    Nat. Rev. Genet.

    (2011)
  • A.S. Belmont et al.

    Visualization of G1 chromosomes: a folded, twisted, supercoiled chromonema model of interphase chromatid structure

    J. Cell Biol.

    (1994)
  • S.J. Boulton et al.

    Components of the Ku-dependent non-homologous end-joining pathway are involved in telomeric length maintenance and telomeric silencing

    EMBO J.

    (1998)
  • M. Braunstein et al.

    Transcriptional silencing in yeast is associated with reduced nucleosome acetylation

    Genes Dev.

    (1993)
  • K. Brogaard et al.

    A map of nucleosome positions in yeast at base-pair resolution

    Nature

    (2012)
  • A. Bucceri et al.

    Rapid accessibility of nucleosomal DNA in yeast on a second time scale

    EMBO J.

    (2006)
  • J.R. Buchberger

    Sir3–nucleosome interactions in spreading of silent chromatin in Saccharomyces cerevisiae

    Mol. Cell. Biol.

    (2008)
  • A.R. Buchman et al.

    Two DNA-binding factors recognize specific sequences at silencers, upstream activating sequences, autonomously replicating sequences, and telomeres in Saccharomyces cerevisiae

    Mol. Cell. Biol.

    (1988)
  • S.W. Buck et al.

    Action of a RAP1 carboxy-terminal silencing domain reveals an underlying competition between HMR and telomeres in yeast

    Genes Dev.

    (1995)
  • L.H. Caro et al.

    In silicio identification of glycosyl-phosphatidylinositol-anchored plasma-membrane and cell wall proteins of Saccharomyces cerevisiae

    Yeast

    (1997)
  • Cited by (32)

    • Breakers and amplifiers in chromatin circuitry: acetylation and ubiquitination control the heterochromatin machinery

      2021, Current Opinion in Structural Biology
      Citation Excerpt :

      H2Bub has also been implicated in heterochromatin protection in S. pombe, where acetylation of the H2B ubiquitin ligase subunit Brl1 by Mst2 regulates H2Bub levels and protects euchromatic genes from heterochromatic silencing by small RNAs [51]. H4K16ac in S. cerevisiae is a classic example of ‘anti-silencing’, which corrals heterochromatin factors into silent regions of the genome to protect active gene transcription from silencing [52]. Even though S. cerevisiae and S. pombe use very different heterochromatin machinery, the functional parallels between scH4K16ac and spH3K14ac are becoming apparent and point to a critical role of specific acetyl marks as repellents in the protection of euchromatin.

    • Stabilization of Sir3 interactions by an epigenetic metabolic small molecule, O-acetyl-ADP-ribose, on yeast SIR-nucleosome silent heterochromatin

      2019, Archives of Biochemistry and Biophysics
      Citation Excerpt :

      The silent heterochromatin of the budding yeast Saccharomyces cerevisiae provides an excellent system for epigenetic studies. In vivo genetics and in vitro biochemical studies have demonstrated that the silent information regulator (Sir) proteins, Sir2, Sir3, and Sir4, are required to establish and maintain silent heterochromatin domains at the telomeres and the mating type loci [8–15]. The Sir proteins form the SIR complex that is capable of nucleosome binding and is important for silent heterochromatin assembly [16–19].

    • Recruitment and allosteric stimulation of a histone-deubiquitinating enzyme during heterochromatin assembly

      2018, Journal of Biological Chemistry
      Citation Excerpt :

      Three general steps in establishing heterochromatin involve the following: 1) recruitment of the silencing complex to the target locus; 2) nucleation via histone-modifying activities; and 3) spreading of the silencing complexes due to iterative cycles of recruitment and nucleation. In budding yeast, silent information regulator (SIR) complex–dependent heterochromatin is a well-studied and simplified silencing model (13–15). The SIR complex is composed of three proteins, Sir2, Sir3, and Sir4.

    • Human sirtuins: Structures and flexibility

      2016, Journal of Structural Biology
      Citation Excerpt :

      In 1987, Rine et al. identified four proteins responsible for the silencing of the Hidden MAT loci of Saccharomyces cerevisiae, implicated in the mating process of the yeast (Rine and Herskowitz, 1987). These four proteins, called silent information regulatory (Sir) proteins, have been found to bind to the nucleosome through interactions with histone proteins H3 and H4, and to repress gene expression (Johnson et al., 1990; Murphy et al., 2013; Cubizolles et al., 2006; Ehrentraut et al., 2011; Oppikofer et al., 2013). Sir proteins have aroused great hope when it was found that they may participate in extending the yeast life span (Lin et al., 2004).

    • The Dynamics of Histone Modifications During Aging

      2016, Epigenomics in Health and Disease
    • Stabilization of mini-chromosome segregation during mitotic growth by overexpression of YCR041W and its application to chromosome engineering inSaccharomyces cerevisiae

      2015, Journal of Bioscience and Bioengineering
      Citation Excerpt :

      Because the stabilizing effect was not observed in the absence of Sir4, however, it seems likely that segregation processes related to telomere function are involved in the stabilization of mini-chromosomes by YCR041W and ZDS1. In the budding yeast, the Sir complex has two physiological roles at the telomere: (i) formation of heterochromatin and silencing (29), and (ii) anchoring telomeres to the nuclear envelope and telomere clustering, thereby stabilizing the chromosome (30). Among the Sir proteins, Sir2 is involved only in heterochromatin formation, whereas Sir3 and Sir4 are involved in both heterochromatin formation and telomere anchoring and clustering (30–32).

    View all citing articles on Scopus
    1

    Current address: Department of Early Discovery Biochemistry, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA 94080, USA.

    2

    Current address: F. Hoffmann-La Roche Ltd. Pharmaceuticals Division, Grenzacherstrasse 124, 4070 Basel, Switzerland.

    View full text