ArticlePDF Available

A dual role of H4K16 acetylation in the establishment of yeast silent chromatin

Authors:

Abstract and Figures

Discrete regions of the eukaryotic genome assume heritable chromatin structure that is refractory to transcription. In budding yeast, silent chromatin is characterized by the binding of the Silent Information Regulatory (Sir) proteins to unmodified nucleosomes. Using an in vitro reconstitution assay, which allows us to load Sir proteins onto arrays of regularly spaced nucleosomes, we have examined the impact of specific histone modifications on Sir protein binding and linker DNA accessibility. Two typical marks for active chromatin, H3K79(me) and H4K16(ac) decrease the affinity of Sir3 for chromatin, yet only H4K16(ac) affects chromatin structure, as measured by nuclease accessibility. Surprisingly, we found that the Sir2-4 subcomplex, unlike Sir3, has higher affinity for chromatin carrying H4K16(ac). NAD-dependent deacetylation of H4K16(ac) promotes binding of the SIR holocomplex but not of the Sir2-4 heterodimer. This function of H4K16(ac) cannot be substituted by H3K56(ac). We conclude that acetylated H4K16 has a dual role in silencing: it recruits Sir2-4 and repels Sir3. Moreover, the deacetylation of H4K16(ac) by Sir2 actively promotes the high-affinity binding of the SIR holocomplex.
Content may be subject to copyright.
A dual role of H4K16 acetylation in the
establishment of yeast silent chromatin
Mariano Oppikofer
1
, Stephanie Kueng
1
,
Fabrizio Martino
2
, Szabolcs Soeroes
3
,
Susan M Hancock
2
, Jason W Chin
2
,
Wolfgang Fischle
3
and Susan M Gasser
1,
*
1
Friedrich Miescher Institute for Biomedical Research, Basel,
Switzerland;
2
Medical Research Council Laboratory of Molecular
Biology, Cambridge, UK and
3
Max Planck Institute for Biophysical
Chemistry, Go
¨ttingen, Germany
Discrete regions of the eukaryotic genome assume herita-
ble chromatin structure that is refractory to transcription.
In budding yeast, silent chromatin is characterized by the
binding of the Silent Information Regulatory (Sir) proteins
to unmodified nucleosomes. Using an in vitro reconstitu-
tion assay, which allows us to load Sir proteins onto arrays
of regularly spaced nucleosomes, we have examined the
impact of specific histone modifications on Sir protein
binding and linker DNA accessibility. Two typical marks
for active chromatin, H3K79
me
and H4K16
ac
decrease the
affinity of Sir3 for chromatin, yet only H4K16
ac
affects
chromatin structure, as measured by nuclease accessibility.
Surprisingly, we found that the Sir2-4 subcomplex, unlike
Sir3, has higher affinity for chromatin carrying H4K16
ac
.
NAD-dependent deacetylation of H4K16
ac
promotes bind-
ing of the SIR holocomplex but not of the Sir2-4 hetero-
dimer. This function of H4K16
ac
cannot be substituted
by H3K56
ac
. We conclude that acetylated H4K16 has a
dual role in silencing: it recruits Sir2-4 and repels Sir3.
Moreover, the deacetylation of H4K16
ac
by Sir2 actively
promotes the high-affinity binding of the SIR holocomplex.
The EMBO Journal (2011) 30, 2610–2621. doi:10.1038/
emboj.2011.170; Published online 10 June 2011
Subject Categories: chromatin & transcription
Keywords: H4K16; histone deacetylation; methylation; silent
chromatin; SIR complex
Introduction
Heterochromatin is a heritable, specialized chromatin struc-
ture that silences discrete regions in eukaryotic genomes.
Among other features, gene silencing within heterochromatic
regions is thought to involve compaction of the chromatin
fibre in order to structurally limit DNA accessibility. In
budding yeast, silent chromatin requires the binding of
Silent Information Regulatory (Sir) proteins to unmodified
nucleosomes. Work of many laboratories has identified Sir2,
Sir3 and Sir4 proteins as the core components of silent
chromatin at telomeres and silent mating type loci (Rine
and Herskowitz, 1987; reviewed in Rusche et al (2003)).
Two-hybrid and protein binding studies suggested that they
form a complex, with Sir4 being a scaffold protein that
bridges between Sir2 and Sir3 (Moazed et al, 1997; Strahl-
Bolsinger et al, 1997; Rudner et al, 2005; Cubizolles et al,
2006). Although initial attempts to purify the Sir proteins
from yeast yielded only an Sir2-4 heterodimer (Ghidelli et al,
2001; Hoppe et al, 2002), a stable Sir2-Sir3-Sir4 heterotrimer
with 1:1:1 stoichiometry (hereafter SIR complex) was purified
from insect cells (Cubizolles et al, 2006). A fourth Sir protein,
Sir1, is important for the establishment of silencing at the
silent mating type loci, but is not required for repression at
telomeres (Pillus and Rine, 1989; Aparicio et al, 1991).
Sir proteins do not bind DNA in a sequence-specific
manner, yet zones of silencing are restricted to specific
domains in the yeast genome. To achieve targeted silencing,
Sir proteins are recruited by bifunctional DNA binding
factors, such as Rap1, Abf1 and Orc1, which bind yeast
silencer elements. The SIR complex then spreads from this
nucleation site for 3–20 kb, depe nding on the abundance and
balance of available Sir proteins (reviewed in Gasser and
Cockell (2001) and Rusche et al (2003)). SIR complex asso-
ciation decreases the ability of enzymes, like DNA methylases
or endonucleases, to access the DNA (Gottschling, 1992; Loo
and Rine, 1994). Transcription in these regions is repressed,
most likely by reducing RNA polymerase II occupancy at
promoters (Chen and Widom, 2005; Lynch and Rusche,
2009), although other studies suggest that Sir protein binding
interferes with RNA polymerase II elongation (Sekinger and
Gross, 2001; Gao and Gross, 2008).
All three Sir proteins, Sir2, Sir3 and Sir4, are essential for
transcriptional repression. Sir3 and Sir4 are primarily thought
to be structural proteins of silent chromatin (Gasser and
Cockell, 2001). SIR3 arose from a duplication of the ORC1
gene, with which it shares an N-terminal BAH domain and a
related AAA þATPase domain (Hickman and Rusche, 2010).
Sir4 is found only in related ascomycetes species, while Sir2 is
a NAD-dependent histone deacetylase conserved from bacteria
to man. Its enzymatic activity is required for gene silencing
(Tanny et al, 1999; Imai et al, 2000; Smith et al, 2000).
The key substrate of Sir2 is histone H4 acetylated on lysine
16 (H4K16
ac
; Imai et al, 2000; Smith et al, 2000; Borra et al,
2004). This mark is found on transcriptionally active chro-
matin in most species and marks early firing origins in yeast
and flies (Kimura et al, 2002; Suka et al, 2002; Schwaiger
et al, 2010). It has been shown that unmodified H4K16
promotes compaction of the chromatin fibre in vitro and
in vivo (Smith et al, 2003; Shogren-Knaak et al, 2006;
Robinson et al, 2008). Consistently, in budding yeast
H4K16
ac
is found throughout the genome except at silent
loci (Suka et al, 2001; Smith et al, 2003).
Recombinant fragments of Sir3 and Sir4 were shown to
bind to the histone H4 tail in vitro, in a manner sensitive to
Received: 17 December 2010; accepted: 28 April 2011; published
online: 10 June 2011
*Corresponding author. Friedrich Miescher Institute for Biomedical
Research, Maulbeerstrasse 66, 4058 Basel, Switzerland.
Tel.: þ41 61 697 5025; Fax: þ41 61 697 3976;
E-mail: susan.gasser@fmi.ch
The EMBO Journal (2011) 30, 2610–2621 |
&
2011 European Molecular Biology Organization |All Rights Reserved 0261-4189/11
www.embojournal.org
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization
EMBO
THE
EMBO
JOURNAL
THE
EMBO
JOURNAL
2610
mutations near K16 and to lysine acetylation (Hecht et al,
1995; Carmen et al, 2002). Using recombinant proteins
and nucleosomal substrates, it was found that the H4K16A
mutation can decrease binding of Sir3 in vitro (Johnson
et al, 2009), while mutations of H4K16 to glycine or gluta-
mate, and to a lesser extent arginine, diminished mating
efficiency in vivo (Johnson et al, 1990; Megee et al, 1990;
Park and Szostak, 1990). Finally it was shown that the
H4K16G phenotype could be suppressed by a compen-
satory mutation in Sir3, suggesting that Sir3 contacts
the H4 tail in an acetylation-sensitive manner (Johnson
et al, 1990).
In yeast, H4K16 is acetylated primarily by the histone
acetyltransferase (HAT) Sas2 (Kimura et al, 2002; Suka
et al, 2002) and secondarily by the essential HAT Esa1,
which also targets H4K5 and H4K12 (Suka et al,2001,
2002; Chang and Pillus, 2009). Similar to conservative muta-
tions in H4K16, the deletion of SAS2 impairs repression of a
reporter gene at telomeres or the HML locus, although the
same mutation favours silencing of a reporter at HMR,
which has much stronger silencer elements (Reifsnyder
et al, 1996; Ehrenhofer-Murray et al, 1997; Meijsing and
Ehrenhofer-Murray, 2001). However, the rate of Sir3 recruit-
ment to HMR was slower in cells that lack Sas2 (Katan-
Khaykovich and Struhl, 2005), as was the establishment of
silencing at HML (Osborne et al, 2009). This, together with
the observation that the catalytic activity of Sir2 is required
for silencing (Tanny et al, 1999; Imai et al, 2000; Smith et al,
2000; Yang and Kirchmaier, 2006; Yang et al, 2008a), suggests
that Sir-mediated deacetylation of H4K16
ac
might have an
active role in the formation of silent chromatin.
The H4K16
ac
mark is also required for efficient methylation
of lysine 79 on the histone H3 (H3K79
me
) by the methyltrans-
ferase Dot1 (Altaf et al, 2007). In vivo H3K79
me
appears to
impair the spreading of the Sir proteins, and is thought to act
by reducing association of Sir3 to chromatin (Ng et al, 2002,
2003; van Leeuwen et al, 2002; Altaf et al, 2007; Onishi et al,
2007). Consistently, a recent study with recombinant proteins
has suggested that both Sir3 and the SIR holocomplex have
lower affinities for reconstituted chromatin bearing H3K79
me
(Martino et al, 2009). This shows that, in addition to the
histone tails, the Sir3 protein also interacts with the nucleoso-
mal core, a property that has been assigned both to the N-
terminal BAH domain (Onishi et al, 2007; Buchberger et al,
2008; Norris et al, 2008; Sampath et al, 2009) as well as the
Sir3 C-terminal region (Altaf et al, 2007).
A further histone modification that interferes with SIR-
mediated repression is the acetylation of K56 on histone H3.
In budding yeast, H3K56
ac
is deposited by Rtt109 during
S phase before the loading of the histones onto DNA, and
therefore serves as a marker for newly assembled nucleo-
somes (Hyland et al, 2005; Masumoto et al, 2005; Han
et al, 2007; Li et al, 2008). A large number of studies have
also linked H3K56
ac
to gene transcription from yeast to man
(Xu et al, 2005, 2007; Schneider et al, 2006; Williams et al,
2008; Yang et al, 2008b; Michishita et al, 2009; Xie et al,
2009). In yeast, amino-acid substitutions at H3K56 severely
disrupt silencing without completely displacing the SIR com-
plex (Xu et al, 2007; Yang et al, 2008b). Moreover, elimination
of the histone deacetylases responsible for removal of
H3K56
ac
, Hst3 and Hst4, disrupts SIR-mediated repression
as well (Yang et al, 2008b). A recent report has shown that
acetylation of H3K56 favours transcriptional elongation
through yeast heterochromatin, generating speculation that
H3K56
ac
promotes the displacement of the Sir proteins (Varv
et al, 2010). However, there is as yet no direct evidence that
the affinity of Sir proteins for nucleosomes is lowered by
H3K56
ac
.
To gain insight into the role played by these histone
modifications in the assembly of silent chromatin, we recon-
stituted SIR-bound chromatin in vitro using nucleosomes
homogeneously modified on only one residue. Our system
recapitulates many of the characteristics of silent chromatin
(Martino et al, 2009) and allows us to probe both Sir protein
binding and accessibility of the linker DNA to micrococcal
nuclease (MNase). We find that both H3K79
me
and H4K16
ac
decrease the affinity of Sir3 for chromatin, while only
H4K16
ac
has an effect on MNase accessibility. Surprisingly,
we found that Sir2-4 prefers to bind to chromatin acetylated
on H4K16. The binding of Sir2-4, in presence of NAD and
Sir3, leads to the removal of the H4K16
ac
mark and couples
stable binding of the Sir2-3-4 complex with a significant
decrease in linker DNA accessibility. On the other hand,
H3K56
ac
slightly increases MNase accessibility and reduces
the interaction of chromatin with the SIR complex.
Importantly, we find that H3K56
ac
is not a substrate for
Sir2-mediated deacetylation. We, thus, show how the anti-
silencing properties of different histone modifications differ-
entially affect silent chromatin. Of particular interest are the
two contradictory roles played by H4K16
ac
, which reduces
the binding of Sir3 and favours the recruitment of Sir2-4. The
acetylation and deacetylation of H4K16 thus appear to or-
chestrate the sequential binding of Sir proteins in order to
establish a stable silent chromatin.
Results
The H4K16
ac
mark differentially affects the binding of
Sir2-4 and Sir3 to chromatin
It is generally accepted that the H4K16
ac
mark plays an
important role in silent chromatin by preventing the ectopic
spread of the Sir proteins from the non-acetylated silent
domains (Kimura et al, 2002; Suka et al, 2002; Millar et al,
2004; Yang et al, 2008a). However, accumulating evidence
suggests that not only the absence of the acetyl mark but its
Sir2-dependent removal may be required for efficient estab-
lishment of silencing (Liou et al, 2005; Yang and Kirchmaier,
2006; Yang et al, 2008a; Martino et al, 2009; Osborne et al,
2009). To shed light on this matter, we analysed in detail the
binding of SIR subcomplexes to nucleosomal arrays bearing a
fully acetylated H4K16.
Nucleosomes were reconstituted with recombinant his-
tones that were either unmodified or fully acetylated on
H4K16. These were generated by expressing a truncated
version of H4 and adding the N-terminal tail by native
chemical ligation (NCL) using a synthetic peptide (Shogren-
Knaak et al, 2006; Supplementary Figure S1). Nucleosomal
arrays were then reconstituted by salt dialysis using a DNA
template containing repeated arrays of a 167-bp histone
octamer positioning sequence (Widom 601) as described
previously (Huynh et al, 2005; Martino et al, 2009).
Recombinant Sir proteins were purified from insect cells
(Figure 2F; Cubizolles et al, 2006; Martino et al, 2009).
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2611
We first compared acetylated and non-acetylated arrays of
a 6mer of nucleosomes by monitoring the accessibility of
linker DNA to MNase in the absence of Sir proteins. Previous
studies have shown that acetylation of H4K16 inhibits chro-
matin compaction both in vitro and in vivo (Shogren-Knaak
et al, 2006; Robinson et al, 2008). By challenging this
chromatin with increasing amounts of MNase, we found
that H4K16
ac
enhances linker DNA accessibility of a chroma-
tin template as short as six nucleosomes (Figure 1A).
Sir3 has been reported to be more susceptible than Sir4 to
modifications on histone tails (Carmen et al, 2002; Johnson
et al, 2009). Therefore, we examined first the effect of
H4K16
ac
on the binding of Sir3 to nucleosomal arrays.
Increasing amounts of Sir3 were titrated onto unmodified or
H4K16
ac
arrays of nucleosomes. The binding was analysed by
gel shift and quantified by scoring the loss of the unbound
6mer. In agreement with previous studies (Carmen et al,
2002; Johnson et al, 2009), we found that H4K16
ac
reduces
the binding affinity of Sir3 to an in vitro reconstituted
nucleosomal array by roughly two-fold (Figure 1B). In con-
trast, the binding affinity of the Sir2-4 heterodimer to chro-
matin was increased nearly two-fold by the presence of the
H4K16
ac
mark (Figure 1C). Superficially, this appears to
contradict the fact that silent chromatin is depleted for this
mark, although it is consistent with the notion that H4K16
ac
is a key substrate of Sir2-4 (see also Johnson et al (2009)).
Therefore, we decided to perform competition experiments in
order to reinforce this observation. The binding of increasing
amounts of Sir2-4 to an unmodified Cy5-labelled array was
competed with a four-fold excess of either unlabelled unmo-
dified or unlabelled H4K16
ac
chromatin. Confirming our
previous results, H4K16
ac
chromatin competed roughly two-
fold more efficiently for the binding of Sir2-4 compared with
unmodified chromatin (Figure 1D).
We previously showed that our recombinant Sir2-4 has
efficient histone deacetylase activity in the presence of its co-
factor NAD (Cubizolles et al, 2006). We reasoned that if
Sir2-4 bound H4K16
ac
with higher affinity because it is a
preferred substrate of Sir2, then the complex should have
less affinity once H4K16
ac
had been deacetylated. To test this,
we quantified the binding of the Sir2-4 heterodimer to
H4K16
ac
chromatin in the absence or presence of NAD.
Confirming our hypothesis, Sir2-4 bound more readily to
acetylated chromatin and less readily following deacetylation
(Figure 1E). Surprisingly, this shows an enhanced affinity
of Sir2-4 for acetylated H4K16, while the opposite is true
for Sir3.
Sir2-dependent deacetylation of H4K16
ac
stabilizes the
association of Sir2-3-4 to chromatin
Removal of H4K16
ac
through the catalytic activity of Sir2 has
been reported to be important for silencing (Johnson et al,
1990; Suka et al, 2001, 2002; Carmen et al, 2002; Kimura et al,
2002). However, it is not clear whether this is due exclusively
Figure 1 Acetylation of H4K16 decreases the binding affinity of
Sir3 but favours the loading of Sir2-4 onto chromatin. (A) Equally
saturated 6mer of 601 nucleosomes with either unmodified or
acetylated H4K16 was digested with increasing amount of MNase,
as indicated. After protein digestion, the denatured DNA was
separated by electrophoresis and visualized by SYBR
s
Safe stain-
ing. The bands showed by an arrow (6-, 5-, 4- and 3mers) were
quantified and normalized to input. The histograms show the ratio
between the amount of intact 6-3mers of H4K16
ac
over unmodified
chromatin for the indicated MNase titration point. The Sir3 protein
(B) or the Sir2-4 heterodimer (C) were titrated into a constant
amount of unmodified or H4K16
ac
6mer of 601 nucleosomes.
Samples were separated by native agarose gel electrophoresis and
visualized by SYBR
s
Safe staining. (D) The binding of increasing
amounts of Sir2-4 to 8 nM of unmodified Cy5-labelled 6mer of
nucleosomes (indicated by the arrowhead) was competed with
32 nM of either unlabelled unmodified or unlabelled H4K16
ac
6mer of nucleosomes. Cy5 fluorescence was used to monitor the
binding of Sir2-4 to the unmodified labelled chromatin. The asterisk
indicates a Cy5-labelled contaminant DNA. (E) The Sir2-4 hetero-
dimer was titrated into a constant amount of H4K16
ac
6mer of 601
nucleosomes. Deacetylation is allowed by the addition of 150 mM
NAD where indicated. Samples were separated and visualized as in
(B,C). The images are representative of at least three independent
experiments, quantifications show the mean value±s.e.m. of the %
of unbound chromatin compared with the input.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization2612
to the generation of unmodified H4K16 or whether it addi-
tionally involves a conformational change coupled to O-
acetyl-ADP-ribose (O-AADPR) production (Liou et al, 2005;
Martino et al, 2009). To gain insight into the molecular
consequences of H4K16
ac
deacetylation on the establishment
of silencing, we compared the binding of the SIR complex
with unmodified or H4K16
ac
chromatin in the presence or
absence of NAD. We first examined the effect of H4K16
ac
on
the binding of the Sir2-3-4 heterotrimer in absence of NAD
(Figure 2A) and found that, similar to Sir2-4 (Figure 1C) but
in a less pronounced manner, the SIR holocomplex bound
slightly better to acetylated chromatin. We then confirmed
that our purified Sir2-3-4 complex was able to efficiently
deacetylate H4K16
ac
within chromatin in the presence of
NAD (Figure 2E), as shown previously for chemically acety-
lated histone octamers (Cubizolles et al, 2006). In the follow-
ing experiments, the term ‘deacetylated chromatin’ will be
used whenever H4K16
ac
marks were actively removed by Sir2
in the presence of NAD, to distinguish it from chromatin
assembled from unmodified histones.
We next compared the binding affinity of Sir2-3-4 with
unmodified or H4K16
ac
chromatin in the presence of NAD.
We found that the active removal of the H4K16
ac
mark
increased the binding affinity of the SIR complex to chroma-
tin by roughly two-fold (Figure 2B). This effect is not caused
by NAD alone as enhanced binding was not observed when
H4K16
ac
chromatin was replaced with unmodified chromatin
(Supplementary Figure S2A). Indeed, in absence of Sir2,
Figure 2 Sir2-dependent deacetylation of H4K16
ac
stabilizes the association of Sir2-3-4 to chromatin. The Sir2-3-4 complex (A,B) or the
catalytically dead Sir2cd-3-4 mutant (C,D) was titrated into a constant amount of unmodified or H4K16
ac
6mer of 601 nucleosomes. Where
indicated, 150 mM NAD was added to the samples. Scatter plot quantifications show the mean value±s.e.m. of the % of unbound chromatin
compared with the input for at least three experiments. (E) Reconstituted chromatin fully acetylated on H4K16 was subjected to NAD-
dependent deacetylation in presence of a 2.5-fold molar excess of the Sir2-3-4 complex or the Sir2cd-3-4 mutant. The acetylation state was then
determined by immunoblotting using acetylation mark-specific antibodies and H3 for loading. (F) Two micrograms of the indicated Sir protein
were denatured in sample buffer and run on a 4–12% NuPAGEsNovexsBis-Tris Gel.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2613
NAD does not affect the acetylation state of chromatin
(Supplementary Figure S2B). This shows that the binding
affinity of the SIR complex for deacetylated chromatin is
higher compared with chromatin assembled from unmodified
histones.
In order to reinforce this finding, we tested whether the
deacetylase activity of Sir2 itself was required for the en-
hanced binding of acetylated template in the presence of
NAD. We generated a catalytic inactive Sir2 (Sir2cd) by
introducing the point mutation N345A, which maps to the
nucleotide binding motif (Rossman fold). This mutation
disrupts Sir2 enzymatic activity in vitro and in vivo (Imai
et al, 2000; Armstrong et al, 2002). The N345A substitution,
however, did not affect the stability of Sir2 or its interaction
with Sir3 and Sir4 and we were able to purify the mutated
Sir2cd-3-4 from insect cells with the same efficiency as for the
Sir2-3-4 complex (Figure 2F). We could furthermore confirm
that this mutant did not retain significant deacetylase activity
(Figure 2E).
In order to confirm that the Sir2cd-3-4 mutant was still able
to recognize its substrate, we monitored the binding of Sir2cd-
3-4 to unmodified or H4K16
ac
chromatinintheabsenceof
NAD. We found that, like Sir2-3-4, Sir2cd-3-4 has a slight
preference for H4K16
ac
chromatin (compare Figure 2A and C).
We then monitored the loading of the Sir2cd-3-4 mutant onto
unmodified or H4K16
ac
chromatin in the presence of NAD.
Unlike the Sir2-3-4 complex, the catalytic dead Sir2cd-3-4
showed the same slight preference for the H4K16
ac
chromatin,
as it did in the absence of NAD (compare Figure 2C and D).
This data reinforce our observation that the deacetylation
reaction has a positive role on the loading of the SIR complex
onto chromatin (Figure 2B). Given that the deacetylation of
H4K16
ac
chromatin reduced the binding affinity of the Sir2-4
heterodimer (Figure 1E), these results suggest that Sir3 and the
deacetylation of H4K16
ac
by Sir2 jointly promote the binding of
the SIR holocomplex to chromatin.
The Sir3 protein and Sir2-dependent deacetylation of
H4K16
ac
are both required to decrease nuclease
accessibility of the linker DNA
SIR complex bound chromatin is thought to have a more
compact structure in vivo as it is less accessible to enzy-
matic attack (Gottschling, 1992; Loo and Rine, 1994). The SIR
complex could compact chromatin in two ways: first by
deacetylating H4K16, and second by binding to chromatin.
We observed that loading of the SIR complex onto unmodi-
fied chromatin greatly reduces the accessibility of the linker
DNA to MNase and the restriction enzyme AvaIin vitro,
consistent with a direct role for binding (Supplementary
Figure S3A and B; Martino et al, 2009). In order to test the
impact of H4K16
ac
deacetylation on compaction in vitro,we
first incubated H4K16
ac
chromatin with the Sir2-4 subcom-
plex in the presence or absence of NAD and then challenged
it with increasing amounts of MNase. We found that the
presence of NAD did not significantly change the accessibility
of the linker DNA (Figure 3A). Since acetylation of chro-
matin usually results in greater accessibility (Figure 1A), this
result is likely a combination of changed accessibility due to
removal of H4K16
ac
and reduced binding affinity of Sir2-4 for
deacetylated chromatin (Figure 1E).
We then performed the same experiment but replaced Sir2-
4 with the SIR holocomplex. The concentration of Sir2-3-4
used resulted in a complete upshift of both acetylated and
unmodified chromatin in a binding assay, ruling out differ-
ential accessibility due to incomplete ligand occupancy.
Interestingly, we observed that in the presence of NAD the
linker DNA was more protected from MNase than SIR-bound
Figure 3 Sir3 is required to translate the Sir2-dependent deacetyla-
tion of H4K16
ac
into a decrease of nuclease accessibility of the linker
DNA. Unmodified or H4K16
ac
6mer of 601 nucleosomes (50 nM)
was incubated with the indicated amount of Sir2-4 (A), Sir2-3-4
(B,Dand E) or Sir2cd-3-4 (C) and was challenged with increasing
amounts of MNase. Where indicated, SIR-bound chromatin was
supplemented with 150 mM NAD and incubated for 15 min at 301C
before MNase digestion. Deproteinated samples were separated by
electrophoresis and the amount of intact 6mer DNA (black arrow)
was quantified and normalized to input. Quantification of at least
three experiments was used to generate the vertical bar charts, data
represent mean value±s.e.m.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization2614
chromatin in the absence of NAD (Figure 3B). Importantly,
the addition of NAD did not change the protection of linker
DNA of an array bound by the catalytic inactive Sir2cd-3-4
(Figure 3C). The same analysis in the absence of Sir3 did
not increase protection against MNase attack (Figure 3A),
arguing that the protective effect of NAD-dependent deacety-
lation of H4K16
ac
by Sir2 requires Sir3 (compare Figures 3A
and B). Finally, the increased linker DNA protection observed
in Figure 3B was not caused by the NAD molecule per se,as
no NAD-dependent differences were scored for linker DNA
accessibility when Sir proteins were bound to unmodified
chromatin (Supplementary Figure S3C). These results sug-
gested that the deacetylation of H4K16
ac
by the SIR complex
promotes linker DNA protection.
When comparing unmodified with acetylated chromatin
bound by Sir2-3-4 in the absence of NAD, we found that the
linker DNA is slightly more protected (Figure 3D), indicating
that at least some of the protection observed in Figure 3B is
due to loss of H4K16
ac
per se. However, given our previous
observation that deacetylated chromatin is bound with higher
affinity than unmodified chromatin (Figure 2B), we decided
to explore the possibility that the deacetylation reaction itself
may also contribute to the increased linker DNA protection
observed in Figure 3B. Therefore, we compared the linker
DNA accessibility of unmodified and H4K16
ac
chromatin in
the presence of Sir2-3-4 and NAD. We found that deacetylated
chromatin is reproducibly more protected from MNase attack
compared with chromatin assembled from unmodified his-
tones (Figure 3E). Together, these results show that both Sir2-
dependent deacetylation of H4K16
ac
and Sir3 are required to
decrease the nuclease accessibility of linker DNA, which
presumably reflects the tighter binding of the SIR holocom-
plex to chromatin. In addition, there may be a conformational
change that enhances linker DNA protection.
H3K56
ac
loosens Sir protein binding to chromatin,
slightly increasing linker DNA accessibility
To ask if our observation for H4K16
ac
can be generalized to
other acetylation marks we tested the effects of H3K56
acetylation, which is found on newly assembled nucleosomes
in S phase. Since there are contradictory reports about which
enzyme deacetylates H3K56
ac
(Xu et al, 2007; Yang et al,
2008b) we first tested if Sir2 can deacetylate H3K56
ac
as
suggested earlier. We incubated H3K56
ac
chromatin with
SIR complex in the presence or absence of NAD. Chromatin
homogenously acetylated at H3K56 was obtained by purify-
ing acetylated H3 from E. coli using an expanded genetic code
strategy (Neumann et al, 2008). Probing the histones with
H3K56
ac
antibodies after incubation showed that, unlike for
H4K16
ac
, the level of H3K56 acetylation remained unchanged
(Figure 4B). We conclude that H3K56
ac
is not a substrate of
the NAD-dependent deacetylase activity of Sir2. This sup-
ports previous work reporting that two Sir2-related enzymes,
Hst3 and Hst4, are required for H3K56
ac
deacetylation in vivo
(Celic et al, 2006; Maas et al, 2006; Yang and Kirchmaier,
2006) and suggests that Hst3 and Hst4 are the exclusive
deacetylases for this residue.
To address whether acetylation of H3K56 has an effect on
Sir protein loading, we compared the binding of the SIR
holocomplex with unmodified and H3K56
ac
chromatin. We
found that H3K56
ac
reduces the affinity of the SIR holocom-
plex for chromatin by roughly two-fold (Figure 4C). The
binding affinity of the Sir2-4 heterodimer was also reduced
in presence of the H3K56
ac
mark (Figure 4D), while the
binding affinity of the Sir3 protein alone was mostly un-
changed (Figure 4E). To explore whether the slight affinity
decrease observed here for the SIR complex could be respon-
sible for the silencing defects seen in vivo, we investigated
whether the SIR complex efficiently protects linker DNA in
chromatin bearing the H3K56
ac
mark. The acetylation on
H3K56 per se has been shown to increase transient unwrap-
ping of the DNA from the histone octamer but not to change
the higher order structure of a 61mer nucleosomal array
(Neumann et al, 2009). Consistently we show, by means of
an MNase digestion assay, a slight increase in linker DNA
accessibility for the chromatin bearing H3K56
ac
over the
unmodified control (Figure 4F). Subsequently, after adding
the SIR complex in saturating concentrations (Supplementary
Figure S4A), the H3K56
ac
chromatin continued to show
slightly higher linker DNA accessibility as compared with
unmodified chromatin (Figure 4G). This is consistent with a
previous in vivo study indicating that H3K56
ac
chromatin
is more sensitive to DNA methylation by an ectopically
expressed bacterial dam methylase (Xu et al, 2007).
Moreover, H3K56 point mutations disrupted silencing at
telomeres without affecting Sir protein spreading (Xu et al,
2007). We conclude that H3K56
ac
does not have a role similar
to that of H4K16
ac
, neither in the recruitment of Sir2-4, nor by
being a substrate for Sir2.
Methylation of H3K79 by Dot1 neither increases linker
DNA accessibility nor reduces Sir2-4 loading
Another mark associated with active chromatin in yeast is
methylation of lysine 79 of histone H3 (H3K79). This methy-
lation is exclusively catalysed by Dot1 and is thought to act as
a boundary for the inappropriate spreading of the Sir proteins
on chromatin (van Leeuwen et al, 2002; Frederiks et al, 2008;
Martino et al, 2009; Verzijlbergen et al, 2009). Moreover,
in vitro studies showed that interaction of the Sir3 protein with
histone peptides was sensitive to the methylation of H3K79
(Altaf et al, 2007; Onishi et al, 2007). Previous work from our
laboratory showed that we can make use of recombinant Dot1
in order to methylate reconstituted nucleosomal arrays in vitro
(Martino et al, 2009). We have previously shown that even
partial methylation of H3K79 decreases the binding affinity of
both the SIR complex and the Sir3 protein alone to chromatin
(Martino et al, 2009). We now provide further evidence that the
lowered affinity indeed affects Sir3 binding, since the Sir2-4
heterodimer associates with unmodified and H3K79
me
chroma-
tin with nearly equal affinity (Figure 5A), while Sir3 clearly
prefers unmodified chromatin (Figure 5B).
We then decided to test whether H3K79
me
also impacts the
structure of SIR-bound or SIR-depleted chromatin. To exam-
ine the potential impact of H3K79
me
on linker DNA protec-
tion, we challenged in vitro methylated chromatin lacking Sir
proteins with increasing amounts of MNase. Unlike the case
for H3K56
ac
, the accessibility of the linker DNA in the
absence of SIR complex was unaffected by H3K79
me
(Figure 5C). However, in the presence of substoichiometric
amounts of Sir2-3-4 (Supplementary Figure S4B), the acces-
sibility of the linker DNA was higher for H3K79
me
chromatin
than for unmodified chromatin (Figure 5D), consistent with
notion that better SIR complex binding enhances linker DNA
protection. When we added additional Sir2-3-4 such that
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2615
we score an equal degree of binding on both substrates
(Supplementary Figure S4C), we observed no difference in
accessibility of linker DNA (Figure 5E). This suggests that
H3K79
me
neither changes chromatin structure nor prevents
the SIR complex from compacting it, but decreases the
affinity of the SIR complex for chromatin. Thus, it antago-
nizes silencing through a mechanism distinct from H3K56
ac
.
Discussion
Silent chromatin in Saccharomyces cerevisiae is the best
studied system of heterochromatic gene silencing, yet we
still do not fully understand the molecular mechanisms of
its assembly and the role of histone modifications in this
process. In vitro binding analysis between Sir protein
domains and histone peptides have been informative, yet
they only reflect a small part of the chromatin template.
To examine the molecular basis of SIR-dependent silen-
cing, we have established a fully recombinant system that
recapitulates key features of silent chromatin in budding
yeast (Martino et al, 2009). Here, we extend this system to
examine how histone modifications participate in the forma-
tion of stable silent and active domains.
We show here that the H4K16
ac
mark has both a positive
and a negative role in SIR binding in a sequential manner (see
Figure 6). Importantly, we show that H4K16
ac
decreases the
binding affinity of Sir3, but, in contrast, promotes the asso-
ciation of the Sir2-4 heterodimer to chromatin. Even the
binding affinity of the SIR holocomplex is slightly increased
by the presence of H4K16
ac
in the absence of NAD (see also
Johnson et al (2009)). This result, while initially counter-
intuitive, helps elucidate the dual role of H4K16
ac
in hetero-
chromatin formation. On one hand, H4K16
ac
prevents the
dispersion of its key ligand, Sir3, into euchromatic chromatin.
On the other hand, the high affinity of Sir2-4 for H4K16
ac
may
help nucleate silent chromatin, since it is likely in yeast that
the targeted nucleosomes are acetylated before SIR complex
loading.
In support of this dual role, it was shown that the sub-
stitution of H4K16 by not only an acetyl-mimicking residue,
but also unacetylatable amino acids, disrupts silencing at
telomeres and mating type loci (Johnson et al, 1990, 1992;
Megee et al, 1990; Park and Szostak, 1990; Aparicio et al,
1991; Millar et al, 2004). Moreover, the deletion of SAS2,
which encodes the HAT responsible for most H4K16
ac
in
yeast, impaired the repression of reporter genes at certain
loci, such as TelVIIL or within the HML locus (Reifsnyder
et al, 1996; Meijsing and Ehrenhofer-Murray, 2001) and led to
the spreading of Sir proteins into subtelomeric regions that
usually lack SIR-mediated repression (Kimura et al, 2002;
Figure 4 H3K56
ac
decreases Sir protein binding affinity and slightly increases linker DNA accessibility. (A) Cartoon representation of the
nucleosome core particle (NCP147; Davey et al, 2002) highlighting the position of H3K56 (black) at the entry/exit point of the DNA around the
histone octamer. (B) Reconstituted chromatin fully acetylated on H3K56 was subjected to NAD-dependent deacetylation in presence of a 2.5-
fold molar excess of the SIR complex. The acetylation state was then determined by immunoblotting using acetylation mark-specific antibodies
and H3 for loading. The SIR complex (C), Sir2-4 heterodimer (D) or Sir3 (E) were titrated into a constant amount of unmodified or H3K56
ac
6mer of 601 nucleosomes. Samples were analysed as in Figure 2. Unmodified or H3K56
ac
6mer of nucleosomes were challenged with an
increasing amount of MNase in absence (F) or presence (G) of the SIR complex. The 6mer, 5mer and 4mer bands (F) or the band corresponding
to the intact 6mer alone (G), shown by black arrows, were quantified and normalized to the input. The histograms show the ratio between the
amounts of quantified DNA from H4K16
ac
chromatin over unmodified for the indicated MNase titration point±s.e.m.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization2616
Suka et al, 2002). In contrast, the repression of a reporter at
HMR was enhanced, probably because this locus has much
stronger silencers, which dominate over an indiscriminate
spreading of Sirs (Ehrenhofer-Murray et al, 1997). Impor-
tantly, both the kinetics of Sir3 recruitment to HMR and the
establishment of silencing at HML were slower in cells that
lack the H4K16-specific HAT, Sas2 (Katan-Khaykovich and
Struhl, 2005; Osborne et al, 2009), suggesting a positive role
for H4K16 acetylation. Collectively, these results support the
model that Sas2-mediated acetylation of H4K16 has more
than one role in silent chromatin formation (see also Zou and
Bi (2008)). Reporter context appears to determine which
role is rate limiting: the recruitment of Sir2-4, or the assembly
and propagation of the Sir3-containing holocomplex along
nucleosomes.
Sequential assembly of nuclease-resistant SIR-bound
chromatin requires H4K16
ac
deacetylation
In vivo the absence of H4K16
ac
from silent chromatin suggests
that it is removed by Sir2 as soon as the SIR complex is
loaded. Moreover, it was shown that in the absence of Sir2
catalytic activity, H4K16
ac
prevents the formation of silent
domains (Yang and Kirchmaier, 2006). On the other hand,
as mentioned above, even conservative substitutions at
H4K16 decrease silencing efficiency at HML and at telomeres
(Johnson et al, 1990; Meijsing and Ehrenhofer-Murray, 2001;
Yang and Kirchmaier, 2006). This supports the notion that not
only the recruitment of Sir2-4 by H4K16
ac
, but the deacetyla-
tion reaction itself helps to seed repression (Johnson et al,
1992; Imai et al, 2000; Millar et al, 2004; Liou et al, 2005;
Yang et al, 2008a; Martino et al, 2009).
Why is the deacetylation reaction important for silencing?
The recapitulation of these steps in vitro helped us to address
this question. Indeed, by adding NAD to SIR-bound H4K16
ac
chromatin, we catalysed deacetylation of H4K16 and in-
creased the binding affinity of the SIR holocomplex to chro-
matin. This shows in a well-defined recombinant system
that removal of the single H4K16
ac
mark by Sir2 increases
SIR complex binding. Even more importantly, we found that
the linker DNA was better protected from MNase digestion
when the SIR complex was assembled on chromatin in the
presence of H4K16
ac
and NAD, as compared with its being
loaded onto unmodified chromatin. This protection nicely
mimics the DNA shielding observed in SIR-silenced chroma-
tin regions in vivo (Gottschling, 1992; Loo and Rine, 1994;
Xu et al, 2007) and argues that the SIR complex may associate
with chromatin in more than one conformation. It was
previously proposed that a by-product of Sir2 NAD-
dependent deacetylation, O-AADPR, might trigger a confor-
mational change of the SIR–chromatin complex to favour
repression (Liou et al, 2005; Onishi et al, 2007; Martino et al,
2009).
Importantly, the deacetylation-dependent increase in affi-
nity of the SIR holocomplex for chromatin, and the increase
in linker DNA protection, depends crucially on the presence
of Sir3. This is consistent with previous results which showed
that exogenously added O-AADPR enhances the binding of
both Sir3 and the SIR holocomplex to chromatin (Martino
et al, 2009). A further study showed that addition of an excess
of acetylated peptides and NAD to SIR–chromatin assemblies
generated a structure that appeared more compact by elec-
tron microscopy (Johnson et al, 2009), perhaps reflecting a
conformational change in the SIR complex (Liou et al, 2005).
Nevertheless, O-AADPR is probably neither absolutely req-
uired for SIR complex loading nor for silencing, since repres-
sion can be achieved in a strain devoid of NAD-dependent
deacetylases if an ectopic HDAC is fused to Sir3 (Chou et al,
2008) or if Sir3 is overexpressed in a H4K16R background
(Yang and Kirchmaier, 2006). Taken together, our data sup-
port a scenario in which the sequential loading of Sir2-4
Figure 5 Methylation of H3K79 by Dot1 affects neither linker DNA
accessibility nor Sir2-4 loading onto chromatin. The Sir2-4 hetero-
dimer (A) or Sir3 (B) was titrated into a constant amount of
unmodified or H3K79
me
6mer of nucleosomes. Samples were ana-
lysed as in Figure 2. Unmodified or H3K79
me
chromatin were
challenged with an increasing amount of MNase in absence (C)or
presence (D,E) of the indicated amount of the SIR complex. The
6mer DNA band alone (C,E) or the 3mer to 6mer bands (D), shown
by black arrows, were quantified and normalized to the input. The
histograms show the ratio between the amount of quantified DNA
from H3K79
me
over unmodified chromatin for the indicated MNase
titration point.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2617
onto nucleosomes containing H4K16
ac
, its NAD-dependent
deacetylation and the loading of Sir3, sequentially promote a
stable assembly that protects linker DNA from exogenous
factors (Figure 6).
Boundary formation and reduction of SIR holocomplex
affinity by histone modifications
The observation that H4K16
ac
might provide a boundary for
heterochromatin spreading (Kimura et al, 2002; Suka et al,
2002) seems counterintuitive given the results described
above. However, H4K16
ac
does affect other processes beyond
SIR complex association to chromatin, most notably, the
recruitment of the histone methyltransferase Dot1 to chro-
matin (Altaf et al, 2007). Given that Sir2-4 preferentially
binds chromatin carrying H4K16
ac
, we propose that Dot1
competes with the recruitment of Sir2-4, and not as proposed
earlier, with Sir3 (Altaf et al, 2007). On the other hand, the
anti-silencing role of the methylation mark itself, H3K79
me
,is
most likely a reflection of reduced interaction between Sir3
and methylated chromatin (Ng et al, 2002, 2003; van
Leeuwen et al, 2002; Altaf et al, 2007; Onishi et al,2007;
Martino et al, 2009). Consistently, we found that nucleo-
somes bearing H3K79
me
neither affect the binding of the
Sir2-4 heterodimer, nor was there an inherent change in
structural properties of H3K79
me
-containing chromatin. This
is consistent with crystallographic analyses which argue that
H3K79
me
does not alter the structure of the nucleosome
(Lu et al, 2008). Given that no enzyme has been found so
far that removes the H3K79
me
mark, depletion of this mark
may depend on histone eviction or on sequential dilution
through rounds of DNA replication. Its slow removal renders
H3K79
me
a more stable barrier to the spreading of the SIR
complex than H4K16
ac
, which instead recruits Sir2-4 and
promotes the spread of repression (Figure 6).
The third histone mark correlated with active chromatin in
yeast is the acetylation of H3 on K56. In contrast with
H3K79
me
, H3K56
ac
is clearly subject to active deacetylation.
Here, we show that Sir2 is unable to remove the H3K56
ac
mark in vitro, which indirectly supports previous work
showing that H3K56
ac
is primarily deacetylated by two Sir2-
related enzymes: Hst3 and Hst4 (Celic et al, 2006; Maas et al,
2006). Indeed, Sir-mediated repression cannot be established
in the absence of these two enzymes, although Sir proteins
still bind telomeres in an hst3Dhst4Dmutant (Yang et al,
2008b). Consistent with our work, this suggests that the
H3K56
ac
mark does not completely block SIR–chromatin
interaction.
How then does H3K56
ac
impair the formation of silent
chromatin? Using our in vitro system, we found that H3K56
ac
affects both the affinity with which SIR complexes bind
chromatin and the formation of a chromatin structure that
is less accessible to MNase attack. The observed drop in
affinity of SIR holocomplex for chromatin agrees with an
in vivo study, which suggested that H3K56
ac
facilitates Sir
protein displacement and RNA polymerase II elongation
within heterochromatin regions (Varv et al, 2010). Our
Figure 6 Combinatorial histone modifications distinguish silent and active chromatin regions. (A) Outline of the role played by different
histone modifications on Sir protein loading and chromatin structure. (B) The Sir proteins are recruited onto chromatin by protein–protein
interactions and bind tightly unmodified nucleosomes driving gene silencing. Spreading of the SIR complex is promoted by H4K16
ac
that
recruits the Sir2-4 heterodimer yet prevents the ectopic spreading of Sir3 alone. The NAD-dependent deacetylation reaction of H4K16
ac
by Sir2
generates a high-affinity binding substrate for Sir3 and the synthesis of O-AADPR favours the tight association of the SIR complex to
unmodified nucleosomes. H3K56
ac
and H3K79
me
generate a boundary to the spreading of the SIR complex mainly by reducing the binding
affinity of the Sir2-4 heterodimer and the Sir3 protein, respectively. The acetylation of H3K56 and H4K16 also enhance the accessibility of the
chromatin fibre, unlike methylation of H3K79.
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization2618
observation that H3K56
ac
increases accessibility of the
linker DNA is consistent with an increase in spontaneous
(but transient) unwrapping of the DNA from the histone
octamer, which may reflect the position of H3K56 at the
entry/exit point of the nucleosomal DNA (Figure 4A;
Neumann et al, 2009). It is striking that even SIR-saturated
arrays showed increased linker DNA accessibility in the
presence of H3K56
ac
, indicating that SIR binding cannot
overcome the effect of H3K56
ac
on nucleosomal structure.
Although it is unclear why this modification reduces SIR
complex binding, this and the increased linker DNA exposure
are likely to account for the anti-silencing effect of the
H3K56
ac
mark.
To conclude, we propose that the euchromatic mark
H4K16
ac
is required for the formation of both active and
silent chromatin. The process of creating stable silent and
active states is not an one-step event, but requires positive
feedback loops. H4K16
ac
may be the starting point for silent
domains, which are reinforced by the Sir2 deacetylation
reaction and possibly the generation of O-AADPR, and active
domains, where it promotes H3K79 methylation. These inter-
dependent pathways are conserved throughout evolution and
mathematical modelling clearly shows that such networks
are required to establish a stable binary switch (Dodd et al,
2007; Mukhopadhyay et al, 2010). Here, we have demon-
strated that H4K16
ac
is actively implicated in the establish-
ment of yeast silent chromatin, being the first histone mark
shown to recruit Sir proteins to chromatin.
Materials and methods
SIR purification and chromatin reconstitution
In vitro reconstitution of SIR-bound chromatin was carried out
essentially as described (Cubizolles et al, 2006; Martino et al, 2009).
Briefly, the Sir proteins were expressed in sf21 insect cells with
baculoviruses generated using BD BaculoGoldTM, BD-Biosciences.
Co-infection was used to produce the Sir2-3-4 complex, the catalytic
dead Sir2cd-3-4 and the Sir2-4 heterodimer, a single infection was
used to produce the Sir3 protein alone (Cubizolles et al, 2006).
Recombinant X. laevis histones were use to reconstitute histone
octamers as described previously (Luger et al, 1997). Chromatin
was assembled in vitro by adding increasing amounts of purified
histone octamer to a constant amount of DNA arrays containing six
601-Widom positioning elements separated by 20 bp of linker DNA,
referred as 601-167-6mer (Lowary and Widom, 1998). An unspecific
DNA sequence of 147 bp (referred as ‘competitor’ on the figures)
was added to the mix in order to bind the excess of histone
octamers subsequent to the saturation of the 601-167-6mer (Huynh
et al, 2005). Cy5-labelled 601-167-6mer was generated by filling the
50overhang-ends of an EcoRI site with Klenow enzyme (NEB,
accordingly to manufacturer’s instruction), using d-CTP-Cy5 (GE
Healthcare). The free nucleotides were then separated from the
Cy5-labelled array using small Bio-spin columns (Bio-Rad). DNA
and histones were mixed in 40 ml of buffer A (10 mM TEA pH 7.4
and 1 mM EDTA) and 2 M NaCl on ice, and chromatin was
reconstituted by step dialysis in buffer A containing 1.2, 1, 0.8 or
0.6 M NaCl for 2 h at 41C and in buffer A overnight (Lee and
Narlikar, 2001). The 601-167-6mer was routinely prepared at a final
nucleosomal concentration of 10
6
M. Increasing amounts of Sir
proteins were added to the 601-167-6mer diluted to 5 10
8
Mor
2.5 10
8
M in 10mM TEA pH 8, 25 mM NaCl, 0.05% Tween-20 on
ice and after 10min incubation the samples were fixed with
0.0025% glutaraldehyde for 10 min on ice. The fixation yields
slightly sharper bands but the results are very similar without
fixation. When chromatin deacetylation was coupled to Sir protein
loading, SIR-bound chromatin was incubated with or without
150 mM NAD for 15 min at 301C before incubation on ice for a
10 min fixation as above. The samples were routinely run at 80 V for
90 min at 41C in a 0.7% agarose gel 0.2 TB: 18 mM Tris, 18 mM
Boric acid. The gel was soaked for 20 min in 1 SYBRsSafe and
the DNA was visualized in a Typhoon 9400 scanner.
Preparation of the histone modifications
Methylation of H3K79 was carried out on reconstituted chromatin
as described before (Martino et al, 2009). Briefly, 0.8 pmol of
recombinant Dot1 was incubated with 8 pmol of reconstituted 601-
167-6mer in 25 mM Tris pH 7.9, 20 mM NaCl, 0.4mM EDTA, with or
without 160 pmol of S-adenosylmethionine (SAM) at 301C for
30 min, then 160 pmol of SAM was added and the reaction was
continued for 30 min. Mass spectrometry analysis showed that
H3K79 is mono-, di- and, to a lesser extent, tri-methylated on at
least 50% of the available K79 residues (Frederiks et al, 2008;
Martino et al, 2009). The chromatin was then stored at 41C.
Homogeneous acetylated histone H3 at the lysine 56 was
obtained using an aminoacyl-tRNA synthetase and tRNA
CUA
pair
created by directed evolution in E. coli (Neumann et al, 2008). An
unmodified control was prepared in parallel. Histone octamers were
assembled as described previously (Luger et al, 1997) and kept at
41C before chromatin reconstitution.
Full acetylation of H4K16 was obtained by NCL as described
previously (Shogren-Knaak et al, 2006). Briefly, the H4 N-terminal
peptide containing residues 1–22 and acetylated lysine at position
16 was synthesized using Fmoc (N-(9-fluorenyl)methoxycarbonyl)-
based solid-phase synthesis and activated at the C-terminus by
thioesterification. Subsequently, the globular X. laevis H4D1-
22,R23C was ligated to the activated H4 peptide and the ligation
product was purified as described previously (Shogren-Knaak
et al, 2006). Identity and purity of the histones were verified by
SDS–PAGE as well as ESI-MS (Supplementary Figure S1). Histone
octamers were assembled as described previously (Luger et al,
1997) and kept at 41C before chromatin reconstitution.
MNase digestion assay
MNase digestion was carried out in 20 ml of 10 mM TEA pH 8,
1.5 mM CaCl
2
, 25 mM NaCl, 0.05% Tween-20. In all, 1 pmol of
601-167-6mer was digested with increasing amounts of MNase, as
detailed in the figures, for 12 min on ice. The digestion was stopped
by adding 10 mM EGTA, proteins were removed by proteinase K
digestion for 15 min at 301C and the samples were run at 65 V for
60 min in a 1.2% agarose gel 1 TBE: 90 mM Tris, 90 mM Boric
acid, 2 mM EDTA. Digestion of SIR-bound chromatin was performed
on 601-167-6mer pre-incubated with the indicated amount of Sir
proteins for 10 min on ice. MNase digestion of deacetylated
chromatin was performed on 601-167-6mer incubated with
0.66 pmol of Sir2-3-4, Sir2cd-3-4 or Sir2-4 for 15 min at 301C with
or without 150 mM NAD and recovered on ice. Concerning the Sir2-
3-4 complex, similar results were obtained by incubating the
nucleosomal array with 0.66 pmol of Sir2-4 at first and adding
0.66 pmol of Sir3 before the recovery on ice. In order to strengthen
our observations, different batches of modified and unmodified
chromatins were compared.
Deacetylation reaction
Deacetylation of 2 pmol of reconstituted chromatin was performed
in 30 ml of 25 mM Tris pH 8, 50 mM NaCl in presence of 5 pmol of
the Sir2-3-4 complex, the Sir2cd-3-4 mutant or Sir2-4 and 150 mM
NAD for 30 min at 301C and stopped by addition of 4 Laemmli
buffer. Similar results were obtained in 25 mM Tris pH 8, 137 mM
NaCl, 2.7 mM KCl and 1 mM MgCl
2
. The acetylation state was
determined by immunoblotting using acetylation mark-specific
antibodies (anti-H3K56
ac
Upstate #07-677, anti-H4K16
ac
Serotec
AHP417) and H3 for loading (anti-H3 Abcam ab1791-100).
Supplementary data
Supplementary data are available at The EMBO Journal Online
(http://www.embojournal.org).
Acknowledgements
We would like to thank the Gasser laboratory and in particular
Helder Ferreira for discussion and support as well as Simon Lattman
for assistance in analysing the data. We thank Heinz Neumann
for providing a preliminary batch of H3K56
ac
histone octamers.
The Gasser laboratory is supported by the Novartis Research
Foundation and the EU network Nucleosome 4D. SK was supported
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2619
by an EMBO long-term fellowship and an FWF Schroedinger
fellowship.
Author contributions: MO, SK and SMG designed the experiments
and interpreted results. MO performed the experiments. SK and
FM contributed reagents. SS and WF contributed the H4K16
ac
histone octamers. SMH and JC contributed the H3K56
ac
histone
octamers. MO, SK and SMG wrote the manuscript. SMG supervised
the work.
Conflict of interest
The authors declare that they have no conflict of interest.
References
Altaf M, Utley RT, Lacoste N, Tan S, Briggs SD, Cote J (2007)
Interplay of chromatin modifiers on a short basic patch of histone
H4 tail defines the boundary of telomeric heterochromatin. Mol
Cell 28: 1002–1014
Aparicio OM, Billington BL, Gottschling DE (1991) Modifiers of
position effect are shared between telomeric and silent mating-
type loci in S. cerevisiae. Cell 66: 1279–1287
Armstrong CM, Kaeberlein M, Imai SI, Guarente L (2002) Mutations
in Saccharomyces cerevisiae gene SIR2 can have differential
effects on in vivo silencing phenotypes and in vitro histone
deacetylation activity. Mol Biol Cell 13: 1427–1438
Borra MT, Langer MR, Slama JT, Denu JM (2004) Substrate speci-
ficity and kinetic mechanism of the Sir2 family of NAD+-depen-
dent histone/protein deacetylases. Biochemistry 43: 9877–9887
Buchberger JR, Onishi M, Li G, Seebacher J, Rudner AD, Gygi SP,
Moazed D (2008) Sir3-nucleosome interactions in spreading of
silent chromatin in Saccharomyces cerevisiae. Mol Cell Biol 28:
6903–6918
Carmen AA, Milne L, Grunstein M (2002) Acetylation of the yeast
histone H4 N terminus regulates its binding to heterochromatin
protein SIR3. J Biol Chem 277: 4778–4781
Celic I, Masumoto H, Griffith WP, Meluh P, Cotter RJ, Boeke JD,
Verreault A (2006) The sirtuins hst3 and Hst4p preserve genome
integrity by controlling histone h3 lysine 56 deacetylation. Curr
Biol 16: 1280–1289
Chang CS, Pillus L (2009) Collaboration between the essential Esa1
acetyltransferase and the Rpd3 deacetylase is mediated by H4K12
histone acetylation in Saccharomyces cerevisiae. Genetics 183:
149–160
Chen L, Widom J (2005) Mechanism of transcriptional silencing in
yeast. Cell 120: 37–48
Chou CC, Li YC, Gartenberg MR (2008) Bypassing Sir2 and O-acetyl-
ADP-ribose in transcriptional silencing. Mol Cell 31: 650–659
Cubizolles F, Martino F, Perrod S, Gasser SM (2006) A homotrimer-
heterotrimer switch in Sir2 structure differentiates rDNA and
telomeric silencing. Mol Cell 21: 825–836
Davey CA, Sargent DF, Luger K, Maeder AW, Richmond TJ
(2002) Solvent mediated interactions in the structure of the
nucleosome core particle at 1.9 a resolution. J Mol Biol 319:
1097–1113
Dodd IB, Micheelsen MA, Sneppen K, Thon G (2007) Theoretical
analysis of epigenetic cell memory by nucleosome modification.
Cell 129: 813–822
Ehrenhofer-Murray AE, Rivier DH, Rine J (1997) The role of Sas2,
an acetyltransferase homologue of Saccharomyces cerevisiae, in
silencing and ORC function. Genetics 145: 923–934
Frederiks F, Tzouros M, Oudgenoeg G, van Welsem T, Fornerod M,
Krijgsveld J, van Leeuwen F (2008) Nonprocessive methylation
by Dot1 leads to functional redundancy of histone H3K79 methy-
lation states. Nat Struct Mol Biol 15: 550–557
Gao L, Gross DS (2008) Sir2 silences gene transcription by targeting
the transition between RNA polymerase II initiation and elonga-
tion. Mol Cell Biol 28: 3979–3994
Gasser SM, Cockell MM (2001) The molecular biology of the SIR
proteins. Gene 279: 1–16
Ghidelli S, Donze D, Dhillon N, Kamakaka RT (2001) Sir2p exists in
two nucleosome-binding complexes with distinct deacetylase
activities. EMBO J 20: 4522–4535
Gottschling DE (1992) Telomere-proximal DNA in Saccharomyces
cerevisiae is refractory to methyltransferase activity in vivo.Proc
Natl Acad Sci USA 89: 4062–4065
Han J, Zhou H, Horazdovsky B, Zhang K, Xu RM, Zhang Z (2007)
Rtt109 acetylates histone H3 lysine 56 and functions in DNA
replication. Science 315: 653–655
Hecht A, Laroche T, Strahl-Bolsinger S, Gasser SM, Grunstein M
(1995) Histone H3 and H4 N-termini interact with SIR3 and SIR4
proteins: a molecular model for the formation of heterochromatin
in yeast. Cell 80: 583–592
Hickman MA, Rusche LN (2010) Transcriptional silencing functions
of the yeast protein Orc1/Sir3 subfunctionalized after gene
duplication. Proc Natl Acad Sci USA 107: 19384–19389
Hoppe GJ, Tanny JC, Rudner AD, Gerber SA, Danaie S, Gygi SP,
Moazed D (2002) Steps in assembly of silent chromatin in yeast:
Sir3-independent binding of a Sir2/Sir4 complex to silencers and
role for Sir2-dependent deacetylation. Mol Cell Biol 22: 4167–4180
Huynh VA, Robinson PJ, Rhodes D (2005) A method for the in vitro
reconstitution of a defined ‘30 nm’ chromatin fibre containing
stoichiometric amounts of the linker histone. J Mol Biol 345:
957–968
Hyland EM, Cosgrove MS, Molina H, Wang D, Pandey A, Cottee RJ,
Boeke JD (2005) Insights into the role of histone H3 and histone
H4 core modifiable residues in Saccharomyces cerevisiae. Mol
Cell Biol 25: 10060–10070
Imai S, Armstrong CM, Kaeberlein M, Guarente L (2000)
Transcriptional silencing and longevity protein Sir2 is an NAD-
dependent histone deacetylase. Nature 403: 795–800
Johnson A, Li G, Sikorski TW, Buratowski S, Woodcock CL, Moazed
D (2009) Reconstitution of heterochromatin-dependent transcrip-
tional gene silencing. Mol Cell 35: 769–781
Johnson LM, Fisher-Adams G, Grunstein M (1992) Identification
of a non-basic domain in the histone H4 N-terminus required
for repression of the yeast silent mating loci. EMBO J 11 :
2201–2209
Johnson LM, Kayne PS, Kahn ES, Grunstein M (1990) Genetic
evidence for an interaction between SIR3 and histone H4 in the
repression of the silent mating loci in Saccharomyces cerevisiae.
Proc Natl Acad Sci USA 87: 6286–6290
Katan-Khaykovich Y, Struhl K (2005) Heterochromatin formation
involves changes in histone modifications over multiple cell
generations. EMBO J 24: 2138–2149
Kimura A, Umehara T, Horikoshi M (2002) Chromosomal gradient
of histone acetylation established by Sas2p and Sir2p functions as
a shield against gene silencing. Nat Genet 32: 370–377
Lee KM, Narlikar G (2001) Assembly of nucleosomal templates by
salt dialysis. Curr Protoc Mol Biol Chapter 21: Unit 21.6
Li Q, Zhou H, Wurtele H, Davies B, Horazdovsky B, Verreault A,
Zhang Z (2008) Acetylation of histone H3 lysine 56 regulates
replication-coupled nucleosome assembly. Cell 134: 244–255
Liou GG, Tanny JC, Kruger RG, Walz T, Moazed D (2005) Assembly
of the SIR complex and its regulation by O-acetyl-ADP-ribose,
a product of NAD-dependent histone deacetylation. Cell 121:
515–527
Loo S, Rine J (1994) Silencers and domains of generalized repres-
sion. Science 264: 1768–1771
Lowary PT, Widom J (1998) New DNA sequence rules for high
affinity binding to histone octamer and sequence-directed nucleo-
some positioning. J Mol Biol 276: 19–42
Lu X, Simon MD, Chodaparambil JV, Hansen JC, Shokat KM, Luger
K (2008) The effect of H3K79 dimethylation and H4K20 trimethy-
lation on nucleosome and chromatin structure. Nat Struct Mol
Biol 15: 1122–1124
Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ (1997)
Crystal structure of the nucleosome core particle at 2.8 A resolu-
tion. Nature 389: 251–260
Lynch PJ, Rusche LN (2009) A silencer promotes the assembly of
silenced chromatin independently of recruitment. Mol Cell Biol
29: 43–56
Maas NL, Miller KM, DeFazio LG, Toczyski DP (2006) Cell cycle and
checkpoint regulation of histone H3 K56 acetylation by Hst3 and
Hst4. Mol Cell 23: 109–119
Martino F, Kueng S, Robinson P, Tsai-Pflugfelder M, van Leeuwen F,
Ziegler M, Cubizolles F, Cockell MM, Rhodes D, Gasser SM (2009)
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
The EMBO Journal VOL 30 |NO 13 |2011 &2011 European Molecular Biology Organization2620
Reconstitution of yeast silent chromatin: multiple contact sites
and O-AADPR binding load SIR complexes onto nucleosomes in
vitro.Mol Cell 33: 323–334
Masumoto H, Hawke D, Kobayashi R, Verreault A (2005) A role for
cell-cycle-regulated histone H3 lysine 56 acetylation in the DNA
damage response. Nature 436: 294–298
Megee PC, Morgan BA, Mittman BA, Smith MM (1990) Genetic
analysis of histone H4: essential role of lysines subject to rever-
sible acetylation. Science 247: 841–845
Meijsing SH, Ehrenhofer-Murray AE (2001) The silencing complex
SAS-I links histone acetylation to the assembly of repressed
chromatin by CAF-I and Asf1 in Saccharomyces cerevisiae.
Genes Dev 15: 3169–3182
Michishita E, McCord RA, Boxer LD, Barber MF, Hong T, Gozani O,
Chua KF (2009) Cell cycle-dependent deacetylation of telomeric
histone H3 lysine K56 by human SIRT6. Cell Cycle 8: 2664–2666
Millar CB, Kurdistani SK, Grunstein M (2004) Acetylation of yeast
histone H4 lysine 16: a switch for protein interactions in hetero-
chromatin and euchromatin. Cold Spring Harb Symp Quant Biol
69: 193–200
Moazed D, Kistler A, Axelrod A, Rine J, Johnson AD (1997) Silent
information regulator protein complexes in Saccharomyces cere-
visiae: a SIR2/SIR4 complex and evidence for a regulatory
domain in SIR4 that inhibits its interaction with SIR3. Proc Natl
Acad Sci USA 94: 2186–2191
Mukhopadhyay S, Nagaraj VH, Sengupta AM (2010) Locus
dependence in epigenetic chromatin silencing. Biosystems 102:
49–54
Neumann H, Hancock SM, Buning R, Routh A, Chapman L, Somers
J, Owen-Hughes T, van Noort J, Rhodes D, Chin JW (2009) A
method for genetically installing site-specific acetylation in re-
combinant histones defines the effects of H3 K56 acetylation. Mol
Cell 36: 153–163
Neumann H, Peak-Chew SY, Chin JW (2008) Genetically encoding
N(epsilon)-acetyllysine in recombinant proteins. Nat Chem Biol
4: 232–234
Ng HH, Ciccone DN, Morshead KB, Oettinger MA, Struhl K (2003)
Lysine-79 of histone H3 is hypomethylated at silenced loci in
yeast and mammalian cells: a potential mechanism for position-
effect variegation. Proc Natl Acad Sci USA 100: 1820–1825
Ng HH, Feng Q, Wang H, Erdjument-Bromage H, Tempst P, Zhang Y,
Struhl K (2002) Lysine methylation within the globular domain of
histone H3 by Dot1 is important for telomeric silencing and Sir
protein association. Genes Dev 16: 1518–1527
Norris A, Bianchet MA, Boeke JD (2008) Compensatory interactions
between Sir3p and the nucleosomal LRS surface imply their direct
interaction. PLoS Genet 4: e1000301
Onishi M, Liou GG, Buchberger JR, Walz T, Moazed D (2007) Role
of the conserved Sir3-BAH domain in nucleosome binding and
silent chromatin assembly. Mol Cell 28: 1015–1028
Osborne EA, Dudoit S, Rine J (2009) The establishment of gene
silencing at single-cell resolution. Nat Genet 41: 800–806
Park EC, Szostak JW (1990) Point mutations in the yeast histone H4
gene prevent silencing of the silent mating type locus HML. Mol
Cell Biol 10: 4932–4934
Pillus L, Rine J (1989) Pigenetic inheritance of transcriptional states
in S. cerevisiae. Cell 59: 637–647
Reifsnyder C, Lowell J, Clarke A, Pillus L (1996) Yeast SAS silencing
genes and human genes associated with AML and HIV-1 Tat inter-
actions are homologous with acetyltransferases. Nat Genet 14:
42–49
Rine J, Herskowitz I (1987) Four genes responsible for a position
effect on expression from HML and HMR in Saccharomyces
cerevisiae. Genetics 116: 9–22
Robinson PJ, An W, Routh A, Martino F, Chapman L, Roeder RG,
Rhodes D (2008) 30 nm chromatin fibre decompaction requires
both H4-K16 acetylation and linker histone eviction. J Mol Biol
381: 816–825
Rudner AD, Hall BE, Ellenberger T, Moazed D (2005) A nonhistone
protein-protein interaction required for assembly of the SIR
complex and silent chromatin. Mol Cell Biol 25: 4514–4528
Rusche LN, Kirchmaier AL, Rine J (2003) The establishment, inheri-
tance, and function of silenced chromatin in Saccharomyces cere-
visiae. Annu Rev Biochem 72: 481–516
Sampath V, Yuan P, Wang IX, Prugar E, van Leeuwen F, Sternglanz R
(2009) Mutational analysis of the Sir3 BAH domain reveals
multiple points of interaction with nucleosomes. Mol Cell Biol
29: 2532–2545
Schneider J, Bajwa P, Johnson FC, Bhaumik SR, Shilatifard A (2006)
Rtt109 is required for proper H3K56 acetylation: a chromatin
mark associated with the elongating RNA polymerase II. J Biol
Chem 281: 37270–37274
Schwaiger M, Kohler H, Oakeley EJ, Stadler MB, Schubeler D (2010)
Heterochromatin protein 1 (HP1) modulates replication timing of
the Drosophila genome. Genome Res 20: 771–780
Sekinger EA, Gross DS (2001) Silenced chromatin is permissive to
activator binding and PIC recruitment. Cell 105 : 403–414
Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL
(2006) Histone H4-K16 acetylation controls chromatin structure
and protein interactions. Science 311: 844–847
Smith CM, Gafken PR, Zhang Z, Gottschling DE, Smith JB, Smith DL
(2003) Mass spectrometric quantification of acetylation at specific
lysines within the amino-terminal tail of histone H4. Anal
Biochem 316: 23–33
Smith JS, Brachmann CB, Celic I, Kenna MA, Muhammad S, Starai
VJ, Avalos JL, Escalante-Semerena JC, Grubmeyer C, Wolberger
C, Boeke JD (2000) A phylogenetically conserved NAD+-depen-
dent protein deacetylase activity in the Sir2 protein family. Proc
Natl Acad Sci USA 97: 6658–6663
Strahl-Bolsinger S, Hecht A, Luo K, Grunstein M (1997) SIR2 and
SIR4 interactions differ in core and extended telomeric hetero-
chromatin in yeast. Genes Dev 11 : 83–93
Suka N, Luo K, Grunstein M (2002) Sir2p and Sas2p opposingly
regulate acetylation of yeast histone H4 lysine16 and spreading of
heterochromatin. Nat Genet 32: 378–383
Suka N, Suka Y, Carmen AA, Wu J, Grunstein M (2001) Highly
specific antibodies determine histone acetylation site usage in
yeast heterochromatin and euchromatin. Mol Cell 8: 473–479
Tanny JC, Dowd GJ, Huang J, Hilz H, Moazed D (1999) An
enzymatic activity in the yeast Sir2 protein that is essential for
gene silencing. Cell 99: 735–745
van Leeuwen F, Gafken PR, Gottschling DE (2002) Dot1p modulates
silencing in yeast by methylation of the nucleosome core. Cell
109: 745–756
Varv S, Kristjuhan K, Peil K, Looke M, Mahlakoiv T, Paapsi K,
Kristjuhan A (2010) Acetylation of H3 K56 is required for RNA
polymerase II transcript elongation through heterochromatin in
yeast. Mol Cell Biol 30: 1467–1477
Verzijlbergen KF, Faber AW, Stulemeijer IJ, van Leeuwen F (2009)
Multiple histone modifications in euchromatin promote
heterochromatin formation by redundant mechanisms in
Saccharomyces cerevisiae. BMC Mol Biol 10: 76
Williams SK, Truong D, Tyler JK (2008) Acetylation in the globular
core of histone H3 on lysine-56 promotes chromatin disassembly
during transcriptional activation. Proc Natl Acad Sci USA 105:
9000–9005
Xie W, Song C, Young NL, Sperling AS, Xu F, Sridharan R, Conway
AE, Garcia BA, Plath K, Clark AT, Grunstein M (2009) Histone h3
lysine 56 acetylation is linked to the core transcriptional network
in human embryonic stem cells. Mol Cell 33: 417–427
Xu F, Zhang K, Grunstein M (2005) Acetylation in histone H3
globular domain regulates gene expression in yeast. Cell 121:
375–385
Xu F, Zhang Q, Zhang K, Xie W, Grunstein M (2007) Sir2 deacety-
lates histone H3 lysine 56 to regulate telomeric heterochromatin
structure in yeast. Mol Cell 27: 890–900
Yang B, Britton J, Kirchmaier AL (2008a) Insights into the impact of
histone acetylation and methylation on Sir protein recruitment,
spreading, and silencing in Saccharomyces cerevisiae. J Mol Biol
381: 826–844
Yang B, Kirchmaier AL (2006) Bypassing the catalytic activity of
SIR2 for SIR protein spreading in Saccharomyces cerevisiae. Mol
Biol Cell 17: 5287–5297
Yang B, Miller A, Kirchmaier AL (2008b) HST3/HST4-dependent
deacetylation of lysine 56 of histone H3 in silent chromatin. Mol
Biol Cell 19: 4993–5005
Zou Y, Bi X (2008) Positive roles of SAS2 in DNA replication and
transcriptional silencing in yeast. Nucleic Acids Res 36: 5189–5200
Dual role of H4K16
ac
in yeast silencing
M Oppikofer et al
&2011 European Molecular Biology Organization The EMBO Journal VOL 30 |NO 13 |2011 2621
... Sir3 prefers to bind to deacetylated H4 tails, specifically amino acid H4K16 [6,152]. In an alternative view, based on the observed affinity of the Sir2/3/4 complex for acetylated H4K16, the complex is thought to bind acetylated nucleosomes first, and then acquire additional stability via deacetylation of H4K16 and docking of Sir3 to the deacetylated tails [153]. As a result of successive spreading, Sir2/3/4 binding, histone deacetylation and interactions between Sir2/3/4 complexes expand the silent chromatin domain until either a barrier is reached, or the pool of free SIR proteins falls below a threshold that facilitates efficient binding. ...
... as well as biochemical studies [149][150][151]153,157,[169][170][171]. The co-crystal structure of the nucleosome and the Sir3 BAH domain [10] visualized how this antagonism by H4K16 acetylation, but also H3K79 methylation would regulate Sir3 association with the nucleosome surface. ...
Article
Full-text available
Heterochromatin is a gene-repressive protein–nucleic acid ultrastructure that is initially nucleated by DNA sequences. However, following nucleation, heterochromatin can then propagate along the chromatin template in a sequence-independent manner in a reaction termed spreading. At the heart of this process are enzymes that deposit chemical information on chromatin, which attracts the factors that execute chromatin compaction and transcriptional or co/post-transcriptional gene silencing. Given that these enzymes deposit guiding chemical information on chromatin they are commonly termed ‘writers’. While the processes of nucleation and central actions of writers have been extensively studied and reviewed, less is understood about how the spreading process is regulated. We discuss how the chromatin substrate is prepared for heterochromatic spreading, and how trans-acting factors beyond writer enzymes regulate it. We examine mechanisms by which trans-acting factors in Suv39, PRC2, SETDB1 and SIR writer systems regulate spreading of the respective heterochromatic marks across chromatin. While these systems are in some cases evolutionarily and mechanistically quite distant, common mechanisms emerge which these trans-acting factors exploit to tune the spreading reaction.
... Deacetylation of histone H4, specifically of the Lysine 16, allows the docking of Sir3 (Aparicio et al., 1991;Hecht et al., 1995Hecht et al., , 1996) that in turn recruits Sir4/Sir2 complex, expanding the growing of the silent chromatin on adjacent histones. Loading of the Sir complex on nucleosomes increases the steric hindrance, as demonstrated by a reduced accessibility for restriction enzyme digestion or Dam DNA methyltransferase (Gottschling, 1992;Oppikofer et al., 2011), resulting in a repression of the underlying genes ( Figure 6). However, analysis of chromatin distribution of Sir proteins at natural budding yeast subtelomeres showed a discontinuous binding of the complex (Ellahi et al., 2015;Radman-Livaja et al., 2011). ...
... It has been proposed that Rpd3L deacetylase complex creates this transition zones devoid of acetylation impairing the binding of the Sir complex. Indeed, active deacetylation by Sir2 has been shown to be necessary for a stabilized binding of Sir complex on nucleosomes (Ehrentraut et al., 2010;Oppikofer et al., 2011;Zhou et al., 2009). In addition, H4 dependent acetylation by Sas2 was shown to be important for the deposition of H2A.Z, a histone variant exhibiting barrier activity (Raisner et al., 2005;Shia et al., 2006). ...
Thesis
Telomeres, the nucleoprotein structures located at the end of eukaryotic DNA, protect chromosomal integrity. These structures undergo changes during development and aging, including length shortening and alterations in the levels of the proteins associated to them called sheltering, all this affecting genome stability as the cells age. However, telomeres also behave as transcriptional regulators acting not only on genes present at subtelomeres but also on more distantly located genes presented throughout the genome. This process is referred as Telomere Position Effect (TPE) and was initially discovered in budding yeast, but also seen in drosophila, fission yeast, plasmodium and more recently in humans. In all these organisms, genes located in the subtelomeres are repressed by an epigenetic mechanism that is dependent on telomere DNA length, telomere nucleoprotein composition and higher order chromatin organization adopted by telomeres and subtelomeres. The TPE mechanism can be described as the spreading of a heterochromatin-like structure toward the centromere most likely accompanied by the formation of large chromatin loops to further extend the transcriptional regulation emanating from a telomere to genes internally located.In this context, the goal of my thesis is to decipher whether telomeres are involved in the transcriptional remodelling occurring in human cellular senescence. For that, we performed RNA-sequencing in young versus replicative senescent lung fibroblast MRC-5 cells. Interestingly, we found an enrichment of upregulated genes in the subtelomeric regions of senescent cells suggesting a TPE alleviation. This alleviation is not homogeneous in the genome, as only some subtelomeres were enriched in upregulated genes at senescence.Finally, we tested the hypothesis that shelterin proteins may also be part of the TPE regulation. For that, we re-stored the levels of the shelterin protein TRF2 (Telomeric Repeat Binding Factor 2) whose expression is decreased as the cells approach senescence. We found that TRF2 is indeed modulating the expression of subtelomeric genes in senescent cells, and this is in part mediated by a long-range chromatin reorganization of subtelomeres as observed by conformation changes in 3D chromatin conformation by FISH.Overall, this work reveals the contribution of telomeres in the transcriptional program of senescent cells and set the basis for the relevance of TPE in the senescence/aging process.
... However, autophagy is a double-edged sword in cerebral ischemia. The acetylation of H4K16 is associated with I/R-induced autophagic activation [212][213][214]. Another study showed that EA can trigger the molecular histone switch of SIRT1 and reduce the acetylation of H4K16, thus promoting the expression of LC3II/I and Beclin1 and reducing I/R damage [215]. ...
Article
Full-text available
Stroke is a major cause of mortality and disability globally, with ischemic stroke (IS) accounting for over 80% of all stroke cases. The pathological process of IS involves numerous signal molecules, among which are the highly conserved nicotinamide adenine dinucleotide (NAD+)-dependent enzymes known as sirtuins (SIRTs). SIRTs modulate various biological processes, including cell differentiation, energy metabolism, DNA repair, inflammation, and oxidative stress. Importantly, several studies have reported a correlation between SIRTs and IS. This review introduces the general aspects of SIRTs, including their distribution, subcellular location, enzyme activity, and substrate. We also discuss their regulatory roles and potential mechanisms in IS. Finally, we describe the current therapeutic methods based on SIRTs, such as pharmacotherapy, non-pharmacological therapeutic/rehabilitative interventions, epigenetic regulators, potential molecules, and stem cell-derived exosome therapy. The data collected in this study will potentially contribute to both clinical and fundamental research on SIRTs, geared towards developing effective therapeutic candidates for future treatment of IS.
... Read-write mechanisms have been defined for heritable histone deacetylation, H3K9 methylation, and H3K27 methylation. Deacetylated nucleosomes of the HM loci or telomeres are recognized by the Sir3 protein, which both binds to unacetylated histone H4 lysine 16 78 and recruits the Sir2 histone deacetylase, facilitating the inheritance and spreading of silencing. Likewise, H3K9 methylation is recognized by both the HP1 protein and a chromodomain within the Clr4 H3K9 methyltransferase. ...
Article
Full-text available
Epigenetic transcriptional regulation frequently requires histone modifications. Some, but not all, of these modifications are able to template their own inheritance. Here, I discuss the molecular mechanisms by which histone modifications can be inherited and relate these ideas to new results about epigenetic transcriptional memory, a phenomenon that poises recently repressed genes for faster reactivation and has been observed in diverse organisms. Recently, we found that the histone H3 lysine 4 dimethylation that is associated with this phenomenon plays a critical role in sustaining memory and, when factors critical for the establishment of memory are inactivated, can be stably maintained through multiple mitoses. This chromatin‐mediated inheritance mechanism may involve a physical interaction between an H3K4me2 reader, SET3C, and an H3K4me2 writer, Spp1⁻ COMPASS. This is the first example of a chromatin‐mediated inheritance of a mark that promotes transcription.
... Introduction Silent information regulator 2 (Sir2) is a conserved NAD + -dependent histone deacetylase with homologues in Archaea, Eubacteria and most if not all eukaryotes [1][2][3]. In Saccharomyces cerevisiae, Sir2 targets lysine residues on both histone H3 and H4 [2,4,5], where deacetylation of lysine 16 (K16) on histone H4 is of particular importance [6]. SIR2 was first found in a screen for mutations that derepressed the cryptic mating-type loci (HMLα and HMRa) in S. cerevisiae [3]. ...
Article
Full-text available
Telomere chromatin structure is pivotal for maintaining genome stability by regulating the binding of telomere-associated proteins and inhibiting the DNA damage response. In Saccharomyces cerevisiae , silent information regulator (Sir) proteins bind to terminal repeats and to subtelomeric X-elements, resulting in transcriptional silencing. Herein, we show that sir2 mutant strains display a specific loss of a nucleosome residing in the X-elements and that this deficiency is remarkably consistent between different telomeres. The X-elements contain several binding sites for the transcription factor Reb1 and we found that Sir2 and Reb1 compete for stabilizing/destabilizing this nucleosome, i.e. inactivation of Reb1 in a sir2 background reinstated the lost nucleosome. The telomeric-repeat-containing RNAs (TERRAs) originate from subtelomeric regions and extend into the terminal repeats. Both Sir2 and Reb1 repress TERRAs and in a sir2 reb1 double mutant, TERRA levels increased synergistically, showing that Sir2 and Reb1 act in different pathways for repressing TERRAs. We present evidence that Reb1 restricts TERRAs by terminating transcription. Mapping the 5′-ends of TERRAs from several telomeres revealed that the Sir2-stabilized nucleosome is the first nucleosome downstream from the transcriptional start site for TERRAs. Finally, moving an X-element to a euchromatic locus changed nucleosome occupancy and positioning, demonstrating that X-element nucleosome structure is dependent on the local telomere environment.
... It has previously been reported that certain HDACs may be recruited by a specific acetyl mark. For instance, H4K16-Ac, a target of Sir2, has been shown to promote the binding of Sir2 to chromatin (65). It is also possible that other acetyl marks may be targeted primarily by Rpd3, potentially including H4K8-Ac, reconfiguring the chromatin so as to interfere with Hst1 activity at different marks, the net result of which might be decreased silencing by Hst1. ...
Article
Full-text available
NAD⁺ is a cellular redox cofactor involved in many essential processes. The regulation of NAD⁺ metabolism and the signaling networks reciprocally interacting with NAD⁺-producing metabolic pathways are not yet fully understood. The NAD⁺-dependent histone deacetylase (HDAC) Hst1 has been shown to inhibit de novo NAD⁺ synthesis by repressing biosynthesis of nicotinic acid (BNA) gene expression. Here, we alternatively identify HDAC Rpd3 as a positive regulator of de novo NAD⁺ metabolism in the budding yeast Saccharomyces cerevisiae. We reveal that deletion of RPD3 causes marked decreases in the production of de novo pathway metabolites, in direct contrast to deletion of HST1. We determined the BNA expression profiles of rpd3Δ and hst1Δ cells to be similarly opposed, suggesting the two HDACs may regulate the BNA genes in an antagonistic fashion. Our ChIP analysis revealed Rpd3 and Hst1 mutually influence each other’s binding distribution at the BNA2 promoter. We demonstrate Hst1 to be the main deacetylase active at the BNA2 promoter, with hst1Δ cells displaying increased acetylation of the N-terminal tail lysine residues of histone H4, H4K5 and H4K12. Conversely, we show that deletion of RPD3 reduces the acetylation of these residues in a Hst1-dependent manner. This suggests Rpd3 may function to oppose spreading of Hst1-dependent heterochromatin and represents a unique form of antagonism between HDACs in regulating gene expression. Moreover, we found that Rpd3 and Hst1 also co-regulate additional targets involved in other branches of NAD⁺ metabolism. These findings help elucidate the complex interconnections involved in effecting the regulation of NAD⁺ metabolism.
... Among the known acetylated lysine residues, histone H4 carries four lysines in its N-terminal tail, namely lysines 5, 8, 12, and 16 (K5, K8, K12, and K16). Acetylation of the H4K16 site was found to regulate important processes such as gene silencing and transcription (Oppikofer et al., 2011). Methylation of specific histone-like residues can turn on or repress transcription in microorganisms (Lämke et al., 2016;Fabrizio et al., 2019). ...
Article
Full-text available
Microbial cell factories have been widely used in the production of various chemicals. Although synthetic biology is useful in improving the cell factories, adaptation is still widely applied to enhance its complex properties. Adaptation is an important strategy for enhancing stress tolerance in microbial cell factories. Adaptation involves gradual modifications of microorganisms in a stressful environment to enhance their tolerance. During adaptation, microorganisms use different mechanisms to enhance non-preferred substrate utilization and stress tolerance, thereby improving their ability to adapt for growth and survival. In this paper, the progress on the effects of adaptation on microbial substrate utilization capacity and environmental stress tolerance are reviewed, and the mechanisms involved in enhancing microbial adaptive capacity are discussed.
Article
Nucleosome dynamics in the coding region of a transcriptionally active locus is critical for understanding how RNA polymerase II progresses through the gene body. Histone acetylation and deacetylation critically influence nucleosome accessibility during DNA metabolic processes like transcription. Effect of such histone modifications is context and residue dependent. Rather than effect of individual histone residues, the network of modifications of several histone residues in combination generates a chromatin landscape that is conducive for transcription. Here we show that in Saccharomyces cerevisiae, crosstalk between deacetylation of the H4 N-terminal tail residue H4K16 and acetylation of the H3 core domain residue H3K56, promotes RNA polymerase II progression through the gene body. Results indicate that deacetylation of H4K16 precedes and in turn induces H3K56 acetylation. Effectively, recruitment of Rtt109, the HAT responsible for H3K56 acetylation is essentially dependent on H4K16 deacetylation. In Hos2 deletion strains, where H4K16 deacetylation is abolished, both H3K56 acetylation and RNA polymerase II recruitment gets significantly impaired. Notably, H4K16 deacetylation and H3K56 acetylation are found to be essentially dependent on active transcription. In summary, H4K16 deacetylation promotes H3K56 acetylation and the two modifications together work towards successful functioning of RNA polymerase II during active transcription.
Article
Full-text available
The formation of heterochromatin at HML , HMR , and telomeres in Saccharomyces cerevisiae involves two main steps: Recruitment of Sir proteins to silencers and their spread throughout the silenced domain. We developed a method to study these two processes at single base-pair resolution. Using a fusion protein between the heterochromatin protein Sir3 and the non-site-specific bacterial adenine methyltransferase M.EcoGII, we mapped sites of Sir3-chromatin interactions genome-wide using long-read Nanopore sequencing to detect adenines methylated by the fusion protein and by ChIP-seq to map the distribution of Sir3-M.EcoGII. A silencing-deficient mutant of Sir3 lacking its Bromo-Adjacent Homology (BAH) domain, sir3-bah∆ , was still recruited to HML , HMR , and telomeres. However, in the absence of the BAH domain, it was unable to spread away from those recruitment sites. Overexpression of Sir3 did not lead to further spreading at HML , HMR , and most telomeres. A few exceptional telomeres, like 6R, exhibited a small amount of Sir3 spreading, suggesting that boundaries at telomeres responded variably to Sir3 overexpression. Finally, by using a temperature-sensitive allele of SIR3 fused to M.ECOGII , we tracked the positions first methylated after induction and found that repression of genes at HML and HMR began before Sir3 occupied the entire locus.
Preprint
Full-text available
The formation of heterochromatin at HML, HMR , and telomeres in Saccharomyces cerevisiae involves two main steps: Recruitment of Sir proteins to silencers and their spread throughout the silenced domain. We developed a method to study these two processes at single base-pair resolution. Using a fusion protein between the heterochromatin protein Sir3 and the non-site-specific bacterial adenine methyltransferase M.EcoGII, we mapped sites of Sir3-chromatin interactions genome-wide using long-read Nanopore sequencing to detect adenines methylated by the fusion protein. A silencing-deficient mutant of Sir3 lacking its Bromo-Adjacent Homology (BAH) domain, sir3-bahΔ , was still recruited to HML, HMR , and telomeres. However, in the absence of the BAH domain, it was unable to spread away from those recruitment sites. Overexpression of Sir3 did not lead to further spreading at HML, HMR , and most telomeres. A few exceptional telomeres, like 6R, exhibited a small amount of Sir3 spreading, suggesting that boundaries at telomeres responded variably to Sir3 overexpression. Finally, by using a temperature-sensitive allele of SIR3 fused to M.ECOGII , we tracked the positions first methylated after induction and found that repression of genes at HML and HMR began before Sir3 occupied the entire locus.
Article
Full-text available
Despite its conservation in organisms from bacteria to human and its general requirement for transcriptional silencing in yeast, the function of the Sir2 protein is unknown. Here we show that Sir2 can transfer labeled phosphate from nicotinamide adenine dinucleotide to itself and histones in vitro. A modified form of Sir2, which results from its automodification activity, is specifically recognized by anti-mono-ADP-ribose antibodies, suggesting that Sir2 is an ADP-ribosyltransferase. Mutation of a phylogenetically invariant histidine residue in Sir2 abolishes both its enzymatic activity in vitro and its silencing functions in vivo. However, the mutant protein is associated with chromatin and other silencing factors in a manner similar to wild-type Sir2. These findings suggest that Sir2 contains an ADP-ribosyltransferase activity that is essential for its silencing function.
Article
Full-text available
Acetylation of histone H4 on lysine 16 (H4-K16Ac) is a prevalent and reversible posttranslational chromatin modification in eukaryotes. To characterize the structural and functional role of this mark, we used a native chemical ligation strategy to generate histone H4 that was homogeneously acetylated at K16. The incorporation of this modified histone into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and impedes the ability of chromatin to form cross-fiber interactions. H4-K16Ac also inhibits the ability of the adenosine triphosphate-utilizing chromatin assembly and remodeling enzyme ACF to mobilize a mononucleosome, indicating that this single histone modification modulates both higher order chromatin structure and functional interactions between a nonhistone protein and the chromatin fiber.
Article
Full-text available
Current biological models of epigenetic switches built on chromatin modifications lead to strong constraints on the repertoire of dynamic behaviors for the system. We use the structure of the bifurcation diagram of the underlying dynamical system to explain the existing single cell data in silencing by the SIR system in yeast.
Article
Full-text available
The replication of a chromosomal region during S phase can be highly dynamic between cell types that differ in transcriptome and epigenome. Early replication timing has been positively correlated with several histone modifications that occur at active genes, while repressive histone modifications mark late replicating regions. This raises the question if chromatin modulates the initiating events of replication. To gain insights into this question, we have studied the function of heterochromatin protein 1 (HP1), which is a reader of repressive methylation at histone H3 lysine 9, in genome-wide organization of replication. Cells with reduced levels of HP1 show an advanced replication timing of centromeric repeats in agreement with the model that repressive chromatin mediates the very late replication of large clusters of constitutive heterochromatin. Surprisingly, however, regions with high levels of interspersed repeats on the chromosomal arms, in particular on chromosome 4 and in pericentromeric regions of chromosome 2, behave differently. Here, loss of HP1 results in delayed replication. The fact that these regions are bound by HP1 suggests a direct effect. Thus while HP1 mediates very late replication of centromeric DNA, it is also required for early replication of euchromatic regions with high levels of repeats. This observation of opposing functions of HP1 suggests a model where HP1-mediated repeat inactivation or replication complex loading on the chromosome arms is required for proper activation of origins of replication that fire early. At the same time, HP1-mediated repression at constitutive heterochromatin is required to ensure replication of centromeric repeats at the end of S phase.
Article
Full-text available
In Saccharomyces cerevisiae SIR proteins mediate transcriptional silencing, forming heterochromatin structures at repressed loci. Although recruitment of transcription initiation factors can occur even to promoters packed in heterochromatin, it is unclear whether heterochromatin inhibits RNA polymerase II (RNAPII) transcript elongation. To clarify this issue, we recruited SIR proteins to the coding region of an inducible gene and characterized the effects of the heterochromatic structure on transcription. Surprisingly, RNAPII is fully competent for transcription initiation and elongation at the locus, leading to significant loss of heterochromatin proteins from the region. A search for auxiliary factors required for transcript elongation through the heterochromatic locus revealed that two proteins involved in histone H3 lysine 56 acetylation, Rtt109 and Asf1, are needed for efficient transcript elongation by RNAPII. The efficiency of transcription through heterochromatin is also impaired in a strain carrying the K56R mutation in histone H3. Our results show that H3 K56 modification is required for efficient transcription of heterochromatic locus by RNAPII, and we propose that transcription-coupled incorporation of H3 acetylated K56 (acK56) into chromatin is needed for efficient opening of heterochromatic loci for transcription.
Article
Full-text available
Lysine acetylation of histones defines the epigenetic status of human embryonic stem cells and orchestrates DNA replication, chromosome condensation, transcription, telomeric silencing, and DNA repair. A detailed mechanistic explanation of these phenomena is impeded by the limited availability of homogeneously acetylated histones. We report a general method for the production of homogeneously and site-specifically acetylated recombinant histones by genetically encoding acetyl-lysine. We reconstitute histone octamers, nucleosomes, and nucleosomal arrays bearing defined acetylated lysine residues. With these designer nucleosomes, we demonstrate that, in contrast to the prevailing dogma, acetylation of H3 K56 does not directly affect the compaction of chromatin and has modest effects on remodeling by SWI/SNF and RSC. Single-molecule FRET experiments reveal that H3 K56 acetylation increases DNA breathing 7-fold. Our results provide a molecular and mechanistic underpinning for cellular phenomena that have been linked with K56 acetylation.
Article
Solvent binding in the nucleosome core particle containing a 147 base pair, defined-sequence DNA is characterized from the X-ray crystal structure at 1.9 Å resolution. A single-base-pair increase in DNA length over that used previously results in substantially improved clarity of the electron density and accuracy for the histone protein and DNA atomic coordinates. The reduced disorder has allowed for the first time extensive modeling of water molecules and ions.Over 3000 water molecules and 18 ions have been identified. Water molecules acting as hydrogen-bond bridges between protein and DNA are approximately equal in number to the direct hydrogen bonds between these components. Bridging water molecules have a dual role in promoting histone–DNA association not only by providing further stability to direct protein–DNA interactions, but also by enabling formation of many additional interactions between more distantly related elements. Water molecules residing in the minor groove play an important role in facilitating insertion of arginine side-chains. Water structure at the interface of the histones and DNA provides a means of accommodating intrinsic DNA conformational variation, thus limiting the sequence dependency of nucleosome positioning while enhancing mobility.Monovalent anions are bound near the N termini of histone α-helices that are not occluded by DNA phosphate groups. Their location in proximity to the DNA phosphodiester backbone suggests that they damp the electrostatic interaction between the histone proteins and the DNA. Divalent cations are bound at specific sites in the nucleosome core particle and contribute to histone–histone and histone–DNA interparticle interactions. These interactions may be relevant to nucleosome association in arrays.
Article
The mechanism by which chromatin is decondensed to permit access to DNA is largely unknown. Here, using a model nucleosome array reconstituted from recombinant histone octamers, we have defined the relative contribution of the individual histone octamer N-terminal tails as well as the effect of a targeted histone tail acetylation on the compaction state of the 30 nm chromatin fiber. This study goes beyond previous studies as it is based on a nucleosome array that is very long (61 nucleosomes) and contains a stoichiometric concentration of bound linker histone, which is essential for the formation of the 30 nm chromatin fiber. We find that compaction is regulated in two steps: Introduction of H4 acetylated to 30% on K16 inhibits compaction to a greater degree than deletion of the H4 N-terminal tail. Further decompaction is achieved by removal of the linker histone.
Article
DNA sequences that position nucleosomes are of increasing interest because of their relationship to gene regulation in vivo and because of their utility in studies of nucleosome structure and function in vitro. However, at present our understanding of the rules for DNA sequence-directed nucleosome positioning is fragmentary, and existing positioning sequences have many limitations. We carried out a SELEX experiment starting with a large pool of chemically synthetic random. DNA molecules to identify those individuals having the highest affinity for histone octamer. A set of highest-affinity molecules were selected, cloned, and sequenced, their affinities (free energies) for histone octamer in nucleosome reconstitution measured, and their ability to position nucleosomes in vitro assessed by native gel electrophoresis. The selected sequences have higher affinity than previously known natural or non-natural sequences, and have a correspondingly strong nucleosome positioning ability. A variety of analyses including Fourier transform, real-space correlation, and direct counting computations were carried out to assess non-random features in the selected sequences. The results reveal sequence rules that were already identified in earlier studies of natural nucleosomal DNA, together with a large set of new rules having even stronger statistical significance. Possible physical origins of the selected molecules' high affinities are discussed. The sequences isolated in this study should prove valuable for studies of chromatin structure and function in vitro and, potentially, for studies in vivo.
Article
The origin recognition complex (ORC) defines origins of replication and also interacts with heterochromatin proteins in a variety of species, but how ORC functions in heterochromatin assembly remains unclear. The largest subunit of ORC, Orc1, is particularly interesting because it contains a nucleosome-binding BAH domain and because it gave rise to Sir3, a key silencing protein in Saccharomyces cerevisiae, through gene duplication. We examined whether Orc1 possessed a Sir3-like silencing function before duplication and found that Orc1 from the yeast Kluyveromyces lactis, which diverged from S. cerevisiae before the duplication, acts in conjunction with the deacetylase Sir2 and the histone-binding protein Sir4 to generate heterochromatin at telomeres and a mating-type locus. Moreover, the ability of KlOrc1 to spread across a silenced locus depends on its nucleosome-binding BAH domain and the deacetylase Sir2. Interestingly, KlOrc1 appears to act independently of the entire ORC, as other subunits of the complex, Orc4 and Orc5, are not strongly associated with silenced domains. These findings demonstrate that Orc1 functioned in silencing before duplication and suggest that Orc1 and Sir2, both of which are broadly conserved among eukaryotes, may have an ancient history of cooperating to generate chromatin structures, with Sir2 deacetylating histones and Orc1 binding to these deacetylated nucleosomes through its BAH domain.