ArticlePDF Available

Insight Into the Mechanism of Nucleosome Reorganization From Histone Mutants That Suppress Defects in the FACT Histone Chaperone

Authors:

Abstract and Figures

FACT (FAcilitates Chromatin Transcription/Transactions) plays a central role in transcription and replication in eukaryotes by both establishing and overcoming the repressive properties of chromatin. FACT promotes these opposing goals by interconverting nucleosomes between the canonical form and a more open reorganized form. In the forward direction, reorganization destabilizes nucleosomes, while the reverse reaction promotes nucleosome assembly. Nucleosome destabilization involves disrupting contacts among histone H2A-H2B dimers, (H3-H4)(2) tetramers, and DNA. Here we show that mutations that weaken the dimer:tetramer interface in nucleosomes suppress defects caused by FACT deficiency in vivo in the yeast Saccharomyces cerevisiae. Mutating the gene that encodes the Spt16 subunit of FACT causes phenotypes associated with defects in transcription and replication, and we identify histone mutants that selectively suppress those associated with replication. Analysis of purified components suggests that the defective version of FACT is unable to maintain the reorganized nucleosome state efficiently, whereas nucleosomes with mutant histones are reorganized more easily than normal. The genetic suppression observed when the FACT defect is combined with the histone defect therefore reveals the importance of the dynamic reorganization of contacts within nucleosomes to the function of FACT in vivo, especially to FACT's apparent role in promoting progression of DNA replication complexes. We also show that an H2B mutation causes different phenotypes, depending on which of the two similar genes that encode this protein are altered, revealing unexpected functional differences between these duplicated genes and calling into question the practice of examining the effects of histone mutants by expressing them from a single plasmid-borne allele.
Content may be subject to copyright.
INVESTIGATION
Insight Into the Mechanism of Nucleosome
Reorganization From Histone Mutants That
Suppress Defects in the FACT Histone Chaperone
Laura McCullough,* Robert Rawlins,* Aileen Olsen,
Hua Xin,* David J. Stillman,
and Tim Formosa*
,1
*Department of Biochemistry and
y
Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
ABSTRACT FACT (FAcilitates Chromatin Transcription/Transactions) plays a central role in transcription and replication in eukaryotes by
both establishing and overcoming the repressive properties of chromatin. FACT promotes these opposing goals by interconverting
nucleosomes between the canonical form and a more open reorganized form. In the forward direction, reorganization destabilizes
nucleosomes, while the reverse reaction promotes nucleosome assembly. Nucleosome destabilization involves disrupting contacts
among histone H2A-H2B dimers, (H3-H4)
2
tetramers, and DNA. Here we show that mutations that weaken the dimer:tetramer
interface in nucleosomes suppress defects caused by FACT deciency in vivo in the yeast Saccharomyces cerevisiae. Mutating the
gene that encodes the Spt16 subunit of FACT causes phenotypes associated with defects in transcription and replication, and we
identify histone mutants that selectively suppress those associated with replication. Analysis of puried components suggests that the
defective version of FACT is unable to maintain the reorganized nucleosome state efciently, whereas nucleosomes with mutant
histones are reorganized more easily than normal. The genetic suppression observed when the FACT defect is combined with the
histone defect therefore reveals the importance of the dynamic reorganization of contacts within nucleosomes to the function of FACT
in vivo, especially to FACTs apparent role in promoting progression of DNA replication complexes. We also show that an H2B mutation
causes different phenotypes, depending on which of the two similar genes that encode this protein are altered, revealing unexpected
functional differences between these duplicated genes and calling into question the practice of examining the effects of histone
mutants by expressing them from a single plasmid-borne allele.
FACT (FAcilitates Chromatin Transcription/Transactions)
is a highly conserved histone chaperone with roles in
both transcription and DNA replication (Reinberg and Sims
2006; Formosa 2008). In Saccharomyces cerevisiae, FACT
is an Spt16-Pob3 heterodimer whose activity is supported
by the High Mobility Group B (HMGB)-like DNA-binding
protein Nhp6 (Brewster et al. 2001; Formosa et al. 2001).
Susceptibility of nucleosomal DNA to digestion by some
restriction endonucleases in vitro increases dramatically in
the presence of FACT (Xin et al. 2009), indicating that
FACT either induces a structural change or stabilizes an ex-
isting alternative nucleosomal structure (Winkler and Luger
2011). We have called this activity nucleosome reorganiza-
tion(Formosa 2008). FACT enhances binding of TATA se-
quences within nucleosomes by TATA-binding protein (TBP/
Spt15) (Biswas et al. 2005), suggesting that increasing ac-
cess to DNA through reorganization is a physiologically im-
portant role of FACT.
FACT can induce displacement of H2A-H2B dimers from
nucleosomes under some conditions in vitro (Belotserkovskaya
et al. 2003; Xin et al. 2009). However, increased nuclease
sensitivity does not require H2A-H2B loss (Xin et al. 2009).
Reorganized nucleosomes instead appear to have the same
composition as canonical nucleosomes, but the components
are associated with one another in a substantially different
manner. This change in structure increases the probability of
H2A-H2B dimer loss, but dimer loss appears to be just one
possible result of reorganization, not its mechanism. The
effect of FACT on nucleosomes is not common to other his-
tone chaperones, and FACT-mediated reorganization does
not require ATP hydrolysis so it is unlike ATP-dependent
Copyright © 2011 by the Genetics Society of America
doi: 10.1534/genetics.111.128769
Manuscript received March 16, 2011; accepted for publication May 18, 2011
Supporting information is available online at http://www.genetics.org/content/suppl/
2011/05/30/genetics.111.128769.DC1.
1
Corresponding author: Department of Biochemistry, University of Utah School of
Medicine, 15 N. Medical Dr. East, Room 4100, Salt Lake City, UT 84112. E-mail: tim@
biochem.utah.edu
Genetics, Vol. 188, 835846 August 2011 835
chromatin remodeling (Orphanides et al. 1998; Clapier and
Cairns 2009). FACT is essential for viability (Formosa 2008;
Lolas et al. 2010), but the detailed nature of reorganized
nucleosomes, the role of FACT in forming and resolving
alternative nucleosome forms, and the importance of H2A-
H2B dimer displacement to FACT activity in vivo remain
poorly understood.
Partial loss-of-function mutations in the genes encod-
ing FACT subunits cause a range of defects in transcrip-
tion, replication, and other processes (Lycan et al. 1994;
ODonnell et al. 2004, 2009; Formosa 2008). Some of
these defects can be enhanced by decreasing histone acety-
lation (Formosa et al. 2002), by blocking histone H3-K4
methylation (Biswas et al. 2006), by mutating histone genes
themselves (Vandemark et al. 2008), or by inactivating the
Hir/Hpc complex that is involved in regulating histone gene
expression and depositing nucleosomes outside of S phase
(Formosa et al. 2002). Some features of chromatin therefore
support FACT activity in vivo, and their loss makes FACT
defects more difcult to tolerate. In contrast, other chroma-
tin factors oppose FACT activity, as the phenotypes caused
by some FACT gene mutations can be suppressed by pre-
venting methylation of H3-K36 (Biswas et al. 2006) or by
inactivating the chromodomain-helicase Chd1 (Biswas et al.
2008). FACT gene mutations can also either enhance or
suppress defects caused by mutating other chromatin factors
such as the histone H3 or the Swi/Snf remodeling complex
(Malone et al. 1991; Duina et al. 2007). Furthermore, phe-
notypes caused by FACT gene mutations can be either en-
hanced or suppressed by altering the ratio of expression of
H2A-H2B relative to H3-H4 (Formosa et al. 2002). The prop-
erties of chromatin therefore affect the efciency of FACT
function in vivo, but it remains unclear how these properties
inuence the central activity of FACT.
To examine the relationship between FACT and nucleo-
some reorganization in vivo, we sought histone gene muta-
tions that could compensate for FACT defects. We reasoned
that deciency for FACT activity could be counterbalanced
by mutating the histones in a way that makes nucleosomes
easier to reorganize. Such mutations would provide insight
into both the functions of FACT in vivo and the nature of
nucleosome reorganization. Here we report that the temper-
ature sensitivity (Ts) and hydroxyurea sensitivity caused by
the spt16-11 allele of FACT can be suppressed by weakening
the H2A-H2B dimer interface with (H3-H4)
2
tetramers
within nucleosomes, but this does not signicantly affect
the Spt
2
phenotype also caused by spt16-11. Puried nucle-
osomes with these mutated histones display elevated rates
of spontaneous reorganization using nuclease sensitivity as
an assay, and this partially compensates for Spt16-11 pro-
teins defect in achieving stable reorganization in vitro.
These results provide new insights into the mechanism of
FACT activity in vivo, supporting an important role for the
stability of the histone dimer:tetramer interface in nucleo-
some reorganization and revealing a potential role for re-
organization in replication fork progression.
Materials and Methods
Strains are listed in Table 1 and in the supporting informa-
tion,Table S1. W303 strains were derived from MSY1905
(kindly provided by M. Mitchell Smith, University of Vir-
ginia) by conversion of the KanMX marker replacing
HHT1-HHF1 with HIS3 followed by standard crosses within
the W303 background to introduce other mutations. A364a
strains were constructed by integrating markers down-
stream of the histone genes and then amplifying the marked
genomic locus by PCR using an upstream primer containing
the desired mutation 30 nucleotides from the 59end and
25 nucleotides from the 39end and a downstream primer
200 bp distal to the marker (the strategy is outlined in
Figure 3; the primers used are listed in Table S2; Toulmay
and Schneiter 2006). The PCR product was used to trans-
form a wild-type strain, selecting for transfer of the marker
and then screening for cotransfer of the mutation by se-
quencing the entire histone gene. Standard crosses within
the A364a background were then performed to obtain com-
binations of mutations.
The S. cerevisiae genome contains two copies of each of
the genes that encode the four core histones (Osley 1991).
Plasmids pJH33, M4958, and M4959 carrying the genes
HTA1-HTB1 and HHT2-HHF2 encoding histones H2A, H2B,
H3, and H4 in vectors pRS316 (URA3), pRS315 (LEU2), or
pRS314 (TRP1), respectively (Sikorski and Hieter 1989),
were kindly provided by M. Mitchell Smith (University of
Virginia) (Ahn et al. 2005). Mutagenized histone genes were
obtained by amplifying M4958 using primers that align
400 bp into the vector sequences anking the histone gene
insert, resulting in a 5279-bp product with 400 bp of ho-
mology at each end to pRS414 or pRS415 (Figure 1). PCR
was performed under standard conditions (100-ml reactions
containing 20 pmol of each primer, 10 ng template, 0.2 mM
each deoxynucleoside triphosphate (dNTP), 10 mMTrisCl
pH 8.3, 1.5 mMMgCl
2
,50mMKCl; 30 cycles of 1 min at 94,
1 min at 54, 5 min at 72) using Pfu polymerase (a small
number of candidates were generated with a 1:200 mixture
of Pfu:Taq polymerases, but most of these contained multi-
ple mutations). Yeast strains DY10003 and DY10004 with
the spt16-11 allele (Table 1) and carrying pJH33 were trans-
formed with this PCR product mixed with vectors pRS414
(TRP1) or pRS415 (LEU2) (Christianson et al. 1992) that
had been linearized with BamHI and HindIII. This yielded
pLM04 (TRP1) and pTF238 (LEU2) derivatives by recombi-
nation in vivo. About 70,000 transformants were obtained
and replica-plated to medium containing 59-FOA to select
for cells lacking the original wild-type histone plasmid
(Boeke et al. 1987), thus demanding that the mutagenized
plasmids expressed histones adequate for supporting viabil-
ity. Strains surviving with only the mutagenized plasmids
were replica-plated to 37or to medium containing 120 mM
hydroxyurea (HU), conditions nonpermissive for growth of
the parent strains. About 2000 candidates were chosen
for retesting, yielding several hundred suppressed strains.
836 L. McCullough et al.
Plasmids were recovered from 100 of the strongest can-
didates, screened for the expected restriction digestion pat-
tern, and used to transform DY10004 pJH33 to establish
linkage of the suppressing mutation with the plasmid.
Twenty-six of these plasmids were found to contain sup-
pressor mutations and were sequenced using four primers
to fully cover all four histone genes on each plasmid (Table
S2). Eighteen of the plasmids had single mutations affect-
ing 1 of 10 residues in H2A or H2B, distributed as shown in
Table2.Someofthe8plasmidswithmultiplemutations
also affected these same residues, and all plasmids had at
least one mutation in H2A or H2B that mapped to the in-
terface between H2A-H2B and H3-H4, but plasmids with
complex mutations were not analyzed further. The screens
for suppression of temperature sensitivity and HU sensitiv-
ity yielded overlapping results, so the resulting plasmids
were combined for further analysis (Figure 1B).
Nucleosomes were reconstituted in vitro with recombi-
nant histones, labeled with uorescent dyes, and tested for
binding afnity with FACT, sensitivity to DraI digestion, and
retention of H2A-H2B dimers as described previously
(Ruone et al. 2003; Rhoades et al. 2004; Xin et al. 2009).
The spt16-11 (T828I, P859S) (Formosa et al. 2002), hta1-
V101I, and htb1-A84D mutations were introduced into
expression constructs using the Quikchange strategy (Stra-
tagene), and the proteins were puried as previously de-
scribed (Ruone et al. 2003; Rhoades et al. 2004; Xin et al.
2009).
Results
Defects caused by FACT mutants can be suppressed by
histone mutants
The spt16-11 allele causes sensitivity to elevated tempera-
tures, sensitivity to the replication toxin HU, and the
Spt
2
phenotype (Formosa et al. 2001). The amount of Spt16
protein in an spt16-11 mutant drops about vefold after a
3-hr incubation at 37(Vandemark et al. 2008), so the Ts
2
phenotype probably reects simple loss of FACT activity.
HU inhibits ribonucleotide reductase (RNR), causing DNA
replication to stall due to the shortage of dNTPs, but also
Table 1 Strains used
Strain Genotype
Figure 1 W303 background
DY10003 MATaade2 can1 his3 leu2 trp1 ura3 spt16-11 hht1-hhf1-D(::HIS3) hht2-hhf2-Δ(::KanMX)
hta1-htb1-Δ(::NatMX) hta2-htb2-Δ(::HphMX) pJH33 (YCp URA3 HHT2-HHF2,HTA1-HTB1)
DY10004 MATaade2 can1 his3 leu2 trp1 ura3 spt16-11 hht1-hhf1-Δ(::HIS3) hht2-hhf2-Δ(::KanMX)
hta1-htb1-Δ(::NatMX) hta2-htb2-Δ(::HphMX) pJH33 (YCp URA3 HHT2-HHF2,HTA1-HTB1)
Figure 3 A364a background
8127-7-4 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@
8500-10-2 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@HTA1(220,His3MX) HTA2(30,URA3)
8541-3-2 MATaura3 leu2 trp1 his3 lys2-128@hta1-V101I(220,His3MX) hta2-V101I(30,URA3)
8262-11-4 MATaura3 leu2 trp1 his3 lys2-128@spt16-11
8554-5-3 MATaura3 leu2 trp1 his3 lys2-128@spt16-11 HTA1(220,His3MX) HTA2(30,URA3)
8541-4-2 MATaura3 leu2 trp1 his3 lys2-128@hta1-V101I(220,His3MX) hta2-V101I(30,URA3) spt16-11
8127-7-4 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@
8500-10-2 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@HTA1(220,His3MX) HTA2(30,URA3)
8541-3-2 MATaura3 leu2 trp1 his3 lys2-128@hta1-V101I(220,His3MX) hta2-V101I(30,URA3)
8324-2-2 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@pob3-Q308K(LEU2)
8500-2-2 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@HTA1(220,His3MX) HTA2(30,URA3) pob3-Q308K(LEU2)
8555-4-2 MATaura3 leu2 trp1 his3 lys2-128@hta1-V101I(220,His3MX) hta2-V101I(30,URA3) pob3-Q308K(LEU2)
Figure 4 A364a background
8483-9-1 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@HTB1(30,URA3) HTB2(30,His3MX)
8606-3-4 MATaura3 leu2 trp1 his3 lys2-128@htb1-A84D(30,URA3) HTB2(30,His3MX)
8868-5-1 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@HTB1(30,URA3) htb2-A84D(30,His3MX)
8442-4-3 MATaura3-Δ0 leu2-Δ0 trp1-Δ2 his3 lys2-128@htb1-A84D(30,URA3) htb2-A84D(30,His3MX)
8482-5-3 MATaura3 leu2 trp1 his3 lys2-128@HTB1(30,URA3) HTB2(30,His3MX) spt16-11
8606-2-1 MATaura3 leu2 trp1 his3 lys2-128@htb1-A84D(30,URA3) HTB2(30,His3MX) spt16-11
8607-1-1 MATaura3 leu2 trp1 his3 lys2-128@HTB1(30,URA3) htb2-A84D(30,His3MX) spt16-11
8455-8-4 MATaura3 leu2 trp1 his3 lys2-128@htb1-A84D(30,URA3) htb2-A84D(30,His3MX) spt16-11
8625-3-4 MATaura3 leu2 trp1 his3 lys2-128@HTB1(30,URA3) hta2-htb2-Δ(::HIS3) spt16-11
8608-5-4 MATaura3 leu2 trp1 his3 lys2-128@htb1-A84D(30,URA3) hta2-htb2-Δ(::KanMX) spt16-11
Table 2 W303 background
DY9999 MATaade2 can1 his3 leu2 trp1 ura3 hht1-hhf1-Δ(::HIS3) hht2-hhf2-Δ(::KanMX3)
hta1-htb1-Δ(::NatMX) hta2-htb2-Δ(::HphMX) pJH33 (YCp URA3 HHT2-HHF2,HTA1-HTB1)
8264-17-3 MATaade2 can1 his3 leu2 trp1 ura3 pob3-Q308K hht1-hhf1-Δ(::HIS3) hht2-hhf2-Δ(::KanMX3)
hta1-htb1-Δ(::NatMX) hta2-htb2-Δ(::HphMX) pJH33 (YCp URA3 HHT2-HHF2,HTA1-HTB1)
FACT Defects Suppressed by Histone Mutants 837
leading to increased transcription of the genes encoding
RNR. At least some FACT mutants retain normal induction
of RNR gene transcription (Biswas et al. 2008; Formosa
2008), so HU sensitivity often reects a defect in DNA rep-
lication, but indirect effects due to awed transcription are
also possible. The Spt
2
phenotype results from inappropri-
ate transcription initiation start-site selection (Clark-Adams
et al. 1988), indicating that spt16-11 causes a defect in tran-
scription even at temperatures permissive for growth where
Spt16 protein levels are normal. spt16-11 was chosen for the
suppressor analysis described here because it causes a broad
range of phenotypes associated with transcription and
replication.
To detect histone gene mutations that can compensate
for the defects caused by spt16-11, we constructed a strain
with this mutation that lacked genomic versions of histone
genes but instead carried a single copy of the genes encod-
ing H2A, H2B, H3, and H4 on a plasmid (Materials and
Methods). PCR amplication of the insert containing all four
histone genes was used to introduce random mutations, and
alleles capable of suppressing the Ts
2
or HU sensitivity phe-
notypes caused by spt16-11 were identied. This approach
yielded 26 suppressing plasmids, 18 of which carried single
mutations that altered 1 of 10 residues in H2A or H2B
(Table 2, Figure 1B). H2B-A84 mutants were isolated ve
times (A84D four times and A84V once), but mutations af-
fecting other residues were recovered only once or twice.
The screen was therefore not saturated, but all of the mu-
tated residues matched the same physical prole in that they
affected residues that are buried within the histone octamer
core on or near the surface of the H2A-H2B dimer that con-
tacts the (H3-H4)
2
tetramer (indicated in Figure 2 as either
magenta or red residues). These results therefore strongly
suggest that FACT activity in vivo involves disruption of the
dimer:tetramer interface, that the spt16-11 mutation causes
a defect in this function, and that weakening the interface by
mutating residues in the interface reduces the requirement
for efcient FACT. It should also be possible to interfere with
this interface by mutating H3 or H4, so it is puzzling that no
such mutations were identied, but it is possible that mu-
tating the interface from the more highly conserved H3 or
H4 protein side leads to inviability.
Suppressors were isolated for the ability to reverse either
the Ts
2
or the HU sensitivity caused by spt16-11, but each
suppressor was found to suppress both phenotypes to vari-
able but similar extents (Figure 1B). This suggests that both
phenotypes have a common underlying defect. Suppression
of spt16-11 was partially allele-specic, as some of the
mutants weakly suppressed a pob3-Q308K mutation affect-
ing the Pob3 subunit of FACT, but others had no effect and
some even enhanced the defects caused by pob3-Q308K
(Table 2, Figure S1A, Figure S4, and Figure 3). spt16-11
and pob3-Q308K therefore cause distinct defects in FACT
function, and weakening the dimer:tetramer interface is
strongly benecial only to the cells with the spt16-11 de-
ciency. The histone gene mutations had little or no effect
in an SPT16 wild-type strain (Table 2, Figure S1B). Altering
the histones therefore caused a signicant enough effect on
nucleosome structure that an spt16-11 mutation was
strongly suppressed, but the change was not sufcient to
cause an obvious defect in an otherwise normal cell.
Integration of histone gene mutations reveals
differences among expression contexts
FACT mutants are sensitive to histone gene copy-number
variation (Formosa et al. 2002), and it is difcult to maintain
plasmids at uniform copy number throughout a population
of cells. Furthermore, HTA1-HTB1 and HTA2-HTB2 encode
slightly different amino acid sequences (Figure S3), tran-
scription of each gene is regulated by different factors (Osley
and Lycan 1987; Xu et al. 1992; Dollard et al. 1994; Hess
and Winston 2005), and HTA1-HTB1 is essential for viability
but HTA2-HTB2 is not (Formosa et al. 2002; Libuda and
Winston 2006). Either the sequence differences or their
transcription proles under different conditions are there-
fore functionally important. To test the effects of the sup-
pressing mutations in a more native context, we integrated
two of the mutations into the genome at the endogenous
Figure 1 Histone gene mutations can suppress defects caused by an
spt16-11 mutation. (A) Scheme for mutagenizing histone genes. A plas-
mid carrying wild-type HHT2-HHF2 (H3-H4) and HTA1-HTB1 (H2A-H2B)
was used as the template for PCR using primers TF04-25 and TF05-28 in
the vector sequence anking the histone gene insert (Table S2). The
product was mixed with linearized vector DNA and used to transform
DY10003 or DY10004 (Table 1). Recombination in vivo produced muta-
genized histone gene plasmids. (B) Candidate plasmids with the single
mutations indicated were recovered and used to transform strain
DY10004 (Table 1), and then isolates lacking the wild-type histone gene
plasmid were derived. Strains with only the mutated plasmid were grown
to saturation, and aliquots of 10-fold serial dilutions were tested on YPAD
(rich medium; Yeast Extract, Peptone, Adenine, Dextrose) at 25or 34or
on HU (60) (YPAD with 60 mMhydroxyurea) at 25.
838 L. McCullough et al.
loci. We chose H2A-V101I and H2B-A84D for this as they
were two of the strongest suppressors of spt16-11 but they
had opposite effects on the pob3-Q308K allele (Table 2).
We inserted selectable markers downstream of each
gene that encodes H2A or H2B in separate wild-type strains
(Figure S2). We then used genomic DNA from these strains
to amplify each gene with the targeted mutation incor-
porated into one PCR primer (Figure 3A) (Toulmay and
Schneiter 2006). The product carrying the desired mutation
was used to transform a wild-type yeast strain, and trans-
formants were screened by sequencing the entire histone
gene to nd strains in which the desired mutation but no
unexpected additional mutation had been integrated into
the genome along with the selectable marker. Finally, stan-
dard crosses were performed to obtain combinations of
mutations.
Figure 3B shows the results obtained with the H2A-V101I
expressed from both HTA1 and HTA2. Neither the integrated
markers themselves (tags) nor the histone mutations af-
fected the phenotypes tested in an SPT16 strain, but the tags
themselves slightly enhanced the HU sensitivity and Spt
2
phenotypes (but not the Ts
2
) caused by spt16-11 (Figure
3B; compare rows 4 and 5: the Spt
2
phenotype is revealed
by growth of these lys2-128@strains on medium lacking
lysine). Marking the wild-type H2A genes therefore causes
a minor defect in their expression; as noted previously and
consistent with these results, a decrease in H2A-H2B expres-
sion is detrimental to some spt16 mutants (Formosa et al.
2002). To control for this effect, subsequent experiments
always include marked versions of the H2A genes, whether
wild type or mutant. This ensures that all comparisons are
between strains that are as genetically matched as possible,
although it remains possible that the markers could have
a differential effect on mutant and wild-type alleles.
The integrated H2A-V101I mutation suppressed both
the Ts
2
and hydroxyurea sensitivity phenotypes caused by
spt16-11, but had only a slight effect on the Spt
2
phenotype
(Figure 3B; compare rows 5 and 6). H2A-V101I therefore
signicantly corrects the spt16-11 defect most closely asso-
ciated with DNA replication, but it has less effect on the
transcription defect. Integrated H2A-V101I had no effect in
a wild-type strain (Table 2, Figure S1B), but partially sup-
pressed the Ts
2
and HU sensitivity caused by pob3-Q308K
(Table 2, Figure S1A, Figure 3C), generally recapitulating
the results from the plasmid-based assays.
In contrast, the locus expressing H2B-A84D expression
signicantly inuenced the effect of this mutant. In a wild-
type strain, expressing H2B-A84D from HTB1 caused a very
mild Spt
2
phenotype (Figure 4A; compare the medium lack-
ing lysine (2lys) growth at 2 and 6 days with the 2-day
incubation of an spt16-11 mutant in Figure 4B). Expressing
H2B-A84D from HTB2 did not cause this phenotype and did
not enhance the effect of htb1-A84D (Figure 4A; compare
rows 2 and 4). In an spt16-11 strain, the htb1-A84D allele
suppressed the Ts
2
and hydroxyurea sensitivity phenotypes
but did not affect the Spt
2
phenotype; this was true in both
an HTA1 htb1-A84D HTA2 HTB2 strain (normal H2B avail-
able from the second copy of the gene) and an HTA1 htb1-
A84D hta2-htb2-Δstrain (only H2B-A84D available; Figure
4B; compare row 1 with 2 or 6). However, the htb2-A84D
allele had no effect when paired with HTB1 (Figure 4B, row
3) and caused variable effects when paired with htb1-A84D
(Figure 4B, rows 3 and 4). (HTB1 is essential for viability
so the effect of htb1-Δhtb2-A84D could not be assessed.)
Relative to htb1-A84D HTB2 spt16-11, the htb1-A84D htb2-
A84D spt16-11 strain displayed markedly reduced suppres-
sion of the Ts
2
phenotype, increased suppression of the
Spt
2
phenotype, and enhanced sensitivity to low concentra-
tions of HU (Figure 4B, rows 2 and 4).
The effects of the source of the H2B-A84D mutation
are therefore quite complex and vary with the phenotype
being observed. When challenged with HU or elevated
Table 2 Effects of spt16-11 suppressors in wild-type and pob3-Q308K strains
Histone Mutation No. of isolates Phenotypes with pob3-Q308K Phenotypes in wild type
H2A hta1-A87T 2
H2A hta1-R89G 2Ts
2
HUs (enhanced defect) Slight Ts
2
, Slight HUs, NaCl-s
H2A hta1-N100D 1 Ts+ HUr (weak suppression)
H2A hta1-V101I 1 Ts+ HUr (weak suppression) Slight Fmd-s
H2B htb1-T55S 2 Fmd-s
H2B htb1-N66S 1 Fmd-s
H2B htb1-F68S 2Ts
2
HUs (enhanced defect)
H2B htb1-T78P 1Ts
2
HUs (enhanced defect)
H2B htb1-A80V 2Ts
2
HUs (enhanced defect)
H2B htb1-A84V 4 Ts+ HUr (weak suppression)
H2B htb1-A84D 1Ts
2
HUs (enhanced defect) Mild Spt
2
H2B htb1-Y86C 1 Mixed weak effects Slight Fmd-s, Slight HUs
H2B htb1-I104F 1 Ts+ HUr (weak suppression)
Strains 8264-17-3 (pob3-Q308K) and DY9999 (wild type) were transformed with LEU2 plasmids carrying the mutation indicated, and then transformants lacking the wild-
type histone gene plasmid were screened for growth at elevated temperatures or on media containing 30150 mMhydroxyurea, 1.2 M NaCl, 3% formamide, 10 mM
caffeine, or 10 mg/ml camptothecin. The pob3-Q308K mutation causes Ts
2
and HUs phenotypes, and these were unaffected, enhanced, or suppressed as indicated. In all
cases, the effects were small compared to other synthetic interactions observed with this allele. Moderate formamide sensitivity was observed where indicated (Fmd-s), with
all other effects in the wild-type strain being weak. The number of times each mutation was isolated in the original screen for spt16-11 suppression is indicated as No. of
isolates.HUs, hydroxyurea sensitive; HUr, hydroxyurea resistant; NaCl-s, NaCl sensitive.
FACT Defects Suppressed by Histone Mutants 839
temperature, expressing H2B-A84D only from the HTB1 lo-
cus was strongly advantageous to an spt16-11 strain (Figure
4B) but had no apparent effect in a wild-type strain (Figure
4A). However, supplying H2B-A84D only from the HTB2
locus under these stress conditions had no effect on either
spt16-11 or wild-type strains. htb1-A84D caused the same
effects whether normal HTB2 was available or not, suggest-
ing that HTB2 expression is irrelevant in these tests. How-
ever, cells with both htb1-A84D and htb2-A84D mutations
displayed distinct phenotypes compared to the single
mutants, so HTB2 expression is important in this context.
While both HTB1 and HTB2 are transcribed at similar levels
under standard growth conditions (David et al. 2006), these
results show that the two loci have distinct functions under
some circumstances, as observed previously for the heat-
shock response (Norris and Osley 1987) and the ability to
suppress the effects of different Ty1 delta-element insertions
(Clark-Adams et al. 1988). These differences may result
from the slightly different proteins produced by the two
genes (Figure S3), or they may indicate that the two genes
are differentially activated by stress conditions. In any case,
this shows that testing variants only from a plasmid-borne
copy of HTB1 can produce an incomplete picture.
Histone mutations that suppress spt16-11 form unstable
nucleosomes in vitro
The clustering of spt16-11 suppressors in the nucleosome
structure (Figure 2) suggests that the Spt16-11 protein is
defective in a process that includes disruption of the H2A-
H2B interface with (H3-H4)
2
tetramers. To investigate this
possibility as well as other potential mechanisms, we recon-
stituted nucleosomes containing H2A-V101I or H2B-A84D
and tested their stability in vitro. A 181-bp DNA fragment
including the 146-bp sea urchin 5S rDNA nucleosome posi-
tioning sequence was labeled with Cy5 and assembled into
nucleosomes using recombinant yeast histones expressed in
bacteria as described previously (Xin et al. 2009). The H2A-
Q114C mutation was introduced into the HTA1 gene to
provide a unique cysteine residue for labeling this subunit
with a maleimide derivative of the uorescent dye Oregon
Green 488 prior to assembly of octamers. The resulting nu-
cleosomes therefore contained two uorescent dyes that
could be detected independently, allowing us to follow the
DNA and H2A-H2B dimer components of the nucleosome
separately.
To test whether suppressor mutations affect the physical
stability of nucleosomes, we used native polyacrylamide gel
electrophoresis to measure the amount of H2A-H2B dis-
placed from the nucleosomes, the amount of tetrasome
formed, and the amount of free DNA released under various
conditions. Over 90% of a sample of wild-type nucleosomes
remained intact by all three measurements after a 1-hr
incubation at 30in 550 mMNaCl (Figure 5, A and B). In
contrast, nucleosomes constructed with H2A-V101I lost
dimers and formed tetrasomes more readily than wild type,
and these effects were even more pronounced with H2B-
A84D. In the latter case, even nucleosomes not exposed to
high salt migrated aberrantly in native polyacrylamide gels,
consistent with the observed spontaneous loss of dimers
during preparation and storage (Figure 5A, lane 5 and not
shown). Incubation of nucleosomes for 1 hr at 65also
caused low levels of tetrasome formation with wild-type
nucleosomes, but substantially elevated levels for each mu-
tant nucleosome (Figure 5, C and D). Thus, both H2A-V101I
and H2B-A84D cause nucleosome instability in vitro, includ-
ing an increase in dimer displacement.
FACT might either induce nucleosomes to reorganize or
selectively bind to and stabilize spontaneously reorganized
nucleosomes. In the rst case, a weakened dimer:tetramer
interface would make it easier to overcome resistance to
reorganization, and in the second case, it would provide
a larger subpopulation of complexes for binding. In either
Figure 2 Histone gene mutations that suppress
spt16-11 map to the dimer:tetramer interface.
Histone residues identied in the suppressor
screen are shown within the structure of a yeast
nucleosome (PDB 1ID3) (White et al. 2001) as
rendered in MacPyMOL (DeLano Scientic).
Two orientations are shown (top and bottom
panels) with a full nucleosome (left), H2A-H2B
removed (center), or only H2A shown (right) to
reveal buried sites. Sites of suppressor mutants
are shown in magenta or red, with the latter
indicating residues H2A-V101 and H2B-A84
that were chosen for further analysis.
840 L. McCullough et al.
case, these results show that the stability of the dimer:
tetramer interface is an important element of the core
reaction promoted by FACT in vivo.
FACT(Spt16-11) has a nucleosome-binding defect
in vitro
We used puried nucleosomes mixed with normal and
mutant FACT complexes to examine the nature of the
suppression observed in vivo.Werst measured the appar-
ent afnity of FACT for nucleosomes using an electropho-
retic mobility shift assay (EMSA) (Rhoades et al. 2004). The
DNA-binding protein Nhp6 is required for complex forma-
tion in this assay (Formosa et al. 2001; Xin et al. 2009), with
50% saturation occurring at 460 nM Nhp6 (Ruone et al.
2003). This same value was observed for both Spt16-Pob3
and (Spt16-11)-Pob3 with wild-type and mutant nucleo-
somes (not shown), so Spt16-11 and Spt16 proteins each
require about the same amount of Nhp6 to support stable
complex formation. In contrast, Spt16-11 was three- to ve-
fold less effective than Spt16 in this assay (Figure 6A). This
could mean that Spt16-11 has a lower afnity for nucleo-
somes, that it is less effective in performing reorganization,
or that it is unable to maintain the reorganized state stably
enough for detection by EMSA. Nucleosomes with H2A-
V101I or H2B-A84D were the same as wild-type H2A in this
assay (Figure S5), so while this reveals a defect in Spt16-11
activity, it does not reveal the mechanism of genetic
suppression.
spt16-11 and histone mutants have opposing effects on
reorganization rates
Reorganized nucleosomes are more prone to losing one or
both H2A-H2B dimers in vitro (Belotserkovskaya et al. 2003;
Xin et al. 2009). Nuclease sensitivity can be detected in
complete octameric nucleosomes, so dimer loss is not a nec-
essary feature of reorganization, but reorganization can lead
Figure 3 H2A-V101I suppresses some phenotypes caused
by two distinct FACT gene mutations. (A) Schematic
showing the method used for integrating the hta2-V101I
mutation into the genome (strategies for other genes
shown in Figure S2 and Table S2). (B and C) Cultures of
strains isogenic with the A364a genetic background and
the additional genotypes shown (see Table 1 for the full
genotypes) were tested as in Figure 1). Cis complete
synthetic medium; HU (90) and HU (150) are YPAD with
90 or 150 mMhydroxyurea. Growth on medium lacking
lysine (2lys) reveals the Spt
2
phenotype in the strains with
the lys2-128@allele (Simchen et al. 1984). Unless noted,
all strains have marker genes (tags) inserted adjacent to
the normal or mutated histone genes (Figure S2). Strains
labeled H2A-V101Iexpress this mutant protein from
both HTA1 and HTA2 loci. Note that the severity of the
Ts
2
and hydroxyurea sensitivity phenotypes caused by
spt16-11 here is lower than in Figure 1 because the initial
screen (Figure 1) was performed in the W303 genetic
background where FACT gene mutations routinely cause
stronger defects, whereas the integrants shown here were
constructed in the A364a genetic background.
FACT Defects Suppressed by Histone Mutants 841
to dimer loss so it serves as an assay for the ability of FACT
to promote this structural change.
We measured the total dimer displacement after a 10-
min incubation. As expected, FACT caused more dimer
displacement than Nhp6 alone, but the levels for Spt16 and
Spt16-11 were comparable (Figure 6B). Histone mutants
displayed increased dimer loss in the presence of Nhp6
alone, consistent with the results described above showing
inherent instability of these nucleosomes. Nucleosomes
with wild-type histones or with the H2A-V101I protein dis-
played slightly lower dimer displacement with Spt16-11
than with Spt16, but nucleosomes with H2B-A84D had
a similarly small but opposite effect. Spt16-11 therefore
appears to have a small effect on dimer loss after a 10-min
incubation.
We next examined the rate of dimer displacement.
Instead of the 181-bp 5S rDNA nucleosomes containing
recombinant yeast histones used above, we used a 255-bp
MMTV (Mouse Mammary Tumor Virus) sequence (Flaus
et al. 2004), recombinant Xenopus laevis histones, and low
Mg
2+
ion concentrations. These conditions increase the total
dimer loss caused by FACT (Xin et al. 2009) and increase the
electrophoretic separation of octameric and tetrameric
nucleosomes. These nucleosomes migrate as one major band
in native gels (Flaus et al. 2004) (Figure 6C, Nuc) along
with several minor translational variants, while recon-
structions show that tetrasomes migrate to four main bands
(Figure 6C, Tet). FACT does not appear to promote trans-
location of nucleosomes (Rhoades et al. 2004), so we infer
that the multiple forms observed with this larger MMTV
fragment are corresponding pairs of octasomal and tetraso-
mal species occupying the same preferred translational
positions.
Samples taken during a 5-min incubation show that
FACT promotes tetrasome formation more rapidly with
mutant histones than with wild-type histones (Figure 6,
CE). Conversely, FACT with Spt16-11 caused displacement
more slowly than FACT with wild-type Spt16 protein (Fig-
ure 6E, Figure S6). Combining histone mutants with the
FACT mutant resulted in a slight suppression of the Spt16-11
defect (Figure 6E, Figure S6), but the amount of compensation
was minimal.
Together, these results show that FACT containing Spt16-11
protein has signicant defects in vitro in forming stable com-
plexes with nucleosomes and in promoting the normal rapid
rate of dimer loss. Mutant histones identied as suppressors
of the spt16-11 allele are lost from nucleosomes at an ab-
normally high rate, but do not strongly reverse the Spt16-11
defects in these assays.
Nuclease sensitivity reveals a potential mechanism
of suppression
We have proposed that FACT promotes equilibration of nu-
cleosomes between canonical and reorganized forms, with
the rate of digestion by nucleases being proportional to the
fraction of time that the nucleosomes spend in the reor-
ganized state (Xin et al. 2009). We therefore used restriction
endonuclease sensitivity to examine whether Spt16-11 and
histone mutants alter the persistence of the reorganized
state.
We measured the rates of DraI digestion of 181-bp 5S
rDNA nucleosomes with yeast histones to probe two distinct
physical contexts [DraI-78, near the dyad of symmetry, and
DraI-140, near an entry/exit point, as described in Xin et al.
(2009)]. Both FACT and FACT(Spt16-11) enhanced the rate
of digestion near the dyad signicantly, although FACT
(Spt16-11) consistently produced less of an effect (Figure
6F). The histone mutants had the opposite effect of increas-
ing the rate of DraI digestion. The same overall pattern was
observed near the entry/exit points, although the rates of
digestion were generally higher (Xin et al. 2009) and the
Figure 4 The source of expression of H2B-A84D affects the resulting
phenotypes. Isogenic strains from the A364a genetic background with
the spt16-11 mutation (Table 1) were tested as in Figure 1, with the
concentration of HU indicated. Panel A shows strains with the WT
SPT16 allele and B shows strains with the spt16-11 mutation. Incubation
times are listed in days to allow comparison of the level of growth at
comparable times. In particular, htb1-A84D causes a weak Spt
2
pheno-
type, as very little growth on medium lacking lysine is observed after
2 days but substantial growth is visible after 6 days. This is weak relative
to the effect of spt16-11, which causes substantial growth on medium
lacking lysine after 2 days. Both HTB1 and HTB2 are tagged in all cases,
whether wild-type (+) or mutant (A84D). Δindicates deletion of both
HTA2 and HTB2.
842 L. McCullough et al.
defect of FACT(Spt16-11) was less signicant. These results
show that FACT(Spt16-11) can reorganize nucleosomes, but
it either produces a structure with less accessibility to the
DNA or fails to achieve or maintain the reorganized state for
the normal length of time.
Notably, the combination of FACT(Spt16-11) with H2B-
A84D nucleosomes produced a rate of digestion that is as
high or higher than the rate with wild-type FACT combined
with wild-type nucleosomes (Figure 6F). This also appears
to be true for FACT(Spt16-11) with H2A-V101I nucleosomes
at the entry/exit point, but not near the dyad. These two
histones behave differently in vivo and may use overlapping
but distinct mechanisms of suppression. These results sug-
gest that FACT with Spt16-11 protein fails to achieve or
maintain the open reorganized nucleosome form long
enough to allow the normal rate of restriction endonuclease
digestion, but this defect is counterbalanced by histone
mutants that achieve this state more rapidly or maintain it
for a longer amount of time than usual.
Discussion
We have shown that histone gene mutations can compen-
sate for defects in FACT activity, with the spt16-11 mutation
being suppressed by H2A-H2B mutants that destabilize the
interface between these dimers and the (H3-H4)
2
tetramer.
In tests with puried components, these histone mutants
caused increased dimer displacement in the absence of
FACT and more rapid or more persistent nucleosome reor-
ganization in the presence of FACT. Genetic suppression
therefore appears to result from combining a FACT complex
that is inefcient at nucleosome reorganization with nucle-
osomes that are more rapidly or more easily reorganized. By
comparing the properties of the same FACT and histone
gene mutations in vivo and in vitro, these results provide
insight into the mechanism of FACT activity in its physiolog-
ical settings. Initial reports indicated that FACT removes one
H2A-H2B dimer from a nucleosome (Belotserkovskaya et al.
2003), and our experiments showed that dimer displace-
ment can be an outcome of FACT action but is not a neces-
sary feature of the mechanism of reorganization (Xin et al.
2009). Both sets of in vitro investigations indicate that FACT
activity involves disruption of the H2A-H2B:(H3-H4)
2
inter-
face, and the results presented here conrm that this is an
important feature of FACT function in vivo.
FACT activity has been implicated in both DNA replica-
tion and transcription (Formosa 2008). FACTs ability to
promote equilibration between stable and dynamic forms of
histone:DNA complexes could reduce the barrier to polymer-
ase progression posed by existing nucleosomes, but it could
also promote establishment of chromatin through the reverse
reaction by converting loosely associated components into
Figure 5 Histone gene mutations destabilize nucleosomes
in vitro. (A) Nucleosomes were constructed using recombi-
nant yeast histones (normal or the mutant indicated) and
a 181-bp 5S rDNA fragment with Cy5 at the 59end and
Oregon Green 488 (Molecular Probes) attached to H2A
residue C114 (originally Q114) (Xin et al. 2009). Samples
were prepared in triplicate and incubated for 1 hr at 30in
100 or 550 mMNaCl and then separated by electropho-
resis through a native 4% polyacrylamide gel in 0.25·TBE
as described (Xin et al. 2009). The single gel was then
scanned to detect the uorescent dyes on the DNA and
the H2A independently using a Typhoon scanner (GE).
One of the three repeats for each condition is shown.
(B) Each lane was scanned and the amount of signal cor-
responding to free DNA, displaced H2A-H2B, or tetrasome
forms was determined as a percentage of the total signal
in the lane. The regions assigned as free DNA and tetra-
somes are shown in the DNA scan in A, as determined
using pure reference samples (tetrasomes migrate as two
main bands using this combination of DNA and histones).
The amount of displaced H2A-H2B was determined by
calculating the signal in the top 40% of the H2A-H2B scan
in A, as described previously (Xin et al. 2009). The change
in the level of each form caused by treatment with high
salt is plotted, with error bars indicating the standard de-
viation among the three samples. (C and D) As in A and B,
except samples were incubated at 30or 65for 1 hr and
only the total amount of tetrasomes detected is displayed.
FACT Defects Suppressed by Histone Mutants 843
mature nucleosomes. Consistent with a nucleosome assem-
bly function, FACT has been implicated in the deposition of
new nucleosomes after DNA replication (Belotserkovskaya
et al. 2003; Vandemark et al. 2006) as well as in the re-
establishment of repressive chromatin after transcription
(Jamai et al. 2009). Defects in this nucleosome deposition
activity are likely to be the cause of cryptic promoter activa-
tion (Kaplan et al. 2003), failed repression of SER3 expres-
sion (Hainer et al. 2010), and the Spt
2
phenotype (Malone
et al. 1991; Rowley et al. 1991). Most FACT gene mutations
(including spt16-11) cause phenotypes associated with chro-
matin quality defects, indicating that maintaining appropri-
ately repressive chromatin throughout the genome requires
optimal levels of FACT activity. The histone mutants de-
scribed in this report did not suppress the Spt
2
phenotype
caused by spt16-11, so by this assay they do not enhance the
ability of the Spt16-11 protein to assemble normal chromatin.
Essentially all known mutations in FACT genes cause the
Spt
2
phenotype, but only a subset of these mutations causes
the HU sensitivity that is likely to reect a defect in replica-
tion fork progression or stability. The results described here
suggest that spt16-11 causes HU sensitivity because Spt16-11
protein is unable to promote nucleosome reorganization rap-
idly enough to allow a normal rate of fork progression
Figure 6 Spt16-11 protein and
its suppressors alter the rates of
reactions on the basis of reorga-
nization in vitro. (A) Nucleosomes
constructed as in Figure 5 were
mixed with different concentra-
tions of Spt16-Pob3 hetero-
dimers containing normal Spt16
or Spt16-11 protein, along with
10 mM Nhp6. Samples were in-
cubated for 10 min at 30and
then separated by native PAGE,
and the fraction of the total
DNA signal in FACTnucleosome
complexes was determined as
described previously (Xin et al.
2009). The nucleosomes shown
contained the H2B-A84D muta-
tion and are typical of results
obtained with wild type and
H2A-V101I. Multiple prepara-
tions of wild-type and mutant
FACT were tested in indepen-
dent experiments; values for half
saturation varied somewhat be-
tween experiments, but the de-
fect for Spt16-11 protein was
reproducible. (B) Nucleosomes
constructed as in Figure 5 were
treated for 10 min at 30with
5mM Nhp6 alone, 5 mM Nhp6
and 200 nM Spt16-Pob3 [FACT
(Spt16)], or 5 mM Nhp6 and
(Spt16-11)-Pob3 [FACT(Spt16-
11)]. Three independent sam-
ples for each condition were
analyzed as in Figure 5, A and
B, with the average and standard
deviation of the three measure-
ments presented. (C and D)
Nucleosomes were constructed
using recombinant histones from
X. laevis (with normal histones or
the mutation indicated) and a 255-bp MMTV DNA fragment (Flaus et al. 2004) labeled with Cy5. Samples were treated with FACT for the amount of
time indicated, excess unlabeled genomic DNA was added to disrupt the FACTnucleosome complexes, and then products were separated by native
PAGE (Rhoades et al. 2004). Conversion from initial forms (Nuc) to the tetrasomal products (indicated by arrows) was quantitated and plotted in D. (E)
As in D, except FACT(Spt16) or FACT(Spt16-11) were mixed with nucleosomes containing H2A or H2A-V101 as indicated. (F) Nucleosomes were
constructed using wild-type or mutant yeast histones and 181-bp 5S rDNA fragments with DraI recognition sites 78 or 140 bp from the left edge of the
nucleosome (Xin et al. 2009). The initial rate of digestion by DraI in the absence of other factors or with wild-type or mutant FACT was determined by
examining samples taken at 8-min intervals with denaturing PAGE (Xin et al. 2009). Each condition was tested in three independent experiments with
the average and standard deviation shown.
844 L. McCullough et al.
through a normal chromatin template. The additional delay
that results from decreased availability of dNTPs when RNR is
inhibited by HU might be intolerable when FACT is defective.
Histone mutants that promote faster nucleosome reorganiza-
tion by destabilizing the histone dimer:tetramer interface
then diminish the impaired progression and restore viability
under HU stress, but do not resolve the defect in chromatin
quality, so the Spt
2
phenotype persists.
Our tests of an H2B mutation integrated into the genome
reveal a clear functional difference between HTB1 and
HTB2. In normal cells, each gene is transcribed at about
the same level on about the same cell cycle schedule. How-
ever, the A84D mutant is effective only at suppressing the
HU sensitivity (likely to be a replication defect) caused by
spt16-11 if it is expressed from HTB1 alone. Curiously, the
effects of htb1-A84D are similar when it is paired with nor-
mal HTB2 or a deletion of HTB2. In contrast, expressing
H2B-A84D from both loci begins to suppress the Spt
2
phe-
notype (transcription defect) caused by spt16-11, but en-
hances the HU sensitivity (probable replication defect).
These differences could be due to the different primary
sequences of Htb1 and Htb2 proteins, or they could indicate
that the two genes have different expression proles under
some circumstances such as nucleosome replacement out-
side of S phase. In either case, Htb1 and Htb2 proteins have
different functional roles, and this becomes particularly im-
portant in an spt16-11 strain. It is therefore necessary to
consider the expression context when examining the effects
of histone gene mutations, especially when they affect H2B.
The results presented here provide support for the model
that FACT activity promotes or requires destabilization of
histone dimer:tetramer interaction. The analysis suggests
that promoting the reversible, rapid oscillation of nucleo-
somes between a stable canonical form and a more open
reorganized form is a key function of FACT in vivo, high-
lighting the dynamic nature of nucleosomes under physio-
logical conditions.
Acknowledgments
We thank Mitch Smith for providing the initial strain used in
our screen, Liz Kendall and Peter Winters for technical
assistance, and members of the Formosa and Stillman labs
for valuable input. This project was supported by a grant
from the National Institutes of Health to T.F. and D.J.S.
Literature Cited
Ahn, S. H., W. L. Cheung, J. Y. Hsu, R. L. Diaz, M. M. Smith, et al.,
2005 Sterile 20 kinase phosphorylates histone H2B at serine
10 during hydrogen peroxide-induced apoptosis in S. cerevisiae.
Cell 120: 2536.
Belotserkovskaya, R., S. Oh, V. A. Bondarenko, G. Orphanides, V.
M. Studitsky, et al., 2003 FACT facilitates transcription-
dependent nucleosome alteration. Science 301: 10901093.
Biswas, D., Y. Yu, M. Prall, T. Formosa, and D. J. Stillman,
2005 The yeast FACT complex has a role in transcriptional
initiation. Mol. Cell. Biol. 25: 58125822.
Biswas, D., R. Dutta-Biswas, D. Mitra, Y. Shibata, B. D. Strahl, et al.,
2006 Opposing roles for Set2 and yFACT in regulating TBP
binding at promoters. EMBO J. 25: 44794489.
Biswas, D., S. Takahata, H. Xin, R. Dutta-Biswas, Y. Yu. et al.,
2008 A role for Chd1 and Set2 in negatively regulating DNA
replication in Saccharomyces cerevisiae. Genetics 178: 649659.
Boeke, J. D., J. Trueheart, G. Natsoulis, and G. R. Fink, 1987
5-Fluoroorotic acid as a selective agent in yeast molecular ge-
netics. Methods Enzymol. 154: 164175.
Brewster, N. K., G. C. Johnston, and R. A. Singer, 2001 A bipartite
yeast SSRP1 analog comprised of Pob3 and Nhp6 proteins mod-
ulates transcription. Mol. Cell. Biol. 21: 34913502.
Christianson, T. W., R. S. Sikorski, M. Dante, J. H. Shero, and P.
Hieter, 1992 Multifunctional yeast high-copy-number shuttle
vectors. Gene 110: 119122.
Clapier, C. R., and B. R. Cairns, 2009 The biology of chromatin
remodeling complexes. Annu. Rev. Biochem. 78: 273304.
Clark-Adams, C. D., D. Norris, M. A. Osley, J. S. Fassler, and F.
Winston, 1988 Changes in histone gene dosage alter transcrip-
tion in yeast. Genes Dev. 2: 150159.
David, L., W. Huber, M. Granovskaia, J. Toedling, C. J. Palm, et al.,
2006 A high-resolution map of transcription in the yeast ge-
nome. Proc. Natl. Acad. Sci. USA 103: 53205325.
Dollard, C., S. L. Ricupero-Hovasse, G. Natsoulis, J. D. Boeke, and
F. Winston, 1994 SPT10 and SPT21 are required for transcrip-
tion of particular histone genes in Saccharomyces cerevisiae. Mol.
Cell. Biol. 14: 52235228.
Duina, A. A., A. Ruange, J. Bracey, J. Hall, A. Nourani, et al.,
2007 Evidence that the localization of the elongation factor
Spt16 across transcribed genes is dependent upon histone H3
integrity in Saccharomyces cerevisiae. Genetics 177: 101112.
Flaus, A., C. Rencurel, H. Ferreira, N. Wiechens and T. Owen-
Hughes, 2004 Sin mutations alter inherent nucleosome mobil-
ity. EMBO J. 23: 343353.
Formosa, T., 2008 FACT and the reorganized nucleosome. Mol.
Biosyst. 4: 10851093.
Formosa, T., P. Eriksson, J. Wittmeyer, J. Ginn, Y. Yu, et al.,
2001 Spt16-Pob3 and the HMG protein Nhp6 combine to form
the nucleosome-binding factor SPN. EMBO J. 20: 35063517.
Formosa, T., S. Ruone, M. D. Adams, A. E. Olsen, P. Eriksson, et al.,
2002 Defects in SPT16 or POB3 (yFACT) in Saccharomyces
cerevisiae cause dependence on the Hir/Hpc pathway: polymer-
ase passage may degrade chromatin structure. Genetics 162:
15571571.
Hainer, S. J., J. A. Pruneski, R. D. Mitchell, R. M. Monteverde, and
J. A. Martens, 2010 Intergenic transcription causes repression
by directing nucleosome assembly. Genes Dev. 25: 2940.
Hess, D., and F. Winston, 2005 Evidence that Spt10 and Spt21 of
Saccharomyces cerevisiae play distinct roles in vivo and function-
ally interact with MCB-binding factor, SCB-binding factor and
Snf1. Genetics 170: 8794.
Jamai, A., A. Puglisi, and M. Strubin, 2009 Histone chaperone
spt16 promotes redeposition of the original h3-h4 histones
evicted by elongating RNA polymerase. Mol. Cell 35: 377383.
Kaplan, C. D., L. Laprade, and F. Winston, 2003 Transcription
elongation factors repress transcription initiation from cryptic
sites. Science 301: 10961099.
Libuda, D. E., and F. Winston, 2006 Amplication of histone
genes by circular chromosome formation in Saccharomyces cer-
evisiae. Nature 443: 10031007.
Lolas, I. B., K. Himanen, J. T. Gronlund, C. Lynggaard, A. Houben,
et al., 2010 The transcript elongation factor FACT affects
Arabidopsis vegetative and reproductive development and ge-
netically interacts with HUB1/2. Plant J. 61: 686697.
Lycan, D., G. Mikesell, M. Bunger, and L. Breeden, 1994 Dif-
ferential effects of Cdc68 on cell cycle-regulated promoters in
Saccharomyces cerevisiae. Mol. Cell. Biol. 14: 74557465.
FACT Defects Suppressed by Histone Mutants 845
Malone, E. A., C. D. Clark, A. Chiang, and F. Winston, 1991 Mu-
tation in SPT16/CDC68 suppress cis- and trans-acting mutations
that affect promoter function in Saccharomyces cerevisiae. Mol.
Cell. Biol. 11: 57105717.
Norris, D., and M. A. Osley, 1987 The two gene pairs encoding
H2A and H2B play different roles in the Saccharomyces cerevi-
siae life cycle. Mol. Cell. Biol. 7: 34733481.
ODonnell, A. F., N. K. Brewster, J. Kurniawan, L. V. Minard, G. C.
Johnston, et al., 2004 Domain organization of the yeast his-
tone chaperone FACT: the conserved N-terminal domain of
FACT subunit Spt16 mediates recovery from replication stress.
Nucleic Acids Res. 32: 58945906.
ODonnell,A.F.,J.R.Stevens,R.Kepkay,C.A.Barnes,G.C.Johnston,
et al., 2009 New mutant versions of yeast FACT subunit Spt16
affect cell integrity. Mol. Genet. Genomics 282: 487502.
Orphanides, G., G. LeRoy, C.-H. Chang, D. S. Luse, and D. Reinberg,
1998 FACT, a factor that facilitates transcript elongation
through nucleosomes. Cell 92: 105116.
Osley, M. A., 1991 The regulation of histone synthesis in the cell
cycle. Annu. Rev. Biochem. 60: 827861.
Osley, M. A., and D. Lycan, 1987 Trans-acting regulatory muta-
tions that alter transcription of Saccharomyces cerevisiae his-
tone genes. Mol. Cell. Biol. 7: 42044210.
Reinberg, D., and R. J. Sims III, 2006 de FACTo nucleosome dy-
namics. J. Biol. Chem. 281: 2329723301.
Rhoades, A. R., S. Ruone, and T. Formosa, 2004 Structural fea-
tures of nucleosomes reorganized by yeast FACT and its HMG
box component, Nhp6. Mol. Cell. Biol. 24: 39073917.
Rowley, A., R. A. Singer, and G. Johnston, 1991 CDC68, a yeast
gene that affects regulation of cell proliferation and transcrip-
tion, encodes a protein with a highly acidic carboxyl terminus.
Mol. Cell. Biol. 11: 57185726.
Ruone, S., A. R. Rhoades, and T. Formosa, 2003 Multiple Nhp6
molecules are required to recruit Spt16-Pob3 to form yFACT
complexes and reorganize nucleosomes. J. Biol. Chem. 278:
4528845295.
Sikorski, R. S., and P. Hieter, 1989 A system of shuttle vectors and
yeast host strains designed for efcient manipulation of DNA
in Saccharomyces cerevisiae. Genetics 122: 1927.
Simchen, G., F. Winston, C. A. Styles, and G. R. Fink, 1984 Ty-
mediated gene expression of the LYS2 and HIS4 genes of Sac-
charomyces cerevisiae is controlled by the same SPT genes. Proc.
Natl. Acad. Sci. USA 81: 24312434.
Toulmay, A., and R. Schneiter, 2006 A two-step method for the
introduction of single or multiple dened point mutations into
the genome of Saccharomyces cerevisiae. Yeast 23: 825831.
VanDemark, A. P., M. Blanksma, E. Ferris, A. Heroux, C. P. Hill,
et al., 2006 The structure of the yFACT Pob3-M domain, its
interaction with the DNA replication factor RPA, and a potential
role in nucleosome deposition. Mol. Cell 22: 363374.
VanDemark, A. P., H. Xin, L. McCullough, R. Rawlins, S. Bentley,
et al., 2008 Structural and functional analysis of the Spt16p
N-terminal domain reveals overlapping roles of yFACT subunits.
J. Biol. Chem. 283: 50585068.
White, C. L., R. K. Suto, and K. Luger, 2001 Structure of the yeast
nucleosome core particle reveals fundamental changes in inter-
nucleosome interactions. EMBO J. 20: 52075218.
Winkler, D. D., and K. Luger, 2011 The histone chaperone FACT:
structural insights and mechanisms for nucleosome reorganiza-
tion. J. Biol. Chem. 286: 1836918374.
Xin, H., S. Takahata, M. Blanksma, L. McCullough, D. J. Stillman,
et al., 2009 yFACT induces global accessibility of nucleosomal
DNA without H2AH2B displacement. Mol. Cell 35: 365376.
Xu, H., U. J. Kim, T. Schuster, and M. Grunstein, 1992 Iden-
tication of a new set of cell cycle-regulatory genes that regulate
S-phase transcription of histone genes in Saccharomyces cerevi-
siae. Mol. Cell. Biol. 12: 52495259.
Communicating editor: F. Winston
846 L. McCullough et al.
GENETICS
Supporting Information
http://www.genetics.org/content/suppl/2011/05/30/genetics.111.128769.DC1
Insight Into the Mechanism of Nucleosome
Reorganization From Histone Mutants That
Suppress Defects in the FACT Histone Chaperone
Laura McCullough, Robert Rawlins, Aileen Olsen, Hua Xin, David J. Stillman, and Tim Formosa
Copyright © 2011 by the Genetics Society of America
DOI: 10.1534/genetics.111.128769
1)!!$)
('$'($#")(($#'##'(&#'1
34(&#@:><29?2;%:;?3'($#2E/%'($#'0!94+'(&#'$&"+(%%!'"'&&-#
(#(:$&:")(($##$(1)!()&'+&&$+#($'()&($##&")"($!!$+!$''$%:;?
&&-#'($##'#("& &/(#982$!'&!!)($#'+&%!$#$"%!('-#((")"$&
")"$#(##=2($(&"#$+$(#!!'!$'(('($##%!'")&#&$+(+($)('!($#$&('
%!'"1!'"!$''''')"($&#$"/'$&'#(-!$=2&''(#(!!'#('(((
")(($#'%!-''-#((&$+((+((")(($#$#(#$"#%!'"/'$'&*$&:2@A#:2@<1
&''(#(!$#'+&(#$'##('($&&$+((;;5$&$#")"$#(##>"-&$,-)&1'("%&()&
''"2%&"''*$&&$+($(''(&#+('($#'/#$(+ ')%%&''$##+ ##"#($
('2%#$(-%+'#$($&&#('($#")(($#'1$#$('($#")(($#'((')%%&''(')'-
!'$')%%&''(')'-/!($)$)(!##('%#$(-%13('$:2@A
#:2@<#('''-&)!(($#(&%&('(''(&#'&%&'#(&&')&**$&'$'!($##(&$&"-
#!)($#!")(($#'$&!(&($#'$,%&''$#41)(($#'(("!$&((#%(9>(&$&#&!!-
,&((#$;+#('(&$"%!'"$#(,(/!($):2989'%!-"!')%%&''$#+##(&(
!!!'+&('(3"#(,(/;41
34(&#AAAA%:;?3/'($#2E/%'($#'0!94+'(&#'$&"+(%%!'"'&&-#
*&$)''($#")(($#'/'$!('! #(%!'"+&$(#-'!($#$#/#'&!!)($#'+&('('
$*1(&#'+&!'$('($&(&&'%$#'($,%$')&($"%($(#/!$"-#/818;D/#)!(&*$!(!(
% 
'(%)$!
%')(#
!%
"&!%!#
! !#
!!%
(""
!  !  !$ ! ! (! ! "" ! '
$! !% !,),-). #!&( !! $!!$ !
! % ,,  $"   ,( !! !
!!! ! !!"(!!0/$ !" 
! ' !,,+$ !$ "  !(! !!" "  $'$!!
!(
("" 
HTA1 MSGGKGGKAGSAAKASQSRSAKAGLTFPVGRVHRLLRRGNYAQRIGSGAPVYLTAVLEYLAAEILE (65)
::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
HTA2 MSGGKGGKAGSAAKASQSRSAKAGLTFPVGRVHRLLRRGNYAQRIGSGAPVYLTAVLEYLAAEILE (65)
V101I
HTA1 LAGNAARDNKKTRIIPRHLQLAIRNDDELNKLLGNVTIAQGGVLPNIHQNLLPKKSAKATKASQEL (131)
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::: ::::::
HTA2 LAGNAARDNKKTRIIPRHLQLAIRNDDELNKLLGNVTIAQGGVLPNIHQNLLPKKSAKTAKASQEL (131)
HTB1p MSAKAEKKPASKAPAEKKPAAKKTSTSTDGKKRSKARKETYSSYIYKVLKQTHPDTGISQKSMSIL (65)
:: ::::::::::::::::::::::: ::::::: ::::::::::::::::::::::::::::::
HTB2p MSSAAEKKPASKAPAEKKPAAKKTSTSVDGKKRSKVRKETYSSYIYKVLKQTHPDTGISQKSMSIL (65)
A84
HTB1p NSFVNDIFERIATEASKLAAYNKKSTISAREIQTAVRLILPGELAKHAVSEGTRAVTKYSSSTQA (130)
:::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
HTB2p NSFVNDIFERIATEASKLAAYNKKSTISAREIQTAVRLILPGELAKHAVSEGTRAVTKYSSSTQA (130)
! "#!!$!!$ ! % ..( " !! 
% &!!'! % &!$
(! #! $'$!! ! !-,-30"!! !&$(!
" ! %"!!!! " (!!!-,-"!!!!" 
 ,21-,21.,21-,2/3).*( "!!! !!#
!  " %!!"!!!!-,-'!!! !#! 
" %!!"!!!!-,-/ !"!! !& "(" '
  "!!! '"! ""!! ! 
' & "!!!("! " !!#!)!
"*!$!!$ ' ! #! !!&! !!&" !"!  (30
"!! $!!" ,204,21,,202,203).*(  
!!'"& !! $ !!#!!$!"
! " ("!  !! &! !! "'#!#!!$!!$
 ! .!!(
*$$
"#!"!!(#(#!##(*""#!"&##
$###!75$##!&!!&#"#$!#)# $#"21+"!$#"
&!#"#!!&#$!##"#*&#$#!&#61(!'($!,41.)46.),61-41.-!
"(##$(",#""(##+#($"(-&!$"*#
"#""#%#($"(,""!%#"+"#"#)32-)$##
#"#*$")'!""#75$##!"!#!##"#!#
'!""!")$"#"#&"!#"#!,#'#)$!5-*3+75'!""!
!"#!#)"$###"!"$#"#%#"##!#$!""#%#(""#%#(*
&%!)3+75"#$"###"$"()"##+#("
$#*
/'!' 
%&! '&&! %! 3&%  &+&& &+!!$ '!%!%/'!%!%)$! %&$'&)&
'!$% &+8>8"$ &! &  &<$%#'  +%&%&! %1977?2)&&$&
909%#' %!$&'&&! %!) /%)$ '&)&($+ !  &$&! %!"&8=0!:)&<@"=-
 '&87 '&%&:74-& %"$&+ &("!+$+&$!"!$%%%%$1977;.
977:2/$&! !%  &$! !&%%!&)&!"*%)%&$  "!&&!$&
!  &$&! % $&0*  $! /!($&! )%!%$(-'& !$"&"&&$ )%&& 
'&"$"&%!%$*"$ &%- & && %&! '!%!%)&&%%&! '&&! %)&&%
""$ & &+% '!%!%)&%&! %/
!
&"%$%%'"%%!
 ' ! 
'!
 
 

 %)"$)"#)$%$#$'(# %"%!$ %"%!%%"' $!
 %)"$)"#)$%$#$'(#'& %"%!$ %"%!%
%"' $!
 %)"$)"#)$%$#$'(# %"%!$'& %"%!%
%"' $!
 %)"$)"#)$%$#$'(#'& %"%!$'& %"%!%
%"' $!
&""

 "  "! "(2*!0*)
-1'/2 . 
-2'/5 . 
-2'/6 / 
-2'0- / 
-2'0. / 
-2'0/ / 
-4/3 1 
-4/4 1 
-4/5 2 
-4/6 2 
-40- 3 

-40. 3 

-405 5 
-414 .- 
-415 .- 
-416 6 
-42- 6 
-42. 4%5 
-42/ 4 
-46. 0 
-46/ 0 

.$ ! ' "!  (.)
/"  ! 
0$ !//-# !!
1$ !0-# !!
2$ !0-# !!
3$ !0-# !!
4!!!!
5!!!!
6!!!!
.-!!!!
1+""%+  
+&&%(*!$($!*
010101010110755; ! 2()%"+*!%$#&%*($)(!&*!%$!$* .)*$%#1
(%*"!:8752:87:1
01101$107559*(+*+("*+()%$+"%)%#)(%($!/..)*$!*)%-
%#&%$$*0 &;1%"""!%"8>5<28>6<1
01011$107558+"*!&" &;%"+")('+!(*%(+!*&*6;2%8*%%(#.
%#&"-)$%($!/+"%)%#)1!%" #9:7==29:7>:1
010101010110755>.!$+)"%"))!!"!*.%$+"%)%#"
,!* %+*727!)&"#$*1%"""8;:28<;1
... Since the spt16 E857K mutation is unable to properly bind to H2A-H2B dimer and reestablish the repressive nucleosome following the elongating RNAPII 32 , this mutation may affect the expression of genes required to mount a response to rapamycin. 34 . We examined whether this spt16-11 mutant would display an altered response to rapamycin. ...
... Spot test analysis revealed that the spt16-11 allele was sensitive to rapamycin, as compared to the parental strain W303 (WT-2) (Fig. 3B). The study by McCullough also isolated several histone mutations, including H2B A84D that strongly suppressed the phenotypes of the spt16-11 mutation 34 . In addition, they showed by in vitro analysis that the H2B A84D mutant caused nucleosome instability by enhancing dimer displacement, thus weakening the histone dimer: tetramer interface, a function attributed to the FACT complex 34 . ...
... The study by McCullough also isolated several histone mutations, including H2B A84D that strongly suppressed the phenotypes of the spt16-11 mutation 34 . In addition, they showed by in vitro analysis that the H2B A84D mutant caused nucleosome instability by enhancing dimer displacement, thus weakening the histone dimer: tetramer interface, a function attributed to the FACT complex 34 . Thus, the H2B A84D mutation may not be able to maintain the nucleosome structure and the repressive chromatin, thus leading to alter gene expression patterns 34 . ...
Article
Full-text available
In yeast Saccharomyces cerevisiae, the immunosuppressant rapamycin inhibits the TORC1 kinase causing rapid alteration in gene expression and leading to G1 arrest. We recently reported the isolation and characterization from the histone mutant collection of a histone H2B R95A mutant that displays resistance to rapamycin. This mutant is defective in the expression of several genes belonging to the pheromone response pathway including STE5 encoding a scaffold protein that promotes the activation of downstream MAP kinases. Cells lacking Ste5 cannot arrest the cell cycle in response to rapamycin and as a consequence exhibit similar resistance to rapamycin as the H2B R95A mutant. Herein, we show that the H2B R95A mutation weakens the association of H2B with Spt16 a component of the FACT complex (FAcilitates Chromatin Transcription), and an essential factor that interacts with the histone H2A-H2B dimer to promote transcription and preserve chromatin integrity. From a collection of spt16 mutants, spt16 E857K and spt16-11 showed striking sensitivity to rapamycin as compared to the parent strain. spt16 E857K and spt16-11 expressed distinct forms of Ste5, while a suppressor mutation H2B A84D of the spt16-11 mutant prevents the expression of Ste5 and confers marked resistance to rapamycin. We interpret these findings to suggest that the Arg95 residue of histone H2B is required to recruit Spt16 to maintain the expression of STE5, which performs a role to arrest cells in the G1 phase in response to rapamycin.
... This suggests an interactive choreography, with FACT promoting some structural maneuvers of histone domains and DNA while blocking others that might be inconsistent with the canonical assembly pathway. Single-particle FRET experiments also support central aspects of this model, showing that FACT reversibly uncoils DNA from nucleosomes and that FACT mutations and histone mutations that suppress their phenotypes in vivo also alter the distribution between canonical and reorganized forms in vitro (49)(50)(51). The kinetics of dissociation of complexes is therefore affected by both the functional integrity of FACT domains and the ability of histones to adopt the canonical nucleosomal structure. ...
... Notably, the homologous residue in human SSRP1 is in a loop that contacts DNA (45), and while both pob3-Q308K and pob3-Q308R were isolated multiple times in a screen for pob3 mutations that cause sensitivity to the DNA replication toxin hydroxyurea, pob3-Q308A had no phenotype (127), suggesting that introduction of a positive charge near the DNA causes the reduced dissociation efficiency. In contrast, FACT with the Spt16-11 mutant subunit binds to and releases from nucleosomes normally but reorganizes them inefficiently (51). The phenotypes caused by these mutations are suppressed and enhanced by different profiles of histone mutations that affect different domains of nucleosomes (51,126). ...
... In contrast, FACT with the Spt16-11 mutant subunit binds to and releases from nucleosomes normally but reorganizes them inefficiently (51). The phenotypes caused by these mutations are suppressed and enhanced by different profiles of histone mutations that affect different domains of nucleosomes (51,126). These and other genetic and biochemical studies suggest that promoting and resolving changes in nucleosome structure have separable functions in distinct processes in vivo. ...
Article
Full-text available
FACT (FAcilitates Chromatin Transcription) has long been considered to be a transcription elongation factor whose ability to destabilize nucleosomes promotes RNAPII progression on chromatin templates. However, this is just one function of this histone chaperone, as FACT also functions in DNA replication. While broadly conserved among eukaryotes and essential for viability in many organisms, dependence on FACT varies widely, with some differentiated cells proliferating normally in its absence. It is therefore unclear what the core functions of FACT are, whether they differ in different circumstances, and what makes FACT essential in some situations but not others. Here, we review recent advances and propose a unifying model for FACT activity. By analogy to DNA repair, we propose that the ability of FACT to both destabilize and assemble nucleosomes allows it to monitor and restore nucleosome integrity as part of a system of chromatin repair, in which disruptions in the packaging of DNA are sensed and returned to their normal state. The requirement for FACT then depends on the level of chromatin disruption occurring in the cell, and the cell's ability to tolerate packaging defects. The role of FACT in transcription would then be just one facet of a broader system for maintaining chromatin integrity.
... Additionally, the dissociation of Atf1 upon starvation triggers the accumulation of histone chaperone FACT [132], which maintains methylated histones by preventing histone turnover [138] (Figure 6). Since FACT potentially recognizes the Q105 site in H2A [83,139,140], it would be intriguing to study whether Atf1 regulates the modification of H2AQ105 in response to nutrient limitation. ...
Article
Full-text available
Restricting ribosome biosynthesis and assembly in response to nutrient starvation is a universal phenomenon that enables cells to survive with limited intracellular resources. When cells experience starvation, nutrient signaling pathways, such as the target of rapamycin (TOR) and protein kinase A (PKA), become quiescent, leading to several transcription factors and histone modification enzymes cooperatively and rapidly repressing ribosomal genes. Fission yeast has factors for heterochromatin formation similar to mammalian cells, such as H3K9 methyltransferase and HP1 protein, which are absent in budding yeast. However, limited studies on heterochromatinization in ribosomal genes have been conducted on fission yeast. Herein, we shed light on and compare the regulatory mechanisms of ribosomal gene transcription in two species with the latest insights.
... The following supporting information can be downloaded at: https: //www.mdpi.com/article/10.3390/cells11192931/s1, Figure S1: Electrophoresis of Hmo1 complexes with nucleosomes, which were constructed with a 181 bp DNA fragment labeled with Cy5 (green) based on the nucleosome positioning sequence from sea urchin 5S rDNA and yeast histones labeled with Oregon Green on H2A-Q114C; Figure S2: Competitor DNA itself does not affect nucleosome structure; Figure S3: FACT itself does not interact with a nucleosome; Figure S4: spFRET analysis of FACT interactions with N35/11 nucleosomes (~1 nM) in the presence of Hmo1 at different Hmo1:FACT ratios; Figure S5: Deletion of HMO1 causes synthetic effects when combined with defective alleles of the FACT subunits. References [63][64][65] ...
Article
Full-text available
Yeast Hmo1 is a high mobility group B (HMGB) protein that participates in the transcription of ribosomal protein genes and rDNA, and also stimulates the activities of some ATP-dependent remodelers. Hmo1 binds both DNA and nucleosomes and has been proposed to be a functional yeast analog of mammalian linker histones. We used EMSA and single particle Förster resonance energy transfer (spFRET) microscopy to characterize the effects of Hmo1 on nucleosomes alone and with the histone chaperone FACT. Hmo1 induced a significant increase in the distance between the DNA gyres across the nucleosomal core, and also caused the separation of linker segments. This was opposite to the effect of the linker histone H1, which enhanced the proximity of linkers. Similar to Nhp6, another HMGB factor, Hmo1, was able to support large-scale, ATP-independent, reversible unfolding of nucleosomes by FACT in the spFRET assay and partially support FACT function in vivo. However, unlike Hmo1, Nhp6 alone does not affect nucleosome structure. These results suggest physiological roles for Hmo1 that are distinct from Nhp6 and possibly from other HMGB factors and linker histones, such as H1.
... Previous work identified the region surrounding H2AQ105 as part of a recognition sequence for the histone chaperone FACT (McCullough et al, 2011). Methylation of Q105 prevents FACT from binding to H2A, impairing its redeposition following transcription and resulting in loss of histones at the rDNA (Tessarz et al, 2014). ...
Article
Full-text available
Ribosome biogenesis is an essential cellular process that requires integration of extracellular cues, such as metabolic state, with intracellular signalling, transcriptional regulation and chromatin accessibility at the ribosomal DNA. Here, we demonstrate that the recently identified histone modification, methylation of H2AQ105 (H2AQ105me), is an integral part of a dynamic chromatin network at the rDNA locus. Its deposition depends on a functional mTor signalling pathway and acetylation of histone H3 at position K56, thus integrating metabolic and proliferative signals. Furthermore, we identify a first epigenetic reader of this modification, the ribonucleoprotein Nhp2, which specifically recognizes H2AQ105me. Based on functional and proteomic data, we suggest that Nhp2 functions as an adapter to bridge rDNA chromatin with components of the small subunit processome to efficiently coordinate transcription of rRNA with its post-transcriptional processing. We support this by showing that an H2AQ105A mutant has a mild defect in early processing of rRNA.
Preprint
Full-text available
Ribosome biogenesis is an essential cellular process that requires integration of extracellular cues, such as metabolic state, with intracellular signaling, transcriptional regulation and chromatin accessibility at the ribosomal DNA. Here, we demonstrate that the recently identified histone modification, methylation of H2AQ105, is an integral part of a dynamic chromatin network at the rDNA locus. Its deposition depends on a functional mTor signaling pathway as well as acetylation of histone H3 at position K56, thus integrating signals from cell cycle, metabolic and proliferative states. Furthermore, we identify a first epigenetic reader of this modification, the ribonucleoprotein Nhp2, which specifically recognizes the methylation on H2AQ105. Based on functional and proteomic data we suggest that Nhp2 functions as an adapter to bridge the rDNA chromatin with components of the small subunit processome and might help to efficiently coordinate transcription of rRNA with its post-transcriptional processing.
Thesis
Full-text available
Chez les eucaryotes, l’organisation de la chromatine est indispensable à la régulation de l’expression des gènes et la stabilité du génome. Les ciliés constituent un excellent modèle d’étude des mécanismes qui permettent de maintenir l’intégrité du génome eucaryote. Notre modèle d’étude, l’eucaryote unicellulaire Paramecium tetraurelia, se distingue par l’élimination massive et reproductible de près de 30% de séquences d’ADN germinal lors du développement du macronoyau somatique après les événements sexuels. Ces séquences sont éliminées grâce à un processus en plusieurs étapes impliquant la formation d’hétérochromatine dirigée par de petits ARN suivi d’excision par la transposase domestiquée Piggy Mac (Pgm) et de réparation. Les mécanismes moléculaires permettant de comprendre la reconnaissance spécifique de ces séquences germinales dans le contexte de la chromatine ainsi que la précision de la coupure demeurent mal compris.Le chaperon d’histone Spt16, associée au partenaire Pob3, fait partie du complexe hétéro-dimérique FACT (FAcilitates Chromatin Transactions). FACT intervient dans de nombreux mécanismes liés au métabolisme de l’ADN tels que la transcription, la réparation, la réplication ou l’accessibilité de la chromatine. Chez Paramecium tetraurelia, on a identifié deux protéines homologues de Spt16 et Pob3 exprimées uniquement durant le développement du macronoyau au moment où se produisent les réarrangements. Les protéines Spt16-1 et Pob3-1 fusionnées à la GFP se localisent dans les macronoyaux en développement. Nous avons montré que la protéine Spt16-1 est essentielle à la production d’une descendance sexuelle viable. Le reséquençage du génome après inactivation de SPT16-1 a montré que Spt16-1 est nécessaire à l‘ensemble des réarrangements du génome et conduit à des défauts semblables à ceux obtenus suite à l’inactivation de PGM. Spt16-1 agit en aval des voies de dépôt des marques d’hétérochromatine guidées par les petits ARNs mais en amont de l’endonucléase Pgm. Nous avons montré que Spt16-1 était nécessaire à la localisation correcte de Pgm responsable de l’introduction des cassures double brin dans les macronoyaux en développement. Nous proposons un modèle dans lequel Spt16-1 favorise l’interaction de la machinerie d’excision avec la chromatine pour rendre accessible les sites de coupure sur l’ADN à Pgm.La méthylation des adénines sur l’ADN (6mA), bien connue chez les bactéries pour son rôle dans le système de restriction modification, a été décrite ces deux dernières années chez plusieurs eucaryotes en faible proportion dans le génome. Cependant son rôle chez les eucaryotes est encore peu compris. D’anciennes analyses par chromatographie à l’aide de nucléotides marqués détectaient de l’ordre de 2,5% des adénines méthylées chez P. tetraurelia mais sa localisation précise n’avait jamais été montrée. Il avait été proposé que cette modification pourrait signaler avec précision les sites de coupures pendant les réarrangements d’ADN où une partie des séquences germinales présentent des bornes AT. L’abondance des adénines méthylées fait de la paramécie un excellent modèle d’étude de cette modification. En combinant des techniques d’Immunofluorescence, d’HPLC-MS et de séquençage, nous avons ainsi décrit la présence et la localisation dans la cellule et au cours du cycle de vie de 6mA chez P. tetraurelia. Ces approches ont également permis de montrer l’absence de cytosine méthylée chez la paramécie. Nous avons montré que la méthylation des adénines était retrouvée majoritairement dans le génome somatique et de manière transitoire dans le génome germinal. Elle est établie au cours du développement du macronoyau somatique quand se produisent les réarrangements du génome. Ces résultats préliminaires permettront de poursuivre l’étude de la méthylation afin d’identifier les enzymes responsables et son rôle dans la cellule.
Article
Full-text available
Monoubiquitination of histone H2B (H2B-Ub) plays a role in transcription and DNA replication, and is required for normal localization of the histone chaperone, FACT. In yeast, H2B-Ub is deubiquitinated by Ubp8, a subunit of SAGA, and Ubp10. Although they target the same substrate, loss of Ubp8 and Ubp10 cause different phenotypes and alter the transcription of different genes. We show that Ubp10 has poor activity on yeast nucleosomes, but that the addition of FACT stimulates Ubp10 activity on nucleosomes and not on other substrates. Consistent with a role for FACT in deubiquitinating H2B in vivo, a FACT mutant strain shows elevated levels of H2B-Ub. Combination of FACT mutants with deletion of Ubp10, but not Ubp8, confers increased sensitivity to hydroxyurea and activates a cryptic transcription reporter, suggesting that FACT and Ubp10 may coordinate nucleosome assembly during DNA replication and transcription. Our findings reveal unexpected interplay between H2B deubiquitination and nucleosome dynamics.
Article
Full-text available
Using a combination of in vitro biochemistry, genetics, and genomics, McCullough and Pham et al. explore how the histone:DNA contact at the entry/exit site of nucleosomes affects the functions of the histone chaperone FACT in the yeast... FACT (FAcilitates Chromatin Transcription/Transactions) is a histone chaperone that can destabilize or assemble nucleosomes. Acetylation of histone H3-K56 weakens a histone–DNA contact that is central to FACT activity, suggesting that this modification could affect FACT functions. We tested this by asking how mutations of H3-K56 and FACT affect nucleosome reorganization activity in vitro, and chromatin integrity and transcript output in vivo. Mimics of unacetylated or permanently acetylated H3-K56 had different effects on FACT activity as expected, but the same mutations had surprisingly similar effects on global transcript levels. The results are consistent with emerging models that emphasize FACT’s importance in establishing global chromatin architecture prior to transcription, promoting transitions among different states as transcription profiles change, and restoring chromatin integrity after it is disturbed. Optimal FACT activity required the availability of both modified and unmodified states of H3-K56. Perturbing this balance was especially detrimental for maintaining repression of genes with high nucleosome occupancy over their promoters and for blocking antisense transcription at the +1 nucleosome. The results reveal a complex collaboration between H3-K56 modification status and multiple FACT functions, and support roles for nucleosome reorganization by FACT before, during, and after transcription.
Article
During S phase, replicated DNA must be assembled into nucleosomes using both newly synthesized and parental histones in a process that is tightly coupled to DNA replication. This DNA replication-coupled process is regulated by multitude of histone chaperones as well as by histone-modifying enzymes. In recent years novel insights into nucleosome assembly of new H3-H4 tetramers have been gained through studies on the classical histone chaperone CAF-1 and the identification of novel factors involved in this process. Moreover, in vitro reconstitution of chromatin replication has shed light on nucleosome assembly of parental H3-H4, a process that remains elusive. Finally, recent studies have revealed that the replication-coupled nucleosome assembly is important for the determination and maintenance of cell fate in multicellular organisms.
Article
Full-text available
Changes in chromatin architecture induced by epigenetic mechanisms are essential for normal cellular processes such as gene expression, DNA repair, and cellular division. Compact chromatin presents a barrier to these processes and is highly regulated by epigenetic markers binding to components of the nucleosome. Histone modifications directly influence chromatin dynamics and facilitate recruitment of additional factors such as chromatin remodelers and histone chaperones. One member of this last class of factors, FACT (facilitates chromatin transcription), is categorized as a histone chaperone critical for nucleosome reorganization during replication, transcription, and DNA repair. Significant discoveries regarding the role of histone chaperones and specifically FACT have come over the past dozen years from a number of independent laboratories. Here, we review the structural and biophysical basis for FACT-mediated nucleosome reorganization and discuss up-to-date models for FACT function.
Article
Full-text available
Transcription by RNA polymerase II is impeded by the nucleosomal organization of DNA; these negative effects are modulated at several stages of nucleosomal DNA transcription by FACT, a heterodimeric transcription factor. At promoters, FACT facilitates the binding of TATA-binding factor, while during transcription elongation FACT mediates the necessary destabilization of nucleosomes and subsequent restoration of nucleosome structure in the wake of the transcription elongation complex. Altered FACT activity can impair the fidelity of transcription initiation and affect transcription patterns. Using reporter genes we have identified new mutant versions of the Spt16 subunit of yeast FACT with dominant negative effects on the fidelity of transcription initiation. Two of these spt16 mutant alleles also affect cell integrity. Cells relying on these spt16 mutant alleles display sorbitol-remediated temperature sensitivity, altered sensitivity to detergent, and abnormal morphologies, and are further inhibited by the ssd1-d mutation. The overexpression of components of protein kinase C (Pkc1) signaling diminishes this spt16 ssd1-d temperature sensitivity, whereas gene deletions eliminating components of Pkc1 signaling further impair these spt16 mutant cells. Thus, the FACT subunit Spt16 and Pkc1 signaling have an overlapping essential function, with an unexpected role for FACT in the maintenance of cell integrity.
Article
Swi4 and Swi6 form a complex which is required for Start-dependent activation of HO and for high-level expression of G1 cyclin genes CLN1 and CLN2. To identify other regulators of this pathway, we screened for dominant, recessive, conditional, and allele-specific suppressors of swi4 mutants. We isolated 16 recessive suppressors that define three genes, SSF1, SSF5, and SSF9 (suppressor of swi four). Mutations in all three genes bypass the requirement for both Swi4 and Swi6 for HO transcription and activate transcription from reporter genes lacking upstream activating sequences (UASs). SSF5 is allelic with SIN4 (TSF3), a gene implicated in global repression of transcription and chromatin structure, and SSF9 is likely to be a new global repressor of transcription. SSF1 is allelic with CDC68 (SPT16). cdc68 mutations have been shown to increase expression from defective promoters, while preventing transcription from other intact promoters, including CLN1 and CLN2. We find that CDC68 is a required activator of both SWI4 and SWI6, suggesting that CDC68's role at the CLN promoters may be indirect. The target of CDC68 within the SWI4 promoter is complex in that known activating elements (MluI cell cycle boxes) in the SWI4 promoter are required for CDC68 dependence but only within the context of the full-length promoter. This result suggests that there may be both a chromatin structure and a UAS-specific component to Cdc68 function at SWI4. We suggest that Cdc68 functions both in the assembly of repressive complexes that form on many intact promoters in vivo and in the relief of this repression during gene activation.
Article
Using a Saccharomyces cerevisiae strain containing an integrated copy of an H2A-lacZ fusion gene, we screened for mutants which overexpressed beta-galactosidase as a way to identify genes which regulate transcription of the histone genes. Five recessive mutants with this phenotype were shown to contain altered regulatory genes because they had lost repression of HTA1 transcription which occurs upon inhibition of chromosome replication (D. E. Lycan, M. A. Osley, and L. Hereford, Mol. Cell. Biol. 7:614-621, 1987). Periodic transcription was affected in the mutants as well, since the HTA1 gene was transcribed during the G1 and G2 phases of the cell cycle, periods in the cell cycle when this gene is normally not expressed. A similar loss of cell cycle-dependent transcription was noted for two of the three remaining histone loci, while the HO and CDC9 genes continued to be expressed periodically. Using isolated promoter elements inserted into a heterologous cycl-lacZ fusion gene, we demonstrated that the mutations fell in genes which acted through a negative site in the TRT1 H2A-H2B promoter.
Article
We have isolated Saccharomyces cerevisiae mutants bearing deletions of one or the other of the two divergently transcribed gene pairs encoding H2A and H2B. The deletions produced diverse effects on the yeast life cycle. Deletion of TRT1, one of the H2A-H2B gene pair sets, affected mitotic growth, sporulation, spore germination, the heat shock response, and exit from the stationary phase; deletion of TRT2, the other H2A-H2B gene pair set, had negligible effects on these same processes. Using a genetic complementation assay, we found that the differential effects of the deletions could be attributed to two features of the gene sets: first, the expression of the TRT1 gene pair, but not the TRT2 gene pair, could compensate for the absence of its partner; second, the protein subtypes encoded by the two gene pairs appear to have different functions in the heat shock response.
Article
Histone mRNA synthesis is tightly regulated to S phase of the yeast Saccharomyces cerevisiae cell cycle as a result of transcriptional and posttranscriptional controls. Moreover, histone gene transcription decreases rapidly if DNA replication is inhibited by hydroxyurea or if cells are arrested in G1 by the mating pheromone alpha-factor. To identify the transcriptional controls responsible for cycle-specific histone mRNA synthesis, we have developed a selection for mutations which disrupt this process. Using this approach, we have isolated five mutants (hpc1, hpc2, hpc3, hpc4, and hpc5) in which cell cycle regulation of histone gene transcription is altered. All of these mutations are recessive and belong to separate complementation groups. Of these, only one (hpc1) falls in one of the three complementation groups identified previously by other means (M. A. Osley and D. Lycan, Mol. Cell. Biol. 7:4204-4210, 1987), indicating that at least seven different genes are involved in the cell cycle-specific regulation of histone gene transcription. hpc4 is unique in that derepression occurs only in the presence of hydroxyurea but not alpha-factor, suggesting that at least one of the regulatory factors is specific to histone gene transcription after DNA replication is blocked. One of the hpc mutations (hpc2) suppresses delta insertion mutations in the HIS4 and LYS2 loci. This effect allowed the cloning and sequence analysis of HPC2, which encodes a 67.5-kDa, highly charged basic protein.
Article
The cell cycle of the budding yeast Saccharomyces cerevisiae has been investigated through the study of conditional cdc mutations that specifically affect cell cycle performance. Cells bearing the cdc68-1 mutation (J. A. Prendergast, L. E. Murray, A. Rowley, D. R. Carruthers, R. A. Singer, and G. C. Johnston, Genetics 124:81-90, 1990) are temperature sensitive for the performance of the G1 regulatory event, START. Here we describe the CDC68 gene and present evidence that the CDC68 gene product functions in transcription. CDC68 encodes a 1,035-amino-acid protein with a highly acidic and serine-rich carboxyl terminus. The abundance of transcripts from several unrelated genes is decreased in cdc68-1 mutant cells after transfer to the restrictive temperature, while at least one transcript, from the HSP82 gene, persists in an aberrant fashion. Thus, the cdc68-1 mutation has both positive and negative effects on gene expression. Our findings complement those of Malone et al. (E. A. Malone, C. D. Clark, A. Chiang, and F. Winston, Mol. Cell. Biol. 11:5710-5717, 1991), who have independently identified the CDC68 gene (as SPT16) as a transcriptional suppressor of delta-insertion mutations. Among transcripts that rapidly become depleted in cdc68-1 mutant cells are those of the G1 cyclin genes CLN1, CLN2, and CLN3/WHI1/DAF1, whose activity has been previously shown to be required for the performance of START. The decreased abundance of cyclin transcripts in cdc68-1 mutant cells, coupled with the suppression of cdc68-1-mediated START arrest by the CLN2-1 hyperactive allele of CLN2, shows that the CDC68 gene affects START through cyclin gene expression.
Article
The Saccharomyces cerevisiae genome contains four loci that encode histone proteins. Two of these loci, HTA1-HTB1 and HTA2-HTB2, each encode histones H2A and H2B. The other two loci, HHT1-HHF1 and HHT2-HHF2, each encode histones H3 and H4. Because of their redundancy, deletion of any one histone locus does not cause lethality. Previous experiments demonstrated that mutations at one histone locus, HTA1-HTB1, do cause lethality when in conjunction with mutations in the SPT10 gene. SPT10 has been shown to be required for normal levels of transcription of several genes in S. cerevisiae. Motivated by this double-mutant lethality, we have now investigated the interactions of mutations in SPT10 and in a functionally related gene, SPT21, with mutations at each of the four histone loci. These experiments have demonstrated that both SPT10 and SPT21 are required for transcription at two particular histone loci, HTA2-HTB2 and HHF2-HHT2, but not at the other two histone loci. These results suggest that under some conditions, S. cerevisiae may control the level of histone proteins by differential expression of its histone genes.
Article
Transcription of non-protein-coding DNA (ncDNA) and its noncoding RNA (ncRNA) products are beginning to emerge as key regulators of gene expression. We previously identified a regulatory system in Saccharomyces cerevisiae whereby transcription of intergenic ncDNA (SRG1) represses transcription of an adjacent protein-coding gene (SER3) through transcription interference. We now provide evidence that SRG1 transcription causes repression of SER3 by directing a high level of nucleosomes over SRG1, which overlaps the SER3 promoter. Repression by SRG1 transcription is dependent on the Spt6 and Spt16 transcription elongation factors. Significantly, spt6 and spt16 mutations reduce nucleosome levels over the SER3 promoter without reducing intergenic SRG1 transcription, strongly suggesting that nucleosome levels, not transcription levels, cause SER3 repression. Finally, we show that spt6 and spt16 mutations allow transcription factor access to the SER3 promoter. Our results raise the possibility that transcription of ncDNA may contribute to nucleosome positioning on a genome-wide scale where, in some cases, it negatively impacts protein-DNA interactions.
Article
The facilitates chromatin transcription (FACT) complex, consisting of the SSRP1 and SPT16 proteins, is a histone chaperone that assists the progression of transcribing RNA polymerase on chromatin templates by destabilizing nucleosomes. Here, we examined plants that harbour mutations in the genes encoding the subunits of Arabidopsis FACT. These experiments revealed that (i) SSRP1 is critical for plant viability, and (ii) plants with reduced amounts of SSRP1 and SPT16 display various defects in vegetative and reproductive development. Thus, mutant plants display an increased number of leaves and inflorescences, show early bolting, have abnormal flower and leaf architecture, and their seed production is severely affected. The early flowering of the mutant plants is associated with reduced expression of the floral repressor FLC in ssrp1 and spt16 plants. Compared to control plants, reduced amounts of FACT in mutant plants are detected at the FLC locus as well as at the locations of housekeeping genes (whose expression is not affected in the mutants), suggesting that expression of FLC is particularly sensitive to reduced FACT activity. Analysis of double mutants that are affected in the expression of both FACT subunits and factors catalysing the mono-ubiquitination of histone H2B (HUB1/2) demonstrates that they genetically interact to regulate various developmental processes (i.e. branching, leaf venation pattern, silique development) but independently regulate the growth of leaves and the induction of flowering.