ArticlePDF Available

Regulation of ATP-dependent chromatin remodelers: Accelerators/brakes, anchors and sensors

Authors:

Abstract and Figures

All ATP-dependent chromatin remodelers have a DNA translocase domain that moves along double-stranded DNA when hydrolyzing ATP, which is the key action leading to DNA moving through nucleosomes. Recent structural and biochemical data from a variety of different chromatin remodelers have revealed that there are three basic ways in which these remodelers self-regulate their chromatin remodeling activity. In several instances, different domains within the catalytic subunit or accessory subunits through direct protein–protein interactions can modulate the ATPase and DNA translocation properties of the DNA translocase domain. These domains or subunits can stabilize conformations that either promote or interfere with the ability of the translocase domain to bind or retain DNA during translocation or alter the ability of the enzyme to hydrolyze ATP. Second, other domains or subunits are often necessary to anchor the remodeler to nucleosomes to couple DNA translocation and ATP hydrolysis to DNA movement around the histone octamer. These anchors provide a fixed point by which remodelers can generate sufficient torque to disrupt histone–DNA interactions and mobilize nucleosomes. The third type of self-regulation is in those chromatin remodelers that space nucleosomes or stop moving nucleosomes when a particular length of linker DNA has been reached. We refer to this third class as DNA sensors that can allosterically regulate nucleosome mobilization. In this review, we will show examples of these from primarily the INO80/SWR1, SWI/SNF and ISWI/CHD families of remodelers.
Structural examples of brakes, accelerators and anchors from different chromatin remodeling families. (A) Shown is the structure of the ISWI subunit with its AutoN (red), NegC (tan) and ATPase domain (green) in its inactive conformation which is overlaid with the active structure of the ATPase domain of Snf2 subunit (purple) when bound to nucleosomes. The AutoN domain serves as a brake by both blocking the DNA-binding cleft of the ATPase domain and distorting the orientation between the two ATPase lobe when compared with Snf2 (PDB# 5JXR; 5XOY). (B) The structure of the tandem chromo domain (orange) and the ATPase domain (green) of Chd1 bound to nucleosomes is shown. The chromodomain binds to nucleosomal DNA adjacent to where the ATPase is bound, thereby promoting the active ATPase domain conformation (PDB# 5O9G). (C) A portion of the SnAC domain (red) binds to one lobe of the ATPase domain of Snf2 (green), the catalytic subunit of SWI/SNF, along with the Brace (yellow) and Post-HSA domains (blue). A large portion of the SnAC domain is not seen in the structure and is represented as a dotted line (PDB#: 5XOY). (D) Shown is the structure of the Arp5 (orange) and Ies6 (blue) subunits of yeast INO80 that binds directly to the H2A-H2B dimer surface and to nucleosomal DNA at SHL −2 and −3. The unresolved regions of Arp5 are shown as dotted lines and are numbered based on their amino acid positions. The H2A and H2B histones are highlighted, respectively, red and yellow with black spheres showing the residues in the acidic pocket (PDB# 6FML).
… 
Content may be subject to copyright.
Review Article
Regulation of ATP-dependent chromatin
remodelers: accelerators/brakes,
anchors and sensors
Somnath Paul
1,2
and Blaine Bartholomew
1,2
1
Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park, Smithville, TX 78957, U.S.A.;
2
Center for Cancer
Epigenetics, MD Anderson Cancer Center, Houston, TX, USA
Correspondence: Blaine Bartholomew (bbartholomew@mdanderson.org)
All ATP-dependent chromatin remodelers have a DNA translocase domain that moves
along double-stranded DNA when hydrolyzing ATP, which is the key action leading to
DNA moving through nucleosomes. Recent structural and biochemical data from a
variety of different chromatin remodelers have revealed that there are three basic ways in
which these remodelers self-regulate their chromatin remodeling activity. In several
instances, different domains within the catalytic subunit or accessory subunits through
direct proteinprotein interactions can modulate the ATPase and DNA translocation prop-
erties of the DNA translocase domain. These domains or subunits can stabilize confor-
mations that either promote or interfere with the ability of the translocase domain to bind
or retain DNA during translocation or alter the ability of the enzyme to hydrolyze ATP.
Second, other domains or subunits are often necessary to anchor the remodeler to
nucleosomes to couple DNA translocation and ATP hydrolysis to DNA movement around
the histone octamer. These anchors provide a xed point by which remodelers can gen-
erate sufcient torque to disrupt histoneDNA interactions and mobilize nucleosomes.
The third type of self-regulation is in those chromatin remodelers that space nucleosomes
or stop moving nucleosomes when a particular length of linker DNA has been reached.
We refer to this third class as DNA sensors that can allosterically regulate nucleosome
mobilization. In this review, we will show examples of these from primarily the INO80/
SWR1, SWI/SNF and ISWI/CHD families of remodelers.
Introduction
Chromatin remodelers physically rearrange nucleosomes using energy derived from ATP hydrolysis
and their remodeling activity can be modulated in response to variations in chromatin such as histone
post-translational modications (i.e. particularly histone tails), histone variants such as H2A.Z or
length of linker DNA separating nucleosomes. Several remodelers, like the ISWI and SWI/SNF fam-
ilies, have been shown to require histone tails for efcient remodeling or recruitment of the remodeler,
and when histones are acetylated or otherwise covalently modied to either enhance or block nucleo-
some remodeling [17]. The interplay of histone modications and chromatin remodelers is an active
area of research and there are likely many of these interactions yet to be discovered that regulate the
activity or recruitment of remodelers. A large number of the pivotal studies have been done with only
the catalytic subunit or with subcomplexes of a limited number of accessory subunits present in the
complex. The INO80, ISWI and CHD families of remodelers are important for regulating nucleosome
spacing at promoters or coding regions of genes, making them crucial for transcription regulation
[815]. These remodelers require not only a minimum length of linker DNA for mobilizing nucleo-
somes but also a sufcient length of linker DNA to be recruited to chromatin. Some chromatin
remodelers target chromatin regions that are marked by the incorporation of histone variants that are
Version of Record published:
22 November 2018
Received: 27 August 2018
Revised: 17 October 2018
Accepted: 18 October 2018
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 1
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
less abundant in the cell [16]. These same remodelers can also change the levels of histone variants incorpo-
rated at distinct regions in the genome. By understanding more fully the various ways in which chromatin
remodelers are regulated, we will learn more about the mechanisms of chromatin remodeling and how these
are disrupted in diseases through mutation of these complexes.
The brakes and accelerators of the ATPase domains
The importance of histone tails for regulating chromatin remodeling is probably best characterized in the ISWI
family. For ISWI complexes, the H4 histone tail has been shown to be required for efcient remodeling and the
H4 tail peptide alone with free DNA to stimulate the ATPase activity of ISWI [57,17]. Histone acetylation can
modulate ISWI chromatin remodeling activity and acetylation of the H4 tail mitigates the stimulatory effect of
the H4 tail with ISWI. Most of the studies with the ISWI remodeler have been done with only the catalytic
subunit, even though these complexes generally have 24 subunits, and have found both negative and positive
regulators of the DNA translocase domain. Deletion analysis of ISWI found two domains that negatively regu-
late ISWI remodeling called the AutoN and NegC domains [1820]. Analysis of the crystal structure of ISWI
with and without H4 peptide found the AutoN to bridge lobes 1 and 2 of the DNA translocation or ATPase
domain, promoting the formation of an inactive ISWI conformation (Figure 1A), and H4 tail relieves inhibition
from the AutoN domain by competitive binding to lobe 2 [21]. The role of the C-terminal NegC domain is
not as clear in the structure because it reaches out to an adjacent ATPase domain and binds to lobe2, which
could be consistent with reports of ISWI forming dimers [21]. These experiments were all performed with only
the ISWI catalytic subunit, which was truncated and lacked the C-terminus containing the HAND, SANT and
SLIDE domains in most experiments, and did not have any of its accessory subunits. From other experiments,
it is evident that the accessory subunit does have a signicant impact on the activity of the catalytic subunit
and the accessory subunit can also bind the H4 tail and affect H4 tail sensing [20]. CHD1 has similar structures
to ISWI in regard to both the AutoN and NegC domains of ISWI acting similar to the chromodomains and
C-terminal bridge of CHD1 (Figure 1B)[22]. The AutoN and NegC domains are therefore examples of
domains acting to brake the ATPase domain (Figure 2A).
Other domains and accessory subunits act as accelerators by positively regulating the activity of the ATPase
domain (Figure 2A). For many of the chromatin remodelers, it is fairly clear that accessory subunits stimulate
the overall activity of complexes. These effects could be due to enhancing substrate binding or recruitment and
not be directly regulating the ATPase activity. One example from yeast SWI/SNF shows that the Snf5 accessory
subunit directly contacts the ATPase domain by mapping intra- and inter-subunit interactions using chemical
cross-linking and mass spectrometry (CX-MS). Snf5 in mammalian SWI/SNF is a tumor suppressor, and in
yeast, SWI/SNF forms a submodule consisting of Snf5, Swp82 and Taf14 (a YEATS domain protein) and the
entire submodule is lost from SWI/SNF when Snf5 is deleted. When this aberrant SWI/SNF complex is fully
saturating nucleosomes, the ATPase and remodeling activities of SWI/SNF are reduced and indicates that the
Snf5 submodule has a catalytic role in SWI/SNF remodeling [23]. The Arp7 and 9 subunits bind to the Snf2
catalytic subunit through an N-terminal domain called the HSA (Helicase/SANT-Associated) domain and have
been shown to genetically interact with the Protusion I region between the two lobes of the ATPase domain
[24,25]. Arp7 and 9 in a minimal RSC complex lacking most of its accessory subunits stimulate its ATPase and
DNA translocation activities, which, in turn, enhances nucleosome remodeling [26].
In the CX-MS study of SWI/SNF, domains within the Snf2 catalytic subunit such as the SnAC (Snf2 ATP
Coupling) domain were also shown to contact the second lobe of the ATPase domain [23]. Earlier studies
found that the SnAC domain is conserved throughout all eukaryotes, is a SWI/SNF specic domain and acti-
vates the ATPase domain as well as serves as a crucial histone anchor needed to couple DNA translocation/
ATPase activity to nucleosome movement [27,28]. The recent cryo-EM structure of the yeast Snf2 subunit con-
rmed that the SnAC domain interacts with lobe 2 of the Snf2 ATPase domain consistent with the earlier
CX-MS data, even though much of the SnAC domain is not observed in this structure because of its inherent
exibility and missing parts of SnAC in the truncated Snf2 used in these studies (Figure 1C and ref. [29]).
Another example comes from CHD1, which is one of the few remodelers in yeast that consists of only one
subunit, the catalytic subunit and no other accessory subunits. The chromodomains of CHD1 appear to
positively regulate the ATPase domain by swinging into a position juxtaposed to the ATPase domain on
nucleosomal DNA, thereby causing closure of the ATPase domain and promoting its translocation along DNA
(Figure 1B and ref. [30]). The CHD1 structure provides compelling evidence for the chromodomains positively
regulating CHD1 through altering the conformation of the ATPase domain.
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society2
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
Torque and nucleosome anchors in chromatin remodeling
Next, we examine the roles of anchors within chromatin remodelers that bind to nucleosomes and work inde-
pendent of regulating either the ATPase or DNA translocation activity of ATPase domains, but are nonetheless
essential for nucleosome mobilization. This type of anchor is thought to be needed in order for the ATPase
domain to translocate on DNA without DNA slipping and releasing the torque needed next to break histone
DNA contacts and move DNA through nucleosomes (Figure 2B). An early example of this type of anchor is
the SnAC domain in SWI/SNF complexes which required the correct conditions such that the nucleosome
anchor activity can be distinguished from the ATPase-stimulating activity of the SnAC domain. Experiments
were done using a higher ATP concentration with SWI/SNF lacking the SnAC domain such that the rate of
ATP hydrolysis with the mutant was equivalent or slightly higher than wild-type SWI/SNF. In these conditions,
SWI/SNF lacking the SnAC domain was found to remodel nucleosomes nearly 200 times slower than wild
type, even though the rate of ATP hydrolysis was slightly higher with the mutant complex [28]. A protein foot-
printing technique in which Fe-EDTA was tethered to the surface of nucleosomes showed that the SnAC
domain of yeast SWI/SNF was located proximal to the histone octamer surface. In tethered single-molecule
B
CD
A
Figure 1. Structural examples of brakes, accelerators and anchors from different chromatin remodeling families.
(A) Shown is the structure of the ISWI subunit with its AutoN (red), NegC (tan) and ATPase domain (green) in its inactive
conformation which is overlaid with the active structure of the ATPase domain of Snf2 subunit ( purple) when bound to
nucleosomes. The AutoN domain serves as a brake by both blocking the DNA-binding cleft of the ATPase domain and
distorting the orientation between the two ATPase lobe when compared with Snf2 (PDB# 5JXR; 5XOY). (B) The structure of the
tandem chromo domain (orange) and the ATPase domain (green) of Chd1 bound to nucleosomes is shown. The
chromodomain binds to nucleosomal DNA adjacent to where the ATPase is bound, thereby promoting the active ATPase
domain conformation (PDB# 5O9G). (C) A portion of the SnAC domain (red) binds to one lobe of the ATPase domain of Snf2
(green), the catalytic subunit of SWI/SNF, along with the Brace (yellow) and Post-HSA domains (blue). A large portion of the
SnAC domain is not seen in the structure and is represented as a dotted line (PDB#: 5XOY). (D) Shown is the structure of the
Arp5 (orange) and Ies6 (blue) subunits of yeast INO80 that binds directly to the H2AH2B dimer surface and to nucleosomal
DNA at SHL 2 and 3. The unresolved regions of Arp5 are shown as dotted lines and are numbered based on their amino
acid positions. The H2A and H2B histones are highlighted, respectively, red and yellow with black spheres showing the
residues in the acidic pocket (PDB# 6FML).
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 3
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
ATP ADP
DNA translocation
Brakes and Accelerators
Nucleosome Anchors
without anchor
with anchor
slips back, releases torsional strain
torsional strain created
translocates
torsional strain created
translocates
DNA moves past barrier
sub-optimal
conformation
DNA Sensors
Nucleosome
with sensor without sensor
A
B
C
optimal
conformation
Figure 2. Modes of regulation for ATP-dependent chromatin remodelers.
(A) The basic properties of ATP hydrolysis and DNA translocation can be either up- or down-regulated to control the extent
of chromatin remodeling. We refer to those subunits or domains that affect remodeling in this way as either brakes or
accelerators. (B) Anchors that attach to the core nucleosome particle are another way in which chromatin remodeling is
regulated. The ATPase domain will have a high tendency to slip on DNA in the presence of barriers such as histoneDNA
interaction without a nucleosome anchor being present due to the torsional strain that is created. (C) Some remodelers will not
move nucleosomes to close together and have some type of sensor that detects when the minimal acceptable length of linker
DNA has been achieved. We refer to the subunits or domains involved in this process as DNA sensors.
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society4
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
experiments, the SnAC mutant SWI/SNF complex was found to translocate as efciently along DNA as
wild-type SWI/SNF, further showing the critical difference in the mutant complex is its histone-anchoring
activity [28].
The other example is from the INO80 remodeling complex, which has 15 different subunits, mobilizes and
spaces nucleosomes, and can potentially exchange H2AH2B dimers [12,31]. Loss of the actin-related protein
(Arp) 5 and Ies6 subunits of human INO80 uncouples the ATPase activity from the nucleosome-mobilizing
activity of INO80 as shown using recombinant INO80. Omission of any one of the two subunits loses both
subunits from the complex [32]. In yeast, deletion of different regions of Arp5 causes the loss of Arp5 and Ies6
from the INO80 complex and these complexes, while the ATPase activity stimulated by the addition of nucleo-
some remained equivalent to wild-type INO80, the mutant INO80 was severely impaired for nucleosome
mobilization [33]. In these experiments, the addition of Arp5/Ies6 to the mutant INO80 complexes was able to
restore the nucleosome-mobilizing activity of INO80, conrming the role of Arp5 and Ies6 for coupling
ATPase activity to nucleosome mobilization [33]. In the recent human and Chaetomium thermophilum INO80
(ctINO80) structures, Arp5 was found to directly contact nucleosomal DNA and the histone octamer close to
the acidic pocket region, consistent with Arp5/Ies6 serving as a nucleosome anchor for INO80 with minimal
interactions detected with the ATPase domain (Figure 1D and refs [34,35]). The Arp5 and Ies6 module is prob-
ably one of the more denitive examples of the important roles nucleosome anchors have in regulating the
efciency of nucleosome movement by the remodeler. Because of where Arp5 binds nucleosomes, Arp5 may be
capable of distinguishing between H2A and H2A.Z where one of the primary differences between these two
histones is the extended acidic patch found in H2A.Z.
DNA sensors for nucleosome spacing-type chromatin
remodelers
At the C-terminus of the ISWI catalytic subunit are located three domains called the HAND, SANT and
SLIDE domains. For the yeast ISW2 remodeling complex, the SLIDE domain was found to bind extranucleoso-
mal DNA 19 bp from the edge of nucleosomes by site-directed DNA cross-linking and peptide mapping [36].
Similarly, the SLIDE and SANT domains of ISW1a have been mapped by a different photocross-linking
approach to 10 and 0 bp from the edge of nucleosomes when ISW1a binds dinucleosomes [37]. The SLIDE
domain is structurally similar to the SANT and MYB DNA-binding domain, except that it has more basic resi-
dues in the appropriate positions to promote DNA binding than the SANT domain [38]. Based on the SLIDE
domains location on extranucleosomal DNA and ISW2 ability to move nucleosomes until the linker DNA has
reached a minimum length of >20 bp [39], the SLIDE domain appeared to be a likely candidate in the ISW2
complex for sensing linker DNA length to regulate the ISW2 chromatin remodeling activity (Figure 2C). The
DNA-binding interface of the SLIDE domain was targeted by mutating four basic residues suggested by
molecular modeling to be critical for binding DNA, which was conrmed by DNA footprinting with hydroxyl-
free radicals [40]. The mutant SLIDE ISW2 complex did, however, bind normally everywhere else on nucleo-
somes like wild-type ISW2. Mutant SLIDE ISW2 hydrolyzes ATP and remodels nucleosomes 69 times slower
than wild-type ISW2, and suggest that stable binding of the SLIDE domain to linker DNA is required for
efcient ISW2 remodeling [40]. Single-molecule FRET experiments showed that mutant SLIDE complexes
were more prone to move DNA bi-directionally, whereas wild-type ISW2 had a strong preference to move
DNA uni-directionally, thereby properly repositioning nucleosomes. These and other data point to the SLIDE
domain being important for promoting DNA entering into the nucleosome as mediated by ISW2 and suggest
that there are co-ordinated actions between the SLIDE and ATPase domains that are both required for efcient
nucleosome movement by ISW2.
The INO80 complex also has DNA sensor activity in that it spaces nucleosomes 30 bp apart and required a
minimum of 3540 bp of extranucleosomal DNA with 70 bp being optimal for binding and mobilizing
nucleosomes [12]. In the recent cryo-EM structure, the part of INO80 binding to the extranucleosomal DNA
was not resolved due to its inherent exibility and does not provide much structural information or insights
into the DNA sensor module of INO80 [34,35]. An alternative approach using site-directed DNA cross-linking
was used instead to map the spatial arrangement of the INO80 subunits bound to linker DNA and by peptide
mapping identied the protein domains or regions that directly bind linker DNA [41]. The Arp8 and Arp4
subunits were found to bind close to each other 3751 bp from the edge of nucleosomes and are at the correct
location to properly sense linker DNA length for the INO80 complex. The non-conserved N-terminus of Arp8
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 5
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
and the actin fold region of Arp4 near its C-terminus were found to contact DNA. Deletion of the N-terminus
of Arp8 eliminated binding of the Arp8Arp4 module to linker DNA without perturbing the complex integrity
of INO80 or its ability to bind nucleosomes or for nucleosomes to stimulate its ATPase activity. Chromatin
remodeling was, however, reduced and shows that deletion of the N-terminus of Arp8 causes ATPase activity
to be uncoupled from nucleosome movement [41]. Additional experiments showed that loss of the N-terminus
of Arp8 and its corresponding loss of binding to linker DNA causes an allosteric effect with the Ino80 catalytic
subunit and its ATPase domain shifting from inside nucleosomes at super helical loop (SHL)-6 to the linker
DNA region 30 bp away from its original position. There is also another allosteric effect in that Arp5s inter-
action with the acidic patch of nucleosomes is reduced, thus mirroring the effect when Arp5 is not present in
the complex [32,33]. For INO80, there is evidence that the DNA sensor consisting of the Arp8Arp4actin
module is an allosteric switch that disengages the critical histone anchor of INO80 required for coupling
ATPase activity to nucleosome movement and causes repositioning of the DNA translocase domain that also
leads to reduced nucleosome movement.
Yeast INO80 appears to have some redundancy in how it senses linker DNA as there is another protein
module besides the Arp8Arp4actin module that binds to linker DNA. Nhp10, an HMG-like protein that
associates with Ies1, 3 and 5, also binds close to the same region as Arp8Arp4actin and remains bound to
this region in the absence of Arp8 and Arp4 binding to this region [41]. In a separate study, deletion of Nhp10
causes the loss of an entire multi-subunit module from the INO80 complex and eliminates the DNA-sensing
capability of INO80 altogether such that it mobilizes nucleosomes regardless of the length of linker DNA
present [42]. The Arp8 and Nhp10 modules appear to work in very distinct ways in that Nhp10 module nega-
tively regulates INO80 remodeling such that it can only work with nucleosomes with particular linker DNA
lengths, whereas binding of Arp8 is required for positively regulating INO80s chromatin remodeling activity.
Perspectives
ATP-dependent chromatin remodelers are key epigenetic factors involved in development and human diseases
that have signicant diversity of operation as well as compositional complexity, making it difcult at times to
study. Much of the efforts in the eld have focused on understanding how the fundamental motors of these
complexes can be regulated, but there is a whole other level of regulation that we are just beginning to appreci-
ate involving the other subunits within these complexes. The recent cryo-EM structures of the human and
yeast INO80 complexes bound to nucleosomes provide important structural information and clues as to how
accessory subunits can potentially regulate chromatin remodeling as critical anchors to the nucleosome core
particle including both histones and nucleosomal DNA. Nevertheless, certain regions within the structure
could not be resolved as exemplied by the cryo-EM structure of INO80, and additional information gained by
other methods such as site-directed DNA and histone cross-linking, chemical CX-MS and other methods are
needed. As the work moves forward, more attention should be given to the regulation of these large
multi-subunit chromatin remodeling complexes not as individual subunits or small subcomplexes, but with
the intact full complex to properly understand how these complexes are regulated and sometimes mis-regulated
in various diseases. It will also be important to nd the interplay and potential overlap in the ways of
altering remodeling activity with brakes/accelerators, anchors and sensors. One domain may work in multiple
ways such as the SnAC domain of SWI/SNF which enhances ATPase activity as well as functions as a
histone anchor.
Abbreviations
Arp, actin-related protein; CX-MS, cross-linking and mass spectrometry; HSA, Helicase/SANT-Associated;
SHL, super helical loop; SnAC, Snf2 ATP coupling.
Competing Interests
The Authors declare that there are no competing interests associated with the manuscript.
References
1 Ferreira, H., Flaus, A. and Owen-Hughes, T. (2007) Histone modications inuence the action of Snf2 family remodelling enzymes by different
mechanisms. J. Mol. Biol. 374, 563579 https://doi.org/10.1016/j.jmb.2007.09.059
2 Chatterjee, N., Sinha, D., Lemma-Dechassa, M., Tan, S., Shogren-Knaak, M.A. and Bartholomew, B. (2011) Histone H3 tail acetylation modulates
ATP-dependent remodeling through multiple mechanisms. Nucleic Acids Res. 39, 83788391 https://doi.org/10.1093/nar/gkr535
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society6
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
3 Chatterjee, N., North, J.A., Dechassa, M.L., Manohar, M., Prasad, R., Luger, K. et al. (2015) Histone acetylation near the nucleosome dyad axis
enhances nucleosome disassembly by RSC and SWI/SNF. Mol. Cell. Biol. 35, 40834092 https://doi.org/10.1128/MCB.00441-15
4 Hassan, A.H., Neely, K.E. and Workman, J.L. (2001) Histone acetyltransferase complexes stabilize SWI/SNF binding to promoter nucleosomes. Cell
104, 817827 https://doi.org/10.1016/S0092-8674(01)00279-3
5 Clapier, C.R., Langst, G., Corona, D.F.V., Becker, P.B. and Nightingale, K.P. (2001) Critical role for the histone H4 N terminus in nucleosome remodeling
by ISWI. Mol. Cell. Biol. 21, 875883 https://doi.org/10.1128/MCB.21.3.875-883.2001
6 Clapier, C.R., Nightingale, K.P. and Becker, P.B. (2002) A critical epitope for substrate recognition by the nucleosome remodeling ATPase ISWI. Nucleic
Acids Res. 30, 649655 https://doi.org/10.1093/nar/30.3.649
7 Corona, D.F., Clapier, C.R., Becker, P.B. and Tamkun, J.W. (2002) Modulation of ISWI function by site-specic histone acetylation. EMBO Rep. 3,
242247 https://doi.org/10.1093/embo-reports/kvf056
8 Ocampo, J., Chereji, R.V., Eriksson, P.R. and Clark, D.J. (2016) The ISW1 and CHD1 ATP-dependent chromatin remodelers compete to set nucleosome
spacing in vivo.Nucleic Acids Res. 44, 46254635 https://doi.org/10.1093/nar/gkw068
9 Smolle, M., Venkatesh, S., Gogol, M.M., Li, H., Zhang, Y., Florens, L. et al. (2012) Chromatin remodelers Isw1 and Chd1 maintain chromatin structure
during transcription by preventing histone exchange. Nat. Struct. Mol. Biol. 19, 884892 https://doi.org/10.1038/nsmb.2312
10 Gkikopoulos, T., Schoeld, P., Singh, V., Pinskaya, M., Mellor, J., Smolle, M. et al. (2011) A role for Snf2-related nucleosome-spacing enzymes in
genome-wide nucleosome organization. Science 333, 17581760 https://doi.org/10.1126/science.1206097
11 Tirosh, I., Sigal, N. and Barkai, N. (2010) Widespread remodeling of mid-coding sequence nucleosomes by Isw1. Genome Biol. 11, R49 https://doi.org/
10.1186/gb-2010-11-5-r49
12 Udugama, M., Sabri, A. and Bartholomew, B. (2011) The INO80 ATP-dependent chromatin remodeling complex is a nucleosome spacing factor. Mol.
Cell. Biol. 31, 662673 https://doi.org/10.1128/MCB.01035-10
13 Krietenstein, N., Wal, M., Watanabe, S., Park, B., Peterson, C.L., Pugh, B.F. et al. (2016) Genomic nucleosome organization reconstituted with pure
proteins. Cell 167, 709721.e12 https://doi.org/10.1016/j.cell.2016.09.045
14 Willhoft, O., McCormack, E.A., Aramayo, R.J., Bythell-Douglas, R., Ocloo, L., Zhang, X. et al. (2017) Crosstalk within a functional INO80 complex dimer
regulates nucleosome sliding. eLife 6, e25782 https://doi.org/10.7554/eLife.25782
15 Zentner, G.E., Tsukiyama, T. and Henikoff, S. (2013) ISWI and CHD chromatin remodelers bind promoters but act in gene bodies. PLoS Genet. 9,
e1003317 https://doi.org/10.1371/journal.pgen.1003317
16 Mizuguchi, G., Shen, X., Landry, J., Wu, W.H., Sen, S. and Wu, C. (2004) ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin
remodeling complex. Science 303, 343348 https://doi.org/10.1126/science.1090701
17 Dang, W., Kagalwala, M.N. and Bartholomew, B. (2006) Regulation of ISW2 by concerted action of histone H4 tail and extranucleosomal DNA. Mol. Cell.
Biol. 26, 73887396 https://doi.org/10.1128/MCB.01159-06
18 Ludwigsen, J., Pfennig, S., Singh, A.K., Schindler, C., Harrer, N., Forné, I. et al. (2017) Concerted regulation of ISWI by an autoinhibitory domain and
the H4 N-terminal tail. eLife 6, e21477 https://doi.org/10.7554/eLife.21477
19 Clapier, C.R. and Cairns, B.R. (2012) Regulation of ISWI involves inhibitory modules antagonized by nucleosomal epitopes. Nature 492, 280284
https://doi.org/10.1038/nature11625
20 Hwang, W.L., Deindl, S., Harada, B.T. and Zhuang, X. (2014) Histone H4 tail mediates allosteric regulation of nucleosome remodelling by linker DNA.
Nature 512, 213217 https://doi.org/10.1038/nature13380
21 Yan, L., Wang, L., Tian, Y., Xia, X. and Chen, Z. (2016) Structure and regulation of the chromatin remodeller ISWI. Nature 540, 466469 https://doi.
org/10.1038/nature20590
22 Manning, B.J. and Peterson, C.L. (2013) Releasing the brakes on a chromatin-remodeling enzyme. Nat. Struct. Mol. Biol. 20,57https://doi.org/10.
1038/nsmb.2482
23 Sen, P., Luo, J., Hada, A., Hailu, S.G., Dechassa, M.L., Persinger, J. et al. (2017) Loss of Snf5 induces formation of an aberrant SWI/SNF complex.
Cell Rep. 18, 21352147 https://doi.org/10.1016/j.celrep.2017.02.017
24 Szerlong, H., Hinata, K., Viswanathan, R., Erdjument-Bromage, H., Tempst, P. and Cairns, B.R. (2008) The HSA domain binds nuclear actin-related
proteins to regulate chromatin-remodeling ATPases. Nat. Struct. Mol. Biol. 15, 469476 https://doi.org/10.1038/nsmb.1403
25 Schubert, H.L., Wittmeyer, J., Kasten, M.M., Hinata, K., Rawling, D.C., Héroux, A. et al. (2013) Structure of an actin-related subcomplex of the SWI/SNF
chromatin remodeler. Proc. Natl Acad. Sci. U.S.A. 110, 33453350 https://doi.org/10.1073/pnas.1215379110
26 Clapier, C.R., Kasten, M.M., Parnell, T.J., Viswanathan, R., Szerlong, H., Sirinakis, G. et al. (2016) Regulation of DNA translocation efciency within the
chromatin remodeler RSC/Sth1 potentiates nucleosome sliding and ejection. Mol. Cell 62, 453461 https://doi.org/10.1016/j.molcel.2016.03.032
27 Sen, P., Ghosh, S., Pugh, B.F. and Bartholomew, B. (2011) A new, highly conserved domain in Swi2/Snf2 is required for SWI/SNF remodeling. Nucleic
Acids Res. 39, 91559166 https://doi.org/10.1093/nar/gkr622
28 Sen, P., Vivas, P., Dechassa, M.L., Mooney, A.M., Poirier, M.G. and Bartholomew, B. (2013) The SnAC domain of SWI/SNF is a histone anchor required
for remodeling. Mol. Cell. Biol. 33, 360370 https://doi.org/10.1128/MCB.00922-12
29 Liu, X., Li, M., Xia, X., Li, X. and Chen, Z. (2017) Mechanism of chromatin remodelling revealed by the Snf2-nucleosome structure. Nature 544,
440445 https://doi.org/10.1038/nature22036
30 Farnung, L., Vos, S.M., Wigge, C. and Cramer, P. (2017) Nucleosome-Chd1 structure and implications for chromatin remodelling. Nature 550, 539542
https://doi.org/10.1038/nature24046
31 Brahma, S., Udugama, M.I., Kim, J., Hada, A., Bhardwaj, S.K., Hailu, S.G. et al. (2017) INO80 exchanges H2A.Z for H2A by translocating on DNA
proximal to histone dimers. Nat. Commun. 8, 15616 https://doi.org/10.1038/ncomms15616
32 Willhoft, O., Bythell-Douglas, R., McCormack, E.A. and Wigley, D.B. (2016) Synergy and antagonism in regulation of recombinant human INO80
chromatin remodeling complex. Nucleic Acids Res. 44, 81798188 https://doi.org/10.1093/nar/gkw509
33 Yao, W., Beckwith, S.L., Zheng, T., Young, T., Dinh, V.T., Ranjan, A. et al. (2015) Assembly of the Arp5 (actin-related protein) subunit involved in distinct
INO80 chromatin remodeling activities. J. Biol. Chem. 290, 2570025709 https://doi.org/10.1074/jbc.M115.674887
34 Eustermann, S., Schall, K., Kostrewa, D., Lakomek, K., Strauss, M., Moldt, M. et al. (2018) Structural basis for ATP-dependent chromatin remodelling
by the INO80 complex. Nature 556, 386390 https://doi.org/10.1038/s41586-018-0029-y
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 7
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
35 Ayala, R., Willhoft, O., Aramayo, R.J., Wilkinson, M., McCormack, E.A., Ocloo, L. et al. (2018) Structure and regulation of the human
INO80-nucleosome complex. Nature 556, 391395 https://doi.org/10.1038/s41586-018-0021-6
36 Dang, W. and Bartholomew, B. (2007) Domain architecture of the catalytic subunit in the ISW2-nucleosome complex. Mol. Cell. Biol. 27, 83068317
https://doi.org/10.1128/MCB.01351-07
37 Yamada, K., Frouws, T.D., Angst, B., Fitzgerald, D.J., DeLuca, C., Schimmele, K. et al. (2011) Structure and mechanism of the chromatin remodelling
factor ISW1a. Nature 472, 448453 https://doi.org/10.1038/nature09947
38 Grüne, T., Brzeski, J., Eberharter, A., Clapier, C.R., Corona, D.F.V., Becker, P.B. et al. (2003) Crystal structure and functional analysis of a nucleosome
recognition module of the remodeling factor ISWI. Mol. Cell 12, 449460 https://doi.org/10.1016/S1097-2765(03)00273-9
39 Kagalwala, M.N., Glaus, B.J., Dang, W., Zofall, M. and Bartholomew, B. (2004) Topography of the ISW2-nucleosome complex: insights into nucleosome
spacing and chromatin remodeling. EMBO J. 23, 20922104 https://doi.org/10.1038/sj.emboj.7600220
40 Hota, S.K., Bhardwaj, S.K., Deindl, S., Lin, Y.C., Zhuang, X. and Bartholomew, B. (2013) Nucleosome mobilization by ISW2 requires the concerted
action of the ATPase and SLIDE domains. Nat. Struct. Mol. Biol. 20, 222229 https://doi.org/10.1038/nsmb.2486
41 Brahma, S., Ngubo, M., Paul, S., Udugama, M. and Bartholomew, B. (2018) The Arp8 and Arp4 module acts as a DNA sensor controlling INO80
chromatin remodeling. Nat. Commun. 9, 3309 https://doi.org/10.1038/s41467-018-05710-7
42 Zhou, C.Y., Johnson, S.L., Lee, L.J., Longhurst, A.D., Beckwith, S.L., Johnson, M.J. et al. (2018) The yeast INO80 complex operates as a tunable DNA
length-sensitive switch to regulate nucleosome sliding. Mol. Cell 69, 677688.e9 https://doi.org/10.1016/j.molcel.2018.01.028
© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society8
Biochemical Society Transactions (2018)
https://doi.org/10.1042/BST20180043
... Unlike DDM1, which has been shown to actively slide nucleosomes in vitro 43 , purified HELLS fails to remodel reconstituted nucleosomes, and has minimal ATPase activity 9,16,17 , although activities are enhanced when complexed with Cell Division Cycle Associated 7 (CDCA7) 9,10 . The lack of intrinsic nucleosome remodeling activity raises the possibility that HELLS is auto-inhibited, and this auto-inhibition is not overcome by the addition of substrate unlike other remodelers including CHD1 and ISWI [44][45][46] . However, the absence of HELLS and DDM1 structures, coupled with limited biochemical and mechanistic studies on this distinct class of SNF2 chromatin remodelers, has impeded our understanding of their biological functions and the design of effective cancer therapeutics targeting HELLS. ...
... Other chromatin remodelers also use different mechanisms for autoinhibition. In CHD1, an Nterminal chromodomain wedges between the catalytic lobes preventing ATP hydrolysis in the absence of DNA and nucleosomes 44 , and ISWI contains AutoN and NegC motifs that negatively regulate ATP hydrolysis in the absence of substrates 45,46 . Here, we have uncovered a novel autoinhibitory element in the coiled-coil domain of HELLS responsible for the inactivity observed in in vitro studies even in the presence of substrates. ...
Preprint
Full-text available
Human HELicase, Lymphoid Specific (HELLS), and plant homolog Deficient in DNA Methylation 1 (DDM1), belong to a distinct class of chromatin remodelers that play important roles in DNA repair, transcription, and maintenance of DNA methylation in heterochromatin. HELLS also promotes the growth of hard-to-treat cancers including glioblastoma and hepatocellular carcinoma. Here, we identify an auto-inhibitory HELLS N-terminal coiled-coil, unravelling a long-standing question of HELLS inactivity in vitro. Using cryo-EM, we determine the 3.5 Å structure of an active DDM1-HELLS chimera in complex with a nucleosome. The structure reveals that a HELLS-specific insertion in the ATPase lobe 2 interacts with the nucleosome acidic patch to enhance chromatin remodeling. At the C-terminus, we resolve a unique motif, and disease hot spot, that binds and stabilizes the ATPase motor of the HELLS family of remodelers. Finally, we provide mechanistic insights for how post-translational modifications in the motor domain and midloop could modulate HELLS activity to regulate cancer stem cell state.
... The catalytic activity of Brg1/Brm and homologs form other species share a common mechanism by which ATP-hydrolysis powers alteration of the path and position of the DNA around a nucleosome particle [1][2][3][4]. Consequently, the diversity of mSWI/SNF complexes likely originated from the need to target the catalytic activity to different places in the genome, as well as provide specialized functions. ...
... SWI/SNF chromatin remodelers are ATP-dependent enzymes that alter nucleosome structure to facilitate or prevent access of regulatory factors to the genome and are conserved throughout the eukaryotic kingdom [1][2][3][4]. Early work in yeast determined that genes encoding proteins involved in mating type switching (SWI) and in sucrose fermentation (SNF) formed a multi-protein complex containing a DNA-dependent ATPase of the SNF2 family [5,6]. ...
Article
Full-text available
Mammalian SWI/SNF (mSWI/SNF) complexes are ATP-dependent chromatin remodeling enzymes that are critical for normal cellular functions. mSWI/SNF enzymes are classified into three sub-families based on the presence of specific subunit proteins. The sub-families are Brm- or Brg1-associated factor (BAF), ncBAF (non-canonical BAF), and polybromo-associated BAF (PBAF). The biological roles for the different enzyme sub-families are poorly described. We knocked down the expression of genes encoding unique subunit proteins for each sub-family, Baf250A, Brd9, and Baf180, which mark the BAF, ncBAF, and PBAF sub-families, respectively, and examined the requirement for each in myoblast differentiation. We found that Baf250A and the BAF complex were required to drive lineage-specific gene expression. KD of Brd9 delayed differentiation. However, while the Baf250A-dependent gene expression profile included myogenic genes, the Brd9-dependent gene expression profile did not, suggesting Brd9 and the ncBAF complex indirectly contributed to differentiation. Baf180 was dispensable for myoblast differentiation. The results distinguish between the roles of the mSWI/SNF enzyme sub-families during myoblast differentiation.
... The mSWI/SNF enzymes utilize the energy obtained from ATP hydrolysis to transiently alter the chromatin configuration on target DNA sequences, which can result in nucleosome sliding or eviction and the stable binding or exclusion of transcription or other regulatory factors. [17][18][19][20][21][22] These enzymes alter local myogenic gene chromatin structure to facilitate the assembly of active transcription complexes that initiate or up-regulate the expression of differentiation-specific genes. 11,23,24 In addition to working within the context of activities by multiple histone modifying enzymes, the mSWI/SNF enzymes themselves are regulated by signaling pathways that involve modification by p38, protein kinase C (PKC) and calcineurin, and CK2, among others. ...
Article
Full-text available
Pyruvate kinase is a glycolytic enzyme that converts phosphoenolpyruvate and ADP into pyruvate and ATP. There are two genes that encode pyruvate kinase in vertebrates; Pkm and Pkl encode muscle‐ and liver/erythrocyte‐specific forms, respectively. Each gene encodes two isoenzymes due to alternative splicing. Both muscle‐specific enzymes, PKM1 and PKM2, function in glycolysis, but PKM2 also has been implicated in gene regulation due to its ability to phosphorylate histone 3 threonine 11 (H3T11) in cancer cells. Here, we examined the roles of PKM1 and PKM2 during myoblast differentiation. RNA‐seq analysis revealed that PKM2 promotes the expression of Dpf2/Baf45d and Baf250a/Arid1A. DPF2 and BAF250a are subunits that identify a specific sub‐family of the mammalian SWI/SNF (mSWI/SNF) of chromatin remodeling enzymes that is required for the activation of myogenic gene expression during differentiation. PKM2 also mediated the incorporation of DPF2 and BAF250a into the regulatory sequences controlling myogenic gene expression. PKM1 did not affect expression but was required for nuclear localization of DPF2. Additionally, PKM2 was required not only for the incorporation of phosphorylated H3T11 in myogenic promoters but also for the incorporation of phosphorylated H3T6 and H3T45 at myogenic promoters via regulation of AKT and protein kinase C isoforms that phosphorylate those amino acids. Our results identify multiple unique roles for PKM2 and a novel function for PKM1 in gene expression and chromatin regulation during myoblast differentiation.
... Achieving such control over individual NTP turnover events would represent a powerful tool for dissecting particularly complex molecular mechanisms. A good example is nucleosome sliding catalyzed by ATP-dependent chromatin remodeling enzymes (remodelers) [31][32][33][34][35][36][37] , where several unstable nucleosome intermediates are formed during multiple consecutive ATP hydrolysis cycles before a nucleosome is stably repositioned 38,39 . For example, the Chd1 (chromodomain helicase DNA-binding protein 1) remodeler ATPase pulls up to four DNA base pairs into the entry side of the nucleosome, forming transient nucleosome structures that buffer the additional DNA, before it is released at the opposite (exit) side 38 . ...
Article
Full-text available
Many essential processes in the cell depend on proteins that use nucleoside triphosphates (NTPs). Methods that directly monitor the often-complex dynamics of these proteins at the single-molecule level have helped to uncover their mechanisms of action. However, the measurement throughput is typically limited for NTP-utilizing reactions, and the quantitative dissection of complex dynamics over multiple sequential turnovers remains challenging. Here we present a method for controlling NTP-driven reactions in single-molecule experiments via the local generation of NTPs (LAGOON) that markedly increases the measurement throughput and enables single-turnover observations. We demonstrate the effectiveness of LAGOON in single-molecule fluorescence and force spectroscopy assays by monitoring DNA unwinding, nucleosome sliding and RNA polymerase elongation. LAGOON can be readily integrated with many single-molecule techniques, and we anticipate that it will facilitate studies of a wide range of crucial NTP-driven processes.
Preprint
Full-text available
Pyruvate kinase is a glycolytic enzyme that converts phosphoenolpyruvate and ADP into pyruvate and ATP. There are two genes that encode pyruvate kinase in vertebrates; Pkm and Pkl encode muscle- and liver/erythrocyte-specific forms, respectively. Each gene encodes two isoenzymes due to alternative splicing. Both muscle-specific enzymes, Pkm1 and Pkm2, function in glycolysis, but Pkm2 also has been implicated in gene regulation due to its ability to phosphorylate histone 3 threonine 11 (H3T11) in cancer cells. Here, we examined the roles of Pkm1 and Pkm2 during myoblast differentiation. RNA-seq analysis revealed that Pkm2 promotes the expression of Dpf2/Baf45d and Baf250a/Arid1A . Dpf2 and Baf250a are subunits that identify a specific sub-family of the mammalian SWI/SNF (mSWI/SNF) of chromatin remodeling enzymes that is required for activation of myogenic gene expression during differentiation. Pkm2 also mediated the incorporation of Dpf2 and Baf250a into the regulatory sequences controlling myogenic gene expression. Pkm1 did not affect expression but was required for nuclear localization of Dpf2. Additionally, Pkm2 was required not only for the incorporation of phosphorylated H3T11 in myogenic promoters, but also for the incorporation of phosphorylated H3T6 and H3T45 at myogenic promoters via regulation of AKT and protein kinase C isoforms that phosphorylate those amino acids. Our results identify multiple unique roles for Pkm2 and a novel function for Pkm1 in gene expression and chromatin regulation during myoblast differentiation.
Article
The packaging of DNA into chromatin in eukaryotes regulates gene transcription, DNA replication and DNA repair. ATP-dependent chromatin remodelling enzymes (re)arrange nucleosomes at the first level of chromatin organization. Their Snf2-type motor ATPases alter histone-DNA interactions through a common DNA translocation mechanism. Whether remodeller activities mainly catalyse nucleosome dynamics or accurately co-determine nucleosome organization remained unclear. In this Review, we discuss the emerging mechanisms of chromatin remodelling: dynamic remodeller architectures and their interactions, the inner workings of the ATPase cycle, allosteric regulation and pathological dysregulation. Recent mechanistic insights argue for a decisive role of remodellers in the energy-driven self-organization of chromatin, which enables both stability and plasticity of genome regulation - for example, during development and stress. Different remodellers, such as members of the SWI/SNF, ISWI, CHD and INO80 families, process (epi)genetic information through specific mechanisms into distinct functional outputs. Combinatorial assembly of remodellers and their interplay with histone modifications, histone variants, DNA sequence or DNA-bound transcription factors regulate nucleosome mobilization or eviction or histone exchange. Such input-output relationships determine specific nucleosome positions and compositions with distinct DNA accessibilities and mediate differential genome regulation. Finally, remodeller genes are often mutated in diseases characterized by genome dysregulation, notably in cancer, and we discuss their physiological relevance.
Article
Full-text available
The SWI/SNF ATP-dependent chromatin remodeler is a master regulator of the epigenome, controlling pluripotency and differentiation. Towards the C-terminus of the catalytic subunit of SWI/SNF is a motif called the AT-hook that is evolutionary conserved. The AT-hook is present in many chromatin modifiers and generally thought to help anchor them to DNA. We observe however that the AT-hook regulates the intrinsic DNA-stimulated ATPase activity aside from promoting SWI/SNF recruitment to DNA or nucleosomes by increasing the reaction velocity a factor of 13 with no accompanying change in substrate affinity (KM). The changes in ATP hydrolysis causes an equivalent change in nucleosome movement, confirming they are tightly coupled. The catalytic subunit’s AT-hook is required in vivo for SWI/SNF remodeling activity in yeast and mouse embryonic stem cells. The AT-hook in SWI/SNF is required for transcription regulation and activation of stage-specific enhancers critical in cell lineage priming. Similarly, growth assays suggest the AT-hook is required in yeast SWI/SNF for activation of genes involved in amino acid biosynthesis and metabolizing ethanol. Our findings highlight the importance of studying SWI/SNF attenuation versus eliminating the catalytic subunit or completely shutting down its enzymatic activity.
Article
Full-text available
Cohesin is a highly conserved, multiprotein complex whose canonical function is to hold sister chromatids together to ensure accurate chromosome segregation. Cohesin association with chromatin relies on the Scc2-Scc4 cohesin loading complex that enables cohesin ring opening and topological entrapment of sister DNAs. To better understand how sister chromatid cohesion is regulated, we performed a proteomic screen in budding yeast that identified the Isw1 chromatin remodeler as a cohesin binding partner. In addition, we found that Isw1 also interacts with Scc2-Scc4. Lack of Isw1 protein, the Ioc3 subunit of ISW1a or Isw1 chromatin remodeling activity resulted in increased accumulation of cohesin at centromeres and pericentromeres, suggesting that ISW1a may promote efficient translocation of cohesin from the centromeric site of loading to neighboring regions. Consistent with the role of ISW1a in the chromatin organization of centromeric regions, Isw1 was found to be recruited to centromeres. In its absence we observed changes in the nucleosomal landscape at centromeres and pericentromeres. Finally, we discovered that upon loss of RSC functionality, ISW1a activity leads to reduced cohesin binding and cohesion defect. Taken together, our results support the notion of a key role of chromatin remodelers in the regulation of cohesin distribution on chromosomes.
Article
Background: Triple-negative breast cancer (TNBC) has the poorest prognosis among all breast cancer subtypes. While several tumor types are excepted to have a curative response to immunotherapy through the AT-rich interaction domain 1A (ARID1A) gene, its role in TNBC remains unclear. Methods: The expression of the ARID1A gene and immune infiltration in TNBC were analyzed by way and function enrichment analysis. Additionally, 27 gene mutations, including ARID1A mutation, were detected in paraffin-embedded TNBC specimens and normal breast specimens using Next Generation Sequencing (NGS). Immunohistochemical staining was employed to detect the expression of AIRD1A, TP53, Ki67, CD4, CD8, and PD-L1 proteins in TNBC and the adjacent normal tissue samples. Results: The bioinformatics analysis revealed that ARID1A was mutated in TNBC and significantly associated with tumor immune infiltration. NGS analysis showed a high mutation rate of ARID1A (35%) in TNBC, but the mutation status of ARID1A was not associated with age at onset, lymph node metastasis, pathological grade, or Ki67 index. Low expression or loss of AIRD1A was more commonly observed in TNBC tissues (36/108) as compared to normal tissues (3/25). Positive expression of CD8 and PD-L1 was observed in TNBC tissues with low ARID1A expression. ARID1A mutation was associated with low protein expression, and patients with ARID1A mutation or low protein expression had shorter progression-free survival. Conclusion: The ARID1A mutation and low expression are associated with poor prognosis and high immune infiltration in TNBC, and might be biomarkers for TNBC prognosis and immunotherapy efficacy.
Article
Full-text available
ISWI-family nucleosome remodeling enzymes need the histone H4 N-terminal tail to mobilize nucleosomes. Here we mapped the H4-tail binding pocket of ISWI. Surprisingly the binding site was adjacent to but not overlapping with the docking site of an auto-regulatory motif, AutoN, in the N-terminal region (NTR) of ISWI, indicating that AutoN does not act as a simple pseudosubstrate as suggested previously. Rather, AutoN cooperated with a hitherto uncharacterized motif, termed AcidicN, to confer H4-tail sensitivity and discriminate between DNA and nucleosomes. A third motif in the NTR, ppHSA, was functionally required in vivo and provided structural stability by clamping the NTR to Lobe 2 of the ATPase domain. This configuration is reminiscent of Chd1 even though Chd1 contains an unrelated NTR. Our results shed light on the intricate structural and functional regulation of ISWI by the NTR and uncover surprising parallels with Chd1.
Article
Full-text available
Nuclear actin and actin-related proteins (Arps) are key components of chromatin remodeling and modifying complexes. Although Arps are essential for the functions of chromatin remodelers, their specific roles and mechanisms are unclear. Here we define the nucleosome binding interfaces and functions of the evolutionarily conserved Arps in the yeast INO80 chromatin remodeling complex. We show that the N-terminus of Arp8, C-terminus of Arp4 and the HSA domain of Ino80 bind extranucleosomal DNA 37-51 base pairs from the edge of nucleosomes and function as a DNA-length sensor that regulates nucleosome sliding by INO80. Disruption of Arp8 and Arp4 binding to DNA uncouples ATP hydrolysis from nucleosome mobilization by disengaging Arp5 from the acidic patch on histone H2A-H2B and the Ino80-ATPase domain from the Super-helical Location (SHL) -6 of nucleosomes. Our data suggest a functional interplay between INO80's Arp8-Arp4-actin and Arp5 modules in sensing the DNA length separating nucleosomes and regulating nucleosome positioning.
Article
Full-text available
DNA in the eukaryotic nucleus is packaged in the form of nucleosomes, ~147 base pairs of DNA wrapped around a histone protein octamer. The position and histone composition of nucleosomes is governed by ATP dependent chromatin remodelers1–3 such as the 15 subunit INO80 complex4. INO80 regulates gene expression, DNA repair and replication by sliding nucleosomes, exchanging histone H2A.Z with H2A, and positioning +1 and -1 nucleosomes at promoter DNA5–8. A structure and mechanism for these remodeling reactions is lacking. Here we report the cryo-electron microscopy structure at 4.3Å resolution, with parts at 3.7Å, of an evolutionary conserved core INO80 complex from Chaetomium thermophilum bound to a nucleosome. INO80core cradles one entire gyre of the nucleosome through multivalent DNA and histone contacts. A Rvb1/2 AAA⁺ ATPase hetero-hexamer is an assembly scaffold for the complex and acts as stator for the motor and nucleosome gripping subunits. The Swi2/Snf2 ATPase motor binds to SHL-6, unwraps ~15 base pairs, disrupts the H2A:DNA contacts and is poised to pump entry DNA into the nucleosome. Arp5-Ies6 grip SHL-2/-3 acting as counter grip for the motor on the other side of the H2A/H2B dimer. The Arp5 insertion domain forms a grappler element that binds the nucleosome dyad, connects the Arp5 core and entry DNA over a distance of ~90Å and packs against histone H2A/H2B near the acidic patch. Our structure together with biochemical data8 suggest a unified mechanism for nucleosome sliding and histone editing by INO80. The motor pumps entry DNA across H2A/H2B against Arp5 and the grappler, sliding nucleosomes as a ratchet. Transient exposure of H2A/H2B by the motor and differential recognition of H2A.Z and H2A may regulate histone exchange during translocation.
Article
Full-text available
Access to DNA within nucleosomes is required for a variety of processes in cells including transcription, replication and repair. Consequently, cells encode multiple systems that remodel nucleosomes. These complexes can be simple, involving one or a few protein subunits, or more complicated multi-subunit machines1. Biochemical studies2–4 have placed the motor domains of several chromatin remodellers in the superhelical location 2 region of the nucleosome. Structural studies of yeast Chd1 and Snf2—a subunit in the complex with the capacity to remodel the structure of chromatin (RSC)—in complex with nucleosomes5–7 have provided insights into the basic mechanism of nucleosome sliding performed by these complexes. However, how larger, multi-subunit remodelling complexes such as INO80 interact with nucleosomes and how remodellers carry out functions such as nucleosome sliding8, histone exchange9 and nucleosome spacing10–12 remain poorly understood. Although some remodellers work as monomers13, others work as highly cooperative dimers11, 14, 15. Here we present the structure of the human INO80 chromatin remodeller with a bound nucleosome, which reveals that INO80 interacts with nucleosomes in a previously undescribed manner: the motor domains are located on the DNA at the entry point to the nucleosome, rather than at superhelical location 2. The ARP5–IES6 module of INO80 makes additional contacts on the opposite side of the nucleosome. This arrangement enables the histone H3 tails of the nucleosome to have a role in the regulation of the activities of the INO80 motor domain—unlike in other characterized remodellers, for which H4 tails have been shown to regulate the motor domains. Cryo-electron microscopy structure of the human IO80 chromatin remodeller in complex with a bound nucleosome reveals that its motor domains are located at the DNA wrap around the histone core.
Article
Full-text available
Chromatin-remodelling factors change nucleosome positioning and facilitate DNA transcription, replication, and repair. The conserved remodelling factor chromodomain-helicase-DNA binding protein 1(Chd1) can shift nucleosomes and induce regular nucleosome spacing. Chd1 is required for the passage of RNA polymerase IIthrough nucleosomes and for cellular pluripotency. Chd1 contains the DNA-binding domains SANT and SLIDE, a bilobal motor domain that hydrolyses ATP, and a regulatory double chromodomain. Here we report the cryo-electron microscopy structure of Chd1 from the yeast Saccharomyces cerevisiae bound to a nucleosome at a resolution of 4.8 Å. Chd1 detaches two turns of DNA from the histone octamer and binds between the two DNA gyres in a state poised for catalysis. The SANT and SLIDE domains contact detached DNA around superhelical location (SHL) -7 of the first DNA gyre. The ATPase motor binds the second DNA gyre at SHL +2 and is anchored to the N-terminal tail of histone H4, as seen in a recent nucleosome-Snf2 ATPase structure. Comparisons with published results reveal that the double chromodomain swings towards nucleosomal DNA at SHL +1, resulting in ATPase closure. The ATPase can then promote translocation of DNA towards the nucleosome dyad, thereby loosening the first DNA gyre and remodelling the nucleosome. Translocation may involve ratcheting of the two lobes of the ATPase, which is trapped in a pre- or post-translocation state in the absence or presence, respectively, of transition state-mimicking compounds.
Article
Full-text available
ATP-dependent chromatin remodellers modulate nucleosome dynamics by mobilizing or disassembling nucleosomes, as well as altering nucleosome composition. These chromatin remodellers generally function by translocating along nucleosomal DNA at the H3–H4 interface of nucleosomes. Here we show that, unlike other remodellers, INO80 translocates along DNA at the H2A–H2B interface of nucleosomes and persistently displaces DNA from the surface of H2A–H2B. DNA translocation and DNA torsional strain created near the entry site of nucleosomes by INO80 promotes both the mobilization of nucleosomes and the selective exchange of H2A.Z–H2B dimers out of nucleosomes and replacement by H2A–H2B dimers without any additional histone chaperones. We find that INO80 translocates and mobilizes H2A.Z-containing nucleosomes more efficiently than those containing H2A, partially accounting for the preference of INO80 to replace H2A.Z with H2A. Our data suggest that INO80 has a mechanism for dimer exchange that is distinct from other chromatin remodellers including its paralogue SWR1.
Article
Full-text available
The SWI/SNF chromatin remodeling complex is highly conserved from yeast to human, and aberrant SWI/SNF complexes contribute to human disease. The Snf5/SMARCB1/INI1 subunit of SWI/SNF is a tumor suppressor frequently lost in pediatric rhabdoid cancers. We examined the effects of Snf5 loss on the composition, nucleosome binding, recruitment, and remodeling activities of yeast SWI/SNF. The Snf5 subunit is shown by crosslinking-mass spectrometry (CX-MS) and subunit deletion analysis to interact with the ATPase domain of Snf2 and to form a submodule consisting of Snf5, Swp82, and Taf14. Snf5 promotes binding of the Snf2 ATPase domain to nucleosomal DNA and enhances the catalytic and nucleosome remodeling activities of SWI/ SNF. Snf5 is also required for SWI/SNF recruitment by acidic transcription factors. RNA-seq analysis suggests that both the recruitment and remodeling functions of Snf5 are required in vivo for SWI/SNF regulation of gene expression. Thus, loss of SNF5 alters the structure and function of SWI/SNF.
Article
The yeast INO80 chromatin remodeling complex plays essential roles in regulating DNA damage repair, replication, and promoter architecture. INO80's role in these processes is likely related to its ability to slide nucleosomes, but the underlying mechanism is poorly understood. Here we use ensemble and single-molecule enzymology to study INO80-catalyzed nucleosome sliding. We find that the rate of nucleosome sliding by INO80 increases ∼100-fold when the flanking DNA length is increased from 40 to 60 bp. Furthermore, once sliding is initiated, INO80 moves the nucleosome rapidly at least 20 bp without pausing to re-assess flanking DNA length, and it can change the direction of nucleosome sliding without dissociation. Finally, we show that the Nhp10 module of INO80 plays an auto-inhibitory role, tuning INO80's switch-like response to flanking DNA. Our results indicate that INO80 is a highly processive remodeling motor that is tightly regulated by both substrate cues and non-catalytic subunits.
Article
Chromatin remodellers are helicase-like, ATP-dependent enzymes that alter chromatin structure and nucleosome positions to allow regulatory proteins access to DNA. Here we report the cryo-electron microscopy structure of chromatin remodeller Switch/sucrose non-fermentable (SWI2/SNF2) from Saccharomyces cerevisiae bound to the nucleosome. The structure shows that the two core domains of Snf2 are realigned upon nucleosome binding, suggesting activation of the enzyme. The core domains contact each other through two induced Brace helices, which are crucial for coupling ATP hydrolysis to chromatin remodelling. Snf2 binds to the phosphate backbones of one DNA gyre of the nucleosome mainly through its helicase motifs within the major domain cleft, suggesting a conserved mechanism of substrate engagement across different remodellers. Snf2 contacts the second DNA gyre via a positively charged surface, providing a mechanism to anchor the remodeller at a fixed position of the nucleosome. Snf2 locally deforms nucleosomal DNA at the site of binding, priming the substrate for the remodelling reaction. Together, these findings provide mechanistic insights into chromatin remodelling.
Article
ISWI is a member of the SWI2/SNF2 family of chromatin remodellers, which also includes Snf2, Chd1, and Ino80. ISWI is the catalytic subunit of several chromatin remodelling complexes, which mobilize nucleosomes along genomic DNA, promoting replication progression, transcription repression, heterochromatin formation, and many other nuclear processes. The ATPase motor of ISWI is an autonomous remodelling machine, whereas its carboxy (C)-terminal HAND–SAND–SLIDE (HSS) domain functions in binding extranucleosomal linker DNA. The activity of the catalytic core of ISWI is inhibited by the regulatory AutoN and NegC domains, which are in turn antagonized by the H4 tail and extranucleosomal DNA, respectively, to ensure the appropriate chromatin landscape in cells. How AutoN and NegC inhibit ISWI and regulate its nucleosome-centring activity remains elusive. Here we report the crystal structures of ISWI from the thermophilic yeast Myceliophthora thermophila and its complex with a histone H4 peptide. Our data show the amino (N)-terminal AutoN domain contains two inhibitory elements, which collectively bind the second RecA-like domain (core2), holding the enzyme in an inactive conformation. The H4 peptide binds to the core2 domain coincident with one of the AutoN-binding sites, explaining the ISWI activation by H4. The H4-binding surface is conserved in Snf2 and functions beyond AutoN regulation. The C-terminal NegC domain is involved in binding to the core2 domain and functions as an allosteric element for ISWI to respond to the extranucleosomal DNA length.