ArticlePDF Available

Structure of nucleosome-bound human BAF complex

Authors:

Abstract and Figures

Architecture of human BAF complex The SWI/SNF family chromatin remodelers regulate chromatin and transcription. The protein complexes BAF and PBAF are mammalian SWI/SNF remodelers that play essential functions in diverse developmental and physiological processes. He et al. determined the structure of the human BAF complex, which contains three modules that bind the nucleosome on the top, bottom, and side, making this nucleosome-recognition pattern distinct from other chromatin remodelers. Mutations in BAF that are frequently associated with human cancer cluster into a nucleosome-interacting region. This structure provides a framework for understanding the BAF-mediated chromatin remodeling mechanism and its dysregulation in cancer. Science , this issue p. 875
Content may be subject to copyright.
STRUCTUR AL BIOLOGY
Structure of nucleosome-bound human BAF complex
Shuang He
1,2
*, Zihan Wu
1,2
*, Yuan Tian
1,2
*, Zishuo Yu
1,2
, Jiali Yu
1,2
, Xinxin Wang
1,2
, Jie Li
1
,
Bijun Liu
1
, Yanhui Xu
1,2,3,4
Mammalian SWI/SNF family chromatin remodelers, BRG1/BRM-associated factor (BAF) and polybromo-
associated BAF (PBAF), regulate chromatin structure and transcription, and their mutations are linked
to cancers. The 3.7-angstrom-resolution cryoelectron microscopy structure of human BAF bound
to the nucleosome reveals that the nucleosome is sandwiched by the base and the adenosine
triphosphatase (ATPase) modules, which are bridged by the actin-related protein (ARP) module. The
ATPase motor is positioned proximal to nucleosomal DNA and, upon ATP hydrolysis, engages with and
pumps DNA along the nucleosome. The C-terminal ahelix of SMARCB1, enriched in positively charged
residues frequently mutated in cancers, mediates interactions with an acidic patch of the nucleosome.
AT-rich interactive domain-containing protein 1A (ARID1A) and the SWI/SNF complex subunit SMARCC
serve as a structural core and scaffold in the base module organization, respectively. Our study provides
structural insights into subunit organization and nucleosome recognition of human BAF complex.
The adenosine triphosphate (ATP)dependent
chromatin remodeling complexes (also
known as chromatin remodelers) reg-
ulate the chromatin packing state by
sliding, ejecting, and restructuring the
nucleosome to enable dynamic regulation of
chromatin structure (1,2). As prototype chro-
matin remodelers, the SWI/SNF complexes
demonstrate nucleosome sliding activity and
distinctive ejection activity, by which they
create nucleosome-depleted regions (NDRs)
that are essential for transcriptional regulation
(310). Mammalian SWI/SNF (mSWI/SNF)
complexes, BRG1/BRM-associated factor (BAF)
and polybromo-associated BAF (PBAF), consist
of up to 15 subunits encoded by more than
29 genes, generating more than 1400 possible
complexes (6,7,11,12). Up to 20% of malig-
nancies contain mutations of BAF and/or PBAF
subunits, making these complexes among the
most frequently dysregulated targets in human
cancer (4,7,13).
Although the compositions and subunit
functions of SWI/SNF complexes have been
extensively studied (8,11,1419), the structural
studies of SWI/SNF complexes have been lim-
ited to the low-resolution electron microscopy
(EM) structures of yeast SWI/SNF complexes
(2023) and structures of isolated domains
(15,2427). A recent study reported an ~7-
Å-resolution structure of RSC (yeast homolog
of PBAF) bound to the nucleosome with the
substrate recruitment module refined to 3.4-Å
resolution (28). However, the molecular mech-
anisms of subunit organization and nucleo-
some recognition of mammalian BAF complex
remain largely unknown.
Structure determination
We reconstituted human BAF complex con-
sisting of the catalytic subunit SMARCA4
(BRG1) and nine auxiliary subunits (Fig. 1A,
figs. S1 and S2, and table S1). The purified
BAF complexes exhibited nucleosome sliding
activities (fig. S1, D and H to J) and were com-
plexed with nucleosome core particle (NCP) in
the absence of ATP and adenosine diphosphate
(ADP) (fig. S1D). The cryo-EM structure of the
BAF-NCP complex (~1.2 MDa) was determined
at an overall resolution of 3.7 Å, with the map of
thebasemodulelocallyrefinedto3.0-Åres-
olution (Figs. 1, B to F, and 2A; figs. S3 and S4;
table S2; and movies S1 to S5). The structural
models were built by fitting available struc-
turesintothecryo-EMmaps(15,24,26,27,29)
followed by manual model building aided by
cross-linking mass spectrometry (XL-MS) (fig.
S2,AandB,anddataS1andS2).The3.7-
Å-resolution map and corresponding model
were used hereafter unless otherwise specified.
BAF sandwiches the nucleosome
ThehumanBAFcomplexsandwichesthenu-
cleosome, and its nucleosome-binding manner
is distinct from that of other representative
chromatin remodelers, including Chd1, SWR1,
and INO80 (3032) (Fig. 1, B to D, and fig. S5).
These remodelers primarily bind nucleosomal
DNA and/or histone tails but have fewer con-
tacts with core histones. The sandwiched
nucleosome binding of BAF may provide a
structural basis to support nucleosome ejec-
tion activity (discussed below). The nucleoso-
mal DNA tends to be detached or disordered
at both entry and exit sites (Fig. 1, E and F,
and movie S5), which may allow the DNA
translocation to occur more efficiently owing
to fewer restraints.
The BAF complex consists of three modules:
the adenosine triphosphatase (ATPase) module,
the actin-related protein (ARP) module, and the
base module (Fig. 1A). A large portion of the
catalytic subunit SMARCA4 (residues 521 to
1647) forms the ATPase module, which grasps
the nucleosome with the ATPase motor par-
tially wrapping around the nucleosomal DNA
(Fig. 1, B to D). Within the ARP module, the
helicase-SANTassociated region (HSA; resi-
dues 446 to 520) of SMARCA4 binds the het-
erodimer formed by ACTL6A (BAF53A) and
ACTB (b-actin) (Fig. 1, A and B). The pre-HSA
(residues 350 to 445) of SMARCA4 is anchored
intothebasemodule,inwhichtheSMARCB1
(BAF47,hSNF5,orINI1)packsagainstthe
bottomsurfaceofthenucleosome(Fig.1,A
and B).
The ARP module bridges the ATPase and
base modules
TheARPmoduleisformedbytheACTL6A-
ACTB heterodimer and the long ahelix of the
HSA of SMARCA4 (Fig. 1G), revealing a fold
similar to the yeast HSA
Snf2
-arp7-arp9-Rtt102
structure (27). The ARP module has no direct
contact with the nucleosome, but it associates
with and bridges the ATPase and base mod-
ules(Fig.1,B,C,andG).Acryo-EMmapre-
veals considerable contacts between lobe 1 of
the SMARCA4 ATPase domain and subdo-
main 4 (residues 266 to 305) of ACTL6A (Fig. 1,
B and G). Moreover, the pre-HSA in the base
moduleandpost-HSAintheATPasemodule
aredirectlyconnectedbytheHSAhelix.Thus,
the ARP module maintains a rigid conforma-
tion of the HSA helix and likely couples the
motions of the ATPase and base modules
during chromatin remodeling. This obser-
vation is consistent with the essential role of
the ARP module both in couplingthe DNA
translocation and ATP hydrolysis and in the
assembly of functional SWI/SNF complexes
(8,18,27).
The ATPase motor engages with
nucleosomal DNA
The cryo-EM density of the ATPase motor is
relatively weak and reveals a tilted and open
conformation (angle of ~90° between the two
ATPase lobes), indicating that the structure
represents a pre-engaged conformation (fig. S6),
which is consistent with the lack of ATP and
ADP in the BAF-NCP structure. The ATPase
motor is positioned at nucleosomal DNA near
superhelical location (SHL) 2.5, and the posi-
tioning is likely guided by the HSA-associated
ARP module and ATPase-associated Snf2 ATP
coupling (SnAC) domain. We observed weak
cryo-EM density stretching across the top
RESEARCH
He et al., Science 367, 875881 (2020) 21 February 2020 1of7
1
Fudan University Shanghai Cancer Center, Institutes of
Biomedical Sciences, State Key Laboratory of Genetic
Engineering, and Shanghai Key Laboratory of Medical
Epigenetics, Shanghai Medical College of Fudan University,
Shanghai 200032, China.
2
The International Co-laboratory of
Medical Epigenetics and Metabolism, Ministry of Science and
Technology, China, Department of Systems Biology for
Medicine, School of Basic Medical Sciences, Shanghai
Medical College of Fudan University, Shanghai 200032,
China.
3
Human Phenome Institute, Collaborative Innovation
Center of Genetics and Development, School of Life
Sciences, Fudan University, Shanghai 200433, China.
4
CAS
Center for Excellence in Molecular Cell Science, Chinese
Academy of Sciences, Shanghai 200031, China.
*These authors contributed equally to this work.
Corresponding author. Email: xuyh@fudan.edu.cn
on February 25, 2020 http://science.sciencemag.org/Downloaded from
surface of the nucleosome (Fig. 1D and fig.
S6A). This region is likely derived from the
SnAC and/or bromodomain, which have
been shown to bind DNA and/or histone
(25,33,34).
The structure of BAF-NCP in the presence of
ADP was refined to 10.3-Å resolution, showing
that the ATPase motor adopts a relatively
closed conformation (~70°) and has tight con-
tacts to nucleosomal DNA around SHL 2 (fig.
S6B and movie S6). Thus, ATP or ADP would
promote conformational transition of the BAF
complex from a pre-engaged to an engaged
state, with the ATPase engaging with nu-
cleosomal DNA and, upon ATP hydrolysis,
pumping DNA toward the nucleosome dyad
and generating DNA translocation.
The organization of the base module
The pre-HSA of SMARCA4 is anchored into
the base module, which consists of seven ad-
ditional auxiliary subunits: two BAF-specific
subunits, ARID1A (BAF250A) and DPF2
(BAF45D); and five BAF/PBAF-shared subunits,
SMARCB1, SMARCD1 (BAF60A), SMARCE1
(BAF57), and two copies of SMARCC (8,11,1519)
(Figs. 1 and 2). These auxiliary subunits exist
exclusively in the base module and account
for ~80% of the total molecular mass of the
BAF complex (fig. S4).
Thebasemodulerevealsacompactfoldand
can be divided into five closely associated sub-
modules: the head, thumb, palm, bridge, and
fingers (Fig. 2). The head and bridge bind the
nucleosome and the ARP module, respectively,
generating intermodular contacts (Fig. 1G).
The thumb is formed by a SANT domain of
SMARCC, the pre-HSA of SMARCA4, and the
C-terminal helices of SMARCD1 (Fig. 2). The
fingers submodule reveals a characteristic
Y-shaped five-helix bundle formed by coiled-
coil (CC) domains of SMARCD1, SMARCE1,
He et al., Science 367, 875881 (2020) 21 February 2020 2of7
Dyad
1.5
2.5
DNA
entry
3.5
WH
SMARCB1
ACTL6A
lobe 2
lobe 1
ACTB
90°
ATPase
ATPase
DNA
Exit
HSA
HSA
DNA
Exit
DNA
Entry
HSA
HSA
ATPase
ATPase
Thumb
Fingers
Palm
Head
Bridge
Fingers
Palm
Thumb
H2A
H2A
/H2B
H2B
pre-HSA
pre-HSA
SHL 2.5
SnAC?
SnAC?
Dyad
1.5
3.5 4.5
Base Module
ARP Module
ATPase Module
1246
ATPase lobe2
SMARCA4
766
ATPase lobe1
ATPase Module
521
ARP Module
BR
SnAc
RPT1 RPT2
SMARCC2
a
SMARCC2
b
SMARCB1
CC-C2
a
CC-C2
b
CC-D2
CC-E1
DPF2
Req
αC
CC-D1
Base Module
SWIB
H2A/H2B
H2B/H2A
H3/H4H4/H3
SMARCD1
pre-HSA
1647
SANT
SWIRM
SMARCA4 ACTL6A
ACTB
SMARCD1
SMARCE1 SMARCC2
a
SMARCB1
DPF2ARID1A
H2A H2B H4H3 DNA
SMARCC2
b
A
Dyad
-1.5
-2.5
-3.5
-4.5
-5.5
DNA
exit
B
D
EF
lobe 2
lobe 1
ACTL6A
WH
SMARCB1
WH
SMARCB1
ACTL6A
αC
ACTL6A
αC
ATPase
ATPase
HSA
G
RPT1
SHL 2.5
Fingers
Thumb
Palm
Head
Bridge
SMARCC2
a
SMARCC2
b
SMARCE1
ACTL6A
ACTB
ARID1A
ARID1A-insert
subdomain 4
(266-305)
90°
C
180°
ARID1A
WH
higher threshold level NCP
NCP
90°
SANT
SWIRM
Fig. 1. Cryo-EM structure of human BAF bound to NCP. (A) Schematic
architecture of BAF complex organization and domain structure of BAF
subunits. Color scheme for BAF subunits is indicated in (G) and used
throughout all figures. (Bto D) The 3.7-Å-resolution cryo-EM map of
BAF-NCP in the absence of ADP in three different views. The map at a
higher threshold level [(B), right panel] reveals more details, with the
ATPase module fading out. The map at lower threshold level [(B), left
panel] reveals density (likely derived from SnAC) packing across the top
surface of nucleosome. The intermodular contacts and BAF-NCP contacts
are indicated with white circles. (Eand F) Two different views of the
cryo-EM map of the nucleosome showing detached and disordered DNA
at entry (E) and exit (F) sites. The positions of nucleosomal DNA are
labeled with SHL numbers. (G) Cartoon model of BAF-NCP structure
shown in two different views. RPT, repeat domain; Req, Requiem
domain;SWIRM,SWI3,RSC8,andMOIRA;SANT,Swi3,Ada2,N-Cor,
and TFIIIB; ARM, armadillo repeats; CC, coiled coil; HSA, helicase-SANT
associated; ARP, actin-related protein; BR, bromodomain; WH, winged
helix; SWIB, SWI/SNF complex B/MDM2.
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
and two SMARCC subunits. The palm con-
nectspre-HSA,SMARCD1,SMARCE1,and
SMARCC, which merge at a four-way junction.
Nucleosome recognition by SMARCB1 in the
head submodule
Theheaddirectlybindsthehistoneoctamer
on the bottom (Fig. 1, B and C, and fig. S7A).
It consists of two repeat (RPT) domains
and a C-terminal a(aC) helix of SMARCB1,
the requiem (Req) domain of DPF2, two SWIRM
domains of SMARCCs, and an insert derived
from ARID1A (ARID1A-insert) (Fig. 3A and
fig. S7B). The two SWIRM domains bind the
RPT1 and RPT2 domains, respectively. The
two RPT-SWIRM complexes adopt a similar
foldandasymmetricallybindeachother,with
the intermolecular interactions buttressed by
the ARID1A-insert (residues 1802 to 1862) and
Req domain (residues 13 to 82) of DPF2 (Fig. 3,
A and B). The head merges with the bridge
and thumb at a three-way junction, and the
interactions are mediated by the interwoven
loops from DPF2, ARID1A, and two SMARCCs
(Fig.3A).TheReqdomainofDPF2isalmost
identical to that of DPF1 and DPF3 in primary
sequence, indicating a similar role of DPF1,
DPF2, and DPF3 in BAF complex organization
(fig. S2D).
SMARCB1 is required for structural integ-
rity and function of the SWI/SNF complex
(16,17,35), and inactivation of SMARCB1
was reported in almost all malignant rhabdoid
tumors (14,3638). The aC helix, which is
enriched in residues that are frequently mu-
tated in human cancers, packs against the
bottom H2A-H2B heterodimer and serves as a
hinge to connect the base module and nu-
cleosome (Fig. 3C and fig. S7A). The four most
frequently mutated arginine residues (R370,
R373, R374, and R377) together form a pos-
itively charged cluster and pack against an
acidic patch on the bottom of the nucleosome
(Fig. 3D). The four arginine residues are in-
variant across species, from yeast to humans
(Fig. 3E), and were clearly visualized in the
cryo-EM map (Fig. 3F). This observation high-
lights the importance of the aChelixof
SMARCB1 in binding of the nucleosome and
is consistent with recent studies (14,28). Al-
though the aC helix of Sfh1 (yeast homo-
log of SMARCB1) is disordered in the yeast
RSC-NCP complex structure, the deletion of
this helix impairs nucleosome ejection activ-
ity (28).
The RPT1 domain of SMARCB1 is in close
proximity to the a2 helix of histone H2A and
likely causes steric clash between the histone
octamer and DNA (Figs. 1, B and G, and 3C;
and fig. S5A), suggesting that RPT1 serves as a
wedgethat favors DNA detachment around
the exit site. The N-terminal winged helix (WH)
domain of SMARCB1 binds the ARM domain
of ARID1A and is located more than 40 Å away
from nucleosomal DNA (Fig. 1, C and G), sug-
gesting a role independent of its previously
reported DNA binding ability (26). The WH
domain is in close proximity to the ARP module,
suggesting a role in regulating ARPbase in-
termodular interactions (Fig. 1, B and C).
ARID1A serves as a rigid core to stabilize the
base module
ARID1A is the largest subunit of the BAF
complex. A large portion of ARID1A, includ-
ing the characteristic AT-rich interacting do-
main (ARID), was invisible in the complex
str ucture, possibly owing to intrinsic flexibility
He et al., Science 367, 875881 (2020) 21 February 2020 3of7
Bridge
Palm
Head
Thumb
Fingers
HSA
HSA
Pillar
Pillar
Req
Req
180°
Bridge
Head
Thumb
Fingers
HSA
HSA
Palm
ARID1A-insert
ARID1A-insert
pre-HSA
pre-HSA
pre-HSA
pre-HSA
Req
Req
αC
SWIB
SWIB
CC-E1
CC-E1
pre-HSA
pre-HSA
HSA
HSA
RPT1
RPT1
RPT2
RPT2
αC
SANT
SANT
a
*
CC-D1
CC-D1
CC-D2
CC-D2
Req
Req
ARM
ARM
Pillar
Pillar
Wedge
Wedge
CC-C2
CC-C2
b
*
SWIRM
SWIRM
a
SWIRM
SWIRM
b
SANT
SANT
b
CC-C2
CC-C2
a
*
A
90°
SMARCA4
ARID1A
SMARCB1
DPF2
SMARCD1
SMARCE1
SMARCC2
a
SMARCC2
b
B
Req
Req
HSA
HSA
RPT1
RPT1
RPT2
RPT2
WH
WH
αC
ARM
ARM
SWIB
SWIB
CC-E1
CC-E1
SWIRM
SWIRM
a
SWIRM
SWIRM
b
CC-C2
CC-C2
a
*
CC-C2
CC-C2
b
*
SANT
SANT
b
4-way junction
4-way junction
Fig. 2. Structure of the base module. (A) The cryo-EM map of the base module locally refined to 3.0-Å resolution. The submodules and elements involved in
complex assembly are indicated. (B) Cartoon model of the base module in two different views. Regions that are involved in nucleosome recognition and complex
formation are indicated. CC-C2
a
* and CC-C2
b
* represent the possibility of two CCs of SMARCC2 because of a disconnected cryo-EM map.
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
(fig. S2D). The modeled C-terminal ARM do-
main consists of seven ARM repeats arranged
in a superhelical conformation and serves as
a rigid core to bind SMARCA4 and all other
base subunits (Fig. 4A). A zinc finger is formed
with a zinc atom coordinated with residues
H2019 and H2021 on ARM3 and residues H2090
and C2094 on ARM4 (Fig. 4B). The zinc finger
stabilizes two ARM repeats and the associ-
ated loop regions. The ARM domain is lo-
cated in the center of the base module, with
the bottom associating with the fingers, one
endjoiningwiththeheadandthumb,and
theotherendcappedbytheSWIBdomainof
SMARCD1. The N-terminal helical turns of
HSA and the preceding loop pack against the
concave surface of the ARM domain. The sur-
face residues of ARM contacting SMARCA4
and SMARCD1 are highly conserved, suggest-
ing evolutionarily conserved functions in sta-
bilizingthebasemodule.
We found that, in a manner consistent with
the central role of the ARM domain in base
module organization, ARID1A bound to other
base subunits and the purified base subcom-
plex (fig. S8, A to C). ARID1A is one of the most
frequently mutated mSWI/SNF subunits in
human cancer, and frameshift mutations occur
near the C terminus (11,39), supporting the im-
portance of the C-terminal ARM domain in BAF
structure and function. The structure showed
that the overall fold of the BAF complex wo uld
not be properly maintained in the absence of
ARID1A (Fig. 4C), although other BAF subunits
would remain associated with SMARCC1 and
SMARCC2 because the BAF
DARID1A
complex sur-
vived ion-exchange chromatography (fig. S1C).
The sliding activity of BAF
DARID1A
was con-
siderably enhanced by the addition of larger
amounts of purified ARID1A (Fig. 4D and
fig. S8D), indicating that ARID1Ais required
for efficient nucleosome sliding activity.
ARID1A and ARID1B share highly similar
primary sequences and are mutually exclusive
in the BAF complex, suggesting that ARID1B-
containing BAF is assembled in a similar
manner (fig. S2D). As a PBAF-specific subunit,
ARID2 is not a paralog of ARID1A or ARID1B
and shows a distinct domain architecture.
However, the N-terminal helical region (res-
idues 150 to 470) of ARID2 is predicted to form
a seven-repeat ARM domain (40). Thus, the
N-terminal ARM domain of ARID2 and the
C-terminal ARM domain of ARID1A may play
a similar role in organizing PABF and BAF,
respectively.
SMARCCs compose base module scaffold, and
SMARCD1 and SMARCE1 facilitate organization
Asthescaffoldofthebasemodule,thetwo
SMARCC subunits bind all other base sub-
units and thread through the head, thumb,
palm, and fingers submodules (Fig. 4C). The
He et al., Science 367, 875881 (2020) 21 February 2020 4of7
E
A
R374
R373
R370
R377
R377
R374
R370
R373
H2B
H2B
C
C
C
C
RPT2
RPT2
RPT1
RPT1
DNAexit
H2A
H2A
Wedge
Wedge
Head
R370
R373
R374
R377
R370
R373
R374
R377
D
C
C
F
Y16
Y17 M21
Q23
H25
Y27
N28
L31
R35
R38 F41
Req
Req
SWIRM
SWIRM
b
b
RPT2
RPT2
B
C
Fig. 3. Structure of the head submodule and role of SMARCB1 in
nucleosome recognition. (A) Close-up view of the head submodule and its
interactions with the bridge and thumb submodules. The sandwiched loops
located in a three-way junction are indicated with a dashed blue circle.
(B) Close-up view of interactions between the Req domain of DPF2 shown in
cartoon and RTP2-SWIRM
b
shown in surface representation (red, negative
charge; blue, positive charge). (C) Close-up view of the contacts between
SMARCB1 and the bottom surface of the nucleosome. Four histone-contacting
arginine residues on the aC helix are shown in stick representation.
Single-letter abbreviations for the amino acid residues are as follows: A, Ala;
C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, Ile; K, Lys; L, Leu; M, Met;
N, Asn; P, Pro; Q, Gln; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr. (D) Basic
residues of the aC helix pack against the acidic surface of the histone
octamer. The positively charged residues are shown as sticks. (E) Conservation
of the aC helix of SMARCB1. The invariant and highly conserved residues are
highlighted with dark-green and green backgrounds, respectively. (F) The
cryo-EM map around the aC helix is shown in mesh in two views. The side chains
of the abovementioned residues are well covered by the map.
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
BAF complexes containing SMARCC1 and
SMARCC2 (BAF-CC1/CC2) and two SMARCC2
subunits (BAF-CC2/CC2) generated almost
identical cryo-EM maps (fig. S3). SMARCC1
and SMARCC2 behave similarly in binding
other BAF subunits and subcomplexes (fig. S9,
D to I, and supplementary text). This obser-
vation is consistent with a previous study
showing that SMARCC1 and SMARCC2 are
functionally similar and play critical roles in the
early stage of mSWI/SNF complex assembly (11).
SMARCD1 adopts an elongated conforma-
tion and runs alongside the two SMARCC
subunits (Fig. 4C). The SWIB domain adopts
a compact globular fold and binds the ARM7
of ARID1A and the CC of SMARCC (Fig. 4A).
The a9 helix of SMARCD1 packs against the
a1 helix of SMARCE1 and a loop of ARID1A,
generating a pillarthat connects to the
bridge (Figs. 2 and 4A). The a10, a11, and
a12 helices of SMARCD1 interact with the
SANT
b
domain and pre-HSA with the a12
helix protruding out of the intersection of the
head,bridge,andthumb(Fig.4,AandC).Thus,
SMARCD1 and SMARCE1 assist SMARCC1 and
SMARCC2 in organizing the base module.
Comparison of BAF-NCP and RSC-NCP
complex structures
The yeast RSC complex is the homolog of the
mammalian PBAF complex, consisting of ARID2
(instead of ARID1A and ARID1B) and a PBAF-
specific subunit, polybromo-1 (PBRM1, or
BAF180). Comparison of our BAF-NCP struc-
ture and a recently published yeast RSC-NPC
structure (28) revealed a similar nucleosome-
binding mode (fig. S10). Unexpectedly, the
nucleosome is mainly bound by the ATPase
domain, and the aC helix of Sfh1 is invisible
in the RSC-NCP structure (28). In contrast,
the nucleosome is sandwiched by BAF with
H2A-H2B dimer stably associated with the aC
helix of SMARCB1. The two complexes may
represent different conformational states, or
this difference might result from different ex-
perimental conditions. Structural comparison
also revealed considerable differences in the
base module organization. ARID1A, SMARCC1,
SMARCC2, SMARCD1, and SMARCE1 exhibit
conformations distinct from their counter-
parts in the RSC complex. The comparison
He et al., Science 367, 875881 (2020) 21 February 2020 5of7
CC-D1
CC-D1
CC-D2
CC-D2
CC-E1
CC-E1
SWIB
SWIB
SANT
SANT
a
*
CC-C2
CC-C2b*
CC-C2
CC-C2
a
*
CC-D1
CC-D1
CC-D2
CC-D2
CC-E1
CC-E1
SWIB
SWIB
CC-C2
CC-C2b*
CC-C2
CC-C2
a
*
SMARCA4
ARID1A
SMARCB1
DPF2
SMARCD1
SMARCE1
SMARCC2
a
SMARCC2
b
SANT
SANT
a
*
SWIRM
SWIRM
a
SWIRM
SWIRM
b
SANT
SANT
b
Bridge
Fingers
Palm
Head
WH
WH
ARM7
ARM7
HSA
HSA
Bridge
SWIB
SWIB
SWIRM
SWIRM
a
SWIRM
SWIRM
b
Req
Req
ARID1A-insert
ARID1A-insert
Thumb
Fingers
Palm
Head
Bridge
HSA
pre-HSA
SWIB
CC-D2
SWIRM
a
CC-D1
ARM7
Req
SWIRM
b
A
100°
45N45 (80nM)
ATP
Time/min
BAF (120nM)
ARID1A (nM)
1 2 3 4 5 6 7 8
SMARCA4
ARM1
ARM1
ARM2
ARM2
ARM4
ARM4
ARM5
ARM5
ARM6
ARM6
ARM7
ARM7
ARM3
ARM3
SMARCD1
Variable Average Conserved
α9
α1
ARM3
ARM3
ARM4
ARM4
H2019
H2090
C2094
H2021
B
D
C
3-way
3-way
junction
junction
zinc finger
Pillar
α
10
10
α
11
11
α
12
12
Thumb
α9
α1
SANT
b
Th
u
m
Fin
ger
s
g
m
H
ea
d
Bri
e
Bri
dge
dg
dg
dg
Bri
e
HSA
pre
-HSA
SWI
B
CC-
D2
SWIRM
a
CC-
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
D1
ARM7
Req
SWI
R
A
zinc finger
Pillar
α
α
α
α
9
α
1
SANT
b
Fig. 4. Subunit organization in the base module. (A) Cartoon model of
interactions between ARID1A and other subunits is shown in two views. (Left middle
panel) The surface of the ARM domain is shown and colored according to the
conservation scores. (Right middle panel) Seven ARM repeats are shown, with front
helices colored in yellow, back helices colored in green, and ridge helices and loops
colored in blue. (B) Close-up view of the zinc finger connecting ARM3 and ARM4,
with zinc-coordinating residues shown in sticks and zinc atom shown as a gray ball.
The location of the zinc finger is indicated with a blue rectangle in the upper panel of
(A). (C)CartoonofthebasemodulewithARID1AandSMARCA4omitted.Thelackof
ARID1A would lead to a clash of the base module, although other subunits remain
associated by the scaffold subunits, SMARCC1 and SMARCC2. The three-pronged
yellow and blue shape indicates a three-way junction of the head, thumb, and bridge.
(D) In vitro nucleosome sliding assay performed using purified BAF
DARID1A
complex
and increasing amounts of purified ARID1A. Note that BAF
DARID1A
demonstrates
nucleosome sliding activity in higher protein concentration (fig. S8D). 45N45, a
center-positioned nucleosome with two 45base pair flanking DNA fragments.
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
also suggested that Rsc7 and Htl1 in RSC are
equivalents of DPF2 and SMARCE1 of BAF,
respectively. BAF has no histone tailbinding
lobe, which may exist exclusively in RSC and
PBAF complexes. Homologs of yeast DNA
interaction subunits, RSC3 and RSC30, do
not exist in mammals, suggesting that PBAF
is also structurally different from RSC.
Model of chromatin remodeling of the
BAF complex
Our structural and biochemical analyses illus-
trated the mechanisms of organization and
nucleosome recognition of the human BAF
complex, the prototype mSWI/SNF remodeler
(Fig. 5A). BAF ejects the nucleosome and cre-
ates and maintains NDRs that are essential
for transcription. Two nonmutually exclusive
models of nucleosome ejection have been
proposed (2). The BAF-NCP structure could
fit both models (Fig. 5B). In the first model,
the BAF-bound histone octamer, and more
possibly the H2A-H2B dimer, could be evicted
because of DNA tension resulting from either
strong DNA translocation (Fig. 5B, model 1a;
independent of the adjacent nucleosome) or
collision with the adjacent nucleosome (Fig. 5B,
model 1b). In the second model, BAF sand-
wiches the nucleosome to ensure that the
ATPase domain stably engages with DNA and
peelsthe DNA off of the adjacent nucleo-
some.Inbothcases,thetwonucleosome-
sandwiching regions, the SnAC domain of
SMARCA4 and the aChelixofSMARCB1,
likely play important roles in nucleosome
ejection, which has been experimentally dem-
onstrated (14,16,28,33). The DNA detachment
may facilitate efficient DNA translocation of
the BAF-associated nucleosome because of
fewer DNAhistone contacts. The chromatin
ejection may also be promoted by the DNA-
interacting ARID domain of ARID1A and/or
acetylated histone tailbinding bromodomain
of SMARCA4, which were not observed in the
cryo-EM map, owing to flexibility.
REFERENCES AND NOTES
1. C. Y. Zhou, S. L. Johnson, N. I. Gamarra, G. J. Narlikar,
Annu. Rev. Biophys. 45, 153181 (2016).
2. C. R. Clapier, J. Iwasa, B. R. Cairns, C. L. Peterson, Nat. Rev.
Mol. Cell Biol. 18, 407422 (2017).
3. L. K. Elfring, R. Deuring, C. M. McCallum, C. L. Peterson,
J. W. Tamkun, Mol. Cell. Biol. 14, 22252234 (1994).
4. J. Masliah-Planchon, I. Bièche, J. M. Guinebretière, F. Bourdeaut,
O. Delattre, Annu. Rev. Pathol. 10,145171 (2015).
5. L. Neigeborn, M. Carlson, Genetics 108, 845858 (1984).
6. W. Wang et al., EMBO J. 15, 53705382 (1996).
7. C. Kadoch, G. R. Crabtree, Sci. Adv. 1, e1500447 (2015).
8. C. R. Clapier et al., Mol. Cell 62, 453461 (2016).
9. M. L. Dechassa et al., Mol. Cell 38, 590602 (2010).
10. H. Boeger, J. Griesenbeck, J. S. Strattan, R. D. Kornberg,
Mol. Cell 14, 667673 (2004).
11. N. Mashtalir et al., Cell 175, 12721288.e20 (2018).
12. L. Ho, G. R. Crabtree, Nature 463, 474484 (2010).
13. C. Kadoch et al., Nat. Genet. 45, 592601 (2013).
14. A. M. Valencia et al., Cell 179, 13421356.e23 (2019).
15. L. Yan, S. Xie, Y. Du, C. Qian, J. Mol. Biol. 429, 16501660
(2017).
16. P. Sen et al., Cell Rep. 18, 21352147 (2017).
17. X. Wang et al., Nat. Genet. 49, 289295 (2017).
18. H. Szerlong et al., Nat.Struct.Mol.Biol.15, 469476
(2008).
19. M. L. Phelan, S. Sif, G. J. Narlikar, R. E. Kingston, Mol. Cell 3,
247253 (1999).
20.M.L.Dechassaet al., Mol. Cell. Biol. 28,60106021
(2008).
21. Y. Chaban et al., Nat. Struct. Mol. Biol. 15, 12721277
(2008).
22. C. L. Smith, R. Horowitz-Scherer, J. F. Flanagan, C. L. Woodcock,
C. L. Peterson, Nat. Struct. Biol. 10,141145 (2003).
23. F. J. Asturias, W. H. Chung, R. D. Kornberg, Y. Lorch, Proc. Natl.
Acad. Sci. U.S.A. 99, 1347713480 (2002).
24. M. Li et al., Nature 567, 409413 (2019).
25. E. A. Morrison et al., Nat. Commun. 8, 16080 (2017).
26. M. D. Allen, S. M. Freund, G. Zinzalla, M. Bycroft, Structure 23,
13441349 (2015).
27. H. L. Schubert et al., Proc. Natl. Acad. Sci. U.S.A. 110,
33453350 (2013).
28. Y. Ye et al., Science 366,838843 (2019).
29. K. Luger, A. W. Mäder, R. K. Richmond, D. F. Sargent,
T. J. Richmond, Nature 389, 251260 (1997).
30. L. Farnung, S. M. Vos, C. Wigge, P. Cramer, Nature 550,
539542 (2017).
31. O. Willhoft et al., Science 362, eaat7716 (2018).
32. R. Ayala et al., Nature 556, 391395 (2018).
He et al., Science 367, 875881 (2020) 21 February 2020 6of7
ACTB
ACTL6A/B
sliding
?
DNA tension is resolved by
ejection of BAF-bound histone
octamer or dimer
ATPase
post-ATPase
SMARCB1
ARP
Base
ATPase
post-ATPase
SMARCB1
ARP
Base
SMARCB1
ARP
Base
ATPase
post-ATPase
ATPase
post-ATPase
SMARCB1
ARP
Base
Model 2Model 1b
ATPase
post-ATPase
SMARCB1
ARP
Base
Free DNA
ARP
ATPase
post-ATPase
SMARCB1
Base
ARP
strong DNA translocation and
high DNA tension lead to
ejection independent
of adjacent nucleosome
Model 1a
ATPase
SMARCB1
Base
Free DNA
DNA peel-off and ejection of
adjacent histone octamer or dimer
adjacent nucleosome collides
AB
ARP
SMARCB1
SMARCE1
ARID1A/B
DPF1/2/3
SMARCD1/2/3
H2A H2B
H3 H4
SMARCE1
ACTB
ACTL6A/B
SMARCA2/4
H2AH2B
H3
H4
SMARCC1/2
SMARCC1/2
Bromodomain PHD finger domain
DNA binding domain (ARID, HMG, WH)
ATP / A D P
Two CCs of SMARCD
Two CCs of SMARCCOne CC of SMARCE1
Rotation
Free DNA
Fig. 5. Models of chromatin remodeling of the BAF complex. (A) BAF
subunits are shown in cartoon model. The indicated paralogs of BAF subunits are
mutually exclusive in the BAF complex. The nucleosome is sandwiched by
SMARCA2/4 and SMARCB1. The ATPase of SMARCA2/4 grasps nucleosomal
DNA and generates DNA translocation in an ATP-dependent manner. PHD, plant
homeodomain; HMG, high-mobility group. (B) The BAF-NCP structure could
fit two nonexclusive models of nucleosome ejection. The histone octamer or
dimer could be ejected from BAF-bound nucleosome (model 1) or the adjacent
nucleosome (model 2). The ejection could occur independent of (model 1a) or
dependent upon (model 1b) the adjacent nucleosome.
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
33. P. Sen et al., Mol. Cell. Biol. 33, 360370 (2013).
34. W. Shen et al., Biochemistry 46, 21002110 (2007).
35. R. T. Nakayama et al., Nat. Genet. 49, 16131623 (2017).
36. I. Versteege et al., Nature 394, 203206 (1998).
37. C. Kadoch, G. R. Crabtree, Cell 153,7185 (2013).
38. M. J. McBride et al., Cancer Cell 33, 11281141.e7 (2018).
39. R. Mathur et al., Nat. Genet. 49, 296302 (2017).
40. L. A. Kelley, S. Mezulis, C. M. Yates, M. N. Wass,
M. J. Sternberg, Nat. Protoc. 10, 845858 (2015).
ACKNOWL EDGME NTS
We thank the Center of Cryo-Electron Microscopy, Fudan
University; the Center of Cryo-Electron Microscopy, ShanghaiTech
University; the Center for Biological Imaging of Institute of
Biophysics (IBP) of Chinese Academy of Sciences (CAS); and
the National Center for Protein Science Shanghai (NCPSS) for
supporting cryo-EM data collection and data analyses. We also
thank the Biomedical Core Facility, Fudan University, for
supporting mass spectrometry analyses. Funding: This work was
supported by grants from the National Key R&D Program of China
(2016YFA0500700), the National Natural Science Foundation of
China (31830107, 31821002, and 31425008), the National Ten-
Thousand Talent Program (Y.X.), the National Program for Support
of Top-Notch Young Professionals (Y.X.), the Shanghai Municipal
Science and Technology Major Project (2017SHZDZX01), and the
Strategic Priority Research Program of the Chinese Academy of
Sciences (XDB08000000). Author contributions: S.H. prepared
the samples for structural and biochemical analyses with help from
J.Y., J.L., X.W., and B.L. Z.W. and Y.T. performed EM analyses
and model building with help from Z.Y. Y.X. and S.H. wrote the
manuscript. Y.X. supervised the project. Competing interests: The
authors declare no competing interests. Data and materials
availability: Cryo-EM maps have been deposited in the Electron
Microscopy Data Bank (EMDB) under accession numbers
EMD-0968 (base module), EMD-0969 (ARP module), EMD-0970
(ATPase-NCP with aC of SMARCB1), EMD-0972 (ATPase-NCP with
detached DNA), EMD-0974 (BAF-NCP, 3.7 Å), EMD-0971 (BAF-NCP,
6.6 Å), and EMD-0973 (BAF
ADP
-NCP). Atomic coordinates for the
base module and BAF-NCP have been deposited in the Protein Data
Bank under IDs 6LTH and 6LTJ, respectively.
SUPPLEMENTARY MATERIALS
science.sciencemag.org/content/367/6480/875/suppl/DC1
Materials and Methods
Supplementary Text
Figs. S1 to S10
Tables S1 and S2
References (4161)
Movies S1 to S6
Data S1 and S2
View/request a protocol for this paper from Bio-protocol.
24 October 2019; resubmitted 10 January 2020
Accepted 22 January 2020
Published online 30 January 2020
10.1126/science.aaz9761
He et al., Science 367, 875881 (2020) 21 February 2020 7of7
RESEARCH |RESEARCH ARTICLE
on February 25, 2020 http://science.sciencemag.org/Downloaded from
Structure of nucleosome-bound human BAF complex
Shuang He, Zihan Wu, Yuan Tian, Zishuo Yu, Jiali Yu, Xinxin Wang, Jie Li, Bijun Liu and Yanhui Xu
originally published online January 30, 2020DOI: 10.1126/science.aaz9761
(6480), 875-881.367Science
, this issue p. 875Science
its dysregulation in cancer.
region. This structure provides a framework for understanding the BAF-mediated chromatin remodeling mechanism and
remodelers. Mutations in BAF that are frequently associated with human cancer cluster into a nucleosome-interacting
nucleosome on the top, bottom, and side, making this nucleosome-recognition pattern distinct from other chromatin
determined the structure of the human BAF complex, which contains three modules that bind theet al.processes. He
PBAF are mammalian SWI/SNF remodelers that play essential functions in diverse developmental and physiological
The SWI/SNF family chromatin remodelers regulate chromatin and transcription. The protein complexes BAF and
Architecture of human BAF complex
ARTICLE TOOLS http://science.sciencemag.org/content/367/6480/875
MATERIALS
SUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/01/29/science.aaz9761.DC1
REFERENCES http://science.sciencemag.org/content/367/6480/875#BIBL
This article cites 60 articles, 11 of which you can access for free
PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions
Terms of ServiceUse of this article is subject to the
is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title
(print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience
Copyright © 2020, American Association for the Advancement of Science
on February 25, 2020 http://science.sciencemag.org/Downloaded from
... Mammalian SWItch/Sucrose Non-Fermentable (mSWI/ SNF) complexes, is a subfamily of ATP-dependent chromatin remodeling complexes. The mSWI/SNF complexes include the BRG1-associated factor (BAF), in its canonical (cBAF) and noncanonical (ncBAF) subtypes, were expressed and purified according to the previous work [34]. Briefly, the plasmids consisting of the BAF subunits were co-transfected to HEK Expi293F cells using PEI, then incubated at 37 °C for 72 h. ...
... A nucleosome sliding assay was performed as previously described [34]. The BAF complexes (100 nM) were incubated with 38N38 nucleosome (100 nM) at 37 °C for 1 h in 20 µL reaction buffer containing 25 mM Tris-HCl, pH 8.0, 30 mM KCl, 5 mM MgCl 2 , 0.1 mg/mL BSA, 2% Glycerol and 3 mM ATP. ...
... To confirm the suitability of ubiquitinated nucleosomes synthesized via NDHOU approach for biochemical studies, we employed them as substrates in a chromatin remodeling assay. BAF complexes, which are part of the human SWI/SNF remodeling family, reposition nucleosomes along DNA in an ATP-dependent mechanism [34]. We produced two variants of the BAF complex (cBAF and ncBAF) and conducted the remodeling assays with both wild-type and ubiquitinated nucleosomes. ...
Article
Full-text available
Background Histone ubiquitination modification is emerging as a critical epigenetic mechanism involved in a range of biological processes. In vitro reconstitution of ubiquitinated nucleosomes is pivotal for elucidating the influence of histone ubiquitination on chromatin dynamics. Results In this study, we introduce a Non-Denatured Histone Octamer Ubiquitylation (NDHOU) approach for generating ubiquitin or ubiquitin-like modified histone octamers. The method entails the co-expression and purification of histone octamers, followed by their chemical cross-linking to ubiquitin using 1,3-dibromoacetone. We demonstrate that nucleosomes reconstituted with these octamers display a high degree of homogeneity, rendering them highly compatible with in vitro biochemical assays. These ubiquitinated nucleosomes mimic physiological substrates in function and structure. Additionally, we have extended this method to cross-linking various histone octamers and three types of ubiquitin-like proteins. Conclusions Overall, our findings offer an efficient strategy for producing ubiquitinated nucleosomes, advancing biochemical and biophysical studies in the field of chromatin biology.
... mutations 5,[8][9][10][11][12][13] . BRG1, the key ATPase subunit of BAF complexes, has been proposed to function in gene regulation by creating nucleosome free regions (NFRs) via nucleosome ejection and/or sliding 4,[14][15][16][17][18][19][20] . Recent studies demonstrated that BRG1 mainly represses transcription from promoters associated with H3K4me3 and narrow NFRs while it counteracts transcriptional repression of promoters associated with H3K4me3 and H3K27me bivalent modifications 10,21 . ...
... All three shift patterns resulted in narrower spacing between the neighboring nucleosomes both at TSSs and enhancers, consistent with decreased accessibility and nascent transcription from these elements. Finally, our data provide direct mechanistic support for working models for RSC complex and SWI/SNF complex functions proposed in recent structural studies 20,44 . In summary, by acute depletion of BRG1, our study suggests that BRG1 primarily functions genome-wide as an activator of gene expression by maintaining chromatin accessibility at gene TSSs and enhancers. ...
Article
Full-text available
The mammalian SWI/SNF-like BAF complexes play critical roles during animal development and pathological conditions. Previous gene deletion studies and characterization of human gene mutations implicate that the complexes both repress and activate a large number of genes. However, the direct function of the complexes in cells remains largely unclear due to the relatively long-term nature of gene deletion or natural mutation. Here we generate a mouse line by knocking in the auxin-inducible degron tag (AID) to the Smarca4 gene, which encodes BRG1, the essential ATPase subunit of the BAF complexes. We show that the tagged BRG1 can be efficiently depleted by osTIR1 expression and auxin treatment for 6 to 10 h in CD4 + T cells, hepatocytes, and fibroblasts isolated from the knock-in mice. The acute depletion of BRG1 leads to decreases in nascent RNAs and RNA polymerase II binding at a large number of genes, which are positively correlated with the loss of BRG1. Further, these changes are correlated with diminished accessibility at DNase I Hypersensitive Sites (DHSs) and p300 binding. The acute BRG1 depletion results in three major patterns of nucleosome shifts leading to narrower nucleosome spacing surrounding transcription factor motifs and at enhancers and transcription start sites (TSSs), which are correlated with loss of BRG1, decreased chromatin accessibility and decreased nascent RNAs. Acute depletion of BRG1 severely compromises the Trichostatin A (TSA) -induced histone acetylation, suggesting a substantial interplay between the chromatin remodeling activity of BRG1 and histone acetylation. Our data suggest BRG1 mainly plays a direct positive role in chromatin accessibility, RNAPII binding, and nascent RNA production by regulating nucleosome positioning and facilitating transcription factor binding to their target sites.
... Knock down of β-actin affects chromatin organization, both in terms of accessibility as well as intra-chromatin interactions and with histones 7 . A recent crystal structure of human BAF bound to a nucleosome 20 showed that BAF contains an actin-related protein (Arp) complex consisting of SMARCA4, β-actin, and Arp4 (ACTL6A/BAF53A) 19 . This complex links the BAF ATPase module, which directly interacts with the nucleosome and the BAF base module; thus changes in actin associations would be predicted to have effects at regulating chromatin states. ...
Article
Full-text available
Polymerized β-actin may provide a structural basis for chromatin accessibility and actin transport into the nucleus can guide mesenchymal stem cell (MSC) differentiation. Using MSC, we show that using CK666 to inhibit Arp2/3 directed secondary actin branching results in decreased nuclear actin structure, and significantly alters chromatin access measured with ATACseq at 24 h. The ATAC-seq results due to CK666 are distinct from those caused by cytochalasin D (CytoD), which enhances nuclear actin structure. In addition, nuclear visualization shows Arp2/3 inhibition decreases pericentric H3K9me3 marks. CytoD, alternatively, induces redistribution of H3K27me3 marks centrally. Such alterations in chromatin landscape are consistent with differential gene expression associated with distinctive differentiation patterns. Further, knockdown of the non-enzymatic monomeric actin binding protein, Arp4, leads to extensive chromatin unpacking, but only a modest increase in transcription, indicating an active role for actin-Arp4 in transcription. These data indicate that dynamic actin remodeling can regulate chromatin interactions.
... The nucleosome sliding activity of the SWI/SNF complex depends on the presence of ARID1A. Loss of ARID1A destabilizes BAF complex folding and decreases nucleosome sliding activity [15]. Thus, ARID1A is a critical transcriptional regulator by modulating access to promoter and enhancer regions of the genome. ...
Article
Full-text available
The highest frequency of genetic alterations in the tumor suppressor ARID1A occurs in malignancies of the female reproductive tract. The prevalence of ARID1A alterations in gynecologic precancers and cancers is summarized from the literature, and the putative mechanisms of tumor suppressive action examined both in benign/precursor lesions including endometriosis and atypical hyperplasia and in malignancies of the ovary, uterus, cervix and vagina. ARID1A alterations in gynecologic cancers are usually loss-of-function mutations, resulting in diminished or absent protein expression. ARID1A deficiency results in pleiotropic downstream effects related not only to its role in transcriptional regulation as a SWI/SNF complex subunit, but also related to the functions of ARID1A in DNA replication and repair, immune modulation, cell cycle progression, endoplasmic reticulum (ER) stress and oxidative stress. The most promising actionable signaling pathway interactions and therapeutic vulnerabilities of ARID1A mutated cancers are presented with a critical review of the currently available experimental and clinical evidence. The role of ARID1A in response to chemotherapeutic agents, radiation therapy and immunotherapy is also addressed. In summary, the multi-faceted role of ARID1A mutation in precancer and cancer is examined through a clinical lens focused on development of novel preventive and therapeutic interventions for gynecological cancers.
... Mammalian cells undergo different forms of cell death, such as apoptosis, necroptosis, and pyroptosis, by two dimers of H2A-H2B [24][25][26]. Variants of H2A, H2B, and H3 have been identified in eukaryotes, and certain variants of H2A have been linked to cancer [27,28]. For instance, H2A.z is believed to have pro-tumor effects [29][30][31], while macroH2A and H2A.x are considered tumor inhibitors [32]. ...
Article
Full-text available
In recent years, there has been an increase in the incidence and mortality rates of prostate cancer (PCa). However, the specific molecular mechanisms underlying its occurrence and development remain unclear, necessitating the identification of new therapeutic targets. Through bioinformatics analysis, we discovered a previously unstudied differential gene called HIST3H2A in prostate cancer. Our study revealed that HIST3H2A is highly expressed in PCa tissues, as confirmed by analysis of both the GEO and UALCAN databases. Further analysis using the KEGG database demonstrated that HIST3H2A regulates the pathway of programmed necroptosis in cells. Additionally, we observed significant up-regulation of HIST3H2A in PCa tissues and cell lines. HIST3H2A was found to regulate cell proliferation, migration, invasion, and the epithelial-mesenchymal transition (EMT) process in tumors. Notably, HIST3H2A’s role in regulating programmed necroptosis in prostate cancer cells differs from its role in apoptosis. In vitro and in vivo experiments collectively support the key role of HIST3H2A in promoting the development of prostate cancer, highlighting its potential as a therapeutic target for patients with PCa.
... These findings were promising, but due to the complexity of SWI/SNF function and unpredictable consequences of loss of this important chromatin regulator, the effect of SWI/SNF loss may have differing effects depending on cell type. Table depicting three SWI/SNF complexes using information from [15], [39] and [40]. ...
Article
Full-text available
The close relationship between chromatin and metabolism has been well-studied in recent years. Many metabolites have been found to be cofactors used to modify chromatin, and these modifications can in turn affect gene transcription. One chromatin-associated factor responsible for regulating transcription is the SWI/SNF complex, an ATP-dependent chromatin remodeler conserved throughout eukaryotes. SWI/SNF was originally described in yeast as regulating genes involved in carbon source metabolism and mating type switching, and its mammalian counterpart has been extensively studied for its role in diseases such as cancer. The yeast SWI/SNF complex is closely associated with activation of stress response genes, many of which have metabolic functions. It is now recognized that this is a conserved function of the complex, and recent work has shown that mammalian SWI/SNF is also a key regulator of metabolic transcription. Emerging evidence suggests that loss of SWI/SNF introduces vulnerabilities to cells due to this metabolic influence, and that this may present opportunities for treatment of SWI/SNF-deficient cancers.
Article
Full-text available
ARID1B haploinsufficiency in humans causes Coffin-Siris syndrome, associated with developmental delay, facial dysmorphism, and intellectual disability. The role of ARID1B has been widely studied in neuronal development, but whether it also regulates stem cells remains unknown. Here, we employ scRNA-seq and scATAC-seq to dissect the regulatory functions and mechanisms of ARID1B within mesenchymal stem cells (MSCs) using the mouse incisor model. We reveal that loss of Arid1b in the GLI1+ MSC lineage disturbs MSCs’ quiescence and leads to their proliferation due to the ectopic activation of non-canonical Activin signaling via p-ERK. Furthermore, loss of Arid1b upregulates Bcl11b, which encodes a BAF complex subunit that modulates non-canonical Activin signaling by directly regulating the expression of activin A subunit, Inhba. Reduction of Bcl11b or non-canonical Activin signaling restores the MSC population in Arid1b mutant mice. Notably, we have identified that ARID1B suppresses Bcl11b expression via specific binding to its third intron, unveiling the direct inter-regulatory interactions among BAF subunits in MSCs. Our results demonstrate the vital role of ARID1B as an epigenetic modifier in maintaining MSC homeostasis and reveal its intricate mechanistic regulatory network in vivo, providing novel insights into the linkage between chromatin remodeling and stem cell fate determination.
Article
Full-text available
Chromatin remodellers include diverse enzymes with distinct biological functions, but nucleosome-sliding activity appears to be a common theme1,2. Among the remodelling enzymes, Snf2 serves as the prototype to study the action of this protein family. Snf2 and related enzymes share two conserved RecA-like lobes³, which by themselves are able to couple ATP hydrolysis to chromatin remodelling. The mechanism by which these enzymes couple ATP hydrolysis to translocate the nucleosome along the DNA remains unclear2,4–8. Here we report the structures of Saccharomyces cerevisiae Snf2 bound to the nucleosome in the presence of ADP and ADP-BeFx. Snf2 in the ADP-bound state adopts an open conformation similar to that in the apo state, and induces a one-base-pair DNA bulge at superhelix location 2 (SHL2), with the tracking strand showing greater distortion than the guide strand. The DNA distortion propagates to the proximal end, leading to staggered translocation of the two strands. The binding of ADP-BeFx triggers a closed conformation of the enzyme, resetting the nucleosome to a relaxed state. Snf2 shows altered interactions with the DNA in different nucleotide states, providing the structural basis for DNA translocation. Together, our findings suggest a fundamental mechanism for the DNA translocation that underlies chromatin remodelling.
Article
Full-text available
COSMIC, the Catalogue Of Somatic Mutations In Cancer (https://cancer.sanger.ac.uk) is the most detailed and comprehensive resource for exploring the effect of somatic mutations in human cancer. The latest release, COSMIC v86 (August 2018), includes almost 6 million coding mutations across 1.4 million tumour samples, curated from over 26 000 publications. In addition to coding mutations, COSMIC covers all the genetic mechanisms by which somatic mutations promote cancer, including non-coding mutations, gene fusions, copy-number variants and drug-resistance mutations. COSMIC is primarily hand-curated, ensuring quality, accuracy and descriptive data capture. Building on our manual curation processes, we are introducing new initiatives that allow us to prioritize key genes and diseases, and to react more quickly and comprehensively to new findings in the literature. Alongside improvements to the public website and data-download systems, new functionality in COSMIC-3D allows exploration of mutations within three-dimensional protein structures, their protein structural and functional impacts, and implications for druggability. In parallel with COSMIC's deep and broad variant coverage, the Cancer Gene Census (CGC) describes a curated catalogue of genes driving every form of human cancer. Currently describing 719 genes, the CGC has recently introduced functional descriptions of how each gene drives disease, summarized into the 10 cancer Hallmarks.
Article
Full-text available
Synovial sarcoma (SS) is defined by the hallmark SS18-SSX fusion oncoprotein, which renders BAF complexes aberrant in two manners: gain of SSX to the SS18 subunit and concomitant loss of BAF47 subunit assembly. Here we demonstrate that SS18-SSX globally hijacks BAF complexes on chromatin to activate an SS transcriptional signature that we define using primary tumors and cell lines. Specifically, SS18-SSX retargets BAF complexes from enhancers to broad polycomb domains to oppose PRC2-mediated repression and activate bivalent genes. Upon suppression of SS18-SSX, reassembly of BAF47 restores enhancer activation, but is not required for proliferative arrest. These results establish a global hijacking mechanism for SS18-SSX on chromatin, and define the distinct contributions of two concurrent BAF complex perturbations. Incorporation of the synovial sarcoma SS18-SSX fusion into BAF complexes results in concomitant eviction of BAF47. McBride et al. show that SS18-SSX retargets BAF complexes from enhancers to polycomb domains to oppose PRC2-mediated repression. Reincorporation of BAF47 upon suppression of SS18-SSX restores enhancer activation but is not required for proliferative arrest.
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
Mechanistic target of rapamycin (mTOR) complex 2 (mTORC2) plays an essential role in regulating cell proliferation through phosphorylating AGC protein kinase family members, including AKT, PKC and SGK1. The functional core complex consists of mTOR, mLST8, and two mTORC2-specific components, Rictor and mSin1. Here we investigated the intermolecular interactions within mTORC2 complex and determined its cryo-electron microscopy structure at 4.9 Å resolution. The structure reveals a hollow rhombohedral fold with a 2-fold symmetry. The dimerized mTOR serves as a scaffold for the complex assembly. The N-terminal half of Rictor is composed of helical repeat clusters and binds to mTOR through multiple contacts. mSin1 is located close to the FRB domain and catalytic cavity of mTOR. Rictor and mSin1 together generate steric hindrance to inhibit binding of FKBP12-rapamycin to mTOR, revealing the mechanism for rapamycin insensitivity of mTORC2. The mTOR dimer in mTORC2 shows more compact conformation than that of mTORC1 (rapamycin sensitive), which might result from the interaction between mTOR and Rictor-mSin1. Structural comparison shows that binding of Rictor and Raptor (mTORC1-specific component) to mTOR is mutually exclusive. Our study provides a basis for understanding the assembly of mTORC2 and a framework to further characterize the regulatory mechanism of mTORC2 pathway.
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
Mammalian switch/sucrose non-fermentable (mSWI/SNF) complexes are multi-component machines that remodel chromatin architecture. Dissection of the subunit- and domain-specific contributions to complex activities is needed to advance mechanistic understanding. Here, we examine the molecular, structural, and genome-wide regulatory consequences of recurrent, single-residue mutations in the putative coiled-coil C-terminal domain (CTD) of the SMARCB1 (BAF47) subunit, which cause the intellectual disability disorder Coffin-Siris syndrome (CSS), and are recurrently found in cancers. We find that the SMARCB1 CTD contains a basic α helix that binds directly to the nucleosome acidic patch and that all CSS-associated mutations disrupt this binding. Furthermore, these mutations abrogate mSWI/SNF-mediated nucleosome remodeling activity and enhancer DNA accessibility without changes in genome-wide complex localization. Finally, heterozygous CSS-associated SMARCB1 mutations result in dominant gene regulatory and morphologic changes during iPSC-neuronal differentiation. These studies unmask an evolutionarily conserved structural role for the SMARCB1 CTD that is perturbed in human disease.
Article
The architecture of the RSC complex RSC is a Snf2-family chromatin remodeler complex that controls the promoter architecture of most of the genes in yeast. Using single-particle cryo–electron microscopy, Ye et al. determined the structure of RSC bound to the nucleosome. The structure reveals the modular architecture of RSC, shows how RSC engages the nucleosome, and explains the remodeling directionality. RSC shows strong similarities to homologous human complexes that are frequently mutated in cancers, and this structure provides valuable information for understanding these systems. Science , this issue p. 838
Article
Mammalian SWI/SNF (mSWI/SNF) ATP-dependent chromatin remodeling complexes are multi-subunit molecular machines that play vital roles in regulating genomic architecture and are frequently disrupted in human cancer and developmental disorders. To date, the modular organization and pathways of assembly of these chromatin regulators remain unknown, presenting a major barrier to structural and functional determination. Here, we elucidate the architecture and assembly pathway across three classes of mSWI/SNF complexes-canonical BRG1/BRM-associated factor (BAF), polybromo-associated BAF (PBAF), and newly defined ncBAF complexes-and define the requirement of each subunit for complex formation and stability. Using affinity purification of endogenous complexes from mammalian and Drosophila cells coupled with cross-linking mass spectrometry (CX-MS) and mutagenesis, we uncover three distinct and evolutionarily conserved modules, their organization, and the temporal incorporation of these modules into each complete mSWI/SNF complex class. Finally, we map human disease-associated mutations within subunits and modules, defining specific topological regions that are affected upon subunit perturbation.
Article
From DNA unwrapping to histone exchange The yeast SWR1 complex, a member of the INO80 family of nucleosome remodelers, exchanges the H2A-H2B histone dimer for the Htz1 variant–containing dimer. Unlike all other remodelers, SWR1 does not translocate the nucleosome. Willhoft et al. applied structural and single-molecule analyses to show that the interaction between SWR1 and the nucleosome destabilizes the DNA wrapped around the histone core. This SWR1-catalyzed partial unwrapping of the DNA was regulated by adenosine triphosphate (ATP) binding but did not require ATP hydrolysis. Science , this issue p. eaat7716