Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

RES EARCH

SPLICEOSOME previously unknown assembly intermediates.

Structural basis of branch site recognition


Our high-resolution reconstructions provide
insight into the architecture and dynamics

by the human spliceosome


of the human U2 snRNP and pre-spliceosome
formation. These new data point at the criti-
cal roles of HTATSF1 and SF3B6 in facilitating
Jonas Tholen1,2†, Michal Razew1, Felix Weis3, Wojciech P. Galej1* pre-mRNA recognition.

Recognition of the intron branch site (BS) by the U2 small nuclear ribonucleoprotein (snRNP) is a critical Results
event during spliceosome assembly. In mammals, BS sequences are poorly conserved, and unambiguous Purification of the 17S U2 snRNP complex
intron recognition cannot be achieved solely through a base-pairing mechanism. We isolated human Existing methods for purification of the U2
17S U2 snRNP and reconstituted in vitro its adenosine 5´-triphosphate (ATP)–dependent remodeling and snRNP use antibodies against SF3a or SF3b
binding to the pre–messenger RNA substrate. We determined a series of high-resolution (2.0 to components (7, 12), which can in principle cap-
2.2 angstrom) structures providing snapshots of the BS selection process. The substrate-bound U2 ture particles in multiple states. To specifically
snRNP shows that SF3B6 stabilizes the BS:U2 snRNA duplex, which could aid binding of introns with select a subset of U2 snRNPs representing a
poor sequence complementarity. ATP-dependent remodeling uncoupled from substrate binding captures single functional state, we used CRISPR-Cas9–

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
U2 snRNA in a conformation that competes with BS recognition, providing a selection mechanism mediated genome editing to introduce a green
based on branch helix stability. fluorescent protein (GFP) tag into the HTATSF1
genomic locus of human embryonic kidney

R
(HEK) 293F cells (fig. S1). Affinity chroma-
emoval of introns from pre-mRNA is point, it is unclear how branch helix stability is tography with anti-GFP nanobodies allowed
catalyzed by a large and dynamic RNA- sensed by the splicing machinery (9, 10). isolation of an intact 17S U2 snRNP, contain-
protein complex known as the spliceo- Upon engagement of U2 snRNA with the ing U2 snRNA and 22 proteins that accounted
some. The spliceosome is assembled de pre-mRNA substrate, a 15–nucleotide (nt)– for a total estimated molecular weight of
novo on each pre-mRNA substrate from long branch helix is formed, adopting helical 1.08 MDa (fig. S1).
five small nuclear ribonucleoprotein (snRNP) geometry even in the absence of full comple-
particles and several dozen protein factors. mentarity. The length of the branch helix is con- High-resolution structure of the 17S U2 snRNP
During spliceosome assembly, three conserved served between yeast and human spliceosomes We determined the cryo-EM structure of the
positions in the pre-mRNA—the 5′ splice site and maintained throughout different stages of 5′ domain of the human 17S U2 snRNP at 2.2-Å
(5′-SS), branch site (BS), and 3′ splice site (3′-SS)— splicing (13–15). In early splicing complexes, resolution, which allowed accurate atomic mod-
are specifically recognized by the components the branch helix is accommodated within a cav- eling of the SF3b complex, SF3A3, HTATSF1RRM,
of the spliceosome, allowing a two-step trans- ity formed by the heteroheptameric SF3b com- and the 5′ end of the U2 snRNA (Fig. 1 and figs.
esterification reaction to occur. plex (7), which contacts the pre-mRNA around S2 to S4). The overall architecture of the com-
In mammalian cells, the BS is initially rec- the BS and stabilizes the U2 snRNA:BS base- plex agrees well with that of previous studies
ognized by SF1 (mBBP) in cooperation with pairing interaction (16, 17). The branchpoint (23, 24), including a recent low-resolution cryo-
U2AF2 (U2AF65), which binds the polypyrim- adenosine (BP-A) is bulged out of the branch EM reconstruction (12). The low-pass–filtered
idine tract (PPT) sequence (1). Concomitantly, helix and binds into a pocket formed by SF3B1 map reveals an unresolved density at the pe-
U1 snRNP binds to the 5′-SS and together they and PHF5A (13, 14). Mutations in SF3B1 asso- riphery, which likely corresponds to the U2
form the first, adenosine 5´-triphosphate (ATP)– ciated with myelodysplastic syndromes have snRNP core (3′ domain) (Fig. 1, B and C). This
independent spliceosome assembly interme- been shown to modulate BS selection (18, 19). domain appears flexible relative to the re-
diate known as complex E (2). The U2 snRNP SF3B6 (p14), which has no homolog in Sac- solved 5′ domain and could not be improved
is loosely associated with complex E (3), and charomyces cerevisiae, was shown to cross-link by further data processing.
its stable incorporation into the prespliceosome to the BP-A in HeLa nuclear extract, indicating a Parts of SF3B2 and SF3A3 have been pre-
(complex A) requires ATP and formation of potential role in BS recognition (20). However, viously observed in cryo-EM maps of mam-
base-pairing interactions between the BS and the position of SF3B6 in human Bact spliceo- malian snRNPs and spliceosomes (21, 25), but
the U2 snRNA (4). somes (21) does not explain the cross-linking owing to limited resolution, they were not in-
In yeast, several factors have been shown to data or its role in splicing. terpreted with atomic coordinates. The high-
facilitate complex A formation. Among them Although parts of the U2 snRNP structure resolution reconstruction provides atomic
are Cus2 (human HTATSF1) (5) and the RNA- have been determined as a component of yeast insights into several interfaces, including
dependent DEAD-box ATPase, Prp5 (human and mammalian spliceosomes, there is no high- HTATSF1RRM:SF3B1 (Fig. 1, A and C) and SF3B2:
DDX46), whose activity is required for Cus2 resolution structural information for the 17S SF3A3 (Fig. 1 and fig. S4), consistent with pre-
displacement (6–8) and fidelity control of BS U2 snRNP and early splicing complexes in vious lower-resolution structures (12, 26).
recognition (9, 10). humans (i.e., E and A). This is of particular
During BS recognition, an evolutionarily interest as sequence conservation and base- In vitro reconstitution of branch site recognition
conserved branchpoint-interacting stem loop pairing potential of human branch sites are by the U2 snRNP
(BSL) presents U2 nucleotides to the intron weak compared with those of yeast (22), and To obtain mechanistic insights into BS recog-
BS for base pairing (11, 12). Although branch the mechanism of BS selection remains elusive. nition by cryo-EM analysis, we reconstituted
helix formation is subject to a fidelity check- Here, we isolated human 17S U2 snRNP and BS recognition in vitro with purified 17S U2
1 reconstituted in vitro its binding to a model snRNP and a model BS oligonucleotide (BPS
European Molecular Biology Laboratory, 71 Avenue des
Martyrs, 38042 Grenoble, France. 2Heidelberg University, BS and the remodeling leading to the dis- oligo). The BPS oligo is complementary to the
Faculty of Biosciences, Heidelberg, Germany. 3European sociation of HTATSF1 from the complex. We positions 27 to 42 of the U2 snRNA and in-
Molecular Biology Laboratory, Structural and Computational determined a series of cryo–electron micros- cludes a bulged-out adenosine, which mimics
Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany.
*Corresponding author. Email: wgalej@embl.fr copy (EM) structures of the U2 snRNP in dif- the BP-A. Similar minimal substrates have been
†Candidate for Joint PhD degree from EMBL and Heidelberg University. ferent conformational states, including two used previously (27, 28).

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 1 of 7


RES EARCH | R E S E A R C H A R T I C L E

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
Fig. 1. High-resolution structure of the human 17S U2 snRNP. (A) Surface (PDB: 6Y5Q). (D) Cryo-EM map of the 17S U2 snRNP filtered and colored by local
representation of the 5′ domain of the 17S U2 snRNP model. (B) Experimental cryo- resolution. (E) The cryo-EM map obtained by merging several U2 snRNP datasets
EM map for the 17S U2 snRNP showing the high-resolution 5′ domain (colored overlaid with the map of the 17S U2 snRNP. (F) Atomic modeling into the highest-
by chain identity) embedded in a low-pass–filtered map showing the position of the resolution region at an interface of SF3B1 and SF3B3. The map was colored by
3′ domain. (C) Pseudo-atomic model for the fully assembled 17S U2 snRNP. The chain identity; water molecules are colored red. Abbreviations for the amino acid
3′ domain was modeled by rigid-body docking of the previously reported coordinates residues are as follows: A, Ala; C, Cys; F, Phe; H, His; P, Pro; and Y, Tyr.

We immobilized 17S U2 snRNP on anti-GFP Next, we investigated the requirements for A-like U2 snRNPs, except for the major dif-
nanobody resin by using a GFP tag on HTATSF1 U2 snRNP engagement with a model substrate. ferences in the 5′ end of the U2 snRNA and
or DDX46 and incubated it under various con- 17S U2 snRNP engages efficiently with the BPS missing SF3B6 (Fig. 2, F and H). Similar to
ditions. In the presence of ATP and the BPS oligo (Fig. 2D), and the binding occurs in a the 17S U2 snRNP, both complexes can be
oligo, U2 snRNP is released from the resin wide range of conditions without requiring divided into a well-resolved 5′ domain and
(Fig. 2A, lane 4) and remains bound to the BPS ATP (fig. S5). No binding was observed when a 3′ domain (Fig. 2, G and I). The 3′ domains
oligo when analyzed by glycerol gradient cen- the remodeled (ATP-treated) U2 snRNP var- remain flexible but occupy different positions
trifugation. These BPS oligonucleotide–bound iant was used in this assay (Fig. 2D). This in- compared to the 17S complex. HTATSF1 and
complexes likely resemble the substrate-bound dicates that displacement of HTATSF1 and the DDX46 helix are missing from these two
U2 snRNP within complex A. Thus, we refer to DDX46 uncoupled from substrate binding reconstructions, consistent with the biochem-
these complexes as “A-like U2 snRNPs” here- leads to the formation of an inhibited confor- ical data and the sample preparation protocol.
after. Addition of ATP alone, without the BPS mation of the U2 snRNP.
oligo, also induces HTATSF1 dissociation at We determined high-resolution cryo-EM Structure and dynamics of the U2 snRNA during
increased temperature (Fig. 2A, lane 3), sug- structures of these two newly identified U2 branch site recognition
gesting that this remodeling can be function- snRNP complexes (Fig. 2, E to I, and figs. S2 SLIIa of the U2 snRNA is resolved to nearly 2 Å
ally uncoupled from substrate binding, leading to S4). in all our reconstructions, which allowed mod-
to the formation of a “remodeled U2 snRNP.” eling of three additional, noncanonical base
We further investigated both reactions bio- Structures of the minimal A-like and remodeled pairs within this stem-loop and its interactions
chemically by probing eluates with antibodies U2 snRNP complexes with the components of the SF3a and SF3b
specific to the core U2 snRNP component, The overall architecture of the A-like U2 snRNP complexes (figs. S4 and S6). These interactions
SNRPB2 (Fig. 2, B and C). Western blotting is in good agreement with the lower-resolution remain unchanged during the transition from
revealed that some HTATSF1 dissociation can descriptions of the U2 snRNP embedded with- the 17S U2 snRNP to the A-like and remodeled
occur spontaneously at increased temperature in the fully assembled human B and Bact U2 snRNP complexes.
(Fig. 2B, lane 2), but it is greatly stimulated spliceosomes (21, 29) (Fig. 2, F and G). How- In the 17S U2 snRNP reconstruction, a helical
by the presence of ATP (Fig. 2B, lanes 3 and 5), ever, the structure exhibits features that are density emerges from the 5′ end of SLIIa and
consistent with previous experiments (8). incompatible with those later splicing com- points toward SF3B1 and PHF5A along the side
The same results were obtained regardless of plexes, indicating that the A-like U2 snRNP of HTATSF1RRM. This density was interpreted
whether the sample was tethered to the resin represents a distinct splicing intermediate. as the BSL and modeled by rigid-body docking
via HTATSF1 (Fig. 2B) or DDX46 (Fig. 2C). The remodeled U2 snRNP closely resembles of an idealized RNA helix with a base-pairing

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 2 of 7


RES EARCH | R E S E A R C H A R T I C L E

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
Fig. 2. Sample preparation and in vitro reconstitution of the branch site Cy5-labeled BPS oligonucleotide binding to the 17S U2 snRNP or remodeled U2
recognition by the U2 snRNP. (A) SDS–polyacrylamide gel electrophoresis snRNP by glycerol gradients. RFU, relative fluorescence units. (E) Schematic
analysis of the eluates from the GFP-HTATSF1–tagged 17S U2 snRNP summarizing the outcome of the in vitro remodeling and substrate binding
immobilized on the GFP nanobody resin and incubated under various conditions. experiments. (F and H) Surface representation of the 5′ domains of the A-like
(B) Western blot analysis of the reconstitution reaction performed as in (A). and Remodeled U2 snRNPs models. (G and I) Experimental cryo-EM maps
Elution and resin fractions were probed with antibodies against SNRPB2, a core of A-like and remodeled U2 snRNPs showing the high-resolution 5′ domain
U2 snRNP component. (C) The same as in (B), but the 17S U2 snRNP sample (colored by chain identity) embedded in a low-pass–filtered (5 Å) map, showing
was immobilized using GFP tag attached to DDX46. (D) Analysis of the positioning of their 3′ domains.

pattern based on previous predictions and the likely stabilized by SF3A2 and its interaction veals that this newly formed stem-loop closely
low-resolution reconstruction of the 17S U2 with the branch helix (Fig. 4A). Most prominent mimics the branch duplex and its interactions
snRNP (12) (Fig. 3A). In contrast to the neigh- of the contacts formed during this transition with the U2 snRNP proteins (Fig. 3, F and G);
boring U2 SLIIa, the BSL density is not well are the interactions of charged amino acids of therefore, we refer to this stem-loop as the
resolved in our map, pointing at the intrinsi- SF3B1 (K1071, R1106, N1107, R1109, K1149) with branch helix–mimicking stem-loop (BMSL).
cally dynamic nature of this structure. Indeed, the BPS RNA phosphate backbone, consistent Formation of the BMSL is mutually exclusive
a three-dimensional (3D) classification focused with previous studies (14, 21). The SF3a com- with the pre-mRNA binding by the U2 snRNP,
on this region allows the separation of the plex is less well resolved in the A-like complex suggesting that the two structures could com-
ensemble structure into at least three distinct assembled in the presence of AMP-PCP, suggest- pete with one another during BS recognition.
conformational states of the BSL (Fig. 3, D and ing that the ATP-dependent remodeling may This finding provides a potential mechanism
E), suggesting a dynamic probing mechanism play a role in facilitating SF3a docking to the for the selection of branch helix stability by the
for BS recognition. complex A (fig. S7). spliceosome and could represent a previously
During transition from 17S to A-like U2 unknown BS fidelity checkpoint.
snRNP, the BSL sequence engages with the 5′ end of the U2 snRNA mimics pre-mRNA
BPS oligo, forming a 12-nt U2 snRNA:BS du- substrate in the absence of HTATSF1 SF3B6 stabilizes the branch helix in the A-like
plex (Fig. 3B). This duplex forms interactions Upon ATP-dependent remodeling and HTATSF1 U2 snRNP
with SF3B1, PHF5A, SF3A2, and SF3A3. The dissociation, U2 snRNA nucleotides 11 to 44 Upon BPS oligo binding to the 17S U2 snRNP,
SF3A2 zinc finger domain binds the branch form a distinct bulged stem-loop structure in an additional density appears near H14 and
helix as previously described (14, 21, 26). SF3A3 the remodeled U2 snRNP complex (Fig. 3C). H15 of SF3B1. This density could be unambig-
becomes more ordered in the A-like complex, Comparison with the A-like U2 snRNP re- uously interpreted by rigid-body docking of

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 3 of 7


RES EARCH | R E S E A R C H A R T I C L E

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022

Fig. 3. Structure and dynamics of the U2 snRNA during branch site dynamics of the BSL visualized directly in the cryo-EM map (see also
recognition. Secondary and tertiary structure of the U2 snRNA in the (A) 17S, movie S2). (F) Adenosine 24 of the U2 snRNA mimics BP-A in the remodeled
(B) A-like, and (C) remodeled U2 snRNPs. (D) BSL is stabilized directly by the U2 snRNP complex. (G) Environment of the BP-A in the A-like U2 snRNP
two domains of HTATSF1 in the 17S U2 snRNP complex. (E) Structural in the same orientation as in (F).

the SF3B6:SF3B1 crystal structure [Protein Data ported structure of the SF3b complex or U2 the 5′ end of the branch helix (Fig. 4A); there-
Bank (PDB): 3LQV (30), fig. S4]. Although it is a snRNP (12, 23) and it differs substantially from fore, it defines the exact position of the bulged
stable component of the SF3b complex, SF3B6 the SF3B6:SF3B1 interface in the Bact spliceo- BP-A relative to the end of the branch helix.
has not been observed in any previously re- some (21). SF3B6 binds to the U2 snRNA at Such an interaction is supported by previous

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 4 of 7


RES EARCH | R E S E A R C H A R T I C L E

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
Fig. 4. SF3B6 stabilizes branch helix in the A-like U2 snRNP while SF3B1 complex (PDB: 6FF7), showing two-step transition from open to closed
HEAT repeats adopt a half-closed conformation. (A) Side view of the SF3B1 conformation. (C and D) Atomic model of the interfaces between
A-like U2 snRNP showing positions of the branch helix and its stabilization by SF3B1HEAT and HTATSF1RRM/LH. (E) Atomic model of the interface of SF3B6
the SF3A2 and SF3B6; yellow arrows indicate U2 snRNA contact points with SF3B1HEAT and U2 snRNA. Abbreviations for the amino acid residues
enforcing helical geometry of the branch helix. (B) Structure of the RNA and are as follows: A, Ala; D, Asp; F, Phe; G, Gly; H, His; Lys; L, Leu; Q, Gln;
HEAT repeats in the 17S U2 snRNP, the A-like U2 snRNP, and the Bact R, Arg; T, Thr; W, Trp; and Y, Tyr.

RNA-protein cross-linking (31, 32) and cross- to the HTATSF1UHM domain that is known to a hinge between HEAT repeats H15 and H16
linking mass spectrometry data (12, 23). A29 bind the SF3B1ULM motif (Fig. 3D) (12, 34, 35). (36). Similar remodeling occurs in our in vitro
of the U2 snRNA inserts into the same pocket Therefore, the two domains of HTATSF1 form system, with no extra factors needed, even
where adenine was placed in the co-crystal stable interfaces with SF3B1 and flank the U2 when the BPS oligo is incubated on ice with
structure (30) and stacks against residue Y22 snRNA BSL from both sides, suggesting a the 17S U2 snRNP in the presence of AMP-PCP,
of SF3B6 (Fig. 4E). Moreover, SF3B6 is oriented direct stabilization mechanism for this tran- a nonhydrolysable ATP analog (table S1 and fig.
in such a way that its disordered N terminus sient RNA secondary structure. S7). This indicates that branch helix formation
points toward BP-A and is close enough to Movement of the BSL correlates with the is the only driving force for the rearrangement
explain previous cross-linking data (20, 33). disappearance of the extra density on top of around the first hinge region and that it does
Our data show that SF3B6 plays a previously HTATSF1RRM and presumably the short vari- not depend on ATP hydrolysis.
unknown role in stabilizing branch helix, ant of the U2 stem-loop I structure (Fig. 3C Although the hinge-like movement of SF3B1
which could be particularly relevant for branch and movie S1). Given the concerted movement is reconstituted in our system, the conformation
sequences with poor complementarity to the with other U2 snRNA elements, we speculate of the N-terminal part of the HEAT repeat
U2 snRNA. that at least part of this density could belong differs appreciably from any of the previously
to the 5′ end of the U2 snRNA, especially that reported states. In the closed conformation,
HTATSF1 stabilizes the BSL in 17S U2 snRNP it occupies the surface that is typically involved SF3B1 helix H1 (residues 509 to 523) inserts
Comparison of the 17S and A-like U2 snRNP in RRM-RNA binding. Indeed, the Y48D mu- into the major groove of the branch helix,
shows that binding of SF3B6 and HTATSF1RRM tation (Y136 in HTATSF1) in the yeast homolog providing additional stabilization for the branch
to SF3B1HEAT are mutually exclusive and Cus2 abolishes U2 snRNA binding (5). Recom- helix, whereas in the A-like complex it remains
HTATSF1RRM needs to be displaced before binant HTATSF1RRM exhibits some nonspecific ~20 Å away from this binding site (Fig. 4B).
stable docking of SF3B6 (Fig. 4B). Our re- affinity for RNA (fig. S8). This supports the hy- We refer to this new SF3B1 conformation as
construction of the 17S U2 snRNP shows that pothesis that interaction between HTATSF1RRM half-closed, in accordance with the previous
HTATSF1RRM binds in a hydrophobic groove and the 5′ end of the U2 snRNA could addi- convention. The movement from half-closed
formed by HEAT repeats H15 and H16 of SF3B1 tionally stabilize the BSL in an indirect manner to closed is different from the hinge-like clo-
(Fig. 4C). The neighboring H16 and H17 repeats by preventing BMSL formation. sure and involves multiple small changes in the
form the interface for the C terminus of the curvature of the HEAT repeats in its N-terminal
HTATSF1 linker helix (HTATSF1LH), comprising Two-step conformational change in SF3B1 upon part (H1-H12) (Fig. 4B). It is possible that bind-
residues 239 to 251 (Fig. 4D). The C terminus of pre-mRNA binding ing of the intron sequence downstream from
HTATSF1LH points toward the BSL and a SF3B1 was previously reported to transition the BS could facilitate complete closure of
globular density nearby, which likely belongs from an open to a closed conformation around the SF3B1.

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 5 of 7


RES EARCH | R E S E A R C H A R T I C L E

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
Fig. 5. Schematic model of branch site recognition by the U2 snRNP pathway (this work). Stable substrates gradually form the branch helix as
based on recent structural data. U2 snRNP associated with spliceosomal shown in the E-to-A (41) and pre-A (42) intermediates. In the absence of
complex E is likely structurally similar to the 17S U2 snRNP described by (12) properly positioned, bulged out BP-A, the pre-A complex is targeted to a
and in this work. Dissociation of HTATSF1 creates competition between the discard pathway. Productive engagement of the branch helix leads to the
formation of a branch helix and the BMSL. Rejection of weak, suboptimal formation of complex A, wherein U2 snRNP is structurally similar to the A-like
substrates results in the remodeled U2 snRNP, which is targeted to a discard U2 snRNP (this work).

Discussion Indeed, in yeast, the ATPase activity of the fission yeast (22), require an additional BP-A
Recognition of the branch point sequences by DDX46 homolog Prp5 is required for complex A positioning mechanism, which is fulfilled by
the U2 snRNP is a critical step of spliceosome formation, but deletion of the HTATSF1 homo- SF3B6. Consequently, SF3B6 is conserved in
assembly. In this work, we used a minimal log Cus2 removes this dependence (8, 34). many species with low BS conservation, but
in vitro system to analyze the structure of the However, the ATP-dependent BS fidelity con- not in S. cerevisiae (fig. S9).
human U2 snRNP and its conformational trol by Prp5 remains unchanged in the absence The emerging data suggest that the tran-
changes upon ATP-dependent remodeling and of Cus2, suggesting a more complex function sition from E to A complex requires ATP-
engagement with the pre-mRNA substrate. of this protein. dependent displacement of HTATSF1, which
The 17S U2 snRNP structure shows that The structure of the A-like U2 snRNP cap- destabilizes the BSL and allows it to probe BS
HTATSF1 and BSL stabilize each other in two tured SF3B6 interacting with the branch helix, sequences (40) (Fig. 5). The absence of HTATSF1
distinct ways: directly, through interactions which has two major implications. First, it pro- creates competition between the branch helix
between BSL and HTATSF1LH/UHM, and in- vides a specific binding site for the U2 snRNA and the BMSL structure within U2 snRNA,
directly, via a possible association of the 5′ in addition to SLIIa and SF3A2ZnF, which im- providing a mechanism for the selection of
end of the U2 snRNA with the HTATSF1RRM, poses helical geometry on the U2 snRNA with- the branch helix stability. Formation of the
which would prevent formation of RNA struc- in the branch helix binding pocket. This provides BMSL would mean rejection of the potential
tures that compete with the BSL (i.e., the long a mechanism for the stabilization of weak branch BS sequences. Therefore, the structure of the
variant of the SLI or the BMSL). point sequences, such as those found in mam- remodeled U2 snRNP likely represents an in-
Our data show that a model BPS oligo- mals, even in the absence of extensive comple- termediate on the discard pathway after sub-
nucleotide can engage in base-pairing interac- mentarity. Second, SF3B6 binds at the junction of optimal substrate rejection. Such a state was
tion with the U2 snRNP without a requirement the branch helix duplex and the single-stranded predicted to exist in the framework of the
for prior remodeling. Although formation of region of the U2 snRNA; therefore, it defines the kinetic proofreading model (9).
complex A has been shown to be ATP depen- length of the branch helix and the exact position BS sequences that withstand competition
dent in HeLa cell nuclear extract, Amin com- of the bulged BP-A relative to its end. It has been with the BMSL would continue to progressively
plex can form without ATP when the sequence previously shown in an orthogonal yeast system form the branch helix through a recently pro-
upstream of the BS is missing (27, 37). This that the position of the BP-A within the branch posed toehold strand invasion mechanism
could be due to the absence of certain BS bind- helix is critical for productive splicing (38). In (41). An intermediate state in this process
ing proteins (e.g., SF1) or lack of topological budding yeast, the BS has evolved to be highly (A3′-SSA complex) was captured by blocking
restraints for branch helix formation. To form conserved, and sequence complementarity be- spliceosome assembly with spliceostatin A
the branch helix, U2 snRNA has to wind around tween BS and U2 snRNA ensures proper po- (SSA) (41), which trapped U2 snRNP with a
the long pre-mRNA substrate, and it is possi- sitioning of the BP-A (39). Weak BS sequence partially formed branch helix, missing bulged
ble that ATP is required to liberate the 5′ end conservation and base-pairing potential in out BP-A. Consequently, the branch helix was
of the U2 snRNA from HTATSF1 to allow that. other organisms, including mammals and not accommodated in its pocket and SF3B1

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 6 of 7


RES EARCH | R E S E A R C H A R T I C L E

remains in the open conformation, resembling served in the Bact complex (21), as its position 35. S. Loerch et al., J. Biol. Chem. 294, 2892–2902 (2019).
that found in the 17S U2 snRNP (Fig. 5). in the A-like U2 snRNP would clash with Prp8 36. C. Cretu et al., Mol. Cell 70, 265–273.e8 (2018).
37. C. M. Newnham, C. C. Query, RNA 7, 1298–1309 (2001).
Without inhibition by SSA, BS sequences and prevent early Bact formation.
38. D. J. Smith, M. M. Konarska, C. C. Query, Mol. Cell 34, 333–343
would continue to fully form the branch helix. Our data provide several high-resolution (2009).
At this point another checkpoint would be snapshots of the complex process of BS rec- 39. J. Sales-Lee et al., Curr. Biol. 31, 1–13 (2021).
reached. If a bulged-out BP-A is present, it will ognition by the U2 snRNP and contribute to 40. C.-Y. Kao, E.-C. Cao, H. L. Wai, S.-C. Cheng, Nucleic Acids Res.
49, 9965–9977 (2021).
bind the pocket in SF3B1, causing transition to a better understanding of the mechanism of
41. C. Cretu et al., Nat. Commun. 12, 4491–15 (2021).
the half-closed conformation and dissociation pre-mRNA splicing in humans. 42. Z. Zhang et al., Nature 596, 296–300 (2021).
of DDX46, as shown in the A-like U2 snRNP.
AC KNOWLED GME NTS
However, in the absence of a properly posi- RE FERENCES AND NOTES We thank M. Pfleiderer and D. Peter for experimental advice;
tioned BP-A, SF3B1HEAT remains in the open 1. J. Valcárcel, R. K. Gaur, R. Singh, M. R. Green, Science 273, A. Fraudeau and E. Marchal for technical assistance; S. Schneider,
conformation and the spliceosome is stalled, 1706–1709 (1996). E. Pellegrini, and W. Hagen for ensuring smooth running of the
as shown in the structure of the yeast pre-A 2. S. Michaud, R. Reed, Genes Dev. 5 (12b), 2534–2546 EMBL cryo-EM facilities; EMBL Proteomic Core Facility for
(1991). performing mass spectrometry experiments; M. Pelosse and
complex (42). In this complex, Prp5 provides 3. R. Das, Z. Zhou, R. Reed, Mol. Cell 5, 779–787 (2000). A. Aubert (EMBL EEF facility) for assistance with cell culture;
steric hindrance for the next step of spliceo- 4. J. Wu, J. L. Manley, Genes Dev. 3, 1553–1561 (1989). M. Pezet (IAB Grenoble Flow Cytometry Platform) for cell sorting;
5. D. Yan et al., Mol. Cell. Biol. 18, 5000–5009 (1998). A. Peuch and the EMBL Grenoble IT team for the support with
some assembly, recruitment of the tri-snRNP 6. C. L. O’Day, G. Dalbadie-McFarland, J. Abelson, J. Biol. Chem.
high-performance computing; C. Query for an insightful discussion;

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
(10). A prolonged block by Prp5 will likely ini- 271, 33261–33267 (1996).
and R. Pillai and S. Fica for critical comments on the manuscript.
tiate a discard pathway. 7. C. L. Will et al., EMBO J. 21, 4978–4988 (2002).
Funding: This project has received funding from the European
8. R. Perriman, I. Barta, G. K. Voeltz, J. Abelson, M. Ares Jr., Proc.
The remodeled U2 snRNP described in this Natl. Acad. Sci. U.S.A. 100, 13857–13862 (2003).
Research Council (ERC) under the European Union’s Horizon 2020
research and innovation programme (grant agreement no. 950278,
work and the pre-A complex (42) are two dis- 9. Y.-Z. Xu, C. C. Query, Mol. Cell 28, 838–849 (2007).
10. W.-W. Liang, S.-C. Cheng, Genes Dev. 29, 81–93 (2015). awarded to W.P.G.). M.R. was supported by a fellowship from the
tinct intermediates that direct suboptimal sub- 11. R. Perriman, M. Ares Jr., Mol. Cell 38, 416–427 (2010). EMBL Interdisciplinary Postdoc (EIPOD) programme under
strates to the discard pathway. They represent 12. Z. Zhang et al., Nature 583, 310–313 (2020). Marie Sklodowska-Curie Actions COFUND (grant agreement
13. R. Rauhut et al., Science 353, 1399–1405 (2016). no. 847543). Author contributions: Conceptualization: J.T.,
different checkpoints in BS fidelity control, 14. C. Yan, R. Wan, R. Bai, G. Huang, Y. Shi, Science 353, 904–911 W.P.G.; Methodology: J.T., M.R., F.W., W.P.G.; Investigation: J.T.,
ensuring both the formation of a stable branch (2016). M.R.; Visualization: J.T., W.P.G.; Funding acquisition: W.P.G.;
helix and the presence of properly positioned 15. W. P. Galej et al., Nature 537, 197–201 (2016). Project administration: J.T., W.P.G.; Supervision: W.P.G.; Writing –
16. O. Gozani, R. Feld, R. Reed, Genes Dev. 10, 233–243 original draft: J.T., W.P.G.; Writing – review & editing: J.T., M.R.,
BP-A (Fig. 5). (1996). F.W., W.P.G. Competing interests: The authors declare that they
Only properly positioned bulged out BP-A 17. C. L. Will et al., EMBO J. 20, 4536–4546 (2001). have no competing interests. Data and materials availability:
can bind the SF3B1-PHF5A pocket and cause 18. Q. Tang et al., Genes Dev. 30, 2710–2723 (2016). Cryo-EM maps were deposited in the EMDB with the following
19. T. J. Carrocci, D. M. Zoerner, J. C. Paulson, A. A. Hoskins, accession codes: EMD-13793 (17S U2 snRNP core); EMD-13810
the transition to the half-closed conformation
Nucleic Acids Res. 45, 4837–4852 (2017). (17S U2 snRNP HEAT repeats); EMD-13811 (A-like U2 snRNP);
of SF3B1, as observed in the A-like U2 snRNP. 20. A. M. MacMillan et al., Genes Dev. 8, 3008–3020 (1994). EMD-13813 (A-like U2 snRNP medium resolution/SF3B6 map);
During this transition, an extensive interac- 21. D. Haselbach et al., Cell 172, 454–464.e11 (2018). EMD-13812 (remodeled U2 snRNP); EMD-13815 (merged datasets –
tion surface forms between the branch helix, 22. A. J. Taggart et al., Genome Res. 27, 639–649 (2017). the highest resolution map); EMD-13814 (AMP-PCP A-like U2
23. C. Cretu et al., Mol. Cell 64, 307–319 (2016). snRNP). Atomic coordinates were deposited in the PDB database
including the bulged BP-A and the HEAT re- with the following accession codes: 7Q3L (17S U2 snRNP); 7Q4O
24. A. Krämer, P. Grüter, K. Gröning, B. Kastner, J. Cell Biol. 145,
peats H15 to H19. Our minimal system shows 1355–1368 (1999). (A-like U2 snRNP); 7Q4P (remodeled U2 snRNP). Materials
that this interaction is the sole driving force 25. X. Zhang et al., Cell Res. 28, 307–322 (2018). generated in this study are available on request from the lead
26. C. Plaschka, P.-C. Lin, K. Nagai, Nature 546, 617–621 contact (wgalej@embl.fr).
for the SF3B1 hinge-like movement. It is not
(2017).
clear which factors are needed for the second
27. C. C. Query, P. S. McCaw, P. A. Sharp, Mol. Cell. Biol. 17,
phase of the transition from half-closed to 2944–2953 (1997). SUPPLEMENTARY MATERIALS
closed SF3B1 conformation and when this 28. E. G. Folco, K. E. Coil, R. Reed, Genes Dev. 25, 440–444 science.org/doi/10.1126/science.abm4245
conformational change occurs. (2011). Materials and Methods
29. X. Zhan, C. Yan, X. Zhang, J. Lei, Y. Shi, Cell Res. 28, 1129–1140 Figs. S1 to S9
Upon A complex formation, poorly defined (2018). Tables S1 and S2
branch sequences would benefit from stabili- 30. M. J. Schellenberg, E. L. Dul, A. M. MacMillan, RNA 17, 155–165 References (43–69)
zation by SF3B6, which enforces helical geom- (2011). MDAR Reproducibility Checklist
31. O. Dybkov et al., Mol. Cell. Biol. 26, 2803–2816 (2006). Movies S1 and S2
etry of the U2 snRNA, even in the absence of
32. R. Spadaccini et al., RNA 12, 410–425 (2006).
extensive branch site complementarity. During 33. M. J. Schellenberg et al., Proc. Natl. Acad. Sci. U.S.A. 103, 17 September 2021; accepted 11 November 2021
subsequent steps of the spliceosome assembly, 1266–1271 (2006). Published online 25 November 2021
SF3B6 has to relocate to its binding site ob- 34. J. Talkish et al., RNA 25, 1020–1037 (2019). 10.1126/science.abm4245

Tholen et al., Science 375, 50–57 (2022) 7 January 2022 7 of 7


Structural basis of branch site recognition by the human spliceosome
Jonas TholenMichal RazewFelix WeisWojciech P. Galej

Science, 375 (6576), • DOI: 10.1126/science.abm4245

Downloaded from https://www.science.org at Indian Institute of Technology, Kharagpur on September 02, 2022
Intron recognition in the spotlight
Excision of noncoding introns from pre–messenger RNAs is catalyzed by the spliceosome, a large RNA-protein
complex that recognizes specific sequences at the exon-intron boundaries (splice sites). These sequences are
highly degenerate in humans, and it has remained elusive how they are recognized by the spliceosome. Tholen et al.
report a series of high-resolution structures of the human U2 small nucleolar ribonucleoprotein, the component of the
spliceosome that recognizes branch sites. The structures explain how SF3B6 helps to stabilize the branch helix in the
absence of extensive sequence complementarity. A newly identified spliceosome assembly intermediate suggests a
mechanism for fidelity control of branch site recognition. —DJ

View the article online


https://www.science.org/doi/10.1126/science.abm4245
Permissions
https://www.science.org/help/reprints-and-permissions

Use of this article is subject to the Terms of service

Science (ISSN ) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW, Washington, DC
20005. The title Science is a registered trademark of AAAS.
Copyright © 2022 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works

You might also like