PIIS0092867424011887
PIIS0092867424011887
Correspondence
[email protected] (M.P.),
[email protected] (N.Z.)
In brief
The stability of BACH1 is controlled by
two F-box proteins, FBXO22 and FBXL17.
There are two forms of the BACH1
quaternary structure degrons that are
encoded by its dimeric BTB domain.
FBXO22 recognizes the intact dimeric
BTB domain, whereas FBXL17 targets
and remodels the BTB domain dimer
when destabilized by S-nitrosylation.
Highlights
d FBXO22 and FBXL17 play non-redundant roles in regulating
the stability of BACH1
ll
OPEN ACCESS
Article
Recognition of BACH1 quaternary structure degrons
by two F-box proteins under oxidative stress
Shiyun Cao,1,2,11 Sheena Faye Garcia,3,4,11 Huigang Shi,1,2,11 Ellie I. James,5,6 Yuki Kito,3,4 Hui Shi,1,2,12 Haibin Mao,1,2
Sharon Kaisari,3,4 Gergely Rona,3,4,7,8 Sophia Deng,3,4 Hailey V. Goldberg,3,4 Jackeline Ponce,3,4,9 Beatrix Ueberheide,3,4,9
Luca Lignitto,3,4,10 Miklos Guttman,5,6 Michele Pagano,3,4,7,* and Ning Zheng1,2,13,*
1Department of Pharmacology, University of Washington, Box 357280, Seattle, WA 98195, USA
2Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
3Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
4Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
5Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA
6Molecular Engineering & Science Institute, University of Washington, Seattle, WA 98195, USA
7Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
8Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
9Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York,
NY 10016, USA
10Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
11These authors contributed equally
12Present address: Biortus Discovery Co., Ltd., 101 South Building, 99-3 Linhu Avenue, Xinwu District, Wuxi, Jiangsu, P.R. China
13Lead contact
SUMMARY
Ubiquitin-dependent proteolysis regulates diverse cellular functions with high substrate specificity, which
hinges on the ability of ubiquitin E3 ligases to decode the targets’ degradation signals, i.e., degrons. Here,
we show that BACH1, a transcription repressor of antioxidant response genes, features two distinct uncon-
ventional degrons encrypted in the quaternary structure of its homodimeric BTB domain. These two degrons
are both functionalized by oxidative stress and are deciphered by two complementary E3s. FBXO22 recog-
nizes a degron constructed by the BACH1 BTB domain dimer interface, which is unmasked from transcrip-
tional co-repressors after oxidative stress releases BACH1 from chromatin. When this degron is impaired by
oxidation, a second BACH1 degron manifested by its destabilized BTB dimer is probed by a pair of FBXL17
proteins that remodels the substrate into E3-bound monomers for ubiquitination. Our findings highlight the
multidimensionality of protein degradation signals and the functional complementarity of different ubiquitin
ligases targeting the same substrate.
INTRODUCTION number of known degrons is still far below the predicted num-
ber of E3 ligases in most, if not all, eukaryotes.
Ubiquitin-dependent protein degradation is a widespread The cullin-RING ubiquitin ligase (CRL) complexes constitute
mechanism regulating virtually every cellular function.1 The the largest superfamily of multi-subunit E3s in humans with
selectivity of this proteolytic process is exquisitely controlled more than 200 family members classified in five subfamilies:
by the specific degradation signal, also known as ‘‘degron,’’ CRL1 (also known as SCF for SKP1, CUL1, F-box protein),
which is encoded in the substrate protein and perceived by CRL2, CRL3, CRL4A/B, and CRL5.6 Built with a modular archi-
its cognate E3 ubiquitin ligase.2,3 Early studies have mapped tecture, each CRL subfamily employs a battery of substrate re-
the degrons of many classical substrate proteins to various ceptor (SR) subunits to recognize the unique degrons of their
short linear ‘‘motifs’’ (SLiMs) with specific consensus se- targets. For example, the CRL1/SCF SRs, b-TrCP and
quences.4 Many of these degrons are recognized by E3 ligases FBXW7, are two F-box proteins well known to recognize phos-
in their native form, while others require modulation by either phorylated SLiM degrons.7,8 A panel of BC-box and DCAF pro-
post-translational modifications or hormonal signal to promote teins act as the CRL2 and CRL4 SRs to recognize various
E3 binding. Although recent proteome-wide studies have C-end degrons found at the extreme C terminus of substrate
further expanded the cellular repertoire of SLiM degrons,5 the polypeptides.9,10
Cell 187, 1–17, December 26, 2024 ª 2024 The Author(s). Published by Elsevier Inc. 1
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Please cite this article in press as: Cao et al., Recognition of BACH1 quaternary structure degrons by two F-box proteins under oxidative
stress, Cell (2024), https://doi.org/10.1016/j.cell.2024.10.012
ll
OPEN ACCESS Article
CRLs also closely participate in redox homeostasis regulation. nature of the BACH1-BTB domain by size-exclusion chromatog-
Oxidative stress results from an accumulation of oxygen and ni- raphy coupled with multi-angle light scattering (SEC-MALS) (Fig-
trogen-derived reactive species (ROS/RNS), which cause direct ure 1C). Despite its expected symmetric architecture, the
damage to cellular components and interfere with redox BACH1 BTB dimer appeared to be associated with only a single
signaling.11,12 To modulate the levels of prooxidant and anti- copy of FBXO22-SKP1.
oxidant species, mammalian cells have evolved two antago- To reveal the mechanism by which FBXO22 recognizes
nizing redox-sensitive transcription factors to dynamically con- BACH1-BTB, we used cryoelectron microscopy (cryo-EM) to
trol the expression of cytoprotective genes. BACH1, also known determine the structure of the SKP1-CUL1-RBX1-FBXO22 com-
as BTB (broad-complex, tramtrack, and bric-à-brac) and CNC plex (also known as SCFFBXO22) bound to the BACH1 BTB dimer
(cap ‘‘n’’ collar) homology 1, is a master transcriptional repressor at 3.9 Å resolution (Figure S1; Table S1). The SCFFBXO22-BACH1
in heme metabolism and redox homeostasis.13 As a member of complex adopts a canonical SCF architecture31 in which the
the basic leucine zipper (bZIP) protein family, BACH1 heterodi- FBXO22-SKP1 SR module, together with the BACH1 substrate,
merizes with small musculoaponeurotic fibrosarcoma (sMAF) is docked to the N-terminal half of the cullin scaffold (Figure 1D).
proteins and binds to the anti-oxidant response elements Unique among F-box proteins, FBXO22 features a C-terminal
(AREs) of many anti-oxidant genes, including heme oxygenase FIST (F-box and intracellular signal transduction proteins)
1 (HMOX1), to suppress their transcription under normal condi- domain responsible for substrate recruitment (Figure 1A).
tions.14–18 Under oxidative stress, a critical transcription acti- Consistent with our SEC-MALS analysis, the F-box protein forms
vator, NRF2 (nuclear factor erythroid-derived 2-like 2), accumu- an asymmetric complex with the BACH1 BTB dimer at a 1:1
lates and competes with BACH1 for binding sMAF and AREs. In molar ratio (Figure 1E).
doing so, NRF2 induces the expression of these genes, whose The FIST domain is found in eukaryotic F-box proteins and pro-
products go on to eliminate reactive species and restore redox karyotic proteins involved in signal transduction.32 It has been pro-
homeostasis.19–22 posed to function as a sensory domain, possibly binding small
A wealth of studies revealed a crucial role of the CRL3 SR molecular ligands, although its fold was previously uncharacter-
KEAP1 (Kelch-like ECH-associated protein 1) in regulating ized. Our structure reveals that the FBXO22 FIST domain is con-
NRF2 activity in a redox-sensitive manner.23 Under steady-state structed by three structural repeats, each consisting of a central
conditions, KEAP1 recognizes two SLiM degrons of NRF2 and four-stranded b-sheet sandwiched by two a-helices on one side
promotes its ubiquitination and degradation.24–26 When cells and a b-hairpin loop on the other (Figure 1F). The central b-sheet
are challenged by oxidative or electrophilic stress, covalent from each repeat packs against the other two repeats, giving rise
modification of highly reactive cysteine residues in KEAP1 im- to a compact globular fold with a pseudo 3-fold symmetry. A Dali
pairs its E3 ligase activity, enabling newly synthesized NRF2 to search identified two classes of unrelated metabolic enzymes,
rapidly accumulate and move to the nucleus to induce the chorismatases and cyanuric acid hydrolases, with a similar overall
expression of anti-oxidant genes.27,28 Recent studies have iden- fold, which could have evolved from monomers of the trimeric
tified two F-box proteins, FBXO22 and FBXL17, which mirror the YjgF superfamily (Figure S2A).33–35 Different from the metabolic
function of KEAP1 to regulate the stability of BACH1 in response enzymes, the FIST domain of FBXO22 does not feature any
to elevated heme and ROS/RNS levels.29,30 Both F-box proteins obvious ligand-binding pocket. We name the three structural re-
have been implicated in oxidative stress-induced degradation of peats of the FIST domain, FIST-1, FIST-2, and FIST-3. Noticeably,
BACH1. However, how FBXO22 and FBXL17 recognize BACH1 FIST-3 is distinguished from the other two repeats by having a
and together regulate its stability remains unknown. longer b-hairpin loop, whose two b-strands, b15 and b16,
In this study, we reveal that the stability of BACH1 is dictated perfectly align with the edge of the central b-sheet and extend it
by two mutually exclusive degrons, both of which are encoded in into a six-stranded sheet (Figures 1F and S2A).
the quaternary structures of its dimeric BTB domain. The two
BACH1 degrons are functionalized by different forms of oxida- BACH1 quaternary structure degron
tive stress and recognized by FBXO22 and FBXL17 via distinct Similar to most BTB dimer structures, the BACH1 BTB domain
and complementary mechanisms. By shedding light on the mo- dimer is characterized by a domain swapping architecture, in
lecular basis of BACH1 regulation, our results define a previously which the two protomers exchange their N-terminal
undescribed class of protein degradation signals, which exploit b-strands.36,37 In BACH1, the N-terminal b-strand (b1) of each
the structural complexity of protein assembly to achieve sub- protomer runs in antiparallel with the C-terminal a-helix (CTH)
strate specificity. (a6) of the opposite protomer, extending the dimer interface
beyond the core BTB domain (Figure 1G). Remarkably, one of
RESULTS these cross-protomer b1-a6 pairs within the BACH1-BTB dimer
makes up the entire interface with the F-box protein by clinging
Structure of the SCFFBXO22-BACH1-BTB complex to the edge of FBXO22 FIST-3 (Figure 2A).
BACH1 contains an N-terminal BTB domain, which mediates its The tri-molecular interface between FBXO22 and the BACH1-
homodimerization and is critical for FBXO22 interaction (Fig- BTB dimer is stabilized by a hybrid of polar and hydrophobic in-
ure 1A).30 In a GST pull-down assay with purified recombinant teractions. Specifically, the b1 strand of one BACH1-BTB chain
proteins, we validated the direct binding between the BACH1 (BTB-B), together with the b5 strand of the second BACH1 BTB
BTB domain and the full-length FBXO22 protein in complex chain, is juxtaposed with the b15-b16 hairpin of FBXO22 FIST-3,
with SKP1 (Figure 1B). We further confirmed the homodimeric creating an intermolecular anti-parallel b-sheet supported by a
ll
Article OPEN ACCESS
A B C
BACH1 - + + GST-FBXO22
BTB LZ + + - + BACH1-BTB 0.08 FBXO22-SKP1
1 130 736
75 GST-FBXO22 62 kDa (± 4%)
FBXO22 63
UV (AU)
0.06
48 BACH1-BTB
F FIST 35
1 25 SKP1 0.04 FBXO22-SKP1 32 kDa (± 2%)
403 -BACH1-BTB
FBXL17 11
BACH1-BTB 0.02 88 kDa (± 6%)
F LRRs
1 701 0.00
GST pull down 5 10 15 20
Volume (ml)
D E
FBXO22
BACH1-BTB(A)
SKP1 FBXO22
BACH1-BTB(A)
BACH1-BTB(B)
F G
FIST-1
FIST
T1 C
N
FIST-2
o α6
90
9
β5
FIST-3 β1
FIST-3
α10 BACH1-BTB
Homodimer
β15
β16
β1 β5
FIST-1
FIST-2 α6
FBXO22 F
FIST Domain
omain C
N
network of backbone hydrogen bonds (Figure 2B). These sec- by two salt bridges and intermolecular van der Waal packing
ondary structure interactions are cemented by an adjacent hy- made by additional residues from the F-box protein (Asp366,
drophobic core, which is nucleated by four amino acids from Arg308, Lys377, and Arg376) and the two protomers of the sub-
the BACH1-BTB b1-a6 pair (Phe9, Tyr11, Cys122, and Phe125) strate (Glu12 and Phe128).
and three residues in the FBXO22 b15-b16 hairpin (Ile368, In support of a critical role played by the N-terminal b1 strand
Phe373, and Leu375). Peripheral to the hydrophobic core, the of BACH1-BTB at the interface, previous studies have shown
FBXO22-BACH1-BTB dimer interface is further strengthened that individual alanine mutations of its two aromatic residues,
ll
OPEN ACCESS Article
A B C
D G H
F I J
K L M
ll
Article OPEN ACCESS
Phe9 and Tyr11, are sufficient to abolish FBXO22-BACH1 inter- Regulation of FBXO22-BACH1 interaction
actions.30 Similarly, replacing Phe125 in the BACH1 a6 helix with Using biolayer interferometry (BLI), we determined the affinity of
alanine or aspartate effectively abrogates E3-substrate complex FBXO22 toward BACH1-BTB, which is 90 nM and on par with
formation (Figure 2C). Therefore, both secondary structure ele- other CRL-substrate interactions (Figure 2G).42 Consistent with
ments of the BACH1-BTB b1-a6 pair, which is shaped by the the lack of any obvious ligand-binding pocket in FBXO22 or
quaternary structure of the BTB dimer, are necessary to define BACH1-BTB, the binding between the two proteins is insensitive
the functional degron. In a second set of mutational analyses, to heme (Figure 2H). Heme, therefore, does not appear to be
we validated that the majority of the FBXO22 residues at the physically required for the productive interaction between
tri-molecular interface are indispensable for binding BACH1, FBXO22 and BACH1 (Figures S2C and S2D) and most likely pro-
suggesting that the edge of the third FBXO22 FIST repeat repre- motes BACH1 recognition by FBXO22 through an indirect mech-
sents the primary docking site for BACH1 (Figure 2D). Notice- anism. In previous studies, heme has been reported to block the
ably, although the symmetric BACH1 BTB domain dimer con- DNA-binding activity of BACH1-sMAF heterodimers indepen-
tains two cross-protomer b1-a6 pairs, two FBXO22 molecules dent of the BACH1 BTB domain.19 This raises the possibility
cannot simultaneously recognize the two identical BACH1 de- that the quaternary structure degron of BACH1 might not be
grons without a severe steric collision (Figure S2B). A single accessible to FBXO22 until the transcription factor is released
SCFFBXO22 E3 complex, therefore, appears responsible for ubiq- from the DNA.
uitinating both protomers of a BACH1 dimer via their BTB do- The BTB domain is widely found in CRL3 SRs and transcrip-
mains (Figures S2C and S2D). tion factors.37 Besides mediating protein oligomerization, the
Mammalian BACH1 has a close paralog, BACH2, which is pri- BTB domain of select transcription repressors, such as PLZF
marily expressed in the brain and spleen with well-documented and BCL6, has been reported to interact with co-repressors.43,44
roles in regulating immune cell differentiation.38–40 The BTB do- In the case of BCL6, distinct SLiMs of SMRT/NCOR2 and BCOR
mains of the two BACH proteins, including their terminal regions, co-repressors have been shown to form the same interface with
are overall 62% identical in sequence (Figure 2E). Their homo- the BCL6 BTB domain dimer, which is highlighted by an inter-
dimer structures can be superimposed with a root mean square molecular b-sheet involving the N-terminal b1-strand of BCL6-
deviation (RMSD) of 0.6 Å (Figure S2E).36,41 Despite their BTB (Figure 2I).45,46 Such a binding mode is remarkably analo-
sequence and structural similarity, only BACH1 but not BACH2 gous to the FBXO22-BACH1-BTB interface (Figure 2J). Similar
can be recognized by FBXO22 (Figure 2F). To identify key ele- to PLZF and BCL6, BACH1 has been previously shown to be
ments in BACH1 conferring this specificity, we made chimeric functionally associated with nuclear co-repressor NCOR1, a
BACH proteins and tested their interactions with the F-box pro- close paralog of SMRT/NCOR2.47 This prompted us to test
tein. Interestingly, replacing Phe9 in the N-terminal b1 strand of whether NCOR1 can compete with FBXO22 for binding the
BACH1 with its counterpart in BACH2 (Tyr12) had little effect BTB domain of BACH1. A central region of NCOR1 has been pre-
on FBXO22 binding. By contrast, swapping the C-terminal a6 he- viously identified to interact with the BTB domain of multiple
lices of the two BACH proteins effectively switched their ability to functionally unrelated transcriptional co-repressors.43 Sequence
engage the E3 (Figure 2F). A close examination of the BACH2 alignment and structure prediction analyses indicate that this re-
BTB domain structure reveals a non-helical nature of its C-termi- gion contains several potential SLiMs conserved among NCOR1
nal region (Figure S2E) distinct from the a6 helix of BACH1. This orthologs, including the one used by NCOR2 for binding BCL6-
structural difference provides a possible explanation for BTB (Figure S2F). When co-expressed and co-purified from
BACH2’s inability to bind FBXO22. E. coli, this 230 amino acid NCOR1 fragment was indeed suf-
Together, these results reinforce the importance of the struc- ficient to form a complex with the BACH1 BTB domain (Fig-
tural integrity of the b1-a6 degron of the BACH1 BTB dimer ure 2K). Importantly, the NCOR1 fragment was able to effectively
for FBXO22 recognition and underscore the capacity of the qua- block FBXO22-BACH1 binding in an AlphaScreen-based
ternary structure degron in conferring high E3-substrate selec- competition assay (Figure 2L), suggesting that the transcriptional
tivity, a previously undescribed characteristic of CRL substrates. co-repressor can occupy the quaternary structure degron of
(D) FLAG immunoprecipitation of FBXO22 mutants. HEK293T cells were transfected with either EV or FLAG-FBXO22 WT or mutants. Cells were then treated with
MLN-4924 prior to FLAG immunoprecipitation. Proteins were immunoblotted as indicated. Non-specific protein bands are labeled with asterisks.
(E) Sequence alignment of the N- and C-terminal regions of BACH1 and BACH2 BTB domains.
(F) FLAG immunoprecipitation of BACH1-BACH2 chimeras. HEK293T cells were transfected with either EV or FLAG-BACH1/BACH2 chimeras as indicated. Cells
were then treated with MG-132 prior to FLAG immunoprecipitation. Proteins were immunoblotted as indicated. Non-specific protein bands are labeled with
asterisks.
(G) Biolayer interferometry (BLI) measurements of FBXO22 and BACH1-BTB interaction. Kd-equilibrium, dissociation constant.
(H) BACH1-BTB pull-down by GST-FBXO22 in the presence and absence of 10 mM hemin. Non-specific protein bands are labeled with asterisks.
(I) Structure of BCL6-BTB dimer (green) in complex with the co-repressor SMRT (purple, PDB:1R2B) or BCOR (slate, PDB: 3BIM).
(J) Structure of BACH1-BTB dimer in complex with FBXO22 b15-b16 hairpin (purple).
(K) BACH1-BTB pull-down by GST-NCOR1.
(L) Inhibition of the FBXO22-BACH1-BTB interaction by NCOR1 fragments measured in AlphaLISA competition assays. Data are presented as mean ± SD. IC50,
half-maximum inhibitory concentration.
(M) Inhibition of SCFFBXO22-catalyzed BACH1 ubiquitination by an 230 amino acid NCOR1 fragment.
See also Figure S2.
ll
OPEN ACCESS Article
BACH1. Interestingly, when the N-terminal and C-terminal the NOR3-S1PC-treated BACH1 BTB domain still retains its ho-
halves of the NCOR1 central fragment were individually tested, modimeric form in solution (Figure S3I), it can no longer interact
each sub-fragment was able to inhibit the interaction between with FBXO22 in a Ni2+ NTA pull-down assay (Figure 3F). By
FBXO22 and the BACH1 BTB dimer, albeit at a much lower po- contrast, FBXL17 showed robust activity in binding the NOR3-
tency (Figure 2L). Therefore, NCOR1 might employ more than S1PC-treated BACH1 BTB domain (Figure 3G). Likewise, NO
one SLiM to simultaneously engage the two identical FBXO22- treatment reduced the amount of FBXO22 that can be co-immu-
binding sites on BACH1-BTB to block the ubiquitination of the noprecipitated by BACH1, while the opposite is true for FBXL17
transcription repressor (Figure 2M). Together, these results imply (Figures 3H and 3I). These results suggest that FBXL17 might
that the NCOR1 co-repressor can shield the quaternary structure have been evolved to recognize and ubiquitinate BACH1
degron of chromatin-bound BACH1 from FBXO22 until BACH1 is when BACH1 is in a form incompatible with FBXO22 binding
released from DNA by heme. (Figures S3J and S3K).
Interestingly, treatment with either an NO donor or hemin
Degradation of S-nitrosylated BACH1 by FBXO22 and induced the release of BACH1 from chromatin (Figure 3J).
FBXL17 Accordingly, sNONO reduced the binding of BACH1 to MafF
FBXL17 is a leucine-rich repeats (LRRs)-containing F-box pro- and NCOR1 in the cell (Figure 3K). Similar to hemin,19 an NO donor
tein (Figure 1A) that, similarly to FBXO22, has been implicated can effectively abrogate the ability of the BACH1 bZIP domain to
in the degradation of BACH1 in response to heme-mediated super-shift sMAF-bound DNA in an electrophoretic mobility shift
oxidative stress.29 Furthermore, it has been proposed that assay (Figure S3L). These results implicate that, in addition to
FBXL17 can recognize non-functional heterodimers of BTB pro- its BTB domain, the bZIP DNA-binding domain of BACH1, which
teins and sequester their monomeric BTB domains to ubiquiti- is rich in cysteine residues, might also be susceptible to
nate and degrade the mismatched substrate proteins.48,49 S-nitrosylation. We speculate that a subpopulation of the
How FBXL17 captures BACH1 upon oxidative stress, presum- BACH1 proteins, depending on their cellular context, might be
ably by recognizing its homodimeric BTB domain, remains un- modified by S-nitrosylation within their bZIP domain but not the
clear. We first investigated the functional relationship between BTB domain, which could explain why BACH1-FBXO22 complex
FBXL17 and FBXO22 in regulating BACH1 stability in the context could still be detected in NO-treated cells (Figure 3H) and, in com-
of BACH1 S-nitrosylation. Nitric oxide (NO) is a free radical in the parison to the parental cells, BACH1 was partially stabilized in
cell that is either produced endogenously or exogenously.50 By FBXO22 KO cells despite NO treatment (Figures 3B–3D).
reacting with cysteine residues of selective targets, NO induces
protein S-nitrosylation and further disulfide formation, which FBXL17 preferentially remodels NO-modified BACH1-
contribute to redox signaling that protects the cell against oxida- BTB dimer
tive stress.51,52 NOR3, an NO donor, has recently been shown From the single time point (30 min) affinity pull-down assay, we
to induce BACH1 degradation, an effect that can be augmented noticed that FBXL17 preferentially interacts with the NO-modi-
by a garlic-derived sulfur amino acid, S-1-propenylcysteine fied BACH1 BTB domain over the native polypeptide (Figure 3G).
(S1PC).53 We first confirmed by biotin switch assay that To further dissect the activity of FBXL17 in BACH1-BTB recogni-
BACH1 can be modified by two additional NO donors, tion, we used BLI to monitor the kinetics of their interaction with
S-nitrosoglutathione (GSNO) and spermine NONOate (sNONO) or without treating BACH1-BTB with NOR3-S1PC. In agreement
(Figure 3A). Using FBXL17 and FBXO22 individual and double with the affinity pull-down results, compound-treated BACH1-
knockout (KO) HCT-116 cells (Figures S3A and S3B), we verified BTB interacted strongly with FBXL17 with an ‘‘apparent Kd’’ of
that both FBXO22 and FBXL17 contribute to BACH1 turnover at 110 nM, whereas the native BACH1-BTB protein only showed
steady state, although FBXO22 appears to play a predominant a detectable affinity toward the F-box protein with an apparent
role (Figures S3C–S3E). By contrast, in response to NO treat- Kd greater than 2.4 mM (Figures 4A and 4B). Noticeably, with
ment, BACH1 degradation is similarly slowed down when either compound-treated BACH1-BTB immobilized on the probe, the
FBXO22 or FBXL17 is missing (Figures 3B–3D, S3F, and S3G). maximum binding signal appeared far from plateauing within
The two F-box proteins, therefore, appear to play nonredundant the time window of the measurement, even though the amplitude
roles in regulating BACH1 stability in response to stress. of the association curves started to show saturation when the
The BTB domain of BACH1 contains five cysteine residues, FBXL17 concentration reached the mM range (Figure 4A). The
two of which, Cys107 and Cys122, are located at the b1-a6 de- same phenomenon held true when we lengthened the time win-
gron recognized by FBXO22 (Figure 3E). Consistent with a recent dow to 10 min (Figure S4A). This feature hinted at a minimally
proteome-wide S-nitrosylation site mapping study,56 mass biphasic nature of the interaction, which might involve complex
spectrometry (MS) analysis of the recombinant BACH1 BTB rearrangement in addition to simple binding equilibrium.
domain treated with the NO donor NOR3-S1PC identified To further characterize the interaction between FBXL17 and
Cys107 but not Cys55 as an S-nitrosylation site (Figure S3H). compound-treated BACH1-BTB, we used SEC-MALS to analyze
Due to incomplete sequence coverage, whether Cys34, samples of the two proteins mixed for different periods of time. In
Cys109, and Cys122 could be modified by NO remained unclear. congruence with the reported activity of FBXL17 in forming stable
As a residue on the a5 helix of BACH1-BTB making direct con- complexes with monomeric BTB domains,48 the F-box protein
tact with the a6 helix (Figure 3E), covalent modification of was found to bind a single copy of BACH1-BTB after the two pro-
Cys107 is expected to perturb the local structure of the b1-a6 teins were mixed for 24 h (Figure 4C). Interestingly, it took a rela-
degron, thereby impairing FBXO22 binding. Indeed, even though tively long incubation time for such a stable 1:1 complex to form,
ll
Article OPEN ACCESS
A HCT-116 B C
sNONO
HCT-116
GSNO
FBXL17 + FBXO22 KO
FBXO22 KO
NT FBXL17 +
WT FBXL17 KO FBXO22 KO FBXO22 KO FBXL17 KO
0 1 2 3 4 5 0 1 2 3 4 5 0 1 2 3 4 5 0 1 2 3 4 5 sNONO (h)
BACH1
AP: biotin
BACH1
KEAP1 (short exp.)
BACH1
SKP1 (long exp.)
Parental
FBXO22
BACH1
WCE
SKP1 GAPDH
F Ni2+ pull down
- - + NOR3-S1PC
Parental FBXO22 KO
C
- + + BACH1-BTB
D FBXL17 KO FBXL17 + FBXO22 KO E N kDa + + + FBXO22-SKP1
α6 α5 BACH1-BTB(A) 100
C107 75
C109 63 GST-FBXO22
48
35
GST-BACH1
25
-BTB
C122 20 His-SKP1
β1
G - - + - NOR3-S1PC
- + + + BACH1-BTB
C55
C34
kDa + + + - FBXL17-SKP1
100
75 MBP-FBXL17
BACH1- 63 *
48 **GST-BACH1
BTB(B)
35
CHX (h) sNONO + CHX (h) -BTB
25
20 His-SKP1
HEK293T
Ni2+ pull down
H
(MLN4924)
EV + + FLAG-BACH1 I HEK293T J K
- + sNONO HCT-116 HEK293T
EV
ll
OPEN ACCESS Article
A B
Apparent Probe: Biotin-BACH1-BTB Probe: Biotin-BACH1-BTB
0.5 (NOR3-S1PC treated) 0.5
Kd = 110 nM (Untreated)
Analyte: FBXL17 Analyte: FBXL17
0.4 0.4
Response (nm)
Response (nm)
[FBXL17]
2000 nM 0.3
0.3 Apparent Kd > 2.4 μM
666 nM
0.2 222 nM 0.2 [FBXL17]
74 nM 2000 nM
0.1 25 nM 0.1 666 nM
222 nM
0.0 0.0 74 nM
0 100 200 300 400 0 100 200 300 400
Time (s) Time (s)
BACH1-BTB 11
treated Untreated NOR3-S1PC treated BACH1-BTB
34kDa
Binding signal (%)
120
E Probe: Biotin-BACH1-BTB
F
0.5
0.5 Probe: Biotin-BACH1-BTB
(Untreated)
0.4 (NOR3-S1PC treated)
Analyte: FBXL17 400nM
Response (nm)
WT
0.3 C55A 0.3 WT
0.2 C107A
0.2 C55A
C109A
0.1 C107A
C122A
0.1 C109A
0.0 C122A
0 100 200 300 400 0.0
Time (s) 0 100 200 300 400
Time (s)
even though BLI could instantaneously detect the interaction be- To fully assess the binding kinetics of FBXL17 and BACH1-
tween FBXL17 and BACH1-BTB. Given that compound-treated BTB, we returned to the affinity pull-down assay and monitored
BACH1-BTB remains a stable dimer even after being diluted to the interaction with a time window up to 1–2 days. As expected
3 mM (Figure S3I), we speculate that the biphasic binding curve from our BLI and SEC-MALS results, it took about 1 h for half of
detected by BLI reflects both the initial contact between FBXL17 the maximal amount of FBXL17 to become stably associated
and dimeric BACH1-BTB and the final complex formation be- with compound-treated BACH1-BTB under the tested experi-
tween FBXL17 and monomeric BACH1-BTB after the BACH- mental condition (Figure 4D). By contrast, the amount of
BTB dimer is remodeled by the F-box protein. FBXL17 captured by untreated BACH1-BTB did not saturate
ll
Article OPEN ACCESS
0 0
NOR3-S1PC treated BACH1-BTB 1 10 102 103 104 105 1 10 102 103 104 105
Exchange (Da)
Tm = 54.9 + 0.2°C 1
4 LSLNDQRK 2
d(Fluorescence)/d(T)
8 2
10000 3 (22-29) 2
DMSO 2 4 7
5 1
5000 NOR3-S1PC (97-101)
1 ILSKE 7
3 6
0 0 0
1 10 102 103 104 105 1 10 102 103 104 105
3 4
-5000 6 (91-96)
30 40 50 60 70 80 90 4 3
2 AYTAKL 4
Temperature (°C) 4 2
2
2 (6-14) 1 (112-119) (123-128)
1
SSVFAYESS 3 FLSVHNIE 5 FQFLKF 6
0 0 0 0
1 10 102 103 104 105 1 10 102 103 104 105 1 10 102 103 104 105 1 10 102 103 104 105
Time (sec)
C 6NSVFAYESSVHSTNVL21 79 VTVKGFEPLIQ 89 113 LSVHNIEES121
Unmod. NO-treated
C
Unmod. NO-treated Unmod. NO-treated
Cent: 864.3895 m/z Cent:864.3907 m/z Cent: 616.1757 m/z Cent: 616.1787 m/z Cent:1028.004 m/z Cent:1028.006 m/z
Undeut.
Cent: 867.003 m/z Cent: 866.9031 m/z Cent: 616.4203 m/z Cent: 617.0101 m/z Cent: 1030.707 m/z Cent: 1030.862 m/z
F-test p = 1.23e-5 F-test p = 2.98e-6
Bimodal 1: Bimodal 1: 64%
21% Bimodal 2:: 36%
Bimodal 2:
79% 3 sec
Relative Intensity
Cent: 869.0092 m/z Cent: 868.7944 m/z Cent: 617.9536 m/z Cent: 618.5192m/z Cent:1032.555 m/z Cent:1032.202 m/z
F-test p = 4.55e-10 F-test p = 1.96e-3 F-test p = 6.21e-8
Bimodal 1: Bimodal 1:
Bimodal 1: 76% 42%
13% Bimodal 2: Bimodal 2:
Bimodal 2: 24% 58% 20 hr
87%
Cent: 869.2066 m/z Cent: 869.2045 m/z Cent: 619.1559 m/z Cent: 619.1489 m/z Cent:1032.766 m/z Cent:1032.892 m/z
Max
deut.
863 867 871 863 867 871 615 617 619 620 623 617 619 620 623 1025 1030 1035 1040 1030 1035 1040
m/z
ll
OPEN ACCESS Article
even after 2 days. Such a strong preference of FBXL17 to cap- FBXO22 but increased its ability to bind FBXL17 (Figures S5E–
ture BACH1-BTB monomers from the compound-treated dimer S5G). This could explain the similar half-lives of the BACH1
over the intact non-treated dimer raised the possibility that NO mutant and the wild-type (WT) protein (Figure S5H), as the
treatment destabilizes BACH1 to promote FBXL17 binding and impairment of its FBXO22-mediated turnover is compensated
subsequent ubiquitination. by FBXL17. Based on these observations, we conclude that
Importantly, this effect of NO is attributable to the modification S-nitrosylation of the two cysteine residues in BACH1 reduces
of two specific cysteine residues on the BACH1 BTB domain. the structural stability of the BACH1-BTB dimer by reconfiguring
Without impacting the resistance of native BACH1-BTB dimer the dimer arrangement, thereby facilitating FBXL17 binding.
to FBXL17 binding (Figure 4E), individual alanine mutations of
Cys107 and Cys122 but not the other three cysteine residues Transient remodeling of BACH1-BTB dimer by a pair of
significantly reduced the ability of FBXL17 to release monomeric SCFFBXL17 proteins
BACH-BTB from its NO donor-treated dimeric form (Figures 4F, To reveal the mechanism by which FBXL17 recognizes and
S4B, and S4C). This result allows us to map Cys122, an amino remodels NO-modified homodimeric BACH1-BTB, we mixed
acid also residing within the b1-a6 degron, as another key resi- NO donor-treated BACH1-BTB with the SCFFBXL17 complex for
due sensitive to S-nitrosylation. different periods of time and subjected the samples to cryo-EM
analysis. 2D classification of the particles imaged at three incuba-
Global destabilization of BACH1-BTB dimer by tion times (30 s, 10 min, and 24 h) indicated that the predominant
S-nitrosylation observable species were either in the form of free SCFFBXL17 or the
To confirm that S-nitrosylation indeed destabilizes BACH1-BTB E3 complex bound to the monomeric BACH1 BTB domain (here-
dimerization, we first used differential scanning fluorometry after referred to as mSCFBTB) (Figures S6A–S6C). Strikingly, a
(DSF) to show that NO treatment reduced the thermostability small fraction of the particles appeared to contain a pair of parallel
of BACH1-BTB by almost 10 degrees Celsius (Figure 5A). Subse- SCFFBXL17 complexes with the FBXL17 LRR domains facing each
quent hydrogen-deuterium-exchange MS (HDX-MS) experi- other, likely sandwiching a BACH1-BTB dimer in the middle (Fig-
ments allowed us to map the secondary structure elements ure 6A). Although these minor classes, which possibly represent a
within the BACH1-BTB dimer that are structurally destabilized. transient intermediate species of the BACH1-BTB dimer remod-
Strikingly, while the a6 and b1 terminal regions are intrinsically eled by the E3 complex, yielded two 7.8 Å 3D reconstructions (Fig-
more susceptible to deuteration in both native and NO donor- ure S6C), the numbers of particles were too limited to determine
treated samples, multiple secondary structure elements within the structures at a higher resolution.
the BTB core domain, including those at the central dimer inter- To overcome the challenge of resolving these presumably un-
face, are impacted the most by NO treatment (Figures 5B, S4D, stable intermediates, we turned to the native BACH1-BTB sam-
and S4E). Because BACH1-BTB remains as a dimer upon NO ple, which is still susceptible to reconfiguration by FBXL17,
treatment (Figure S3I), these HDX results hint at an altered topol- except at a much slower rate (Figure 4D). We reasoned that the
ogy of the BTB dimer induced by S-nitrosylation. In support of higher resistance of the native BACH1-BTB sample to FBXL17-
this notion, several peptides detected in our HDX-MS analysis mediated monomerization might offer an experimental window
displayed a bimodal exchange profile in the NO-treated samples to capture the intermediate steps of the reaction. Remarkably,
(Figures 5C and S4F), indicating that the BACH1-BTB dimer cryo-EM analysis of a sample of SCFFBXL17 incubated with native
might exist in two distinct populations where one population un- BACH1-BTB unveiled four species, free SCFFBXL17, mSCFBTB,
derwent faster deuterium exchange than the other. For some and two distinct dimeric SCFFBXL17-BACH1-BTB super-assemblies
peptides, the two species were also detectable in the native (hereafter referred to as dSCFBTB-I and dSCFBTB-II) (Figure S7).
sample. NO treatment appeared to shift the equilibrium between The structure of the mSCFBTB complex resolved at 3.2 Å resolu-
the two populations, increasing the fast exchange species, tion is highly analogous to the previously reported crystal struc-
which is structurally less stable. These results are in full agree- ture of FBXL17 in complex with the monomeric mutant KEAP1
ment with the global destabilizing effect of NO treatment de- BTB domain,48 except that no clear density is present for the
tected by the DSF thermal shift assay (Figure 5A). b1 strand of the BACH1 BTB domain (Figure 6B). In both struc-
By directly detecting two S-nitrosylated peptides from our LC- tures, the canonical dimer interface of the BTB domains is
MS analysis of the NOR3-S1PC-treated sample, we further blocked by the FBXL17 CTH. Importantly, both Cys107 and
confirmed that C107 and C122 are the only two NO-modified Cys122 of BACH1-BTB are solvent-exposed and far away from
amino acids within the BACH1 BTB domain (Figures S5A– the BACH1-FBXL17 interface, indicating that they are not directly
S5C). Akin to NO treatment, a C122E mutation in BACH1 recognized by the F-box protein. Modification of these two resi-
mimicking the effect of S-nitrosylation reduced the thermosta- dues in the NO donor-treated BACH1 sample, therefore, likely
bility of the BTB domain (Figure S5D). Both in vitro and in cells, accelerated FBXL17 binding by destabilizing the BACH1-
the BACH1 C122E mutant was no longer recognized by BTB dimer.
that was slightly more protected in the NO-treated sample. White indicates no observable differences. Individual uptake plots show the exchange for unmodified
(blue) and NO modified (red). Data are presented as mean ± SD.
(C) Peptides 6–21 and 79–89 exhibit bimodal exchange behavior but with different extents in the unmodified and NO-modified states. Individual populations
within the spectra are shown in purple and green. By contrast, peptides 113–121 do not exhibit bimodal profiles.
See also Figures S4 and S5.
ll
Article OPEN ACCESS
BACH1-
Anti-parallel dimer BTB(B)
FBXL17
LRR Domain
C dSCFFBXL17-BTB-I dSCFFBXL17-BTB-II E
BACH1- Free form dSCFFBXL17-bound form
SKP1 BACH1-
BTB BTB Partially α6 BACH1- BACH1-
Melt BTB(A) (β1) (α6) BTB(A)
β1 1 2
β5 Disordered β5
BACH1- BACH1- α5 β5
FBXL17 BTB FBXL17 BTB Disordered
β1 α5
α6
β5 BACH1- (α6) (β1)
CUL1 CUL1 BTB(B)
BACH1- α5
RBX1 BTB(B) Partially Melt
RBX1
F HEK293T G H
WCE IP: α-FLAG 1.5 8 24 hours 1.5 8 24 hours
EV - - + + + EV - - + + + FLAG-FBXL17 + - + - + - Biotin-BACH2-BTB + - + - + - Biotin-BACH1-BACH2
+--+- +--+- HA-FBXL17 - + - + - + Biotin-BACH1-BTB - + - + - + Biotin-BACH1-BTB
- +- - + - +- - + HA-FBXL17 ΔCTH + + + + + + MBP-FBXL17-SKP1 + + + + + + MBP-FBXL17-SKP1
kDa kDa
HA-FBXL17 (α-HA) 75 75 MBP-FBXL17
MBP-FBXL17
HA-FBXL17 ΔCTH (α-HA) 63 63
48 48
35 35
FLAG-FBXL17 (α-FLAG) 25 SKP1 25 SKP1
17 17
SKP1 11
Biotin-BACH1/2-BTB Biotin-BACH1-BTB
11 BACH2-BTB
GAPDH Biotin pull down Biotin pull down
ll
OPEN ACCESS Article
Although the 3D reconstructions of dSCFBTB-I and dSCFBTB-II BTB domain and a mutant BTB defective for binding FBXL17
suffered from a lower resolution (4.4 Å) (Figure 6C), local refine- (Figures 6J and 6K). These results strongly suggest that the
ment improved the density map and enabled us to resolve the homodimeric BACH1 BTB dimer is indeed inspected by a pair
secondary structures of the two BACH1 BTB domains in the mid- of SCFFBXL17 proteins, which is distinct from SCFFBXO22. If the
dle of the dimeric SCF (Figures 6D, S7C, and S7D). In dSCFBTB-I, structural integrity of the substrate dimer is compromised by
FBXL17 is engaged with BACH1-BTB through the same interface oxidative modifications, such as S-nitrosylation, SCFFBXL17 can
as found in mSCFBTB. However, in addition to the b1 strand, the a6 efficiently seize monomeric BACH1.
helix in each BACH1-BTB protomer is also structurally disordered.
Remarkably, the b5 strands of the two protomers join each other DISCUSSION
and form an intermolecular anti-parallel b sheet, structurally
mimicking the b1–b5 cross-protomer two-stranded b sheet found Degrons beyond SLiMs
in the isolated BACH1-BTB dimer (Figure 6E). This new dimer Ubiquitin ligases have been well known to recruit substrates by
interface is further substantiated by the cross-promoter packing recognizing their SLiM degrons.2 Although portability has long
of a coiled region transformed from the mostly melt a5 helices. been implicated as an inherent property of these degradation
A similar dimer interface, albeit being slightly twisted, is found in signals, it is not an essential feature originally used to define
dSCFBTB-II. As a result, the C-terminal lobes of the two CUL1- the term degron.3 Recent studies have shown that the targeting
RBX1 catalytic scaffolds become separate from each other (Fig- signals of a number of CRL-substrate proteins are constructed
ure 6C). The dimeric arrangement of these dSCFBTB complexes, by their tertiary folds.57,58 This study expands the degron space
therefore, is predominantly, if not solely, stabilized by the into the quaternary structures of protein oligomers, which allow
BACH1 BTB domains. Importantly, the two structure models, E3s to differentiate targets based on signature motifs encrypted
namely dSCFBTB-I and dSCFBTB-II, derived from the native only in high-order assemblies. In the case of the b1-a6 degron of
BACH1-BTB sample, fit snuggly into the 7.8 Å 3D reconstruction the dimeric BACH1-BTB recognized by FBXO22, substrate
maps obtained with the NO-treated sample, indicating that they specificity is dictated by the proper association of the two iden-
belong to common species found in the two specimens tical subunits of the substrate. By contrast, the degradation
(Figures S6D and S6E). Taken together, we postulate that these signal of the BACH1-BTB dimer decoded by FBXL17 is deter-
dimeric SCFFBXL17-BACH1-BTB super-assemblies represent inter- mined by the strength of protein-protein interaction within the
mediate states of BACH1-BTB dimer remodeling, which is simul- homodimeric assembly, a physical property that can be affected
taneously catalyzed by two SCFFBXL17 E3 complexes. This notion by post-translation modifications associated with oxidative
is further supported by co-immunoprecipitation analysis, which stress. These examples highlight the diverse nature of degrada-
revealed physical association between two differentially tagged tion signals recognized by ubiquitin ligases and underscore the
FBXL17 when co-expressed in the cell (Figures 6F and S5I). Their complex mechanisms governing E3-substrate specificity in pro-
complexation was largely abrogated when the CTH, which is tein degradation. We speculate that the number of degrons with
responsible for BTB binding, was removed from one of the high-order structural complexity beyond simple primary amino
FBXL17 constructs (Figure 6F), suggesting that the association acid sequence will continue growing, especially as more poten-
of the two FBXL17 proteins is dependent on BTB binding. tial CRL E3 substrates are deconvoluted.59–61
To assess whether efficient substrate remodeling indeed re-
quires the synergy of two copies of the E3 complexes, we iso- Substrate repertoire of FBXO22 and FBXL17
lated a BACH1-BACH2 BTB domain heterodimer with tandem FBXO22 has been suggested to ubiquitinate several substrate
affinity purification upon co-expression. Lacking the key resi- proteins with no structural similarity to BACH1.62 How the F-box
dues for FBXL17 binding,48 homodimeric BACH2-BTB is protein recognizes these targets, presumably also through its
completely resistant to remodeling by the F-box protein (Fig- FIST domain, remains to be determined. With a role in protein
ure 6G). Interestingly, even with an incubation time of about dimerization quality control, FBXL17 has been implicated in ubiq-
1 day, FBXL17 can barely capture BACH1-BTB from the uitinating a subset of BTB-domain-containing proteins when they
BACH1-BACH2 heterodimer, which is in stark contrast to the ho- form mismatched heterodimers through their BTB domains.48,49
modimeric BACH1-BTB (Figure 6H). Of note, because the However, only the structure of FBXL17 in complex with a mono-
BACH1-BACH2 heterodimer retains a functional b1-a6 quater- meric BTB domain has been captured so far, leading to a paradox
nary structure degron (Figure 2F), it can still be recognized by in the mechanism of action of this E3. We propose that it is not the
FBXO22 (Figure 6I). Similarly, FBXL17 can sequester WT mismatched nature of the BTB domain heterodimers but the
BACH1-BTB from a WT BACH1-BTB homodimer far more effi- strength of protein-protein interaction at the dimeric interface
ciently than a BACH1-BTB heterodimer consisting of a WT that is perceived by FBXL17. This distinction explains why the
(H) A comparison of the remodeling efficiency of BACH1-BTB homodimer and BACH1-BTB-BACH2-BTB heterodimer by FBXL17 as monitored by the time
course of BACH1-BTB-MBP-FBXL17 association.
(I) Binding of the BACH1-BTB-BACH2-BTB heterodimer to FBXO22 by MBP pull-down assay. MSB is a protein solubilizing tag unrelated to MBP.
(J) FBXL17 pull-down by biotinylated wild-type (WT) BACH1-BTB but not a mutant (MT) defective in FBXL17 binding.
(K) A comparison of the remodeling efficiency of WT BACH1-BTB homodimer and a WT and mutant BACH1-BTB heterodimer by FBXL17 as monitored by the
time course of BACH1-BTB-MBP-FBXL17 association.
See also Figures S6 and S7 and Table S1.
ll
Article OPEN ACCESS
ll
OPEN ACCESS Article
BACH1, including several in the bZIP domain, have been identi- d Any additional information required to reanalyze the data reported in this
fied by proteome-wide analyses to be sensitive to modifications paper is available from the lead contact upon request.
by H2O2 and NO donors.56,63 It is plausible that these cysteine
residues of BACH1 could also serve as sensors for a variety of ACKNOWLEDGMENTS
pro-oxidants and possibly affect the function and stability of
the transcriptional repressor via unknown mechanisms.64,65 The authors would like to thank R. Yan, X. Zhao, J. Jung, and Z. Yu at the Cryo-
Beyond its role in the oxidative stress response pathway, EM Facility on the Janelia Research Campus of the Howard Hughes Medical
BACH1 also targets genes involved in cell cycle control, apoptosis, Institute and J.D. Quispe and S. Dickinson at the Arnold and Mabel Beckman
Cryo-EM Center at the University of Washington for their assistance in electron
and metabolism.16,47,66 By positively regulating the expression of
microscopy data acquisition. The authors thank Mengxi Liu for her contribution
multiple pro-metastatic genes, BACH1 has been shown to pro- to this work. We also thank the NYU Proteomics Lab (supported in part by the
mote cancer cell invasiveness.67–69 Our studies suggest that com- NYU School of Medicine and the Laura and Isaac Perlmutter Cancer Center
pounds that could modify the BTB or DNA-binding domains of Support grant P30CA016087 from the National Cancer Institute). In addition,
BACH1 have the potential to promote the ubiquitination and the mass spectrometric experiments were supported with a shared instrumen-
degradation of BACH1 by FBXL17 or FBXO22, thereby amelio- tation grant from the NIH (1S10OD010582-01A1 for the purchase of an Orbi-
trap Eclipse) for S-nitrosylation site-mapping experiments. The authors also
rating BACH1-mediated cancer metastasis. Similar strategies to
thank T.R. Hinds for help on BLI data analysis and members of the Zheng
reshape or re-surface domains and complexes of disease-pro- Lab and Pagano Lab for discussion. N.Z. and M.P. are Howard Hughes Med-
moting target proteins with small molecules hold the promise to ical Institute Investigators. This work is also supported by NIH grant
create ‘‘neo’’ tertiary or quaternary structure degrons for targeted GM136250 to M.P. and NIH NIGMS grant S10OD030237 and the National Sci-
protein degradation as an emerging therapeutic modality.70,71 ence Foundation award 2304707 to M.G.
AUTHOR CONTRIBUTIONS
Limitations of the study
The affinity of the central NCOR1 fragment toward the BACH1- S.C., S.F.G., M.P., and N.Z. conceived the project. S.C. constructed and pu-
BTB domain is much lower than that of FBXO22, raising the ques- rified all protein samples in this study with inputs from S.F.G. and H.M. S.C.
tion as to how the transcriptional co-repressor could compete performed BLI, AlphaScreen, EMSA, DSF, and in vitro pull-down and ubiquiti-
with the E3 ubiquitin ligase before BACH1 is released from chro- nation experiments and prepared the sample for cryo-EM analysis. S.F.G.
generated CRISPR KO cell lines, designed plasmid constructs, and performed
matin. We hypothesize that binding of NCOR1 to BACH1 might
all of the cell-based co-immunoprecipitations and protein stability assays, with
be reinforced by multivalent interactions between other subunits notable contributions from Y.K., H.V.G., S.D., S.K., G.R., and L.L. J.P. and B.U.
of the co-repressor complex and chromatin. The nature of these performed the mass spectrometric sample preparation, data acquisition, and
interactions remains to be elucidated. Future studies are also analysis. Huigang Shi performed the majority of cryo-EM grid preparation,
needed to dissect the sub-populations of BACH1, which might specimen screening, data collection, and processing tasks with early contribu-
be differentially modified by S-nitrosylation at different sites and tions from Hui Shi and S.C. E.I.J. and M.G. designed and performed the HDX-
at different times after induction of oxidative stress. Validation of MS experiments. The manuscript was written with inputs from all authors. The
order of the second and third co-first authors was chosen randomly.
this hypothesis could help explain why, in addition to FBXL17,
FBXO22 also plays a role in promoting the degradation of DECLARATION OF INTERESTS
BACH1 upon NO treatment, despite its inability to bind the
S-nitrosylated BACH1 BTB domain. Finally, future structural N.Z. and M.P. are scientific cofounders of SEED Therapeutics with financial in-
studies might reveal additional intermediate states of BACH1- terests. They also received research funding from and are shareholders in Ky-
BTB dimer recognized by FBXL17. mera Therapeutics. N.Z. serves on the scientific advisory boards of Synthex,
Molecular Glue Labs, and Differentiated Therapeutics with financial interests.
M.P. is a consultant and/or serves on the scientific advisory boards of CullGen,
RESOURCE AVAILABILITY
Sibylla Biotech, and Triana Biomedicines with financial interests. The findings
presented in this manuscript were not shared with these companies.
Lead contact
Requests for further information and resources should be directed to and will
STAR+METHODS
be fulfilled by the lead contact, Ning Zheng ([email protected]).
Detailed methods are provided in the online version of this paper and include
Materials availability the following:
All unique/stable reagents generated in this study are available upon request. d KEY RESOURCES TABLE
d EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
Data and code availability B Cell lines
d Original western blot images have been deposited at Mendeley at B Source organism
https://dx.doi.org/10.17632/vf73rr8p5n.1 and are publicly available as d METHOD DETAILS
of the date of publication. B Protein preparation
d The atomic coordinates and EM maps of protein complexes have been B Affinity pull-down assay
deposited at the Protein Data Bank (PDB) and the Electron Microscopy B BioLayer interferometry
Data Bank (EMDB) with accession codes listed in the key resources ta- B AlphaLISA
ble. They are publicly available as of the date of publication. B SEC–MALS
d HDX-MS data have been deposited at ProteomeXchange as PXD053923. B Differential scanning fluorimetry (DSF)
Proteomics data have been deposited at ProteomeXchange as B In vitro ubiquitination assay
PXD056513. They are publicly available as of the date of publication. B Electrophoretic mobility shift assay (EMSA)
d This paper does not report original code. B Cryo-EM sample preparation and data collection
ll
Article OPEN ACCESS
B Image processing and 3D reconstruction 13. Igarashi, K., Nishizawa, H., Saiki, Y., and Matsumoto, M. (2021). The tran-
B Model building and refinement scription factor BACH1 at the crossroads of cancer biology: From epithe-
B Plasmids and antibodies lial-mesenchymal transition to ferroptosis. J. Biol. Chem. 297, 101032.
B Drug Treatment Procedures https://doi.org/10.1016/j.jbc.2021.101032.
B Co-immunoprecipitation assay
14. Oyake, T., Itoh, K., Motohashi, H., Hayashi, N., Hoshino, H., Nishizawa, M.,
B Cell-based protein stability assay
Yamamoto, M., and Igarashi, K. (1996). Bach proteins belong to a novel
B CRISPR-Cas9 genome editing
family of BTB-basic leucine zipper transcription factors that interact with
B Chromatin Fractionation Assay
MafK and regulate transcription through the NF-E2 site. Mol. Cell. Biol.
B Biotin Switch Assay
16, 6083–6095. https://doi.org/10.1128/MCB.16.11.6083.
B S-nitrosylation site mapping
15. Sun, J., Hoshino, H., Takaku, K., Nakajima, O., Muto, A., Suzuki, H., Ta-
B HDX-MS analyses
shiro, S., Takahashi, S., Shibahara, S., Alam, J., et al. (2002). Hemoprotein
d QUANTIFICATION AND STATISTICAL ANALYSIS
Bach1 regulates enhancer availability of heme oxygenase-1 gene. EMBO
J. 21, 5216–5224. https://doi.org/10.1093/emboj/cdf516.
SUPPLEMENTAL INFORMATION
16. Warnatz, H.J., Schmidt, D., Manke, T., Piccini, I., Sultan, M., Borodina, T.,
Supplemental information can be found online at https://doi.org/10.1016/j.cell. Balzereit, D., Wruck, W., Soldatov, A., Vingron, M., et al. (2011). The BTB
2024.10.012. and CNC homology 1 (BACH1) target genes are involved in the oxidative
stress response and in control of the cell cycle. J. Biol. Chem. 286,
23521–23532. https://doi.org/10.1074/jbc.M111.220178.
Received: October 12, 2023
Revised: August 25, 2024 17. Hintze, K.J., Katoh, Y., Igarashi, K., and Theil, E.C. (2007). Bach1 repres-
Accepted: October 10, 2024 sion of ferritin and thioredoxin reductase1 is heme-sensitive in cells and
Published: November 5, 2024 in vitro and coordinates expression with heme oxygenase1, beta-globin,
and NADP(H) quinone (oxido) reductase1. J. Biol. Chem. 282, 34365–
REFERENCES 34371. https://doi.org/10.1074/jbc.M700254200.
18. Dhakshinamoorthy, S., Jain, A.K., Bloom, D.A., and Jaiswal, A.K. (2005).
1. Hershko, A., and Ciechanover, A. (1998). The ubiquitin system. Annu. Rev. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant
Biochem. 67, 425–479. https://doi.org/10.1146/annurev.biochem.67. response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1
1.425. gene expression and induction in response to antioxidants. J. Biol.
2. Zheng, N., and Shabek, N. (2017). Ubiquitin Ligases: Structure, Function, Chem. 280, 16891–16900. https://doi.org/10.1074/jbc.M500166200.
and Regulation. Annu. Rev. Biochem. 86, 129–157. https://doi.org/10. 19. Ogawa, K., Sun, J., Taketani, S., Nakajima, O., Nishitani, C., Sassa, S.,
1146/annurev-biochem-060815-014922. Hayashi, N., Yamamoto, M., Shibahara, S., Fujita, H., and Igarashi, K.
3. Varshavsky, A. (1991). Naming a targeting signal. Cell 64, 13–15. https:// (2001). Heme mediates derepression of Maf recognition element through
doi.org/10.1016/0092-8674(91)90202-A. direct binding to transcription repressor Bach1. EMBO J. 20, 2835–
2843. https://doi.org/10.1093/emboj/20.11.2835.
4. Mészáros, B., Kumar, M., Gibson, T.J., Uyar, B., and Dosztányi, Z. (2017).
Degrons in cancer. Sci. Signal. 10, eaak9982. https://doi.org/10.1126/sci- 20. Reichard, J.F., Sartor, M.A., and Puga, A. (2008). BACH1 is a specific
signal.aak9982. repressor of HMOX1 that is inactivated by arsenite. J. Biol. Chem. 283,
5. Zhang, Z., Sie, B., Chang, A., Leng, Y., Nardone, C., Timms, R.T., and El- 22363–22370. https://doi.org/10.1074/jbc.M801784200.
ledge, S.J. (2023). Elucidation of E3 ubiquitin ligase specificity through 21. Reichard, J.F., Motz, G.T., and Puga, A. (2007). Heme oxygenase-1 induc-
proteome-wide internal degron mapping. Mol. Cell 83, 3377–3392.e6. tion by NRF2 requires inactivation of the transcriptional repressor BACH1.
https://doi.org/10.1016/j.molcel.2023.08.022. Nucleic Acids Res. 35, 7074–7086. https://doi.org/10.1093/nar/gkm638.
6. Harper, J.W., and Schulman, B.A. (2021). Cullin-RING Ubiquitin Ligase 22. Namgaladze, D., Fuhrmann, D.C., and Brüne, B. (2022). Interplay of Nrf2
Regulatory Circuits: A Quarter Century Beyond the F-Box Hypothesis. and BACH1 in inducing ferroportin expression and enhancing resistance
Annu. Rev. Biochem. 90, 403–429. https://doi.org/10.1146/annurev-bio- of human macrophages towards ferroptosis. Cell Death Discov. 8, 327.
chem-090120-013613. https://doi.org/10.1038/s41420-022-01117-y.
7. Welcker, M., and Clurman, B.E. (2008). FBW7 ubiquitin ligase: a tumour 23. Baird, L., and Yamamoto, M. (2020). The Molecular Mechanisms Regu-
suppressor at the crossroads of cell division, growth and differentiation. lating the KEAP1-NRF2 Pathway. Mol. Cell. Biol. 40, e00099-20. https://
Nat. Rev. Cancer 8, 83–93. https://doi.org/10.1038/nrc2290. doi.org/10.1128/MCB.00099-20.
8. Bi, Y., Cui, D., Xiong, X., and Zhao, Y. (2021). The characteristics and roles 24. Itoh, K., Wakabayashi, N., Katoh, Y., Ishii, T., O’Connor, T., and Yama-
of b-TrCP1/2 in carcinogenesis. FEBS Journal 288, 3351–3374. https:// moto, M. (2003). Keap1 regulates both cytoplasmic-nuclear shuttling
doi.org/10.1111/febs.15585. and degradation of Nrf2 in response to electrophiles. Genes Cells 8,
9. Koren, I., Timms, R.T., Kula, T., Xu, Q., Li, M.Z., and Elledge, S.J. (2018). 379–391. https://doi.org/10.1046/j.1365-2443.2003.00640.x.
The Eukaryotic Proteome Is Shaped by E3 Ubiquitin Ligases Targeting 25. Kobayashi, A., Kang, M.I., Okawa, H., Ohtsuji, M., Zenke, Y., Chiba, T.,
C-Terminal Degrons. Cell 173, 1622–1635.e14. https://doi.org/10.1016/j. Igarashi, K., and Yamamoto, M. (2004). Oxidative stress sensor Keap1
cell.2018.04.028. functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal
10. Lin, H.C., Yeh, C.W., Chen, Y.F., Lee, T.T., Hsieh, P.Y., Rusnac, D.V., Lin, degradation of Nrf2. Mol. Cell. Biol. 24, 7130–7139. https://doi.org/10.
S.Y., Elledge, S.J., Zheng, N., and Yen, H.S. (2018). C-Terminal End- 1128/MCB.24.16.7130-7139.2004.
Directed Protein Elimination by CRL2 Ubiquitin Ligases. Mol. Cell 70, 26. Tong, K.I., Katoh, Y., Kusunoki, H., Itoh, K., Tanaka, T., and Yamamoto, M.
602–613.e603. https://doi.org/10.1016/j.molcel.2018.04.006. (2006). Keap1 recruits Neh2 through binding to ETGE and DLG motifs:
11. Hayes, J.D., Dinkova-Kostova, A.T., and Tew, K.D. (2020). Oxidative characterization of the two-site molecular recognition model. Mol. Cell.
Stress in Cancer. Cancer Cell 38, 167–197. https://doi.org/10.1016/j. Biol. 26, 2887–2900. https://doi.org/10.1128/MCB.26.8.2887-2900.2006.
ccell.2020.06.001. 27. McMahon, M., Lamont, D.J., Beattie, K.A., and Hayes, J.D. (2010). Keap1
12. Forman, H.J., and Zhang, H. (2021). Targeting oxidative stress in disease: perceives stress via three sensors for the endogenous signaling molecules
promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 20, nitric oxide, zinc, and alkenals. Proc. Natl. Acad. Sci. USA 107, 18838–
689–709. https://doi.org/10.1038/s41573-021-00233-1. 18843. https://doi.org/10.1073/pnas.1007387107.
ll
OPEN ACCESS Article
28. Kobayashi, A., Kang, M.I., Watai, Y., Tong, K.I., Shibata, T., Uchida, K., Mol. Cell. Biol. 22, 1804–1818. https://doi.org/10.1128/MCB.22.6.1804-
and Yamamoto, M. (2006). Oxidative and electrophilic stresses activate 1818.2002.
Nrf2 through inhibition of ubiquitination activity of Keap1. Mol. Cell. Biol. 44. Huynh, K.D., and Bardwell, V.J. (1998). The BCL-6 POZ domain and other
26, 221–229. https://doi.org/10.1128/MCB.26.1.221-229.2006. POZ domains interact with the co-repressors N-CoR and SMRT. Onco-
29. Tan, M.K., Lim, H.J., Bennett, E.J., Shi, Y., and Harper, J.W. (2013). Paral- gene 17, 2473–2484. https://doi.org/10.1038/sj.onc.1202197.
lel SCF adaptor capture proteomics reveals a role for SCFFBXL17 in NRF2 45. Ahmad, K.F., Melnick, A., Lax, S., Bouchard, D., Liu, J., Kiang, C.L., Mayer,
activation via BACH1 repressor turnover. Mol. Cell 52, 9–24. https://doi. S., Takahashi, S., Licht, J.D., and Privé, G.G. (2003). Mechanism of SMRT
org/10.1016/j.molcel.2013.08.018. corepressor recruitment by the BCL6 BTB domain. Mol. Cell 12, 1551–
30. Lignitto, L., LeBoeuf, S.E., Homer, H., Jiang, S., Askenazi, M., Karakousi, 1564. https://doi.org/10.1016/S1097-2765(03)00454-4.
T.R., Pass, H.I., Bhutkar, A.J., Tsirigos, A., Ueberheide, B., et al. (2019). 46. Ghetu, A.F., Corcoran, C.M., Cerchietti, L., Bardwell, V.J., Melnick, A., and
Nrf2 Activation Promotes Lung Cancer Metastasis by Inhibiting the Degra- Privé, G.G. (2008). Structure of a BCOR corepressor peptide in complex
dation of Bach1. Cell 178, 316–329.e18. https://doi.org/10.1016/j.cell. with the BCL6 BTB domain dimer. Mol. Cell 29, 384–391. https://doi.
2019.06.003. org/10.1016/j.molcel.2007.12.026.
31. Zheng, N., Schulman, B.A., Song, L., Miller, J.J., Jeffrey, P.D., Wang, P., 47. Dohi, Y., Ikura, T., Hoshikawa, Y., Katoh, Y., Ota, K., Nakanome, A., Muto,
Chu, C., Koepp, D.M., Elledge, S.J., Pagano, M., et al. (2002). Structure A., Omura, S., Ohta, T., Ito, A., et al. (2008). Bach1 inhibits oxidative stress-
of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature induced cellular senescence by impeding p53 function on chromatin. Nat.
416, 703–709. https://doi.org/10.1038/416703a. Struct. Mol. Biol. 15, 1246–1254. https://doi.org/10.1038/nsmb.1516.
32. Borziak, K., and Zhulin, I.B. (2007). FIST: a sensory domain for diverse 48. Mena, E.L., Jevtic, P., Greber, B.J., Gee, C.L., Lew, B.G., Akopian, D., No-
signal transduction pathways in prokaryotes and ubiquitin signaling in eu- gales, E., Kuriyan, J., and Rape, M. (2020). Structural basis for dimerization
karyotes. Bioinformatics 23, 2518–2521. https://doi.org/10.1093/bioinfor- quality control. Nature 586, 452–456. https://doi.org/10.1038/s41586-
matics/btm384. 020-2636-7.
33. Sinha, S., Rappu, P., Lange, S.C., Mäntsälä, P., Zalkin, H., and Smith, J.L. 49. Mena, E.L., Kjolby, R.A.S., Saxton, R.A., Werner, A., Lew, B.G., Boyle,
(1999). Crystal structure of Bacillus subtilis YabJ, a purine regulatory pro- J.M., Harland, R., and Rape, M. (2018). Dimerization quality control en-
tein and member of the highly conserved YjgF family. Proc. Natl. Acad. Sci. sures neuronal development and survival. Science 362, eaap8236.
USA 96, 13074–13079. https://doi.org/10.1073/pnas.96.23.13074. https://doi.org/10.1126/science.aap8236.
34. Volz, K. (1999). A test case for structure-based functional assignment: the 50. Hess, D.T., Matsumoto, A., Kim, S.O., Marshall, H.E., and Stamler, J.S.
1.2 A crystal structure of the yjgF gene product from Escherichia coli. Pro- (2005). Protein S-nitrosylation: purview and parameters. Nat. Rev. Mol.
tein Sci. 8, 2428–2437. https://doi.org/10.1110/ps.8.11.2428. Cell Biol. 6, 150–166. https://doi.org/10.1038/nrm1569.
35. Burman, J.D., Stevenson, C.E., Sawers, R.G., and Lawson, D.M. (2007). 51. Fernando, V., Zheng, X., Walia, Y., Sharma, V., Letson, J., and Furuta, S.
The crystal structure of Escherichia coli TdcF, a member of the highly (2019). S-Nitrosylation: An Emerging Paradigm of Redox Signaling. Anti-
conserved YjgF/YER057c/UK114 family. BMC Struct. Biol. 7, 30. https:// oxidants 8, 404. https://doi.org/10.3390/antiox8090404.
doi.org/10.1186/1472-6807-7-30.
52. Wolhuter, K., Whitwell, H.J., Switzer, C.H., Burgoyne, J.R., Timms, J.F.,
36. Ito, N., Watanabe-Matsui, M., Igarashi, K., and Murayama, K. (2009). Crys- and Eaton, P. (2018). Evidence against Stable Protein S-Nitrosylation as
tal structure of the Bach1 BTB domain and its regulation of homodimeriza- a Widespread Mechanism of Post-translational Regulation. Mol. Cell 69,
tion. Genes Cells 14, 167–178. https://doi.org/10.1111/j.1365-2443.2008. 438–450.e5. https://doi.org/10.1016/j.molcel.2017.12.019.
01259.x.
53. Tsuneyoshi, T., Kunimura, K., and Morihara, N. (2019). S-1-Propenylcys-
37. Bonchuk, A., Balagurov, K., and Georgiev, P. (2023). BTB domains: A teine augments BACH1 degradation and heme oxygenase 1 expression
structural view of evolution, multimerization, and protein-protein interac- in a nitric oxide-dependent manner in endothelial cells. Nitric Oxide 84,
tions. BioEssays 45, e2200179. https://doi.org/10.1002/bies.202200179. 22–29. https://doi.org/10.1016/j.niox.2019.01.003.
38. Muto, A., Hoshino, H., Madisen, L., Yanai, N., Obinata, M., Karasuyama, 54. Fourquet, S., Guerois, R., Biard, D., and Toledano, M.B. (2010). Activation of
H., Hayashi, N., Nakauchi, H., Yamamoto, M., Groudine, M., and Igarashi, NRF2 by nitrosative agents and H2O2 involves KEAP1 disulfide formation.
K. (1998). Identification of Bach2 as a B-cell-specific partner for small maf J. Biol. Chem. 285, 8463–8471. https://doi.org/10.1074/jbc.M109.051714.
proteins that negatively regulate the immunoglobulin heavy chain gene 30 55. Li, C.Q., Kim, M.Y., Godoy, L.C., Thiantanawat, A., Trudel, L.J., and Wo-
enhancer. EMBO J. 17, 5734–5743. https://doi.org/10.1093/emboj/17. gan, G.N. (2009). Nitric oxide activation of Keap1/Nrf2 signaling in human
19.5734. colon carcinoma cells. Proc. Natl. Acad. Sci. USA 106, 14547–14551.
39. Muto, A., Tashiro, S., Nakajima, O., Hoshino, H., Takahashi, S., Sakoda, https://doi.org/10.1073/pnas.0907539106.
E., Ikebe, D., Yamamoto, M., and Igarashi, K. (2004). The transcriptional 56. Mnatsakanyan, R., Markoutsa, S., Walbrunn, K., Roos, A., Verhelst,
programme of antibody class switching involves the repressor Bach2. Na- S.H.L., and Zahedi, R.P. (2019). Proteome-wide detection of
ture 429, 566–571. https://doi.org/10.1038/nature02596. S-nitrosylation targets and motifs using bioorthogonal cleavable-linker-
40. Roychoudhuri, R., Hirahara, K., Mousavi, K., Clever, D., Klebanoff, C.A., based enrichment and switch technique. Nat. Commun. 10, 2195.
Bonelli, M., Sciumè, G., Zare, H., Vahedi, G., Dema, B., et al. (2013). https://doi.org/10.1038/s41467-019-10182-4.
BACH2 represses effector programs to stabilize T(reg)-mediated immune 57. Xing, W., Busino, L., Hinds, T.R., Marionni, S.T., Saifee, N.H., Bush, M.F.,
homeostasis. Nature 498, 506–510. https://doi.org/10.1038/nature12199. Pagano, M., and Zheng, N. (2013). SCF(FBXL3) ubiquitin ligase targets
41. Rosbrook, G.O., Stead, M.A., Carr, S.B., and Wright, S.C. (2012). The cryptochromes at their cofactor pocket. Nature 496, 64–68. https://doi.
structure of the Bach2 POZ-domain dimer reveals an intersubunit disulfide org/10.1038/nature11964.
bond. Acta Crystallogr. D Biol. Crystallogr. 68, 26–34. https://doi.org/10. 58. Wang, H., Shi, H., Rajan, M., Canarie, E.R., Hong, S., Simoneschi, D., Pa-
1107/S0907444911048335. gano, M., Bush, M.F., Stoll, S., Leibold, E.A., et al. (2020). FBXL5 Regu-
42. Cao, S., Kang, S., Mao, H., Yao, J., Gu, L., and Zheng, N. (2022). Defining lates IRP2 Stability in Iron Homeostasis via an Oxygen-Responsive
molecular glues with a dual-nanobody cannabidiol sensor. Nat. Commun. [2Fe2S] Cluster. Mol. Cell 78, 31-41.e35. https://doi.org/10.1016/j.mol-
13, 815. https://doi.org/10.1038/s41467-022-28507-1. cel.2020.02.011.
43. Melnick, A., Carlile, G., Ahmad, K.F., Kiang, C.L., Corcoran, C., Bardwell, 59. Lumpkin, R.J., Baker, R.W., Leschziner, A.E., and Komives, E.A. (2020).
V., Prive, G.G., and Licht, J.D. (2002). Critical residues within the BTB Structure and dynamics of the ASB9 CUL-RING E3 Ligase. Nat. Commun.
domain of PLZF and Bcl-6 modulate interaction with corepressors. 11, 2866. https://doi.org/10.1038/s41467-020-16499-9.
ll
Article OPEN ACCESS
60. Sherpa, D., Chrustowicz, J., Qiao, S., Langlois, C.R., Hehl, L.A., Gotte- 75. Emsley, P., Lohkamp, B., Scott, W.G., and Cowtan, K. (2010). Features
mukkala, K.V., Hansen, F.M., Karayel, O., von Gronau, S., Prabu, J.R., and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66,
et al. (2021). GID E3 ligase supramolecular chelate assembly configures 486–501. https://doi.org/10.1107/S0907444910007493.
multipronged ubiquitin targeting of an oligomeric metabolic enzyme. 76. Adams, P.D., Grosse-Kunstleve, R.W., Hung, L.W., Ioerger, T.R., McCoy,
Mol. Cell 81, 2445–2459.e13. https://doi.org/10.1016/j.molcel.2021. A.J., Moriarty, N.W., Read, R.J., Sacchettini, J.C., Sauter, N.K., and Ter-
03.025. williger, T.C. (2002). PHENIX: building new software for automated crystal-
61. Teng, F., Wang, Y., Liu, M., Tian, S., Stjepanovic, G., and Su, M.Y. (2023). lographic structure determination. Acta Crystallogr. D Biol. Crystallogr. 58,
Cryo-EM structure of the KLHL22 E3 ligase bound to an oligomeric meta- 1948–1954. https://doi.org/10.1107/S0907444902016657.
bolic enzyme. Structure 31, 1431–1440.e5. https://doi.org/10.1016/j.str.
77. Afonine, P.V., Poon, B.K., Read, R.J., Sobolev, O.V., Terwilliger, T.C., Urz-
2023.09.002.
humtsev, A., and Adams, P.D. (2018). Real-space refinement in PHENIX
62. Cheng, J., Lin, M., Chu, M., Gong, L., Bi, Y., and Zhao, Y. (2020). Emerging for cryo-EM and crystallography. Acta Crystallogr. D Struct. Biol. 74,
role of FBXO22 in carcinogenesis. Cell Death Discov. 6, 66. https://doi. 531–544. https://doi.org/10.1107/S2059798318006551.
org/10.1038/s41420-020-00303-0.
78. Guttman, M., Weis, D.D., Engen, J.R., and Lee, K.K. (2013). Analysis of
63. Zhang, J., Simpson, C.M., Berner, J., Chong, H.B., Fang, J., Ordulu, Z., overlapped and noisy hydrogen/deuterium exchange mass spectra.
Weiss-Sadan, T., Possemato, A.P., Harry, S., Takahashi, M., et al. J. Am. Soc. Mass Spectrom. 24, 1906–1912. https://doi.org/10.1007/
(2023). Systematic identification of anticancer drug targets reveals a nu- s13361-013-0727-5.
cleus-to-mitochondria ROS-sensing pathway. Cell 186, 2361–2379.e25.
https://doi.org/10.1016/j.cell.2023.04.026. 79. Suloway, C., Pulokas, J., Fellmann, D., Cheng, A., Guerra, F., Quispe, J.,
Stagg, S., Potter, C.S., and Carragher, B. (2005). Automated molecular mi-
64. Zenke-Kawasaki, Y., Dohi, Y., Katoh, Y., Ikura, T., Ikura, M., Asahara, T.,
croscopy: the new Leginon system. J. Struct. Biol. 151, 41–60. https://doi.
Tokunaga, F., Iwai, K., and Igarashi, K. (2007). Heme induces ubiquitina-
org/10.1016/j.jsb.2005.03.010.
tion and degradation of the transcription factor Bach1. Mol. Cell. Biol.
27, 6962–6971. https://doi.org/10.1128/MCB.02415-06. 80. Mastronarde, D.N. (2005). Automated electron microscope tomography
using robust prediction of specimen movements. J. Struct. Biol. 152,
65. Hira, S., Tomita, T., Matsui, T., Igarashi, K., and Ikeda-Saito, M. (2007).
36–51. https://doi.org/10.1016/j.jsb.2005.07.007.
Bach1, a heme-dependent transcription factor, reveals presence of multi-
ple heme binding sites with distinct coordination structure. IUBMB Life 59, 81. Wu, C., Huang, X., Cheng, J., Zhu, D., and Zhang, X. (2019). High-quality,
542–551. https://doi.org/10.1080/15216540701225941. high-throughput cryo-electron microscopy data collection via beam tilt
66. Matsumoto, M., Kondo, K., Shiraki, T., Brydun, A., Funayama, R., Na- and astigmatism-free beam-image shift. J. Struct. Biol. 208, 107396.
kayama, K., Yaegashi, N., Katagiri, H., and Igarashi, K. (2016). Genome- https://doi.org/10.1016/j.jsb.2019.09.013.
wide approaches for BACH1 target genes in mouse embryonic fibroblasts 82. Guo, H., Franken, E., Deng, Y., Benlekbir, S., Singla Lezcano, G., Janssen,
showed BACH1-Pparg pathway in adipogenesis. Genes Cells 21, B., Yu, L., Ripstein, Z.A., Tan, Y.Z., and Rubinstein, J.L. (2020). Electron-
553–567. https://doi.org/10.1111/gtc.12365. event representation data enable efficient cryoEM file storage with full
67. Wiel, C., Le Gal, K., Ibrahim, M.X., Jahangir, C.A., Kashif, M., Yao, H., Zie- preservation of spatial and temporal resolution. IUCrJ 7, 860–869.
gler, D.V., Xu, X., Ghosh, T., Mondal, T., et al. (2019). BACH1 Stabilization https://doi.org/10.1107/S205225252000929X.
by Antioxidants Stimulates Lung Cancer Metastasis. Cell 178, 330– 83. Punjani, A., Rubinstein, J.L., Fleet, D.J., and Brubaker, M.A. (2017). cryo-
345.e22. https://doi.org/10.1016/j.cell.2019.06.005. SPARC: algorithms for rapid unsupervised cryo-EM structure determina-
68. Liang, Y., Wu, H., Lei, R., Chong, R.A., Wei, Y., Lu, X., Tagkopoulos, I., tion. Nat. Methods 14, 290–296. https://doi.org/10.1038/nmeth.4169.
Kung, S.Y., Yang, Q., Hu, G., and Kang, Y. (2012). Transcriptional network 84. Zhang, K. (2016). Gctf: real-time CTF determination and correction.
analysis identifies BACH1 as a master regulator of breast cancer bone J. Struct. Biol. 193, 1–12. https://doi.org/10.1016/j.jsb.2015.11.003.
metastasis. J. Biol. Chem. 287, 33533–33544. https://doi.org/10.1074/
85. Bepler, T., Morin, A., Rapp, M., Brasch, J., Shapiro, L., Noble, A.J., and
jbc.M112.392332.
Berger, B. (2019). Positive-unlabeled convolutional neural networks for
69. Yun, J., Frankenberger, C.A., Kuo, W.L., Boelens, M.C., Eves, E.M.,
particle picking in cryo-electron micrographs. Nat. Methods 16, 1153–
Cheng, N., Liang, H., Li, W.H., Ishwaran, H., Minn, A.J., and Rosner,
1160. https://doi.org/10.1038/s41592-019-0575-8.
M.R. (2011). Signalling pathway for RKIP and Let-7 regulates and predicts
metastatic breast cancer. EMBO J. 30, 4500–4514. https://doi.org/10. 86. Punjani, A., Zhang, H., and Fleet, D.J. (2020). Non-uniform refinement:
1038/emboj.2011.312. adaptive regularization improves single-particle cryo-EM reconstruction.
Nat. Methods 17, 1214–1221. https://doi.org/10.1038/s41592-020-
70. Verma, R., Mohl, D., and Deshaies, R.J. (2020). Harnessing the Power of
00990-8.
Proteolysis for Targeted Protein Inactivation. Mol. Cell 77, 446–460.
https://doi.org/10.1016/j.molcel.2020.01.010. 87. Punjani, A., and Fleet, D.J. (2021). 3D variability analysis: Resolving contin-
71. Teng, M., and Gray, N.S. (2023). The rise of degrader drugs. Cell Chem. uous flexibility and discrete heterogeneity from single particle cryo-EM.
Biol. 30, 864–878. https://doi.org/10.1016/j.chembiol.2023.06.020. J. Struct. Biol. 213, 107702. https://doi.org/10.1016/j.jsb.2021.107702.
72. Zheng, S.Q., Palovcak, E., Armache, J.P., Verba, K.A., Cheng, Y., and 88. Rohou, A., and Grigorieff, N. (2015). CTFFIND4: Fast and accurate defocus
Agard, D.A. (2017). MotionCor2: anisotropic correction of beam-induced estimation from electron micrographs. J. Struct. Biol. 192, 216–221.
motion for improved cryo-electron microscopy. Nat. Methods 14, https://doi.org/10.1016/j.jsb.2015.08.008.
331–332. https://doi.org/10.1038/nmeth.4193. 89. Murphree, T.A., Vorauer, C., Brzoska, M., and Guttman, M. (2020). Imida-
73. Mirdita, M., Schütze, K., Moriwaki, Y., Heo, L., Ovchinnikov, S., and Stei- zolium Compounds as Internal Exchange Reporters for Hydrogen/
negger, M. (2022). ColabFold: making protein folding accessible to all. Nat. Deuterium Exchange by Mass Spectrometry. Anal. Chem. 92, 9830–
Methods 19, 679–682. https://doi.org/10.1038/s41592-022-01488-1. 9837. https://doi.org/10.1021/acs.analchem.0c01328.
74. Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt, 90. Watson, M.J., Harkewicz, R., Hodge, E.A., Vorauer, C., Palmer, J., Lee,
D.M., Meng, E.C., and Ferrin, T.E. (2004). UCSF Chimera–a visualization K.K., and Guttman, M. (2021). Simple Platform for Automating Decoupled
system for exploratory research and analysis. J. Comput. Chem. 25, LC-MS Analysis of Hydrogen/Deuterium Exchange Samples. J. Am. Soc.
1605–1612. https://doi.org/10.1002/jcc.20084. Mass Spectrom. 32, 597–600. https://doi.org/10.1021/jasms.0c00341.
ll
OPEN ACCESS Article
STAR+METHODS
ll
Article OPEN ACCESS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Dimeric FBXL17LRR-BACH1-BTB structure This paper PDB: 8UBV
EM map of FBXO22-BACH1-BTB This paper EMDB: EMD-42049
EM map of SCFFBXO22-BACH1-BTB This paper EMDB: EMD-42051
EM map of monomeric FBXL17-BACH1-BTB This paper EMDB: EMD-42064
EM map of monomeric SCFFBXL17-BACH1-BTB This paper EMDB: EMD-42102
EM map of dimeric SCFFBXL17-BACH1-BTB This paper EMDB: EMD-42105
close conformation
EM map of dimeric FBXL17LRR-BACH1-BTB This paper EMDB: EMD-42106
EM map of dimeric SCFFBXL17-BACH1-BTB open conformation This paper EMDB: EMD-42115
HDX-MS data This paper ProteomeXchange: PXD053923
Original images of western blot data This study; Mendeley Data https://dx.doi.org/10.17632/vf73rr8p5n.1
Proteomic data This study ProteomeXchange: PXD056513
Experimental models: Cell lines
HEK293T (female) ATCC Cat# CRL-3216; RRID: CVCL_0063
HCT-116 (male) ATCC Cat# CCL-247; RRID: CVCL_0291
FBXO22 -/- HCT-116 (male) This paper N/A
FBXL17 -/- HCT-116 (male) This paper N/A
FBXO22 -/-; FBXL17 -/- HCT-116 (male) This paper N/A
Spodoptera frugiperda Sf9 Life Technologies Cat# B825-01
Trichoplusia ni Hi5 Life Technologies Cat# B85502
Oligonucleotides
Full list of oligos See Table S2 N/A
Recombinant DNA
pcDNA3.1 2xFLAG-2xSTREP-BACH1 Lignitto et al.30 N/A
pcDNA3.1 HA-BACH1 This paper N/A
pcDNA3.1 2xFLAG-2xSTREP-BACH2 This paper N/A
pcDNA3.1 2xFLAG-2xSTREP-FBXO22 Lignitto et al.30 N/A
pcDNA3.1 HA-FBXO22 This paper N/A
pcDNA3.1 FLAG-FBXL17 This paper N/A
pcDNA3.1 HA-FBXL17 This paper N/A
pSpCas9(BB)-2A-GFP (px458) Addgene Plasmid# 48138
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (D366A) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (D366R) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (I368A) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (N372A) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (F373D) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (K377A) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (K377D) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (L375A) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP FBXO22 (L375D) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP-BACH1 (F9Y) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP-BACH1 (a6-BACH2) This paper N/A
pcDNA3.1 2xFLAG-2xSTREP-BACH2 (a6-BACH1) This paper N/A
pcDNA3.1 HA-BACH1 (E12A) This paper N/A
pcDNA3.1 HA-BACH1 (C122E) This paper N/A
pcDNA3.1 HA-BACH1 (F125A) This paper N/A
pcDNA3.1 HA-BACH1 (F125D) This paper N/A
pcDNA3.1 HA-BACH1 (F128A) This paper N/A
(Continued on next page)
ll
OPEN ACCESS Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
pcDNA3.1 HA-BACH1 (F128D) This paper N/A
pcDNA3.1 HA-BACH1 BTB (K129*), *: stop codon This paper N/A
pcDNA3.1 HA-FBXL17 DCTH (T675*), *: stop codon This paper N/A
pFB-GST-FBXO22 This paper N/A
pFB-His-SKP1 This paper N/A
pACE-His-MBP-FBXL17 This paper N/A
pAL-His-BACH1-BTB This paper N/A
pET-GST-BACH1-BTB This paper N/A
pET-GST-Avi-BACH1-BTB This paper N/A
pAL-His-Avi-BACH1-BTB This paper N/A
pAL-His-Avi-BACH1-BTB (C34A) This paper N/A
pAL-His-Avi-BACH1-BTB (C55A) This paper N/A
pAL-His-Avi-BACH1-BTB (C107A) This paper N/A
pAL-His-Avi-BACH1-BTB (C109A) This paper N/A
pAL-His-Avi-BACH1-BTB (C122A) This paper N/A
pAL-His-Avi-BACH1-BTB (C122E) This paper N/A
pET-His-Avi-BACH1-BTB This paper N/A
pET-His-Avi-BACH1-BTB (S17C/V20I/V32I/S50A/H60W/S61Q) This paper N/A
pAL-MBP-BACH1-BTB This paper N/A
pAL-MBP-BACH1-BTB (S17C/V20I/V32I/S50A/H60W/S61Q) This paper N/A
pET-GST-Avi-BACH2-BTB This paper N/A
pET-GST-BACH2-BTB This paper N/A
pAL-His-BACH2-BTB This paper N/A
pAL-MBP-BACH2-BTB This paper N/A
pET-MSB-BACH1-BTB This paper N/A
pACE-His-Venus-BACH1-FL This paper N/A
pET-MSB-FLAG-BACH1-BTB This paper N/A
pET-GST-MAF This paper N/A
pAL-His-MBP-BACH1-bZIP This paper N/A
pET-GST-NCOR1 This paper N/A
pET-GST-GLMN This paper N/A
pET-His-RBX1-His-CUL1 This paper N/A
Software and algorithms
ImageJ Software 2.0 NIH https://imagej.net/software/imagej2/
GraphPad Prism v9.5.1 Graphpad Software https://www.graphpad.com
MotionCor2 Zheng et al.72 https://emcore.ucsf.edu/ucsf-software
AlphaFold-Multimer in Google ColabFold Mirdita et al.73 https://colab.research.google.com/
github/sokrypton/ColabFold/blob/
main/AlphaFold2.ipynb
UCSF Chimera Pettersen et al.74 https://www.cgl.ucsf.edu/chimera/
CryoSPARC Structura Biotechnology Inc. https://cryosparc.com
75
Coot Emsley et al. https://www2.mrc-lmb.cam.ac.uk/
personal/pemsley/coot/
PHENIX Adams et al.76 https://phenix-online.org
Afonine et al.77
PyMOL Schrödinger https://www.pymol.org
HDExaminer v3 Sierra Analytics https://massspec.com/hdexaminer/
HXExpress v2 Guttman et al.78 https://www.hxms.com/HXExpress/
(Continued on next page)
ll
Article OPEN ACCESS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Other
HiTrap Q HP GE Healthcare Cat# 17115401
Superdex 200 Increase 10/300 GL GE Healthcare Cat# 28990944
Quantifoil R1.2/1.3 200 mesh copper grid Electron Microscopy Sciences Cat# Q250-CR1.3
Cell lines
Cell lines were purchased from ATCC. HCT-116 (male, ATCC CCL-247) cells were propagated in McCoy’s 5A medium (Gibco).
HEK293T (female, ATCC CRL-3216) cells were propagated in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco). Tissue culture
media were supplemented with 10 % fetal bovine serum (FBS) (Corning Life Sciences) and 1 % penicillin-streptomycin (Corning Life
Sciences). Cells were maintained at 37 C and 5 % CO2 in a humidified atmosphere. Cells were routinely monitored for Mycoplasma
contamination using the MycoStrip - Mycoplasma Detection Kit (Invivogen). No cell lines used in this study were found in the data-
base of commonly misidentified cell lines that is maintained by ICLAC and NCBI Biosample. Specific details about cell lines used are
provided in the key resources table.
Source organism
For DNA extraction, E. coli DH5a, NEB5a, and NEB stable was used. For bacmid production, E. coli DH10Bac was used. For bacu-
lovirus production and amplification, Sf9 insect cells were used. For protein expression, both E. coli BL21 (DE3) and Hi5 insect cells
were used. E. coli grew at 37 C in Luria-Bertani medium (Fisher). Protein expression was induced by 0.2-0.4 mM Isopropyl b-D-1-
thiogalactopyranoside (IPTG) at 16 C overnight. Insect cells grew at 27 C in Grace’s Insect Medium (Gibco) supplemented with
10 % fetal bovine serum (Cytiva) and 1 % penicillin-streptomycin-glutamine (Gibco).
METHOD DETAILS
Protein preparation
Human FBXO22 (N-terminally GST-tagged) and SKP1 (N-terminally His-tagged) were co-expressed and purified from Hi5 insect cells
using glutathione affinity chromatography followed by TEV cleavage to remove the tags if necessary. The tag-free FBXO22-SKP1
complex was further purified by anion exchange and size exclusion chromatography. The wild-type and mutant BACH1-BTB domain
(amino acids 7-128) or BACH2-BTB domain (amino acids 9-131) constructs were expressed with an N-terminal His- or GST-tag in
E. coli BL21. The full-length BACH1 protein was constructed with an N-terminal His-Venus tag and expressed in Hi5 insect cells. His
or GST-tagged BACH1 or BACH2 was purified by Ni-NTA or glutathione resin with subsequent TEV protease treatment to remove the
tags if necessary. BACH1 or BACH2 was further purified by anion exchange and/or size exclusion chromatography. To prepare
biotin-tagged BACH1-BTB or BACH2-BTB, an additional Avi-tag was designed after the TEV cleavage site. After TEV cleavage of
the His- or GST-tag, Avi-BACH1-BTB or Avi-BACH2-BTB was generated and subsequently biotinylated in a biotin ligase BirA-cata-
lyzed biotinylation reaction. Biotinylated protein was further purified by size exclusion chromatography. To make the BACH1-BTB-
BACH2-BTB heterodimer, BACH1-BTB and BACH2-BTB with different tags were co-expressed in E. coli BL21 and isolated by
tandem purification. For example, MBP-tagged BACH2-BTB/MSB-tagged BACH1-BTB were purified by amylose resin (for MBP-
tagged protein) and anion exchange (for MSB-tagged protein, eluted at high salt 450 mM NaCl). In another case, GST-Avi-
BACH1-BTB-His-BACH2-BTB were tandemly purified, biotinylated and TEV treated to generate the biotin-BACH1-BTB-
tag-free BACH2-BTB heterodimer. The FBXL17-binding defective BACH1-BTBMT (S17C/V20I/V32I/S50A/H60W/S61Q) was used
to prepare the BACH1-BTBWT-BACH1-BTBMT heterodimer. MBP-tagged BACH1-BTBMT and His-Avi-tagged BACH1-BTBWT
were co-expressed in E. coli BL21, purified by Ni-NTA resin and subject to biotinylation. Amylose resin was subsequently used to
purify the MBP-BACH1-BTBMT-His-biotin-BACH1-BTBWT heterodimer and remove extra biotin. The MBP- BACH1-BTBWT-His-
biotin-BACH1-BTBWT heterodimer was made by the same method. GST-tagged NCOR1 was expressed in E. coli BL21 and purified
by glutathione resin and size exclusion chromatography. GST-MAF (254-361) and MBP-BACH1-bZIP (510-658) were co-expressed
in E. coli BL21 and subject to tandem affinity purification. GLMN with an N-terminal GST tag was expressed and purified from E. coli
BL21 using glutathione affinity chromatography followed by TEV cleavage to remove the tag. Tag-free GLMN was further purified by
anion exchange and size exclusion chromatography. RBX1 (amino acids 16-108) and CUL1 with two short unstructured segments
(amino acids 1-12 and 58-81) truncated were constructed with an N-terminal His-tag followed by a TEV cleavage site. After co-
expression in E. coli BL21(DE3), the CUL1-RBX1 complex was purified by a Ni-NTA affinity column followed by TEV cleavage to
remove the tag. The tag-free CUL1-RBX1 was further purified by gel filtration. Separately purified BACH1-BTB, FBXO22-SKP1,
CUL1-RBX1, and GLMN were mixed at the molar ratio of 3:1:1:1.5 with BACH1 and GLMN at extra to assemble and purify the
GLMN-SCFFBXO22-BACH1 complex on size exclusion chromatography for cryo-EM analysis. FBXL17 (amino acids 310–701) with an
ll
OPEN ACCESS Article
N-terminal His-MBP-tag and SKP1 with an N-terminal His-tag were co-expressed and purified from Hi5 insect cells using Ni-NTA
affinity chromatography followed by TEV cleavage to remove the tags if necessary. GST-BACH1-BTB without or with NOR3-
S1PC treatment and FBXL17-SKP1 were incubated for 16 hours or at various times, respectively, and purified by a glutathione affinity
column. Thereafter, CUL1-RBX1 and GLMN were loaded on the above column to form the GLMN- SCFFBXL17-BACH1-BTB complex.
After on-column cleavage by TEV, the tag-free complex was collected in the flow-through of the glutathione affinity column and
was further purified by size exclusion chromatography for cryo-EM analysis.
BioLayer interferometry
The binding affinity between FBXO22 (or FBXL17) and BACH1-BTB was measured using the Octet Red 96 (ForteBio, Pall Life Sci-
ences) following the manufacturer’s procedures. The optical probes were coated with streptavidin, loaded with 200 nM biotinylated
BACH1-BTB (without or with NOR3-S1PC treatment), or anti-GST probes loaded with 200 nM GST-FBXO22-SKP1. Subsequently,
the probes were quenched with 0.5 mM biocytin or 1 mM GST protein prior to kinetic binding analysis. The reactions were carried out
in black 96 well plates maintained at 30 C. The reaction volume was 200 mL in each well. The binding buffer contained 25 mM HEPES,
pH 7.4, 100 mM NaCl, 1 mM TCEP, 0.1% Tween-20, and 0.05 mg/mL Bovine Serum Albumin. As the analyte, BACH1-BTB or
FBXL17-SKP1 was tested at various concentrations. There was no binding of the analyte to the unloaded probes. Binding kinetics
of the analyte at different concentrations were measured simultaneously. The data were analyzed by the Octet data analysis soft-
ware. The dissociation constant (Kd) was determined from the steady-state equilibrium measurements. All BLI experiments have
been repeated at least 2-3 times.
AlphaLISA
Amplified luminescent proximity homogeneous assays for measuring protein-protein interactions were performed using EnSpire
reader (PerkinElmer). Biotinylated BACH1-BTB was immobilized to streptavidin-coated AlphaScreen donor beads. His-SKP1-
FBXO22 was bound to anti-His acceptor beads. The acceptor and donor beads were brought into proximity by the interactions be-
tween the E3 and its substrate. Excitation of the donor beads by a laser beam of 680 nm promotes the formation of singlet oxygen.
When an acceptor bead is in close proximity, the singlet oxygen reacts with thioxene derivatives in the acceptor beads and causes
the emission of 520-620 nm photons, which are detected as the binding signal. If the beads are not in close proximity to each other,
the oxygen will return to its ground state and the acceptor beads will not emit light. Competition assays were performed by titrating
GST-tagged NCOR1 fragments or GST alone at various concentrations. To establish the binding signal, the concentration of tagged
E3 and biotinylated BACH1-BTB were first titrated with fixed amount of donor and acceptor beads. The concentration of each
component was selected based on the rising AlphaScreen signal before the beads became saturated. 25 nM His-SKP1-FBXO22
and 8.3 nM biotinylated BACH1-BTB were used. The binding assay buffer contained 25 mM HEPES, pH 7.4, 100 mM NaCl,
1 mM TCEP, 0.1% Tween-20, and 0.05 mg/mL Bovine Serum Albumin. 5 mg/mL donor and acceptor beads were used in the assays.
The experiments were performed in three or four replicates. IC50 was determined using non-linear curve fitting of the dose response
curves generated with Prism (GraphPad).
SEC–MALS
Size-exclusion chromatography coupled with light scattering, refractive index, and ultraviolet absorption (SEC-LS-RI-UV) was done
using a SEC-MALS system consisting of an Agilent HPLC pump with a 1260 Infinity lamp, a Dawn light scattering instrument (Wyatt),
an OptiLab refractive index instrument (Wyatt), and the Superdex 200 Increase 10/300 GL (GE Healthcare). The column was equil-
ibrated with 20 mM Tris-HCl pH 8.0, 150 mM NaCl, and 0.25 mM TCEP. Tag-free FBXO22-SKP1, His-BACH1-BTB and their mixture
were analyzed at room temperature. Similarly, His-MBP-FBXL17-SKP1, NOR3-S1PC-treated His-BACH1-BTB and their mixture
with different incubation times were analyzed.
ll
Article OPEN ACCESS
2.5 mg of each protein was mixed with Protein Thermal Shift Dye for analysis. The temperature increment was 0.3 C, recording
fluorescence from 35 C to 90 C. The data was analyzed by Protein Thermal Shift software 1.4. The melting temperature (Tm) was
determined based on the peak of the first derivative of the fluorescence vs temperature plots. The experiments were performed in
three replicates.
ll
OPEN ACCESS Article
picker and two rounds of 2D classification sequentially. 497,102 out of 3,087,124 particles were picked for heterogeneous refinement
using reconstructions from the previous dataset as templates. 267,523 good particles from the refinement were selected and further
cleaned by 2D classification. 84,154 particles were chosen as training particles for Topaz particle picking. 7,309,475 particles were
picked with Topaz and subjected to two rounds of 2D classification. 1,099,238 particles were selected for ab-initio reconstruction
and heterogeneous refinement sequentially. 364,786 particles from one good class were applied to non-uniform refinement. The
overall resolution of the complex in the monomeric form was refined to 3.1 Å. To improve the local density of FBXL17-BACH1-
BTB, all particles were subjected to 3D Variability. 193,908 particles were selected for local refinement to generate the SKP1-
FBXL17-BACH1-BTB subcomplex with a resolution of 3.2 Å. In addition to the above complex in the monomeric form, the dimeric
GLMN-SCFFBXL17-BACH1-BTB complex was detected as well. 30,123 particles of the dimer were selected as training particles for
Topaz particle picking. 244,189 out of 2,969,728 particles were chosen for heterogeneous refinement. Two good classes represent-
ing different conformations, named dSCFBTB-I and dSCFBTB-II, were kept with 54,546 particles and 47,336 particles, respectively. The
C2 symmetry was imposed for non-uniform refinement. The overall resolution of the two classes reached 4.4 Å and 4.5 Å, respec-
tively. To overcome the pseudo-symmetry of the dimeric reconstructions, all particles of dSCFBTB-I were subjected to symmetry
expansion in CryoSPARC. Afterward, a soft mask focused on the leucine-rich repeat domain of FBXL17 and BACH1-BTB was
applied to subtract the signal from raw particles. These subtracted particles were used for local refinement. The density of the
FBXL17-LRR-BACH1-BTB structure was refined to 4.1 Å. See Figure S7 for more details.
For the compound-treated GLMN-SCFFBXL17-BACH1-BTB sample, the collected data was processed with CryoSPARC in a similar
manner. The data processing workflow was shown in Figure S6. Briefly, 22,720 EER movies were collected and subjected to patch
motion correction and patch CTF estimation sequentially. After filtering with CTF parameters, 20,028 micrographs were kept for the
subsequent blob picker and 2D classification. Representative particles were selected as the training data for Topaz particle picking.
After heterogeneous refinement, a monomeric SCFFBXL17-BACH1 class yielded a 3D reconstruction at 5.7 Å resolution. A dimeric
SCFFBXL17-BACH1 with 461 particles was selected and employed for Topaz particle picking. After 2D classification and heterogeneous
refinement, two good classes of dimeric SCFFBXL17-BACH1 were kept. 10,173 particles and 7,944 particles from dimeric reconstruc-
tions were subjected to non-uniform refinement with C2 symmetry imposed. After refinement, the overall resolution of two classes
both reached 7.8 Å.
Co-immunoprecipitation assay
HEK293T cells were transiently transfected using polyethyleneimine (Polysciences); HCT-116 cells were transiently transfected with
Lipofectamine 3000 (Thermo Fisher Scientific). Where indicated, 48 h after transfection, cells were treated with 2.5 mM MLN-4924 for
4 h before further treatment with either vehicle (DMSO) or the indicated NO donor. Cells were collected by scraping and washed with
ll
Article OPEN ACCESS
PBS prior to lysis in IP lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 5 mM MgCl2, 0.2% NP-40)
supplemented with protease inhibitor (Complete ULTRA, Roche) and phosphatase inhibitor (Phosphatase Inhibitor Cocktail 2,
Sigma-Aldrich) immediately before the experiment. The supernatant was clarified by centrifugation at 21, 000 x g for 10 minutes
at 4 C. For FLAG-tagged proteins, the supernatant was incubated with FLAG-M2 agarose beads (Sigma-Aldrich) for 2h at 4 C.
For HA-tagged proteins, the supernatant was incubated with Pierce Anti-HA magnetic beads (Thermo Fisher Scientific) for 2h at
4 C. The beads were then washed 5 times in lysis buffer before incubating the beads in NuPAGE LDS Sample Buffer (Thermo Fisher
Scientific) supplemented with b-mercaptoethanol (Sigma-Aldrich) at 95 C for 10 minutes. For Western blotting, samples of the whole
cell extract and immunoprecipitates were separated by SDS-PAGE and transferred onto 0.45 mm Immobilon-P PVDF membranes
(Millipore Sigma). Membranes were then blocked in 5% nonfat dried milk / PBST for 30 minutes at room temperature and incubated
with the indicated primary antibodies at 4 C overnight. After washing the membranes, secondary antibodies conjugated with horse-
radish peroxidase were applied. Immunoreactive bands were visualized via application of enhanced chemiluminescence (ECL) re-
agent (Thermo Fisher Scientific). Each Western blot was performed no less than three times to ensure results exhibit a high degree
of reproducibility. Representative Western blots have been selected for each figure.
ll
OPEN ACCESS Article
HDX-MS analyses
Stock concentrations of BACH1 (unmodified or NO-treated, 0.2 mg/mL) in Tris buffer (20 mM tris, 150 mM NaCl, 0.25 mM TCEP,
pH 8.0) were diluted into 90 mL of deuterated PBS buffer (20 mM phosphate, 150 mM NaCl, 0.02% sodium azide, 1 mM EDTA
pH* 7.77, 85%D final) containing 0.2 nM bradykinin and incubated for either 3 seconds, 1 minute, 30 minutes, or 20 hours at
21 C. Each starting stock also included a mixture of imidazolium compounds to serve as exchange reference standards.89 At the
desired time point the sample was rapidly mixed with an equal volume of ice cold 0.2% formic acid and 0.1% trifluoroacetic acid
(TFA) for a final pH of 2.5. Samples were then immediately frozen on ethanol/dry ice and stored at -80 C until LC-MS analysis.
Undeuterated samples were prepared the same way but with undeuterated buffer for each step. Maximally deuterated samples
were prepared by collecting the Nepenthesin II digested peptides after they were trapped and resolved as described below. These
samples were dried down, resuspended in 10 mL of undeuterated buffer, incubated in deuterated PBS for 2 hours at 21 C, then
quenched and frozen as described above. Some late-eluting peptides were outside of the collection window and therefore are absent
in the maximally deuterated data.
Samples were thawed at 5 C for 4.5 minutes and injected using a custom LEAP robot integrated with an LC-MS system.90 The
protein was first passed over a Nepenthesin II column (2.1 x 30 mm; AffiPro) at 400 mL/min for inline digestion with the protease col-
umn held at 20 C. Peptides were then trapped on a Waters XSelect CSH C18 trap cartridge column (2.1 x 5 mm 2.5 mm) and resolved
over a CSH C18 column (1 x 50 mm 1.7 mm 130Å) using linear gradient of 5 to 35% B (A: 0.1% FA, 0.025% TFA, 5% ACN; B: ACN with
0.1% FA) over 10 minutes and analyzed on a Thermo Orbitrap Ascend mass spectrometer at a resolution setting of 120,000. A series
of washes over the trap and Nepenthesin II columns was used between injections to minimize carry-over.90 Data dependent MS/MS
acquisition was performed on an undeuterated sample of either condition using rapid CID and HCD scans and processed in Byonic
(Protein Metrics) with a score cutoff of 150 to identify peptides. S-nitrosylation and cysteine mono- and di-oxidation were included as
potential variable modifications. Deuterium incorporation was analyzed using HDExaminer v3 (Sierra Analytics). Peptide spectra
ll
Article OPEN ACCESS
were exported from HDExaminer and screened for multimodal mass envelopes with PyHXExpress (https://github.com/tuttlelm/
pyHXExpress). Peptides of interest were then analyzed with HXExpress.78 Data are available via ProteomeXchange with identifier
PXD053923.
Resolution estimations of cryo-EM density maps are based on the Fourier Shell Correlation (FSC) curves at the gold-standard
threshold of 0.143. Protein quantification was done using Bio-rad Protein Assay Dye and comparing readings against a standard
curve of BSA. Quantification of protein bands in Western blots and SDS-PAGE gels were done using ImageJ. Data were analyzed
and plotted by Prism (GraphPad). Unless otherwise noted, data were presented as mean ± SD of n = 3 biologically independent
samples.
Supplemental figures
A B
C Template picker
MotionCor2 CTFFIND4 with cryoSPARC 2D classification
6,131 movies 6,131 micrographs 4,991 micrographs 2,888,035 particles 243,712 particles
Manually Curate
Topaz Picking
with cryoSPARC
with cryoSPARC
Remove duplicates 2D classification
949,569 selected particles 1,384,053 particles 3,645,675 particles
Ab-initio reconstruction
Hetergenous refinement
D
Particle
3D Variability Analysis Particle
Substraction
with cryoSPARC Substraction
171,378 particles
Local Local refinement
refinement with cryoSPARC 3.8 Å
3.6 Å with cryoSPARC
3.9 Å
413,179 particles F
Figure S1. The schematic workflow of single-particle reconstruction of the SCFFBXO22-BACH1complex, related to Figure 1
(A) A representative cryo-EM micrograph.
(B) Typical 2D averages of the cryo-EM dataset. Scale bar, 10 nm.
B C
D E
Figure S2. Structural and biochemical analysis of FBXO22-BACH1 interaction, related to Figures 1 and 2
(A) A comparison of the three FIST domains of FBXO22 and homologous structure of YabJ from the YjgF superfamily (PDB: 1QD9).
(B) Steric hindrance prevents the formation of a BACH1-BTB-FBXO22 complex with a 2:2 ratio. The asymmetric complex formed between a BACH1 dimer
(protomer A, light blue; subunit B, orange) and FBXO22 (purple) is shown as cartoon diagram. A second copy of FBXO22 shown in slate surface representation is
modeled onto the BACH1-BTB dimer and is in clash with the other FBXO22 macromolecule.
(C and D) In vitro ubiquitination of BACH1 and BACH1-BTB domain by SCFFBXO22.
(E) Superposition of the crystal structures of BACH1-BTB (PDB: 2IHC) and BACH2-BTB (PDB: 3OHU). BACH2 C-terminal region is disordered and highlighted
in red.
(F) Sequence alignment of five NCOR1 vertebrate orthologs and human NCOR2. Highly conserved short linear motifs (SLiMs) are underlined with gray bars. The
BCL6 BTB-interacting motif found in PDB: 1R2B is underlined with a purple bar.
ll
Article OPEN ACCESS
A B E
FBXL17 + FBXO22 KO
FBXO22 KO
HCT-116 HCT-116
FBXL17 KO
FBXL17 Parental
FBXO22 wild type
wild type
representative
representative FBXL17
FBXO22 knock-out CHX (h)
knock-out
C HCT-116 D HCT-116
FBXL17 + 0 3 6 CHX (h) 0 3 6 CHX (h)
Parental FBXL17 KO FBXO22 KO FBXO22 KO 1 2 3 1 2 3 1 2 3 replicate 1 2 3 1 2 3 1 2 3 replicate
0 3 6 0 3 6 0 3 6 0 3 6 CHX (h)
FBXO22 KO
BACH1 BACH1
Parental
FBXO22 KO
FBXL17 KO
cyclin D1
FBXL17 +
FBXO22 FBXO22
actin actin
F HCT-116 G HCT-116
FBXL17 + FBXL17
Parental FBXL17 KO FBXO22 KO FBXO22 KO Parental FBXL17 KO FBXO22 KO + FBXO22 KO
CHX (h) sNONO
0 6 0 6 0 6 0 6 0 6 0 6 0 6 0 6 + CHX (h)
1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 replicate 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 1 2 3 replicate
BACH1
* BACH1
FBXO22
*
FBXO22
cyclin D1 cyclin D1
GAPDH GAPDH
H I
1.2e+04
8765 43 2 1 NOR3-S1PC-treated
ENVDEVC107K BACH1-BTB
1.0e+04 2.0
1234567 8 10-fold diluted
Relative Intensity
UV x 10-2(AU)
8.0e+03 1.5
BACH1-BTB
6.0e+03 34 kDa (± 25%)
1.0
4.0e+03
0.5
2.0e+03
0.0
6 9 12 15 18
0.0e+00 Volume (ml)
200 400 600 m/z 800 1000 1200
+ - + FBXL17-SKP1 MAF-DNA
(Ub)n~MSB-Flag-BACH1-BTB kDa - + + Ubiquitin MAF-DNA
245 (Ub)n~BACH1-FL
180 dsDNA dsDNA
135 Venus-BACH1-FL ssDNA ssDNA
MSB-Flag-BACH1-BTB 100 NOR3-S1PC treated
Figure S3. Nonredundant roles of FBXO22 and FBXL17 in regulating BACH1 stability, related to Figures 3 and 4
(A and B) Characterization of FBXO22 KO, FBXL17 KO, and FBXL17/FBXO22 double KO cell lines.
(C and D) Cycloheximide (CHX) chase assays monitoring the stability of BACH1 in parental, FBXL17 KO, FBXO22 KO, and FBXL17/FBXO22 double KO HCT-116
cells.
(E) Quantification of the data in (D) with each data point representing mean densities of BACH1 relative to untreated at 0 h. Data are presented as mean ± SD.
(F and G) BACH1 CHX chase assays performed with and without sNONO treatment. The data were used to create the graph in Figure 3D.
(H) An MS/MS fragmentation spectrum assigned to the S-nitrosylated peptide from BACH1-BTB containing Cys107 together with the assignments of the
fragmentation series to the sequence of the modified peptide. The B and Y ions represent the two halves of the peptide formed by splitting the original peptides
between different amino acids. The split sites are indicated by the red and blue ‘‘L’’-shaped marks lying above and below the peptide sequence. The B ions are the
products when the charge is retained on the N terminus, and the Y ions are the product when the charge is retained at the C terminus. The peaks of the products
are labeled accordingly within the spectrum. The measured m/z value of the y2 ion is consistent with the modification of Cys107 by 3-(N-maleimidopropionyl)-
biocytin with a monoisotopic mass of 523 Da.
(I) SEC-MALS analyses of BACH1-BTB treated with NOR3-S1PC with experimentally determined molecular weight. The theoretical molecular weight of BACH1-
BTB is 17 kDa for monomer or 34 kDa for dimer.
(J) Inhibition of in vitro SCFFBXO22-catalyzed BACH1 ubiquitination by NO treatment.
(K) In vitro ubiquitination of NO-treated BACH1 by SCFFBXL17.
(L) NO treatment abrogates the DNA-binding activity of BACH1 bZIP but not sMAF as detected by electrophoresis mobility shift assay (EMSA).
ll
Article OPEN ACCESS
A B C
0.5 Probe: Biotin-BACH1(NOR-3/S1PC) 0.5 Probe: Biotin-BACH1-BTB 1.0
Analyte: FBXL17 (Untreated)
0.4 Apparent Kd = 57 nM 0.4 WT
Analyte: FBXL17 400nM 0.8
Response (nm)
Response (nm)
Response (nm)
C34A
0.3 [FBXL17] 0.3 WT 0.6
1700 nM Probe: Biotin-BACH1-BTB
0.2 567 nM C34A (NOR3/S1PC treated)
0.2 0.4
189 nM Analyte: FBXL17 400nM
0.1 63 nM 0.1 0.2
21 nM
0.0
0.0 0.0
0 200 400 600 800 1000 1200 0 100 200 300 400 0 100 200 300 400
Time (s) Time (s) Time (s)
D Unmodified
< 10%
< 20%
E NO-treated <
<
<
30%
40%
50%
< 60%
< 70%
10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 125 < 80%
6 < 90%
SSVFAYESSVHSTNVLLSLNDQRKKDVLCDVTIFVEGQRFRAHRSVLAACSSYFHSRIVGQADGELNITLPEEVTVKGFEPLIQFAYTAKLILSKENVDEVCKCVEFLSVHNIEESCFQFLKF > 90%
3s
1m
30m
20h
F 22
LSLNDQRKK30 45
FRAHRSVL52 59
FHSRIVGQADGELN 72
Unmod. NO-treated Unmod. NO-treated Unmod. NO-treated.
1×10 8 1×10 8 2×10 9 8×10 8 2×10 8 1×10 8
Cent: 551.6003 m/z Cent: 493.5491 m/z Cent: 493.5464 m/z Cent: 772.2515 m/z z2 Cent: 772.3086 m/z
8×10 7 8×10 7
1.5×10 9 6×10 8 1.5×10 8 7.5×10 7
6×10 7 6×10 7
Not detected 1×10 9 4×10 8 1×10 8 5×10 7 Undeut.
4×10 7 4×10 7
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
1.5×10 8 5×10 7 8×10 8 3×10 7 1×10 8 5×10 7
Cent: 551.7519 m/z Cent: 552.3394 m/z Cent: 493.6878 m/z Cent: 493.7668 m/z Cent: 775.1324 m/z Cent: 775.1757 m/z
F-test p = 1.1e-5 F-test p = 0.0873
4×10 7 Bimodal 1: 72% Bimodal 1: 85%
6×10 8 7.5×10 7 3.75×10 7
Bimodal 2: 28% Bimodal 2: 15%
1×10 8 2×10 7
3×10 7
2×10 7
4×10 8 5×10 7 2.5×10 7
3 sec
5×10 7 1×10 7
2×10 8 2.5×10 7 1.25×10 7
1×10 7
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
1.5×10 8 4×10 7 5×10 8 1.5×10 8 1×10 8 6×10 7
Cent: 551.873 m/z Cent: 552.4223 m/z Cent: 493.7357 m/z Cent: 493.8935 m/z Cent: 775.6057 m/z Cent: 775.6356 m/z
F-test p = 2.19e-6 F-test p = 0.127
Bimodal 1: 73% 4×10 8 Bimodal 1: 80%
3×10 7 7.5×10 7 4.5×10 7
Bimodal 2: 27% Bimodal 2: 20%
1×10 8 1×10 8
3×10 8
2×10 7 5×10 7 3×10 7 60 sec
2×10 8
5×10 7 5×10 7
1×10 7 2.5×10 7 1.5×10 7
1×10 8
Intensity
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
8×10 7 3×10 7 5×10 8 2×10 7 5×10 7 5×10 7
Cent: 552.339 m/z Cent: 552.8369 m/z Cent: 494.0153 m/z Cent: 494.2374 m/z Cent: 776.0401 m/z Cent: 776.1068 m/z
F-test p = 1.31e-5 F-test p = 0.0066
Bimodal 1: 69% 4×10 8 Bimodal 1: 83%
6×10 7 1.5×10 7 3.75×10 7 3.75×10 7
Bimodal 2: 31% Bimodal 2: 17%
2×10 7
3×10 8
4×10 7 1×10 7 2.5×10 7 2.5×10 7 30 min
2×10 8
1×10 7
2×10 7 5×10 6 1.25×10 7 1.25×10 7
1×10 8
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
6×10 7 2.5×10 7 2.5×10 8 5×10 7 4×10 7 6×10 7
Cent: 553.0741 m/z Cent: 555.5091 m/z Cent: 494.4945 m/z Cent: 494.7657 m/z Cent: 776.1102 m/z Cent: 776.0953 m/z
F-test p = 9.53e-5 F-test p = 5.38e-7 F-test p = 0.0703 F-test p = 2.66e-4
Bimodal 1: 65% 2×10 7 Bimodal 1: 30% 2×10 8 Bimodal 1: 74% 4×10 7 Bimodal 1: 45% 3×10 7 4.5×10 7
Bimodal 2: 35% Bimodal 2: 70% Bimodal 2: 26% Bimodal 2: 55%
4×10 7
1.5×10 7 1.5×10 8 3×10 7
2×10 7 3×10 7 20 hr
1×10 7 1×10 8 2×10 7
2×10 7
1×10 7 1.5×10 7
5×10 6 5×10 7 1×10 7
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
4×10 7 4×10 7 2.5×10 8 4×10 7 1.6×10 7 8×10 6
Cent: 554.1212 m/z Cent: 554.1219 m/z Cent: 495.3677 m/z Cent: 495.2787 m/z Cent: 776.2465 m/z Cent: 776.2316 m/z
2×10 8
3×10 7 3×10 7 3×10 7 1.2×10 7 6×10 6
1.5×10 8
2×10 7 2×10 7 2×10 7 8×10 6 4×10 6 Max deut.
1×10 8
0 0 0 0 0 0
551 553 555 557 559 551 553 555 557 559 492 494 496 498 500 492 494 496 498 500 770 772 774 776 778 780 770 772 774 776 778 780
m/z
Figure S4. Analysis of non-treated and NO-treated BACH1-BTB, related to Figures 4 and 5
(A) BLI measurements of the binding between FBXL17 and NOR3-S1PC-treated BACH1-BTB with a 10-min association step. Kd, dissociation constant.
(B and C) BLI measurements of the interaction between FBXL17 and BACH1-BTB (WT and C34A mutant) untreated or treated with NOR3-S1PC. In the absence of
compound treatment, C34A enhanced FBXL17 binding. This effect is exaggerated upon compound treatment. The C34 residue, therefore, is not required for
S-nitrosylation.
(D and E) Heat map summary of HDX data for unmodified (D) and NO-treated (E) BACH1-BTB. Colors under the primary sequence show deuterium uptake
from <10% (blue) to >90% (red) for each time point. Bars above the protein sequence show peptide coverage; green bars represent high-confidence peptides,
yellow bars represent medium-confidence peptides as assigned in HDExaminer v3. Data for the NO-treated sample reflect the population of peptides containing
unmodified cysteines.
(F) Bimodal and unimodal hydrogen-deuterium exchange profiles of additional BACH1-BTB peptides. As observed with peptides 79–89, both unmodified and
NO-modified BACH1 exhibit bimodal exchange profiles at peptides 22–30 and 45–52. With the NO modification, the less-protected population (green) appears
much earlier and is the predominant population by the longest (20 h) time point. Note for peptides 45–52: the 3 and 60 s NO-modified time points do not pass the
F-test but have been plotted as two populations due to the presence of statistically significant bimodal fits for the 30 min and 20 h time points. The peptide-based
maximally deuterated peptides 45–52 control for the unmodified condition eluted late and were not collected for preparation of the maximally deuterated
standard. Peptides 59–72 appear unimodal with either condition throughout the HDX time course. Importantly, the maximally deuterated controls do not show
any bimodal profiles, confirming that the bimodality is not a spectral artifact.
ll
Article OPEN ACCESS
A B
D E H
I
F G
Figure S5. Analysis of NO-treated BACH1-BTB and C122E mutant, related to Figure 5
(A and B) Peptide mapping experiments to identify and quantify levels of S-nitrosylation. Extracted ion chromatograms are shown for the monoisotopic peak of
the 2+ charge state of peptide LSVHNIEESC and ILSKENVDEVCK as unmodified (blue), nitrosylated (+29, orange), and deoxidized (+32, purple). For
LSVHNIEESC, an additional species that is trioxidized (+48, cyan) was detected and is also shown. The traces are shown for both the treated protein and the
untreated sample. * The signal at the unmodified m/z corresponds to some in-source loss of the nitroso group. ** The third isotopic peak from the nitrosylated
(+29) peptide overlaps with the deoxidized (+32) m/z peak. *** Unrelated peptide that is present in both treated and untreated samples.
(C) The annotated MS/MS spectra identifying unmodified and nitrosylated peptides extracted from Byonic (Protein Metrics Inc). Positions of peptide frag-
mentation resulting in b/y ions are shown on each sequence in the inset.
(D) Thermostability of WT BACH1-BTB and the C122E mutant determined by differential scanning fluorimetry. Tm is melting temperature and is presented as
mean ± SD.
(E) Affinity pull-down assay showing the loss of FBXO22 binding in BACH1-BTB C122E mutant.
(F) The time-courses of the interactions between FBXL17 and WT BACH1-BTB vs. C122E mutant as assessed by affinity pull-down assay.
(G) Co-immunoprecipitation analysis of WT BACH1 vs. C122E mutant for FBXO22 and FBXL17 binding.
(H) CHX chase assay assessing the stability of FLAG-tagged WT BACH1 vs. C122E mutant.
(I) FLAG co-immunoprecipitation assay detecting physical association between two differentially tagged FBXL17. Three unrelated F-box proteins (FBXL1,
FBXL11, and FBXL15) were included as negative controls.
ll
Article OPEN ACCESS
A B
C
Patch Motion
Correction Manual curate Blob picker
22,720 movies 22,720 micrographs 20,028 micrographs 7,148,295 particles
Two rounds
Ab-initio 2D classification
Topaz picking reconstruction
6,019,230 particles 660,991 particles
2D classification 143,997 particles
Ab-initio
reconstruction
718,765 particles
Topaz
183,814 picking
484,750 particles particles 156,298 particles
Heterogeneous 2D classification
refinement
213,113 particles 461 particles
5.7 Å Two rounds
7,944 10,173 2D classification
particles particles Ab-initio
reconstruction
38,168 particles
Heterogeneous
Non uniform Non uniform refinement
refinement refinement
with C2 symmetry with C2 symmetry
7.8 Å 7.8 Å
BACH1-
BACH1-
FBXL17 BTB
FBXL17 BTB
CUL1 CUL1
RBX1 RBX1
Figure S6. Schematic workflow of single-particle reconstruction of NO-treated SCFFBXL17-BACH1 complex, related to Figure 6
(A) A representative cryo-EM micrograph for the sample containing a mixture of SCFFBXL17 and BACH1-BTB treated with NOR3-S1PC.
(B) Typical 2D averages.
(C) The flowchart of single particle analysis of the sample containing a mixture of SCFFBXL17 and BACH1-BTB treated with NOR3-S1PC.
(D and E) Fitting of the WT dSCFFBXL17-BACH1-I and dSCFFBXL17-BACH1-II structures in the 3D reconstructions obtained with the NO-treated BACH1-BTB sample.
ll
OPEN ACCESS Article
A B
4.5 Å Local
refinement 3.2 Å
4.1 Å
D E G
Figure S7. Cryo-EM single-particle analysis workflow of native SCFFBXL17-BACH1, related to Figure 6
(A) A representative cryo-EM micrograph.
(B) Typical 2D averages revealing the most populated monomeric complex.
(C) The flowchart of single particle analysis of the sample containing a mixture of SCFFBXL17 and BACH1-BTB.
(D) Local resolution map of the BACH1-BTB dimer flanked by the LLR domains of two FBXL17 ranging from 3 to 8 Å.
(E) The particle angular distribution and FSC curves of dSCFFBXL17-BACH1-I with local refinement.
(F) The particle angular distribution and FSC curves of dSCFFBXL17-BACH1-II.
(G) The angular distribution and FSC curves of the monomeric SCFFBXL17-BACH1 complex.