You are on page 1of 8

Computers in Biology and Medicine 149 (2022) 106006

Contents lists available at ScienceDirect

Computers in Biology and Medicine


journal homepage: www.elsevier.com/locate/compbiomed

An order-to-disorder structural switch regulates HIF-1 transcription


through S247 phosphorylation in the HIF1α PAS-B domain
Chia-Hung Hsu a, Ya-Jyun Chen b, Chia-Ning Yang b, *
a
Department of Internal Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
b
Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan

A B S T R A C T

Hypoxia-inducible factor 1 (HIF-1), a transcriptional activator that mediates cellular responses to hypoxic stress, is essential for tumor progression. It is a heterodimer
comprising HIF1α and HIF1β, with multiple interfaces among their PAS-A, PAS-B, and bHLH domains. HIF1β is also known as aryl hydrocarbon receptor nuclear
translocator (ARNT). Casein kinase 1δ–dependent phosphorylation of the solvent-front residue S247 on the HIF1α PAS-B domain interrupts HIF1α–ARNT complex
formation and reduces HIF-1 transcription activity. However, S247 is involved in neither HIF1α–ARNT complex formation nor stabilization of the relative orientation
between the HIF1α PAS-A and PAS-B domains. To uncover the underlying allosteric mechanism, we conducted Gaussian accelerated molecular dynamics simulations
and identified two distinct conformations of the pS247-carrying HIF1α PAS-B domain: H291-in and H291-out. The H291-in structure can associate with the HIF1α
PAS-A domain and form a V-shaped pouch to accommodate the ARNT PAS-A domain, but it cannot associate with the ARNT PAS-B domain. By contrast, the H291-out
structure can bind to the ARNT PAS-B domain, but its association with the HIF1α PAS-A domain leads to an unsuitable relative orientation to accommodate the ARNT
PAS-A domain. Both conformations were also collected in parallel simulations of the unphosphorylated PAS-B domain. Both structures manage to associate with the
ARNT PAS-B and HIF1α PAS-A domains; thus, they are adequate for HIF1α–ARNT complex formation. The domain–domain contact pattern in a phosphorylated
variant is shuffled by an order-to-disorder structural switch, triggered by the newly formed K251–pS247 interaction.

1. Introduction HIF1α, and the hydroxylated HIF1α is recognized by the von Hip­
pel–Lindau protein, which then activates the ubiquitin ligase system for
Hypoxia-inducible factors (HIFs) are transcriptional activators that proteasomal degradation. However, PHD is inactive under hypoxic
regulate cellular oxygen sensing and homeostasis. These regulations are conditions; therefore, HIF1α is stabilized for dimerization with ARNT.
pivotal to tumor growth and progression; therefore, HIFs have become Both HIF1α and ARNT use their bHLH, PAS-A, and PAS-B domains for
attractive cancer therapeutic targets [1–10]. To execute their function, dimerization, as indicated in Fig. 1(A), where two crossed bHLH do­
HIFs form HIF1α–HIF1β and HIF2α–HIF1β heterodimers; HIF1β is also mains form a reading head to bind to the regulatory hypoxia-response
called an aryl hydrocarbon receptor nuclear translocator (ARNT). In this elements (HREs) in the promoter region of HIF target genes. More­
study, we focused on the HIF1α–ARNT system. In HIF1α, the N-terminal over, the HIF1α–ARNT heterodimer binds to CBP/p300 to promote the
region comprises a basic helix–loop–helix domain (bHLH), which context-dependent expression of multiple genes that offset cellular
functions as part of a DNA reading head. It also has two consecutive hypoxia [11–14].
Per-ARNT-Sim (PAS) homology domains—PAS-A and PAS-B—which are HIF1α transcriptional activity is also subject to oxygen-independent
required for dimerization with ARNT, and an oxygen-dependent regulation. Kalousi et al. observed that casein kinase 1δ (CK1δ)-depen­
degradation domain essential for oxygen-dependent hydroxylation and dent serine 247 (S247) phosphorylation on the HIF1α PAS-B domain has
degradation. Moreover, the C-terminal region contains an N-terminal a structural impact, interrupting HIF1α–ARNT dimerization and,
transactivation domain (N-TAD) and a C-TAD, which are separated by accordingly, abolishing transcriptional activity [15]. The hypoxic
an inhibitory domain. These TADs are responsible for transcriptional HRE-dependent transcriptional activity of HIF1α was found to be greatly
activity under hypoxia. The transcriptional activity of the C-TAD reduced due to CK1δ overexpression; furthermore, S247 on the HIF1α
domain is associated with the binding of the transcriptional coac­ PAS-B domain was the target site for modification by CK1δ. However,
tivators: CREB-binding protein (CBP) and p300. Under normoxic con­ CK1δ inhibition—through CK1δ inhibitor PF-670462 [16] or siRNA
ditions, prolyl hydroxylase (PHD) hydroxylates two proline residues of knockdown—enhanced HIF1α transcriptional activity under hypoxia.

* Corresponding author. No. 70 Lien-hai Road, Kaohsiung, 80424, Taiwan.


E-mail address: cnyang@mail.nsysu.edu.tw (C.-N. Yang).

https://doi.org/10.1016/j.compbiomed.2022.106006
Received 24 February 2022; Received in revised form 11 August 2022; Accepted 14 August 2022
Available online 19 August 2022
0010-4825/© 2022 Published by Elsevier Ltd.
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

Moreover, compared with the association of ARNT with wild-type Because of their strong ability to sample conformational changes,
HIF1α, the association was stronger when S247 of HIF1α was replaced molecular dynamics simulations are useful for relating protein struc­
by alanine and weaker when HIF1α harbored phosphomimetic mutant tures to their functions. Gaussian accelerated molecular dynamics
S247D. Together, these results indicate that HIF1α S247 phosphoryla­ (GaMD) offers simultaneous, unconstrained, enhanced conformational
tion inhibits HIF1α–ARNT heterodimerization, thus impairing tran­ sampling and free energy landscape construction for biomolecules; this
scriptional activity. is achieved by considering a harmonic boost potential to smooth the
The present study demonstrated that structural variance originating biomolecular potential energy surface [17–19]. We employed GaMD
from phosphorylated S247 (pS247) interrupts HIF1α–ARNT dimeriza­ simulations to collect conformations of the HIF1α PAS-B domain in the
tion. For the HIF1α–ARNT–HRE complex structure retrieved from unphosphorylated wild type (WT) and the pS247-carrying variants to
PDB:4ZPR, each HIF1α and ARNT has bHLH, PAS-A, and PAS-B do­ determine how pS247 modifies the HIF1α PAS-B domain and interferes
mains; these domains are intertwined when associating with HREs with HIF1α–ARNT dimerization. High linkage flexibility, even in the
(Fig. 1(A)). Fig. 1(B) presents the secondary structure arrangement of complex structure, suggests the independent movement of each domain
the HIF1α PAS-B domain, showing the location of S247 on the C-ter­ when in free HIF1α and ARNT. Accordingly, the pS247-induced struc­
minal end of the Aβ strand in the domain, with its hydrophilic hydroxyl tural change can be investigated within the HIF1α PAS-B domain alone;
group in the solvent front and not in contact with the ARNT PAS-B thus, the following strategy was adopted. First, for each PAS-B of the
domain. Moreover, the HIF1α PAS-B domain (green) is surrounded by unphosphorylated WT and pS247-carrying variants, three independent
ARNT PAS-A (gray), ARNT PAS-B (gray), and HIF1α PAS-A (blue). simulation runs were performed to cluster conformation ensembles and
Notably, the coordinates of a short I235–D238 segment (linking the extract representative structures. Furthermore, each pS247 representa­
HIF1α PAS-A and PAS-B domains) and of a rather long V345–P360 tive conformation was superimposed on a complex structure comprising
segment (connecting the ARNT PAS-A and PAS-B domains) were not the HIF1α PAS-B and ARNT PAS-B domains to produce an initial
determined due to high mobility. PAS-B/PAS-B structure for further binding examination. The process

Fig. 1. HIF1α structure in dimerization with ARNT retrieved from PDB:4ZPR. (A) Complex structure of the HIF1α–ARNT heterodimer associated with the HRE
sequence. Dashed lines represent the missing segments connecting the bHLH and PAS-A domains or connecting the PAS-A and PAS-B domains. (B) Arrangement of
the secondary structural elements in the HIF1α PAS-B domain. The residue S247, which is to be phosphorylated, is specified with an all-atom style. (C) Interface
interactions between the HIF1α PAS-A domain (blue) and the PAS-B domain (green). (D) Interface interactions between the HIF1α PAS-B domain (green) and the
ARNT PAS-B domain (gray).

2
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

was repeated to produce an initial HIF1α PAS-A/PAS-B structure and to ensemble at 1 atm and 300 K for 40 ps with the same restraints as in the
examine the structure’s binding strength. The structure was later used to NVT run, followed by a density procedure at 300 K for 100 ps and
determine whether the pocket formed by the HIF1α PAS-A and PAS-B constant equilibration at 300 K for 800 ps. A 25-ns conventional mo­
domains could accommodate the ARNT PAS-A domain, as indicated in lecular dynamics simulation was performed to collect potential energy
Fig. 1(C). With this work, we intended to determine the structural statistics, including the maximum, minimum, average, and standard
impact that results in a weak HIF1α–ARNT association. However, the deviation. Another GaMD-based 25-ns equilibration was performed
whole HIF1α (formed by their bHLH, PAS-A, and PAS-B domains) or the after the boost potential had been applied. For both WT and pS247, three
HIF1α–ARNT complex were not simulated because these structures may independent GaMD production simulations were performed in 500 ns
not yield sufficient conformational adjustment that can be attributed to with different randomized initial atomic velocities. Simulation frames
the phosphate group introduced on S247. For a parallel comparison, all were saved every 1 ps for analysis. In total, 50,000 snapshots were
the aforementioned modeling procedures were also performed for the retrieved from each trajectory, and 150,000 snapshots were used to plot
WT. In this work, we identified two conformation clusters: H291-in and a free energy landscape against carefully selected coordinates. For each
H291-out. Without S247 phosphorylation, H291 is interconvertible be­ conformation cluster, 500 conformations centered at low potential en­
tween H291-in and H291-out when in PAS-B alone and when associated ergy were collected for structural analysis.
with its PAS-A and ARNT PAS-B. By contrast, the H291-in and H291-out Furthermore, to examine the ability of the HIF1α PAS-A domain to
conformations in the pS247 variant restrain PAS-B-binding behavior. associate with the low-energy conformations sampled from GaMD sim­
That is, when adopting H291-in, HIF1α PAS-B’s facade for association ulations, we extracted the structures of the HIF1α PAS-A and PAS-B
with ARNT PAS-B is impaired, whereas that for association with HIF1α domains from PDB:4ZPR. and superposed the sampled low-energy
PAS-A is retained; the opposite is observed for H291-out. Altogether, conformations onto PAS-B, and replaced the X-ray-solved PAS-B
phosphorylated S247 interferes with the HIF1α–ARNT association. domain to generate the initial HIF1α PAS-A/PAS-B structures. To
examine the binding behavior of the HIF1α PAS-B and ARNT PAS-B
2. Methods domains, the same procedure was used to generate the initial HIF1α
PAS-B/ARNT PAS-B structures. The HIF1α–ARNT complex is formed by
2.1. System setup multiple interfaces (Fig. 1(A)). Because the ARNT PAS-A and PAS-B
domains are largely separated, whereas the HIF1α PAS-A and PAS-B
The initial structure of the WT PAS-B domain was extracted from the domains are in contact during heterodimerization, the HIF1α PAS-A
crystal structure of the HRE-bound human HIF1α–ARNT complex (PDB and PAS-B domains likely form a V-shape pouch first and consecu­
code: 4ZPR) [20]. Because S247 was in the solvent front, S247 was tively accommodate ARNT PAS-A and PAS-B in any order. Therefore, we
manually phosphorylated to generate pS247 without spatial conflict. examined the association events between HIF1α PAS-A and PAS-B and
The two simulated systems were immersed in a cubic TIP3P water box in between HIF1α PAS-B and ARNT PAS-B.
which the distance separating any protein atom from the water box wall
was ≥12 Å. To neutralize the protein systems, two and four Na+ ions 3. Results and discussion
were added to the WT and pS247 variants, respectively. Any neigh­
boring TIP3P water molecules within 1.5 Å of the PAS-B domain were 3.1. Conformations of the HIF1α PAS-B domain in the WT and pS247
discarded to prevent the formation of biased hydrogen bonds between variants
PAS-B and TIP3P water molecules prior to modeling. Each PAS-B system
was energy-minimized in three stages, each using 25,000 steps of both For each of the studied HIF1α PAS-B domains in the WT and pS247
the steepest descent algorithm and conjugate gradient algorithm. In variants, the 150,000 conformations collected from three replicas were
stage one, PAS-B was restrained to allow reorientation of the added plotted as a free-energy landscape, where the centroid separation dis­
TIP3P water molecules. In stage two, the PAS-B backbone was restrained tance H291–D249 was used as the x-axis and H291–E277 was used as
to optimize the interplay between the amino acid side chains and water the y-axis. As indicated in Fig. 1(D), the E277 and D249 residues of the
molecules. In stage three, the entire solvated system was minimized with HIF1α PAS-B domain electrostatically interact with the R366’ and N448’
no restraints. residues of the ARNT PAS-B domain, respectively, whereas H291 and its
For each HIF1α PAS-B system, three independent GaMD simulations nearby H292 and F295, all on the C-terminal portion of Fα, form a hy­
were performed using the AMBER 18 software package [21]. The stan­ drophobic dent to accommodate the P449’ residue of the ARNT PAS-B
dard protocol covering gradual heating, density, equilibration, and domain. In Fig. 2(A), HIF1α PAS-B domain conformations in the WT
production procedures in the isothermal isobaric ensemble (P = 1 atm are clustered in two low-energy basins—H291-out and H291-in—with a
and T = target temperature) was employed. A minimized solvated sys­ similar H291–E277 separation distance of approximately 15 Å. In the
tem served as the initial structure for subsequent GaMD simulations. A H291-out cluster, the imidazole moiety of H291 moves out of the hy­
cutoff of 8 Å was applied to treat nonbonding interactions, such as drophobic core and moves close to D249, leading to a short H291–D249
short-range electrostatics and van der Waals interactions, whereas the separation distance of approximately 8 Å. In the H291-in conformation,
particle mesh Ewald method [22] was used to treat long-range electro­ the imidazole ring is slightly embedded inside the hydrophobic core and
static interactions. All hydrogen atom bonds were constrained using the shifted away from D249, thereby increasing the H291–D249 separation
SHAKE algorithm [23,24]. distance up to 11 Å. Fig. 2(A) also presents the PAS-B domain structure
(denoted with an asterisk) in the HIF1α–ARNT–HRE complex as deter­
2.2. GaMD-enhanced sampling simulations mined through crystallography. The complexed PAS-B structure is be­
tween the two clusters, implying that both H291-in and H291-out
GaMD simulations were employed to collect the conformations of the conformations are slightly rearranged after dimerization.
native and phosphorylated PAS-B domains. With a harmonic boost po­ Similarly, Fig. 2(B) presents two clusters, with their representative
tential to smooth the potential energy surface and reduce energy bar­ structures showing moving out and embedded H291 for the pS247
riers, GaMD can enhance conformational sampling [18,25]. The variant. Fα deformation and Eα downward movement result in the
aforementioned minimized WT and pS247 structures were heated from H291–E277 separation distance (13.5–14 Å) being shorter than that in
0 to 300 K in a 60-ps simulation by applying 10 kcal/(mol⋅Å2) harmonic the WT (14.5–15 Å). In the H291-in conformation, H291 is shielded by
position restraints to protein heavy atoms in the constant num­ the M250 aliphatic chain (on Bβ), whereas in the H291-out conforma­
ber–volume–temperature (NVT) ensemble. Each system was further tion, the H291 ring moiety and M250 aliphatic chain are stacked in
re-equilibrated using the constant number–pressure–temperature parallel for insertion into the hydrophobic core of the HIF1α PAS-B

3
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

Fig. 2. Two-dimensional free energy profiles according to the centroid separation distances of H291–D249 as the x-axis and of H291–E277 as the y-axis; confor­
mations obtained from the GaMD result for the HIF1α PAS-B domain in the (A) WT and (B) pS247 variants. Representative conformations are on the sides.

domain. variants). Fig. 4(C) also illustrates the relative positions of H291 and
We used the dictionary of secondary structures of proteins (DSSP) M250 for H291-in and H291-out.
method to evaluate Fα (residues D285–F295) intactness. The Fα helical
character is well-preserved in both the H291-in and H291-out clusters in 3.2. HIF1α PAS-B binding with ARNT PAS-B
the WT PAS-B (Fig. 3(A) and 3(B)). In H291-out in the pS247 variant, the
D285–H291 segment exhibits mixed helix and loop/turn characteristics We retrieved HIF1α PAS-B and ARNT PAS-B domains from PDB:4ZPR
(Fig. 3(C)). Regarding H291-in in the pS247 variant, the D285–T288 as a template structure and replaced the HIF1α PAS-B with the H291-in
segment has mixed loop/turn and strand characteristics (Fig. 3(D)). and H291-out conformations obtained from the WT and pS247 trajec­
To elucidate the phosphate group–triggered structural change, we tories as determined using X-ray crystallography. Four initial complex
performed a structural analysis of the WT. In Fig. 4(A) for the retrieved structures for HIF1α PAS-B were thus generated to associate with the
HIF1α PAS-B domain, a buried hydrophobic cavity present above the ARNT PAS-B domain, with each structure submitted for three indepen­
β-sheet is maintained by a balanced spatial arrangement of M250 on Aβ- dent GaMD simulation runs. Plotted in Fig. 5(A) and (B) are free-energy
Bβ; Y276 on Eα; and H291, H292, and F295 on Fα. Moreover, the landscapes for ARNT PAS-B binding to HIF1α PAS-B in the WT, where
K251–R273 and K251–S274 electrostatic interactions stabilize Bβ and H291-out and H291-out conformations were used as the initial struc­
DEβ, further securing Y276. In the pS247 variant, K251 electrostatically tures. These landscapes are plotted against the ARNT Y450′ –HIF1α L248
interacts with the phosphate group. The consequence of this K251 centroid separation distance (as the y-axis) and H291–D249 centroid
movement toward the phosphate group is twofold. One is the rear­ separation distance (as the x-axis). Fig. 1(C) indicates a hydrophobic
rangement of DEβ and reorientation of the Y276 aromatic ring moiety interaction between ARNT Y450′ and HIF1α L248 in the complex
(see Fig. 4(B) for a comparison of the WT and pS247 variants). Another structure; thus, their separation indicates an association event between
is the repositioning of M250, which also affects the H291 ring moiety the two PAS-B domains. Fig. 5(A) and (B) suggest that the H291-in- or
(see Fig. 4(C) for a comparison of the WT and pS247 variants). Overall, H291-out-carrying WT can associate with the ARNT PAS-B domain.
the aforementioned balanced arrangement in the WT, built by M250, Notably, the imidazole ring is free to flip in and out regardless of the
Y276, and H291, is crumpled in the pS247 variant. Several N-terminal initial spatial state of H291, as evidenced by the two basins centered at 8
Fα residues sink into the buried cavity, thereby deforming the N-ter­ Å (for H291-out) and 11 Å (for H291-in) along the H291–D249 sepa­
minal Fα and the shrunk cavity, as estimated using a bio-tool in CAVER ration distance (Fig. 5(A) and (B)).
Analyst [26] (see Fig. 4(D) for a comparison of the WT and pS247 Fig. 5(C) presents the landscape plotted for the pS247 variant when

4
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

Fig. 3. DSSP-based analysis of secondary structural elements (loop/turn, helix, and strand) on HIF1α PAS-B domain’s Fα helix for the (A) H291-out cluster in the WT,
(B) H291-in cluster in the WT, (C) H291-out in the pS247 variant, and (D) H291-in in the pS247 variant.

Fig. 4. Structural details of WT and pS247. (A) The HIF1α PAS-B domain has a cavity (gray). Structure extracted from PDB:4ZPR. (B) Relative orientation of K251
and Y276 in different conformations. (C) Relative orientation of M250 and H291 in different conformations. (D) Comparison of the cavity in different conformations.
WT in H291-out and H291-in maintained a shrunk cavity. In the pS247 variant, having either conformation, the cavity is embedded by the surrounding amino
acid residues.

5
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

Fig. 5. Two-dimensional free energy profiles according to the H291–D249 centroid separation distance as the x-axis and the Y450′ –L248 centroid separation distance
as the y-axis; conformations obtained from the GaMD result for the ARNT PAS-B domain associating with the HIF1α PAS-B domain in (A) using the H291-out cluster
in the WT, (B) using the H291-in cluster in the WT, and (C) using the H291-out cluster in the pS247 variant, and (D) using the H291-in cluster in the pS247 variant
with a representative conformation on the side.

using the H291-out conformation as the initial complex structure. The depends on the H291 conformation prior to this association, and only
distance separating the centroids of ARNT Y450′ and HIF1α L248 is H291-out has the opportunity to grasp the ARNT PAS-B domain.
approximately 5 Å, which is identical to that observed in Fig. 5(A) and
(B), suggesting the formation of a complex of two PAS-B domains.
3.3. HIF1α PAS-B binding with HIF1α PAS-A
Notably, the H291–D249 distance indicates that the H291 movement is
restricted in the H291-out conformation and that it is not allowed to flip
HIF1α PAS-A and PAS-B domains that are linked by a flexible
in. As presented in Fig. 5(D), when the initial pS247 variant is in H291-
I235–D238 loop (whose coordinates are not solved in the HIF1α–ARNT
in, the complex structure is not stable since the ARNT Y450′ -HIF1α L248
complex structure) are likely to move independently when not associ­
separation distance is populated between 4.5 and 10 Å. Again, the H291
ated with ARNT. As indicated in Fig. 1(A), the HIF1α PAS-A domain
position is locked in the H291-in confirmation, as indicated by the
(blue) and PAS-B domain (green) form a V-shaped pouch to accommo­
H291–D249 distance centered at 12 Å. To summarize, the association
date the ARNT PAS-A domain. The appropriate opening of the V-shaped
between ARNT PAS-B and HIF1α PAS-B domains in the pS247 variant
pouch is determined using a salt bridge formed by PAS-A’s E143 and

Fig. 6. Two-dimensional free energy profiles according to the Y92–W316 centroid separation distance as x-axis and the E143–K297 centroid separation distance as y-
axis; conformations obtained from the GaMD result for the HIF1α PAS-A domain associating with PAS-B in (A) using the H291-out cluster in the WT, (B) using the
H291-in cluster in the WT, (C) using the H291-out cluster in the pS247 variant, and (D) using the H291-in cluster in the pS247 variant with a representative
conformation on the side.

6
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

PAS-B’s K297 and can be monitored in accordance with the separation structural impact of accommodating an artificial ligand in the PAS-B
distance between PAS-A’s Y92 and PAS-B’s W316, as depicted in Fig. 1 domain [34,35]. For instance, Masetti’s group evaluated the
(C). Both Y92 and W316 exhibit nonnegligible hydrophobic interactions ligand-bound and ligand-unbound states of the HIF2α PAS-B domain and
with the ARNT PAS-A domain. To elucidate how pS247 causes a struc­ a complex formed by the HIF2α PAS-B and ARNT PAS-B domains, where
tural change to weaken HIF1α–ARNT complex formation, we also the considered ligand was a prototypical disrupting ligand [34]. Chong
examined the interaction between HIF1α PAS-A and PAS-B domains. and Anderson characterized the structural stabilization attributed to
The structure of these domains recovered from PDB:4ZPR was used as a eight ordered water molecules in the druggable pocket of the HIF2α
template structure where the HIF1α PAS-B determined through X-ray PAS-B domain [35]. Our group previously conducted a series of classical
crystallography was replaced by the H291-in and H291-out conforma­ molecular dynamics simulations on the HIF-2α PAS-A and PAS-B do­
tions from WT and pS247 trajectories; this resulted in four initial HIF1α mains of the WT and of three mutants carrying R171A and/or V192D for
PAS-A bindings with the HIF1α PAS-B domain without the flexible the HIF-2α–ARNT dimer destabilization [36]. For both R171 and V192
linkage. Fig. 6(A) and (B) show free energy landscapes obtained when in the interface of the HIF-2α PAS-A and PAS-B domains, mutations alter
using the PAS-B domain with H291-out and H291-in, respectively, the relative domain–domain orientation and accordingly damage the
plotted against the K297–E143 and Y92–W316 center-of-mass separa­ V-shaped pouch that can hold the ARNT PAS-A domain.
tion distances for the aforementioned reasons. The narrow K297–E143 Through S247 phosphorylation, HIF1α PAS-B experiences an order-
span (8.5–9.5 Å) means that the salt bridge must be formed in a precise to-disorder structural switch, which leads to an inability to dimerize
orientation. By contrast, the rather wide Y92–W316 span (21–33 Å) with ARNT, thus interrupting the transcription activity. Such an order-
denotes the staunch K297–E143 salt bridge that enables open-to-closed to-disorder structural switch was also observed for the regulation of
movement of two PAS domains until the ARNT PAS-A domain is FoxM1 transcription factor, whose transcription activity is inhibited by
accessed, indicated by an asterisk sign in Fig. 6(A), which displays the the association of its transactivation domain (TAD) to its negative reg­
corresponding separation distances in the HIF1α–ARNT complex ulatory domain (NRD). Upon phosphorylation of TAD’s S715, TAD
structure. dissociates from NRD and turns into a disordered conformation, which is
Fig. 6(C) illustrates the association between PAS-A and PAS-B do­ able to bind to its transcriptional co-activator CBP [37].
mains when the pS247 H291-out conformation is used as the initial Fig. 7 summarizes the structural impact of pS247. In the unphos­
structure. H291 stays out in all three replica trajectories. Owing to the phorylated WT, there is a minor structural difference between the H291-
extensive Fα distortion, as indicated in Fig. 3(C), the misplaced E143 in and H291-out conformations, and H291 is interconvertible between
extensively disrupts the interface between the HIF1α PAS-A and PAS-B the two states when alone and upon associating with HIF1α PAS-A and
domains, resulting in disintegrated clusters. In a cluster with ARNT PAS-B. However, in the pS247 variant, the H291-in and H291-out
K297–E143 span of 8.5–9.5 Å and Y92–W316 span of 33–43 Å, the conformations rigidly restrict PAS-B-binding behavior. When in H291-
separation between Y92 and W316 is too large to undergo induced-fit in, the facade for association with ARNT PAS-B is impaired, whereas
when the ARNT PAS-A domain is encountered. Regarding H291-in that for association with HIF1α PAS-A is retained; the opposite is
(Fig. 6(D)), although the population is diffusing along the y-axis, a observed in H291-out. Taken together, this allosteric mechanism ex­
cluster with a K297–E143 span of 8.5–9.5 Å and a Y92–W316 span of plains the diminished HIF1α–ARNT heterodimerization.
22–31 Å is likely for HIF1α PAS-B to first associate with the HIF1α PAS-A In this study, we provide structural clues to link a local structural
domain and subsequently bind to the ARNT PAS-A domain. Meanwhile, variance caused by the phosphorylated Ser247 toward the interface
the slightly broad K297–E142 separation in Fig. 6(D) is attributable to between the HIF1α PAS-B and ARNT PAS-B domains and the interface
Fα distortion. In summary, our simulation results suggest that only the shared by the HIF1α PAS-A, HIF1α PAS-B, and ARNT PAS-A domains.
H291-in conformation of the pS247 variant has the opportunity to Because all the concluding remarks were based on molecular modeling,
associate with HIF1α PAS-A and then accommodate the ARNT PAS-A further analysis of protein–protein association events among the PAS-A
domain. and PAS-B domains of HIF1α and ARNT are required to confirm our
results. Because HIF1α-ARNT is a heterodimer with six interfaces [20],
4. Conclusion as indicated in Fig. 1(A), our model of the propagated structural disorder
suggests that HIF1α PAS-B plays a critical role in heterodimer formation.
Enhanced conformational sampling through GaMD revealed two However, these six interfaces certainly impede propagation of the
structures in terms of the H291-in and H291-out conformations for the structural influence from the phosphorylated site as if we had used the
HIF1α PAS-B domain. The unphosphorylated WT structure maintains a intertwined HIF1α-ARNT complex in the beginning for structural eval­
flexible hydrophobic facade held by structured Fα, inflexible Eα, and Aβ- uation. The fact that the segment connecting the HIF1α PAS-A and
Bβ loop to enable the interconversion of H291 between the H291-in and PAS-B domains was too flexible to be solved [20] suggests that these two
H291-out positions without the whole conformation being disturbed. PAS domains tend to move independently when not associated with
Moreover, K251 points toward the solvent and occasionally electro­ ARNT. On the basis of these facts, we characterized conformational
statically interacts with S274 near Eα and D249 on the tip of the Aβ-Bβ features of the phosphorylated and unphosphorylated HIF1α PAS-B
loop. Upon S247 phosphorylation, K251 is driven away toward the alone, instead of the entire HIF1α-ARNT complex, to shed light on the
phosphate group. Such a keystone displacement consecutively de­ structural change that occurs prior to the binding event, and we further
stabilizes Eα and the Aβ-Bβ loop and disrupts the N-terminal Fα, ulti­ surmised its possible binding behaviors with other domains. Such a
mately deforming the hydrophobic facade stabilized by M250, Y276, bottom-up strategy should be useful for studies on protein–protein
and H291 in the WT. recognition.
Many inhibitors bind to an internal water-filled cavity of the HIF2α
PAS-B domain to interrupt the interaction between the HIF2α PAS-B and CRediT authorship contribution statement
ARNT PAS-B domains for therapeutic purposes [27–33]. HIF2α PAS-B
domain-bound inhibitors cause conformational changes in M252, Chia-Hung Hsu: Project discussion and administration; Conceptual­
Y278, and H293 (M250, Y276, and H291 in HIF1α PAS-B) to interfere ization. Ya-Jyun Chen: Data curation and validation. Chia-Ning Yang:
with HIF2α–ARNT dimerization [27,30,31]. This is consistent with our Draft writing, reviewing, and editing; Investigation; Conceptualization.
finding that these three residues, when spatially rearranged, reshape the
interaction pattern between the HIF1α PAS-B and ARNT PAS-B domains. Declaration of competing interest
Along with research on drug development in relation to HIFs, mo­
lecular modeling studies on HIFs have been conducted to identify the The authors declare that they have no known competing financial

7
C.-H. Hsu et al. Computers in Biology and Medicine 149 (2022) 106006

Fig. 7. Summary of the difference caused by pS247. In the WT, the H291-in and H291-out conformations are observed and interconvertible upon association with the
ARNT PAS-B and HIF 1α PAS-A domains. In the pS247 variant, the H291-in and H291-out conformations are not interconvertible. Moreover, H291-in conformation
cannot associate with the ARNT PAS-B domain but can appropriately associate with the HIF 1α PAS-A domain and further accommodate the ARNT PAS-A domain. By
contrast, H291-out conformation can appropriately bind to the ARNT PAS-B domain but not to the HIF 1α PAS-A domain.

interests or personal relationships that could have appeared to influence [17] J. Wang, et al., Gaussian accelerated molecular dynamics: principles and
applications, Wiley Interdiscip. Rev. Comput. Mol. Sci. 11 (2021), e1521.
the work reported in this paper.
[18] A.-h. Wang, Z.-c. Zhang, G.-h. Li, Advances in enhanced sampling molecular
dynamics simulations for biomolecules, Chin. J. Chem. Phys. 32 (2019) 277.
Acknowledgment [19] Y. Miao, Acceleration of biomolecular kinetics in Gaussian accelerated molecular
dynamics, J. Chem. Phys. 149 (2018), 072308.
[20] D. Wu, et al., Structural integration in hypoxia-inducible factors, Nature 524
The authors thank Zuoying Branch of Kaohsiung Armed Forces (2015) 303–308.
General Hospital, Kaohsiung, Taiwan (grant number: ZBH 107-06), for [21] I. Case, et al., AMBER 2018, University of California, San Francisco, 2018.
the financial support. [22] R.W. Hockney, J.W. Eastwood, Computer Simulation Using Particles, Crc, 1988.
Press.
[23] W. Van Gunsteren, H.J. Berendsen, Algorithms for macromolecular dynamics and
References constraint dynamics, Mol. Phys. 34 (1977) 1311–1327.
[24] J.-P. Ryckaert, G. Ciccotti, H.J. Berendsen, Numerical integration of the cartesian
[1] C.M. Bailey, et al., Targeting HIF-1α abrogates PD-L1–mediated immune evasion in equations of motion of a system with constraints: molecular dynamics of n-alkanes,
tumor microenvironment but promotes tolerance in normal tissues, J. Clin. Invest. J. Comput. Phys. 23 (1977) 327–341.
132 (2022). [25] A. Bhattarai, Y. Miao, Gaussian accelerated molecular dynamics for elucidation of
[2] Y. Shirai, et al., An overview of the recent development of anticancer agents drug pathways, Expet Opin. Drug Discov. 13 (2018) 1055–1065.
targeting the HIF-1 transcription factor, Cancers 13 (2021) 2813. [26] A. Jurcik, et al., CAVER Analyst 2.0: analysis and visualization of channels and
[3] A. Lequeux, et al., Targeting HIF-1 alpha transcriptional activity drives cytotoxic tunnels in protein structures and molecular dynamics trajectories, Bioinformatics
immune effector cells into melanoma and improves combination immunotherapy, 34 (2018) 3586–3588.
Oncogene 40 (2021) 4725–4735. [27] M.C. Ghosh, et al., Therapeutic inhibition of HIF-2α reverses polycythemia and
[4] C.E. McAleese, et al., Hypoxia-mediated drug resistance in breast cancers, Cancer pulmonary hypertension in murine models of human diseases, Blood, The Journal
Lett. 502 (2021) 189–199. of the American Society of Hematology 137 (2021) 2509–2519.
[5] W.S.W. Choi, J. Boland, J. Lin, Hypoxia-inducible factor-2α as a novel target in [28] Y. Yu, Q. Yu, X. Zhang, Allosteric inhibition of HIF-2α as a novel therapy for clear
renal cell carcinoma, Journal of Kidney Cancer and VHL 8 (2021) 1. cell renal cell carcinoma, Drug Discov. Today 24 (2019) 2332–2340.
[6] X. Sun, et al., Hypoxia-mediated cancer stem cell resistance and targeted therapy, [29] Z. Li, Q. You, X. Zhang, Small-molecule modulators of the hypoxia-inducible factor
Biomed. Pharmacother. 130 (2020), 110623. pathway: development and therapeutic applications, J. Med. Chem. 62 (2019)
[7] T.K. Choueiri, W.G. Kaelin, Targeting the HIF2–VEGF axis in renal cell carcinoma, 5725–5749.
Nat. Med. 26 (2020) 1519–1530. [30] A. Poch, et al., Molecular modelling predicts that 2-methoxyestradiol disrupts HIF
[8] M.C. Bosco, G. D’Orazi, D. Del Bufalo, Targeting hypoxia in tumor: a new function by binding to the PAS-B domain, Steroids 144 (2019) 21–29.
promising therapeutic strategy, J. Exp. Clin. Cancer Res. 39 (2020) 1–3. [31] K.D. Courtney, et al., Phase I dose-escalation trial of PT2385, a first-in-class
[9] M. Liu, et al., Novel therapeutic targets for hypoxia-related cardiovascular diseases: hypoxia-inducible factor-2α antagonist in patients with previously treated
the role of HIF-1, Front. Physiol. 11 (2020) 774. advanced clear cell renal cell carcinoma, J. Clin. Oncol. 36 (2018) 867.
[10] W. Tang, G. Zhao, Small molecules targeting HIF-1α pathway for cancer therapy in [32] E.M. Wallace, et al., A small-molecule antagonist of HIF2α is efficacious in
recent years, Bioorg. Med. Chem. 28 (2020), 115235. preclinical models of renal cell carcinoma, Cancer Res. 76 (2016) 5491–5500.
[11] T. Kumar, R. Pandey, N.S. Chauhan, Hypoxia inducible factor-1α: the curator of gut [33] J. Key, et al., Principles of ligand binding within a completely buried cavity in
homeostasis, Front. Cell. Infect. Microbiol. 10 (2020) 227. HIF2α PAS-B, J. Am. Chem. Soc. 131 (2009) 17647–17654.
[12] G.L. Semenza, The genomics and genetics of oxygen homeostasis, Annu. Rev. [34] M. Masetti, F. Falchi, M. Recanatini, Protein dynamics of the HIF-2α PAS-B domain
Genom. Hum. Genet. 21 (2020) 183–204. upon heterodimerization and ligand binding, PLoS One 9 (2014), e94986.
[13] A. Barzegari, et al., Nesting and Fate of Transplanted Stem Cells in Hypoxic/ [35] A.S. Chong, P.C. Anderson, Molecular dynamics simulations of the hypoxia-
ischemic Injured Tissues: the Role of HIF1α/sirtuins and Downstream Molecular inducible factor PAS-B domain confirm that internally bound water molecules
Interactions, Biofactors, 2020. function to stabilize the protein core for ligand binding, Biochemistry 59 (2019)
[14] Q. Yao, M. Parvez-Khan, E. Schipani, In vivo survival strategies for cellular 450–459.
adaptation to hypoxia: HIF1α-dependent suppression of mitochondrial oxygen [36] Y.-J. Chen, B.-Y. Huang, C.-N. Yang, Molecular modeling on HIF-2α–ARNT dimer
consumption and decrease of intracellular hypoxia are critical for survival of destabilization caused by R171A and/or V192D mutations in HIF-2α, J. Mol.
hypoxic chondrocytes, Bone 140 (2020), 115572. Graph. Model. 79 (2018) 35–45.
[15] A. Kalousi, et al., Casein kinase 1 regulates human hypoxia-inducible factor HIF-1, [37] A.H. Marceau, et al., An order-to-disorder structural switch activates the FoxM1
J. Cell Sci. 123 (2010) 2976–2986. transcription factor, Elife 8 (2019), e46131.
[16] L. Badura, et al., An inhibitor of casein kinase Iε induces phase delays in circadian
rhythms under free-running and entrained conditions, J. Pharmacol. Exp.
Therapeut. 322 (2007) 730–738.

You might also like