You are on page 1of 33

Article

Structures of the entire human opioid receptor


family
Graphical abstract Authors
Yue Wang, Youwen Zhuang,
Jeffrey F. DiBerto, ..., Yi Jiang,
Bryan L. Roth, H. Eric Xu

Correspondence
zhuangyouwen@simm.ac.cn (Y.Z.),
bryan_roth@med.unc.edu (B.L.R.),
eric.xu@simm.ac.cn (H.E.X.)

In brief
Structures of all four human opioid
receptors with the endogenous or
exogenous opioid peptides are reported,
which reveal the conserved mechanisms
of opioid receptor activation and specific
recognition of opioid peptides by the
receptors.

Highlights
d Structures of the entire family of human opioid receptors with
opioid peptides

d The N-terminal ‘‘YGGF’’ message motif of opioid peptides


drives receptor activation

d The C-terminal sequence variations of opioid peptides


address receptor selectivity

d Extracellular loop 2 is key for opioid receptors to filter


specific opioid peptides

Wang et al., 2023, Cell 186, 413–427


January 19, 2023 ª 2022 Elsevier Inc.
https://doi.org/10.1016/j.cell.2022.12.026 ll
ll

Article
Structures of the entire
human opioid receptor family
Yue Wang,1,2,8 Youwen Zhuang,1,8,* Jeffrey F. DiBerto,3,8 X. Edward Zhou,4 Gavin P. Schmitz,3 Qingning Yuan,1,5
Manish K. Jain,3 Weiyi Liu,1,2 Karsten Melcher,4 Yi Jiang,1,6 Bryan L. Roth,3,* and H. Eric Xu1,2,7,9,*
1The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica,

Chinese Academy of Sciences, Shanghai 201203, China


2University of Chinese Academy of Sciences, Beijing 100049, China
3Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
4Department of Structural Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
5The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai

201203, China
6Lingang Laboratory, Shanghai 200031, China
7School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
8These authors contributed equally
9Lead contact

*Correspondence: zhuangyouwen@simm.ac.cn (Y.Z.), bryan_roth@med.unc.edu (B.L.R.), eric.xu@simm.ac.cn (H.E.X.)


https://doi.org/10.1016/j.cell.2022.12.026

SUMMARY

Opioids are effective analgesics, but their use is beset by serious side effects, including addiction and res-
piratory depression, which contribute to the ongoing opioid crisis. The human opioid system contains four
opioid receptors (mOR, dOR, kOR, and NOPR) and a set of related endogenous opioid peptides (EOPs),
which show distinct selectivity toward their respective opioid receptors (ORs). Despite being key to the
development of safer analgesics, the mechanisms of molecular recognition and selectivity of EOPs to
ORs remain unclear. Here, we systematically characterize the binding of EOPs to ORs and present five
structures of EOP-OR-Gi complexes, including b-endorphin- and endomorphin-bound mOR, deltorphin-
bound dOR, dynorphin-bound kOR, and nociceptin-bound NOPR. These structures, supported by
biochemical results, uncover the specific recognition and selectivity of opioid peptides and the conserved
mechanism of opioid receptor activation. These results provide a structural framework to facilitate rational
design of safer opioid drugs for pain relief.

INTRODUCTION various physiological effects including intrinsic analgesia.14–16


Nearly all known EOPs are derived from proteolytic cleav-
The endogenous opioid system (EOS), composed of four opioid age of the inactive protein precursors, including proenke-
receptors (ORs) and their endogenous opioid peptide (EOP) phalin (PENK), proopiomelanocortin (POMC), prodynorphin
ligands, is an evolutionarily conserved innate response system (PDYN), and prepronociceptin (PNOC), by endopeptidases and
for nociception. The EOS is widely distributed in the central and carboxypeptidases.1,16 Among them, the well-known b-endor-
peripheral nervous systems, as well as in the immune system.1,2 phin, which preferentially binds to mOR and dOR, is the proteo-
In addition to pain control, the EOS also participates in a broad lytic product of POMC; dynorphins, which have high selectivity
range of important biological activities such as reward process- for kOR, are the hydrolysis products of PDYN; and nociceptin/
ing,3,4 perception,5 mood control,6 responses to stress,2,7 regula- orphanin FQ (NFQ), which selectively targets NOPR, is the
tion of food intake,8 and modulation of immune response.9 ORs cleavage product of PNOC.17–19 Endomorphins, such as
are a group of G protein-coupled receptors (GPCRs), comprising endomorphin-1 (YPWF), display high specificity toward mOR
mOR, dOR, kOR, and nociceptin receptor (NOPR), all of over the other ORs,20 but the precursors of endomorphins
which transduce signaling primarily through the inhibitory Gi/o pro- remain unknown.16 A common feature of EOPs is that most of
teins.10 ORs can be stimulated not only by EOPs but also by exog- them share a conserved N-terminal opioid motif of YGGF, except
enously administered opioid compounds, thus blocking pain for NFQ having a specific FGGF motif, to mediate the ‘‘message-
signal transduction in both the central and peripheral nervous sys- address-efficacy’’ mechanism upon ligand recognition.18,21,22 In
tems, resulting in the ultimate analgesic effect.11–13 addition to EOPs, ORs also respond to exogenous opioid pep-
EOPs, including enkephalins, endorphins, dynorphins, endo- tides, such as casomorphin from food or deltorphins from
morphins, and nociceptin, act as neurotransmitters to execute amphibian skin.23–25

Cell 186, 413–427, January 19, 2023 ª 2022 Elsevier Inc. 413
ll
Article

Clinically used opioid drugs, which mimic the analgesic func- NFQ showed over a hundred times selectivity to their corre-
tions of EOPs, continue to be the most effective and widely used sponding receptors, mOR, dOR, kOR, and NOPR, respectively,
therapeutics for analgesia, especially those targeting mOR such over other ORs (Figures 1A–1C), which is consistent with radio-
as morphine, oxycodone, and fentanyl. However, their clinical ligand binding data (Figure S1B). However, b-endorphin showed
application is heavily restricted due to the severe side effects comparable potency toward mOR and dOR and about ten times
of respiratory depression and addiction, which have led to the less potency toward kOR and NOPR. These data are consistent
widespread ‘‘opioid crisis’’ in the USA.26,27 Many efforts have with previous studies on the selective activation of divergent
been made to develop effective opioid analgesics without un- ORs by these opioid peptides.17,18 We then individually assem-
wanted side effects.28 Attention has also been focused on selec- bled the ORs-Gi signaling complexes with their selective opioid
tive agonists of kOR and dOR, although the development of peptide agonists, purified the complexes to homogeneity, and
opioid drugs targeting kOR and dOR was halted owing to the determined the structures by using the single-particle cryo-EM
serious side effects, including hallucinations associated with method (Figures 1D–1H and S1). The single-chain antibody
kOR5,29 and convulsions and seizures with dOR.30 It was sug- scFv16 was used to stabilize the Gai and Gb interface. The struc-
gested that the G protein-biased opioids were ideal analgesics tures of b-endorphin- and endomorphin-bound mOR-Gi, deltor-
with fewer side effects;28 however, this hypothesis was chal- phin-bound dOR-Gi, dynorphin-bound kOR-Gi, and NFQ-bound
lenged by the findings that side effects like respiratory depres- NOPR-Gi were determined at the resolution range of 3.0–3.3 Å
sion remained in animal studies and clinical trials using G pro- (Figures S2 and S3). The EM maps enabled us to unambiguously
tein-biased mOR agonists.31,32 Peptide drugs have been model side chains for most portions of ORs, Gi proteins, scFv16
developed to target many GPCRs.33,34 However, the develop- antibody, b-endorphin residues from Y1 to A21, dynorphin resi-
ment of peptide drugs of ORs with potent pain-relief activities dues Y1 to I8, NFQ residues F1 to L14, and the whole molecules
is hindered by an incomplete molecular understanding of the of endomorphin and deltorphin. Interestingly, we found that both
interplay among opioid peptides and their receptors. mOR-Gi and dOR-Gi complexes formed anti-parallel homo-
In addition, selective NOPR agonists and bi-functional ago- dimers but with distinct interaction patterns in the dimer inter-
nists acting on both mOR and NOPR have shown promise as faces (Figure S4A). In the mOR homodimer, the dimer interface
non-addictive analgesics.35,36 However, the mechanism of is surrounded by six cholesterols, which interacted with residues
NOPR activation remains elusive, impeding the discovery of from TM4, TM5, ICL2, and ECL2, with an orientation angle at
NOPR-based opioid drugs. Here, we report human mOR-Gi com- about 180 between two monomers (Figure S4B). In contrast,
plex structures bound to the endogenous non-selective peptide the dimer interface of the dOR homodimer was mainly packed
b-endorphin1–31, the longest form of b-endorphin, as well as the through residues from TM5 and TM6 without additional choles-
mOR-selective agonist endomorphin-1. To clarify the selective terols, and the two monomers adopted a clockwise rotation rela-
mechanisms of OR subtype activation, we also determined tive to each other of 160 when measured according to the
structures of the dynorphin A1–13-bound kOR-Gi complex, the conformational change of TM5 (Figure S4C). These anti-parallel
NFQ-bound NOPR-Gi complex, as well as the [D-Ala2] deltorphin homodimers are presumably not physiologically relevant but
II-bound dOR-Gi complex. (For simplicity, we used b-endorphin, possibly a result of in vitro complex assembly.
endomorphin, deltorphin, dynorphin and NFQ referring to
b-endorphin1–31, endomorphin-1, [D-Ala2] deltorphin II, dynor- Distinct binding of b-endorphin and endomorphin to mOR
phin A1–13, and nociception/orphanin FQ.) These structures pro- As endogenous opioids, endomorphin showed much improved
vide molecular insights into how endogenous and exogenous selectivity and potency to mOR when compared with b-endor-
opioid peptides selectively recognize and activate ORs. phin. In our structures, both b-endorphin and endomorphin in-
serted deeply into the orthosteric binding pocket (OBP) with
RESULTS the N terminus-inside mode, identical to that of DAMGO (Fig-
ure 2A). Despite the fact that the structures of b-endorphin-
Overall structures of opioid-peptide-bound ORs and endomorphin-bound mOR are quite similar, with root-
We first characterized the binding and activation profiles of the mean-square deviation (RMSD) at 0.4 Å for the Ca atoms of
opioid peptides used in this study for the four human ORs, mOR, the interaction patterns of these two peptides with mOR
mOR, dOR, kOR, and NOPR, through radio-ligand binding assays are distinct.
and cell-based cAMP inhibition assays combined with biolumi- b-endorphin formed extensive amphipathic interactions with
nescence resonance energy transfer (BRET) assays to measure mOR from the bottom of the OBP to the top extracellular regions,
heterotrimeric G protein dissociation. These peptides comprised including residues from TM1/2/3, TM5/6/7, and ECL1/2. Only
b-endorphin, endomorphin, deltorphin, dynorphin, and NFQ. densities of Y1-A21 of b-endorphin could be observed, indi-
[D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO), a selective cating the flexible conformation of the remaining C-terminal frag-
mOR agonist, was utilized as a reference compound. All of these ment. The N-terminal fragment, known as the ‘‘opioid motif’’
peptides are endogenous except for deltorphin; thus, we also (YGGF), was well overlaid with DAMGO (Figures 2A and 2B).
measured the activities of the reference ligands Leu/Met- The opioid motif mediated the majority of interactions with the
enkephalin (Figure S1), which are endogenous but not selective bottom OBP within the TMD, whereas the other residues M5-
for dOR. For kOR and NOPR, dynorphin and NFQ used here A21 stretched out of the TMD core and occupied the top region
served as appropriate endogenous references. In both cAMP of the OBP. Residues M5-A21 could further be divided into two
and Gi BRET assays, endomorphin, deltorphin, dynorphin, and parts: the C-terminal part of P13-A21 formed an a-helix turn at

414 Cell 186, 413–427, January 19, 2023


ll
Article

Figure 1. Cryo-EM structures of opioid peptide-bound ORs


(A) The pEC50 values of opioid peptides to induce cAMP inhibition in HEK293 cells expressing mOR, dOR, kOR, and NOPR. N.A., not active. The opioid motif is
highlighted in bold.
(B) Dose-dependent response curves of opioid receptors activated by opioid peptide ligands measured by cAMP accumulation assays.
(C) Gi1 dissociation concentration-response curves of peptide ligands at dOR, kOR, mOR, and NOPR. See also Table S2.
(D–H) Cryo-EM maps and corresponding models of the opioid peptide-or-Gi complex, b-endorphin-bound mOR-Gi complex (D), endomorphin-bound mOR-Gi
complex (E), deltorphin-bound dOR-Gi complex (F), dynorphin-bound kOR-Gi complex (G), and NFQ-bound NOPR-Gi complex (H). The densities of respective
ligands have been extracted and shown in surface presentation. See also Figures S2 and S3 and Table S1.

the extracellular end of TM1/2 cleft and the N-terminal part of opioid motif and the C-terminal a-helix turn (Figure 2A). The
M5-T12, which are highly hydrophilic residues and interact extended interactions formed by b-endorphin M5-A21 residues
mainly with ECL2, served as a linker between the N-terminal were absent in DAMGO and the non-peptide agonist

Cell 186, 413–427, January 19, 2023 415


ll
Article

Figure 2. Binding modes of b-endorphin and endomorphin to mOR


(A) Cross-section of the mOR binding pockets occupied by b-endorphin, endomorphin, and DAMGO.
(B) Structure superposition of b-endorphin with DAMGO (PDB: 8EFQ) and BU72 (PDB: 5C1M) within the mOR OBP revealed similar binding modes of DAMGO and
BU72 relative to the opioid motif.
(C) Polar interactions between b-endorphin and mOR.
(D) Hydrophobic interactions of the opioid motif of b-endorphin with mOR.
(E) Interactions of the C-term a helix of b-endorphin with mOR.
(F) Different binding modes between b-endorphin and endomorphin with W3 in endomorphin overlapping F4 of b-endorphin.
(G) Interactions of F4 in endomorphin with the extended hydrophobic pocket of mOR formed by residues from TM1, TM2, and TM7.
(H) Differential effects of mutations in the extended pocket that accommodates F4 of endomorphin on cAMP inhibition induced by b-endorphin and endomorphin.
Responses were normalized to 100% for the wild type (WT).
(I and J) Mutational effects on cAMP, Gi BRET, and b-arrestin2 recruitment of mOR induced by b-endorphin and endomorphin. Data shown are means ± SEM from
three independent experiments performed in technical duplicates. N.A., not active, refers to both no response and responses that can be observed but for which
reliable parameter estimates cannot be established over the tested concentration range. See also Figure S5 and Tables S3 and S4.

BU72-bound mOR structures.37,38 From the bottom to the top re- Q1262.60 and Y3287.43 (Figure 2B). It is worth noting that such
gion of the OBP, the primary amino group in Y1 of the opioid a polar interaction network is highly conserved in ligand recogni-
motif formed a salt bridge (4.5 Å) with D1493.32 (superscript tion of ORs.40–44 Indeed, mutations of D1493.32A, Q1262.60A, or
according to Ballesteros-Weinstein numbering,39 hereafter), of Y3287.43A nearly abolished downstream signaling including Gi
which the side chain is stabilized by hydrogen-bonds with and arrestin signal pathways (Figures 2 and S5; Table S3). The

416 Cell 186, 413–427, January 19, 2023


ll
Article

main-chain carbonyl of G2 and side chains of E8 and K9 hydrogen-bond interactions with Y1293.33 (Figure 3F). Spatially,
formed hydrogen bonds with W3207.35, R213ECL2/Q214ECL2, E4 occupied the same space as M5 of b-endorphin, which is
and D218ECL2, respectively (Figure 2C). Mutations of these resi- docked into a hydrophobic pocket in mOR (Figure 3F). Thus,
dues to the non-polar residue alanine showed compromised the distinct interaction patterns of E4 in deltorphin and M5 in
mOR activation by b-endorphin (Figure 2; Table S3), supporting b-endorphin might contribute to their selectivity toward dOR
the binding pose of b-endorphin in our structure. In addition to and mOR, respectively. In the C-terminal region of deltorphin,
polar interactions, at the bottom region, the side chain of Y1 the side chains of V5 and V6 are orientated toward the highly hy-
made a hydrophobic interaction network with nearby residues drophobic extracellular ends of TM6/7 and ECL3, accommoda-
Y1503.33, M1533.36, V2385.42, I2986.51, and V3026.55. The ben- ting well the hydrophobic pocket formed by residues V2816.55,
zene moiety of F4 was inserted toward TM2/3 and packed W2846.58, L3007.35, H3017.36, and I3047.39 (Figure 3G). In addi-
against residues Q1262.60, W135ECL1, V1453.28, and I1463.29 (Fig- tion, the main-chain carbonyl of V5 made direct ionic interaction
ure 2D). At the top region, the a-helix turn formed hydrophobic with R291ECL3, leading to the more inward movement of the
interactions with M671.29, I711.33, L1312.65, and M1322.66 extracellular ends of TM6/7 and ECL3 to the OBP compared
(Figure 2E). with the reported dOR crystal structures in complex with selec-
Endomorphin is a tetrapeptide agonist of YPWF that is tive agonists DPI-287 and KGCHM07 (Figure S4D). Unlike the
different from ‘‘traditional’’ EOPs in its amino acid length and bulkier benzamide moiety in DPI-287 or the bistrifluoromethy-
sequence. In the structure, the tetrapeptide YPWF of endomor- lated benzyl moiety in KGCHM07, the hydrophobic side chains
phin docked into the mOR orthosteric pocket and showed of V5 and V6 of deltorphin showed less space constraints to
several differences compared with b-endorphin (Figure 2A). TM6/7, which may facilitate deltorphin to induce inward move-
First, the orientation of the whole peptide was redirected by ment of the extracellular sides of TM6/7 (Figure S4D).
the bulky side chain of W3, leading to a closer salt bridge The C-terminal EVVG fragment of deltorphin formed a short
connection (3.5 Å) between the primary amine group of helix below ECL3 of dOR, which contributed to the selectivity
Y1 and D1493.32 (Figure 2F). Next, residue W3 but not F4 of of deltorphin to dOR over mOR (Figure 3H). Structural compari-
endomorphin engaged in more extensive hydrophobic interac- son of deltorphin-bound dOR with b-endorphin-bound mOR re-
tions with the hydrophobic pocket between TM2 and TM3, vealed that the positively charged K3056.58 and the rigid ECL3
similar to that formed by F4 of b-endorphin (Figure 2F). In addi- of mOR would be energetically unfavorable for deltorphin binding
tion, F4 of endomorphin formed an extended hydrophobic (Figure 3H). Consistently, mutations of K3056.58I/W or replace-
pocket composed of residues Y771.39, Y1302.64, H3217.48, and ment of ECL3 with that of dOR in mOR increased the potency
I3247.39, which was not occupied by the opioid motif of b-endor- of deltorphin to induce Gi activation of the mutated mOR receptor
phin, and mutants of the extended pocket have more effects on (Figure 3I). Moreover, at the top region of dOR OBP, the main-
endomorphin than b-endorphin (Figures 2G and 2H). Together, chain carbonyl of V6 and the basic amine group of amidated
these factors contribute to the higher potency and selectivity of G7 made additional ionic interactions with K1082.63 and the
endomorphin toward mOR than that of b-endorphin. main-chain carbonyl of C198ECL2, respectively, facilitating the
potent binding of deltorphin to dOR (Figure 3H).
Unique binding of deltorphin to dOR
Although several structures of dOR bound with either antagonist Full activation state of dOR by deltorphin
or agonist were reported,41,45–46 the molecular features of dOR Our structure of deltorphin-bound, Gi-coupled dOR revealed
activated by subtype-selective opioid peptides were poorly several noticeable differences in ligand-induced conformational
investigated. The exogenous opioid peptide [D-Ala2] deltorphin changes at the cytoplasmic regions from the dOR structures
II, referred as deltorphin in this paper, is a derivative of deltor- determined by crystallography45,48 (Figure S4D). Compared
phins, the naturally occurring opioid peptides from the skin of with the crystal structure of agonist KGCHM07-bound dOR,77
Phyllomedusa species. Deltorphin showed high selectivity the intracellular ends of TM5 and TM6 in our structure adopted
toward dOR over the other ORs.47 The overall structure of deltor- a 3.5 Å less outward shift in TM5 and 6.9 Å greater outward
phin-bound dOR was similar to b-endorphin and endomorphin- movement in TM6 as measured at the Ca atoms of R2415.66
bound mOR (Figure 3A), especially in the intracellular parts of and K2526.26 (Figure S4G). Moreover, we also observed that
TM5/6 (Figure 3B), whose conformational changes were pivotal the cytoplasmic end of TM7 of our structure, especially in the
for activation of GPCRs. Despite the structural similarity, the NP7.50xxY motif, adopted an apparent counter-clock rotation
OBP of dOR (702 Å3) was smaller than that of mOR (839 Å3) relative to that of KGCHM07-bound dOR. The more pronounced
(Figure 3C). TM7 rotation might be caused by the distinct rearrangement of
The whole deltorphin molecule was buried deeply in the OBP the polar network of the DQY motif (Figure S4F). As a result,
of dOR, with each of its residues forming extensive interactions the Y3187.53 in the NP7.50xxY motif swings its side chain upward
with dOR. Recognition of deltorphin in the dOR OBP could be near TM6, thus opening the space for the bulky side chain of
separated into two regions, of which the N-terminal Y[D-Ala]F R1463.50 from the conserved DRY motif. The much more rotated
fragment occupied the bottom pocket similar to YGGF of the NP7.50xxY motif in TM7, together with the reconstruction of the
opioid motif in mOR and the C-terminal EVVG fragment sat on DRY motif, led to the greater outward movement at the cyto-
the top region formed by the extracellular ends of TM2/6/7 and plasmic end of TM6 and larger opening of the intracellular cavity
ECL2/3 (Figures 3D, 3E, and 3J). In the C-terminal region of del- for Gi anchoring (Figure S4H). Thus, we propose that the deltor-
torphin, E4 formed close ionic interactions with K2145.39 and phin-bound dOR structure was in full active conformation,

Cell 186, 413–427, January 19, 2023 417


ll
Article

Figure 3. Interaction and selectivity of deltorphin toward dOR


(A–C) Structure alignment of b-endorphin-/endomorphin-bound mOR with deltorphin-bound dOR. (A) overall superposition of mOR and dOR structures; (B and C),
structural comparison of the extracellular regions (B) and the cytoplasmic regions (C) of mOR and dOR.
(D) The binding mode of deltorphin in the dOR pocket, which is superposed to that of the YGGY motif of b-endorphin in the mOR pocket. The red dashed circle
indicates the Y[D-Ala]F fragment of deltorphin, which is well overlaid with the YGGF motif in b-endorphin.
(E) Detailed interactions of the N-terminal Y[D-Ala]F fragment of deltorphin with dOR.
(F) Comparison of the interactions of E4 in deltorphin with dOR to the interactions of M5 in b-endorphin with mOR.
(G) Detailed interactions of the C-terminal fragment EVVG of deltorphin with dOR.
(H) Specific interactions of deltorphin with ECL3 and W2846.58 of dOR (pink). Corresponding interactions were not observed in mOR (cyan).
(I) Effects of wild-type and mutated mOR with K3056.58I/W mutations or with dOR ECL3 swap on cAMP inhibition induced by deltorphin. Data shown are means ±
SEM from three independent experiments performed in technical duplicate. N.A., not active, refers to both no response and responses that can be observed but
for which reliable parameter estimates cannot be established over the tested concentration range. See also Figure S5 and Table S5.

whereas the crystal structures, which had less outward move- ments of dynorphin had comparable potency as the N-terminal
ment of TM6 and less rotation of TM7, were in the intermediate 13 residue fragment in activation of kOR.21,49 Compared with
active state. the crystal structure of kOR bound to opioid-like agonist
MP1104,50 the dynorphin-bound kOR structure adopts a highly
Recognition of dynorphin by kOR similar receptor conformation with an RMSD of 0.8 Å for the
Dynorphin is an EOP whose N-terminal 13 residue fragment can entire receptor (Figure 4A). The N-terminal YGGF motif of dynor-
activate the OR family except for NOPR (Figures 1A–1C), never- phin occupied the similar bottom pocket of kOR as that by
theless, it has at least 100-fold higher potency toward kOR over MP1104. The primary amine group of Y1 mimicked the role of
other ORs. In the dynorphin-bound kOR structure, the entire 13 the tertiary amine group of MP1104, forming an extensive polar
residue peptide is organized vertically into the OBP of kOR, with network with the DQY motif and T1112,51 (Figure 4B). Unlike
only the N-terminal eight residues visible in the EM density map. MP1104, the C-terminal LRRI fragment of dynorphin made addi-
This is consistent with the fact that the first eight residue frag- tional interactions with the top region of the kOR OBP, with its

418 Cell 186, 413–427, January 19, 2023


ll
Article

Figure 4. Recognition of dynorphin by kOR


(A) Structure superposition of dynorphin-kOR with MP1104-kOR (PDB: 6B73).
(B) The conserved polar interactions between DQY motif of kOR with dynorphin and MP1104.
(C) The polar interactions between R6 in dynorphin with kOR.
(D) The interactions between R7-I8 in dynorphin with kOR.
(E) Electrostatic surface representation of the opioid peptides and extracelluar parts of receptors.
(F) The effects of kOR mutations in the extended binding pocket for dynorphin on cAMP inhibition induced by dynorphin. Data shown are means ± SEM from three
independent experiments performed in technical duplicate.
(G) The increased effects of K3056.58E of mOR mutation on cAMP inhibition induced by dynorphin. Data shown are means ± SEM from three independent ex-
periments performed in technical duplicate.
(H) Mutational effects on cAMP, Gi BRET, and b-arrestin2 recruitment of kOR induced by dynorphin. Data shown are means ± SEM from three independent
experiments performed in technical duplicate. N.A., not active, refers to both no response and responses that can be observed but for which reliable parameter
estimates cannot be established over the tested concentration range. See also Figure S5 and Table S6.
(I) Steric clash of Y3137.38 in kOR with V6 in deltorphin and F4 in b-endorphin.
(J) Steric clash of R7 in dynorphin with K3056.58 in mOR and W2846.58 in dOR.
Responses were normalized to 100% for kOR WT.

two positively charged residues R6 and R7 forming direct salt kOR might explain its high potency for kOR despite the fact
bridges with E209ECL2 and E2976.58, which are at opposite sides that dynorphin has a relatively flexible N-terminal YGGF motif
of the extracellular vestibule of kOR (Figures 4B–4D). Mutations compared with the rigid chemical scaffold of the morphinan
of E209ECL2 and E2976.58 to alanine or lysine, which disrupted the agonist MP1104.
ionic interactions, compromised the ability of dynorphin to The dynorphin-bound kOR structure also explained the
induce Gi activation by kOR, corroborating the effect of alanine 100-fold selectivity of dynorphin for kOR over mOR and dOR.
substitutions of R6 and R7 of dynorphin52 (Figures 4F and 4H; All three ORs shared the same set of residues in the bottom re-
Table S6). The more extended interactions of dynorphin with gions of OBP occupied by their respective subtype-selective

Cell 186, 413–427, January 19, 2023 419


ll
Article

peptide agonists. However, their extracellular vestibules, which iations from that of the other ORs, contributing to the selectivity
constituted the top region of the OBP, were non-conserved of NFQ toward NOPR. The T5-L14 region of NFQ made both hy-
and differed in charge distribution, especially in the ECL2/3 drophobic and electrostatic interactions with the extracellular
and extracellular ends of TM6/7 regions (Figure 4E). A structural side of NOPR (Figures 5E–5G). The bulky side chains of posi-
alignment revealed that the extracellular regions of mOR and tively charged residues of NFQ, including R8, K9, R12, and
dOR, corresponding to the R6–R7 binding pockets in kOR, K13, stretched toward TM2 and ECL2 regions of NOPR and
were positively charged or highly hydrophobic, which are formed extensive salt bridge interactions with negatively
incompatible for dynorphin binding. Consistently, the K3056.58E charged residues D1102.63, E194ECL2, and E199ECL2(Figure 5F).
mutation in mOR increased the potency of dynorphin to mOR by Individual mutations of these three residues to alanine dimin-
100-fold (Figure 4G). Moreover, the replacement of ECL2 of ished NFQ-induced NOPR activation (Figures 5N and S5;
kOR with that of mOR decreased both the potency and activation Table S7).
efficacy of dynorphin (Figure 4F). The bulky side chain of The ECL2 region is important for the selectivity of kOR and
Y3137.36 in kOR is predicted to have steric clashes with endo- NOPR to dynorphin and NFQ. Despite the common negatively
morphin and deltorphin, which is highly selective for mOR and charged feature, the distribution of acidic residues of ECL2 in
dOR, respectively. It should be noted that the bulky side chains kOR and NOPR differs, leading to the divergent placement of
of ECL3 residues in dOR would have steric clashes with the the long-extended side chains of basic residues in dynorphin
C terminus of dynorphin (Figures 4I and 4J). Collectively, we pro- and NFQ near the ECL2 region. In addition, ECL2 of kOR is three
pose that the ECL2 and extracellular ends of TM6/7 serve as residues longer and much more inwardly shifted than that of
important selectivity filters of ORs to distinct opioid peptides. NOPR. The extra ECL1/2 residues in kOR, including N122ECL1
and R202ECL2, could have steric clashes with the bulky side
Selectivity of NFQ toward NOPR chains from the NFQ C terminus, partly accounting for its low af-
NOPR is the last discovered member of the OR family and dis- finity to NFQ (Figures 5I and 5J). Furthermore, several other non-
plays strict selectivity for its endogenous agonist NFQ over conserved residues in the extracellular part of NOPR also play a
other opioid peptides. NFQ is a highly basic opioid peptide vital role in the selectivity of NFQ, including D1102.63 in NOPR,
(Figure 5A) similar to dynorphin in amino acid composition, which makes ionic interaction with R8 of NFQ (Figure 5I). Consis-
including the C-terminal ‘‘address’’ sequence. Likewise, both tently, the V1182.63D mutation in kOR greatly increased the po-
NOPR and kOR contain 30% acidic residues in ECL2 (Fig- tency of NFQ to kOR (Figure 5K). Similarly, for mOR and dOR,
ure 5B), indicating the potentially important role of ECL2 in the topologies and charge properties of their extracellular sides
the high-affinity binding of NFQ to NOPR rather than mOR formed by ECL2, ECL3, and residue 2.63 may serve as selective
and dOR. However, NFQ differs from all the other known determinants toward NFQ (Figure 5L). For example, dOR has
EOPs in the first N-terminal residue, which is phenylalanine large, charged residues in ECL3 and greater inward movements
rather than tyrosine. Y1 is a universal feature of classical of ECL2 and ECL3, which lead to a narrower extracellular gate
EOPs that participates in a water-mediated polar network than the corresponding regions of other ORs (Figure 5M), there-
including residues Y3.33 and H6.52 in other ORs, thus facilitating fore preventing the access of longer opioid peptides, such as
the stable binding of EOPs.38,48 Nevertheless, the phenylala- NFQ and dynorphin, to the OBP of dOR.
nine substitution of tyrosine in NFQ suggested that Y1 is not
necessary for the high potency of NFQ to NOPR.53 Conserved OR chamber for opioid motif recognition
In our structure, NFQ, similar to dynorphin in kOR, is vertically Canonical EOPs, such as enkephalins, endorphins, and dynor-
inserted into the OBP of NOPR, with its N terminus toward the phins, follow the formulated ‘‘message-address’’ concept in
bottom of the OBP (Figure 5C). The NFQ fragment G6-K14 amino acid sequences.21,56,57,58 The N-terminal tetrapeptide
formed a short stable helix, consistent with a helical conforma- YGGF, known as the opioid motif, was hypothesized to be the
tion of this region as reported by NMR and modeling studies.54,55 ‘‘message region’’ and the minimum structural element for
EM densities were clearly observed for the first 14 residues of the EOP recognition and OR activation. In our structures, both the
NFQ molecule, consistent with that NFQ1–13 had comparable ac- YGGF motif from b-endorphin and dynorphin adopt a similar
tivity with the full-length NFQ.56,51 The FGGF fragment in NOPR conformation and occupy a highly aromatic pocket with nearly
shared a similar conformation with the opioid motif in other ORs the same residue composition from TM2/3/5/6/7 in the bottom
and occupied the corresponding amphipathic bottom region of transmembrane part of OBPs in mOR or kOR. Similar to the
the NOPR pocket. The conformation of F1 was stabilized YGGF fragment, we observed that YPW of endomorphin, Y
by hydrophobic interactions with Y1313.33, M1343.36, I2195.42, [D-Ala]F of deltorphin, and FGGF of NFQ all share such highly
V2796.51, and V2836.55, among which Y1313.33 formed p-stack- aromatic pocket in mOR, dOR, and NOPR. Mutations in most
ing interactions with the benzene ring of F1 (Figure 5D). Point mu- residues around the conserved hydrophobic pocket dramatically
tations of these residues largely decreased NFQ-induced NOPR diminish the potency of peptide ligands to their respective
activation (Figures 5N and S5; Table S7), consistent with the ORs (Figure 6E). In addition, the synthesized YGGF peptide
result of a previous mutagenesis study.42 The primary amine alone can activate all the four types of ORs with various
group of F1, similar to Y1 in other EOPs, formed a salt bridge potencies (Figure 6F). We thus propose that the common hydro-
with the conserved residue D3.32 from the DQY motif (Figure 5D). phobic pocket in the bottom region of the OBP of ORs serves as
In contrast to the conserved bottom opioid pocket, the extracel- a conserved chamber for opioid ligand recognition and activa-
lular part of NOPR occupied by T5-L14 of NFQ showed great var- tion of ORs.

420 Cell 186, 413–427, January 19, 2023


ll
Article

Figure 5. Selectivity of NFQ toward NOPR


(A) Electrostatic surface representation of NOPR and NFQ.
(B) Sequence alignment of ECL2 of opioid receptors. Positively and negatively charged amino acids are shown in blue and red, respectively.
(C) Structure of NFQ-bound NOPR.
(D–G) Detailed interactions from N terminus to C terminus of NFQ with NOPR. Hydrogen bonds are shown as yellow dashed lines.
(H–J) Superposition of dynorphin-bound kOR with NFQ-bound NOPR showed that ECLs of kOR cause steric clashes with NFQ. (H) Overall steric collision of NFQ
with kOR. (I and J) specific collision of NFQ with kOR in ECL1 (I) and in ECL2 (J).
(K) Effects of kOR mutations with corresponding residues of NOPR on cAMP inhibition induced by NFQ. Data shown are means ± SEM from three independent
experiments performed in technical duplicate.
(L and M) Extracellular view of mOR and dOR shows a steric hindrance with NFQ that prevents NFQ binding, similar to the steric hindrance of NFQ with kOR. The
pink dashed oval highlights the narrower gate that is formed by dOR.
(N) Mutational effects on cAMP, Gi BRET, and b-arrestin2 recruitment of NOPR induced by NFQ. Data shown are means ± SEM from three independent ex-
periments performed in technical duplicate. N.A., not active, refers to both no response and responses that can be observed but for which reliable parameter
estimates cannot be established over the tested concentration range. See also Figure S5 and Table S7.

Consensus activation mechanism of ORs tion mechanism of NOPR. Structural alignment revealed relative
Since the antagonist-bound NOPR structure was available,42 the conformational changes similar to that observed in mOR, dOR,
active NFQ-NOPR structure enabled us to investigate the activa- and kOR, mainly characterized by the inward movement of

Cell 186, 413–427, January 19, 2023 421


ll
Article

Figure 6. The conserved chambers for opioid peptide recognition in ORs


(A–D) Conformational changes of the conserved motifs during opioid receptor activation. Upper panel: active opioid receptor structures determined in this study.
Lower panel: inactive opioid receptor structures determined previously. mOR (PDB: 4DKL), dOR (PDB: 4N6H), kOR (PDB: 4DJH), and NOPR (PDB: 4EA3).
(E) The fold of changes of EC50 and Emax of mutations on extremely conserved and important residues interacting with the YGGF motif. Data shown are means ±
SEM from three independent experiments performed in technical duplicate. N.A., not active, refers to both no response and responses that can be observed but
for which reliable parameter estimates cannot be established over the tested concentration range.
(F) cAMP inhibition curves of four opioid receptors induced by a synthetic YGGF peptide. Data shown are means ± SEM from three independent experiments
performed in technical duplicate.

TM5 and outward movement of TM6 in their intracellular parts tein. The overall structures of OR-Gi complexes are quite
and clockwise rotations of TM7 in the active NOPR structure similar to each other, especially the Gi coupling interfaces.
relative to the inactive structure. It was suggested for mOR, The intracellular parts of Gi coupled ORs were well overlapped
kOR, and dOR that such obvious conformational differences (Figures 7A and 7B), which was expected from the highly
could be attributed to the ligand-induced collapse of the poten- similar activation chamber in OBPs and conserved activation
tial Na+ pocket and reconstruction of the polar network beneath mechanism of ORs. The interaction interfaces of ORs and Gi
the OBP, the toggle switch in CWxP motif, and the rearrange- all shared a similar set of residue composition, including resi-
ment of the PIF core triad and subsequent conformational dues in TM3, TM5, TM6, ICL2, and ICL3 of ORs, and residues
changes in NPxxY motif of TM7 and DRY motif of TM3 in aN, b2-b3 loop, b6, and a5 helix of Gai (Figure S6), as
(Figures 6A–6D). Superposition of the active structures of all observed in the DAMGO-bound mOR-Gi structure.38 The a5 he-
four subtypes of ORs showed that the residue conformation of lix of Gai inserted into the intracellular cavities of ORs, providing
these conserved motifs, which are important for receptor activa- the major interface module for receptor binding, in a manner
tion, overlapped well. In addition, the intracellular parts of all ORs resembling other structures of GPCR-G protein complexes.
could also be well aligned. These structural observations indi- The above observations suggested that ORs adopt a highly
cate that the ORs share a similar consensus activation conserved mode of coupling to Gi. Two Gi conformational
mechanism. states, the canonical and non-canonical states, were observed
in previously reported NTSR1-Gi59 and GHSR-Gi structures.60
G protein coupling of ORs In our OR-Gi structures, the Gi binding modes are all in the ca-
Structure superimposition enabled the elucidation of the nonical state as observed in most of the published GPCR-Gi
coupling features of ORs with the downstream Gi signaling pro- signaling complexes.

422 Cell 186, 413–427, January 19, 2023


ll
Article
Figure 7. G protein coupling of opioid re-
ceptors
(A) Superposition of four opioid receptors coupled
with opioid peptides and Gi heterotrimer.
(B) Intracellular views of the opioid receptors.
(C) Conformational shift of Ga5 (left) and GaN (right)
of opioid receptor-bound Gai.
(D) Hydrophobic interactions of ICL2 of opioid re-
ceptors with Gai.
(E) Ionic patch of R56.50 and K6.26 of opioid re-
ceptors with Gai.
(F) Alignment of opioid receptor residues that in-
teracted with Gai.

pharmacological understanding of ligand


recognition and receptor activation by
small opioid molecules and related anal-
gesic medications.18,19,63 As endogenous
analgesics of ORs, the EOPs represent
an alternative strategy for drug develop-
ment to manage pain and other diseases
caused by EOS dysfunction, such as
drug abuse and mood disorders. Despite
the fact that the binding of EOPs to ORs
has been widely investigated since the first
Despite the high similarities in structure and residue composi- discovery of EOPs in the 1970s,16 a structural understanding of
tions in the intracellular regions of ORs, structural differences how EOPs preferentially recognize and activate their respective
were observed in the Gi conformation, characterized by an about ORs has remained elusive. In this study, we have reported the
2 Å shift of the a5 helix (Figure 7C) (measured at the Ca atoms of structures of ORs-Gi signaling complexes activated by opioid
N331G.H5.03, superscripts refer to the common Ga numbering peptides, including four EOPs, b-endorphin, endomorphin, dy-
[CGN] system, hereafter61) and an about 3.0 Å to 6.0 Å shift of norphin, and NFQ, and the exogenous opioid peptide deltorphin.
the aN helix (Figure 7C) (measured at the Ca atoms of Structural comparisons indicated that all the opioid peptides
K10G.HN.34). The structural changes of Gi protein varying from occupy a highly conserved activation chamber at the bottom
OR subtypes may be due to the subtle conformational differ- of the OBPs of ORs, which is engaged by the opioid motif,
ences in the intracellular ends of TM5/6 and ICL2/3 induced by YGGF. Mutations of residues of the activation chamber dis-
opioid peptide ligands. played much more significant effects on receptor activation
ORs were known to preferentially activate G proteins of the Gi/o compared with other residues in the OBPs, suggesting the indis-
family over other G protein subtypes, such as Gs and Gq.62 In pensable role of the conserved chamber in the recognition and
addition to the steric effect of the narrow intracellular cavity with pharmacological activity of ORs. The extracellular vestibules of
the bulky C terminus of Gas, it was shown that the ICL2 and ORs, including ECL2/3 and the extracellular ends of TM2/6/7,
ICL3 regions of mOR also served as important filters for its selec- are not conserved in either sequence or charged residue distri-
tivity toward Gi over Gs.38 In our structures, we found that the ICL2 bution, serving as selectivity filters of ORs for different opioid
and ICL3 of ORs shared highly similar interaction profiles with Gai. peptides and regulating the efficacies of OR activation by opioid
In ICL2, P34.50 and V/I34.51 formed a hydrophobic network with peptides. These structural findings support and extend the
L194G.S3.01, F336G.H5.08, T340G.H5.12, I343G.H5.15, and I344G.H5.16 previous message-address hypothesis for EOP binding to
from Gai (Figure 7D). The positively charged ICL3 and the nearby ORs.21,56,57,58 The extracellular vestibule is an important regu-
TM5/6 residues formed ionic patch with the negatively charged lator of ligand binding properties of many GPCRs. It was shown
residues from the a4-b6 loop and a5 helix of Gai (Figure 7E). It is that the electrostatic and conformational differences in the extra-
worth noting that the a4-b6 loop is non-conserved in the distribu- cellular regions, including ECL2 and ECL3, were critical for the
tion of charged residues among Gi, Gs, and Gq (Figures S7A– selective recognition of formyl peptide receptors, FPR1 and
S7C); thus, the charged complementation of ICL3 and intracellular FPR2, toward formyl peptide ligands.64 Moreover, in aminergic
ends of TM5/6 with Gi could partially account for the G protein GPCRs, the extended binding pockets located in the less
selectivity of ORs toward Gi over other G protein subtypes. conserved extracellular vestibules were suggested to be deter-
minants for ligand selectivity and affinity of these receptors.65–67
DISCUSSION Therefore, the extracellular vestibule may act as a common regu-
lator for efficient and selective ligand binding of GPCRs.
ORs represent the major drug targets for pain-relief treatment.13 The structure of NFQ-bound NOPR reported in this study
Long-lasting efforts have been made to clarify the structural and enabled a comprehensive understanding of the activation

Cell 186, 413–427, January 19, 2023 423


ll
Article

mechanisms of ORs. Structural comparisons showed that the the opioid YGGF motif and unique structural features of ECL2
important structural motifs related to OR activation, including and ECL3 for the selectivity of the extended opioid peptides.
the CWxP motif, PIF core triad, sodium pocket, DRY motif, and The structures also reveal a universal activation mechanism for
NPxxY motif, underwent nearly the same conformational all four opioid ORs and specific structural components such as
changes from inactive to active states. The conformations of ICL3 for their selective coupling to the Gi protein subtype.
these motifs could be superimposed in all four ORs, except Together, our work provides a rational framework for under-
that the NPxxY of dOR has a larger rotation relative to that of standing biology of the opioid system as well as structural tem-
mOR, kOR, and NOPR. These observations indicated that ORs plates for ligand design.
share a conserved activation mechanism, including the confor-
mational change of toggle switch residue W6.48 and rearrange- Limitations of the study
ment of DRY and NPxxY motifs, which ultimately lead to the There are several limitations in our study. First, the cryo-EM
outward movement of TM6 for G protein coupling. structures capture relatively static snapshots of ligand binding
ORs show high selectivity toward Gi/o coupling rather than poses within the receptors. The dynamic aspects of ligand bind-
other G protein subtypes. Structure alignments indicated that ing, for example, the process of ligand entry and release from the
the ICL3 of ORs existed as stable a-helix turn in Gi binding state receptor pockets, remain largely unanswered in our study.
(Figure 7E). The stable interaction of ICL3 with Gai is attributed to Future NMR studies and molecular simulations will be required
not only the extended hydrophobic interactions but also the to study these dynamic aspects of ORs. Second, the develop-
electrostatic interactions from the charged patch formed by ment of peptide drugs is limited by instability, low bioavailability,
the abundant positively charged residues in ICL3 and intracel- and poor blood-brain barrier penetration of peptide ligands.
lular ends of TM5/6 to the negatively charged residues of Gai in Efforts have been focused on chemical modification of peptide
a4-b6 loop and a5 helix regions. ICL3 is also visible in many other ligands or specific small molecule ligands. However, due to the
GPCR-Gi complexes, including Gi complexes of FPR268 and plasticity of ORs, the binding pockets of peptide ligands re-
CB269,70 (Figure S7A). However, since other G protein isoforms, ported in our structures may not be a good representation for
such as Gs and Gq/11, do not have the same charge properties as modified peptide ligands or small molecule ligands. To facilitate
Gi, even if ICL3 of Gs/Gq/11-coupled receptors have positively structure-based drug discovery, many more structures with
charged amino acids, they cannot form electrostatic interactions specific modified peptides or small molecule ligands will be
with Gs/Gq/11. This unstable interaction makes the structure required.
of ICL3 missing in many Gs- and Gq-coupled receptors
(Figures S7B and S7C).71–73 Thus, in addition to ICL2 and the
STAR+METHODS
outward movement of TM6, the specific ionic patch of ICL3 pro-
vides additional selectivity for ORs to couple primarily with Gi/o.
Detailed methods are provided in the online version of this paper
The structures of mOR bound to morphine and fentanyl,74 as
and include the following:
well as with lofentanil and mitragynine pseudoindoxyl,75 have
recently been determined. The overall mOR structures with d KEY RESOURCES TABLE
different ligands are nearly identical when mOR bound to d RESOURCE AVAILABILITY
morphine, fentanyl, endomorphin, or b-endorphin with the B Lead contact
RMSD of the whole receptor Ca atoms less than 0.4 Å (Fig- B Materials availability
ure S7D). In cAMP assays, fentanyl has greater potency than B Data and code availability
DAMGO, whereas endomorphin and b-endorphin are weaker d EXPERIMENTAL MODEL AND SUBJECT DETAILS
than DAMGO (Figures S7E and S7F). Structural analysis reveals d METHOD DETAILS
that fentanyl binds to an additional pocket of mOR that is not B Constructs design
occupied by endomorphin and b-endorphin, perhaps providing B Preparation of scFv16 antibody
a higher potency of fentanyl to mOR (Figures S7G and S7H). B Expression and purification of opioid receptor- Gi
Finally, ORs are reported to form homodimers or heterodimers complexes
between receptor subtypes.76,77,79 Although anti-parallel homo- B Cryo-EM grid preparation and data collection
dimers of mOR and dOR might be not physiologically relevant, it is B Image processing and 3D reconstruction
interesting to see how these dimers can form in our biochemical B Model building, structure refinement, and figure
samples. The mOR anti-parallel dimer interface is mediated by preparation
TM4 and TM5 with six cholesterols in between (Figure S4), which B cAMP accumulation assays
is different from the parallel homodimers observed in mOR crystal B Radio-ligand binding assays
structures.40 The dOR anti-parallel dimer interface is mediated B Bioluminescence resonance energy transfer assays
by TM5 and TM6, similar to the TM5-TM6 dimer interface used B Enzyme-Linked Immunosorbent Assay
in the parallel dimer of mOR, suggesting that parallel homodimers d QUANTIFICATION AND STATISTICAL ANALYSIS
of dOR or heterodimers of mOR and dOR could use the same
TM5-TM6 dimer interface.
SUPPLEMENTAL INFORMATION
In summary, we systematically characterized the binding of
several EOPs to all four ORs and determined their Gi complex Supplemental information can be found online at https://doi.org/10.1016/j.cell.
structures. Our structures reveal both a conserved pocket for 2022.12.026.

424 Cell 186, 413–427, January 19, 2023


ll
Article
ACKNOWLEDGMENTS stress response. Prog. Neuropsychopharmacol. Biol. Psychiatry 25,
729–741.
The cryo-EM data were collected at the Cryo-Electron Microscopy Research 8. Gosnell, B.A., and Levine, A.S. (2009). Reward systems and food intake:
Center, Shanghai Institute of Materia Medica (SIMM), and Advanced Center role of opioids. Int. J. Obes. (Lond) 33, S54–S58.
for Electron Microscopy, Shanghai Institute of Materia Medica (SIMM). We
9. Eisenstein, T.K. (2019). The role of opioid receptors in immune system
sincerely thank all the staff at both institutions for their assistance. This work
function. Front. Immunol. 10, 2904.
was partially supported by grants from the Ministry of Science and Technology
(China) grants (2018YFA0507002 to H.E.X.); the National Natural Science 10. McDonald, J., and Lambert, D.G. (2013). Opioid mechanisms and opioid
Foundation of China (82121005 to H.E.X. and Y.J., 32130022 to H.E.X., drugs. Anaesth. Intensive Care Med. 14, 505–509.
32171187 to Y.J.), the Shanghai Municipal Science and Technology Major 11. Bourgoin, S., Benoliel, J.J., Collin, E., Mauborgne, A., Pohl, M., Hamon,
Project (2019SHZDZX02 to H.E.X.); Shanghai Municipal Science and Technol- M., and Cesselin, F. (1994). Opioidergic control of the spinal release of
ogy Major Project (H.E.X.); the CAS Strategic Priority Research Program neuropeptides. Possible significance for the analgesic effects of opioids.
(XDB37030103 to H.E.X.); and the Special Research Assistant Project of Chi- Fundam. Clin. Pharmacol. 8, 307–321.
nese Academy of Sciences (to Y.Z.). B.L.R. is supported by the Michael Hook- 12. Yaksh, T.L. (1997). Pharmacology and mechanisms of opioid analgesic
er Distinguished Professorship, the NIMH Psychoactive Drug Screening Pro- activity. Acta Anaesthesiol. Scand. 41, 94–111.
gram and NIH Grant RO1DA055656.
13. Che, T., and Roth, B.L. (2021). Structural insights accelerate the discovery
of opioid alternatives. Annu. Rev. Biochem. 90, 739–761.
AUTHOR CONTRIBUTIONS
14. Feng, Y., He, X., Yang, Y., Chao, D., Lazarus, L.H., and Xia, Y. (2012). Cur-
rent research on opioid receptor function. Curr. Drug Targets 13, 230–246.
H.E.X. and Y.Z. initialed the project. Y.Z. and Y.W. designed and screened the
expression constructs of hORs and optimized the purification conditions of 15. Kaur, J., Kumar, V., Sharma, K., Kaur, S., Gat, Y., Goyal, A., and Tanwar, B.
protein complexes. Y.W. prepared protein samples of endomorphin-mOR-Gi- (2020). Opioid peptides: an overview of functional significance. Int. J. Pept.
scFv16, deltorphin-dOR-Gi-scFv16, and dynorphin-kOR-Gi-scFv16 com- Res. Ther. 26, 33–41.
plexes for cryo-EM grid making and data collection, performed cryo-EM 16. Fricker, L.D., Margolis, E.B., Gomes, I., and Devi, L.A. (2020). Five decades
structure determination of these protein complexes, performed cAMP assays, of research on opioid peptides: current knowledge and unanswered ques-
prepared the figures, and participated in manuscript editing. Y.Z. prepared tions. Mol. Pharmacol. 98, 96–108.
b-endorphin-mOR-Gi-scFv16 and NFQ-NOPR-Gi-scFv16 complex for cryo- 17. Corbett, A.D., Paterson, S.J., and Kosterlitz, H.W. (1993). Selectivity of
EM data collection and performed structure determination, prepared the draft ligands for opioid receptors. In Opioids (Springer), pp. 645–679.
of the manuscript with the input of Y.W., and participated in figure preparation.
18. Corbett, A.D., Henderson, G., McKnight, A.T., and Paterson, S.J. (2006).
J.F.D. performed radio-ligand binding assay, Gi BRET and arrestin BRET as-
75 years of opioid research: the exciting but vain quest for the Holy Grail.
says with the assistance of G.P.S. and M.K.J. X.E.Z. modeled and refined all
Br. J. Pharmacol. 147, S153–S162.
the structures. Q.Y. assisted in cryo-EM data collection. W.L. assisted with
cellular experiments for the cAMP assay. Y.J. supervised W.L. K.M. super- 19. James, A., and Williams, J. (2020). Basic opioid pharmacology - an up-
vised X.E.Z. and participated in manuscript editing. B.L.R. supervised J.F.D., date. Br. J. Pain 14, 115–121.
evaluated data and participated in manuscript editing. H.E.X. conceived and 20. Zadina, J.E., Hackler, L., Ge, L.J., and Kastin, A.J. (1997). A potent and se-
supervised the project and participated in manuscript editing. H.E.X. and lective endogenous agonist for the mu-opiate receptor. Nature 386,
Y.Z. wrote the manuscript with input from Y.W. and B.L.R. 499–502.
21. Chavkin, C., and Goldstein, A. (1981). Specific receptor for the opioid pep-
DECLARATION OF INTERESTS tide dynorphin: structure–activity relationships. Proc. Natl. Acad. Sci. USA
78, 6543–6547.
The authors declare no competing interests. 22. Mansour, A.,M.T., Hoversten, M.T., Taylor, L.P., Watson, S.J., and Akil, H.
(1995). The cloned mu, delta and kappa receptors and their endogenous
Received: May 31, 2022 ligands: evidence for two opioid peptide recognition cores. Brain Res.
Revised: October 11, 2022 700, 89–98.
Accepted: December 13, 2022
23. Xu, X., and Lai, R. (2015). The chemistry and biological activities of
Published: January 12, 2023
peptides from amphibian skin secretions. Chem. Rev. 115,
1760–1846.
REFERENCES
24. Vardy, E., Sassano, M.F., Rennekamp, A.J., Kroeze, W.K., Mosier, P.D.,
Westkaemper, R.B., Stevens, C.W., Katritch, V., Stevens, R.C., Peterson,
1. Benarroch, E.E. (2012). Endogenous opioid systems: current concepts
R.T., et al. (2015). Single amino acid variation underlies species-specific
and clinical correlations. Neurology 79, 807–814.
sensitivity to amphibian skin-derived opioid-like peptides. Chem. Biol.
2. Toubia, T., and Khalife, T. (2019). The endogenous opioid system: role and 22, 764–775.
dysfunction caused by opioid therapy. Clin. Obstet. Gynecol. 62, 3–10.
25. Stefanucci, A., Mollica, A., Macedonio, G., Zengin, G., Ahmed, A.A., and
3. Wang, S. (2019). Historical review: opiate addiction and opioid receptors. Novellino, E. (2018). Exogenous opioid peptides derived from food pro-
Cell Transplant. 28, 233–238. teins and their possible uses as dietary supplements: A critical review.
4. Darcq, E., and Kieffer, B.L. (2018). Opioid receptors: drivers to addiction? Food Rev. Int. 34, 70–86.
Nat. Rev. Neurosci. 19, 499–514. 26. Chan, H.C.S., McCarthy, D., Li, J., Palczewski, K., and Yuan, S. (2017).
5. Roth, B.L., Baner, K., Westkaemper, R., Siebert, D., Rice, K.C., Steinberg, Designing safer analgesics via mu-opioid receptor pathways. Trends
S., Ernsberger, P., and Rothman, R.B. (2002). Salvinorin A: A potent natu- Pharmacol. Sci. 38, 1016–1037.
rally occurring nonnitrogenous kappa opioid selective agonist. Proc. Natl. 27. Del Vecchio, G., Spahn, V., and Stein, C. (2017). Novel opioid analgesics
Acad. Sci. USA 99, 11934–11939. and side effects. ACS Chem. Neurosci. 8, 1638–1640.
6. Lutz, P.E., and Kieffer, B.L. (2013). Opioid receptors: distinct roles in mood 28. Manglik, A., Lin, H., Aryal, D.K., McCorvy, J.D., Dengler, D., Corder, G.,
disorders. Trends Neurosci. 36, 195–206. Levit, A., Kling, R.C., Bernat, V., Hübner, H., et al. (2016). Structure-based
7. Drolet, G., Dumont, E.C., Gosselin, I., Kinkead, R., Laforest, S., and Trot- discovery of opioid analgesics with reduced side effects. Nature 537,
tier, J.F. (2001). Role of endogenous opioid system in the regulation of the 185–190.

Cell 186, 413–427, January 19, 2023 425


ll
Article
29. Wang, Y.H., Sun, J.F., Tao, Y.M., Chi, Z.Q., and Liu, J.G. (2010). The role of d-opioid receptor crystal structure with peptide and small-molecule ago-
kappa-opioid receptor activation in mediating antinociception and addic- nists. Sci. Adv. 5, eaax9115.
tion. Acta Pharmacol. Sinica 31, 1065–1070. 49. Schwarzer, C. (2009). 30 years of dynorphins–new insights on their func-
30. Günther, T., Dasgupta, P., Mann, A., Miess, E., Kliewer, A., Fritzwanker, S., tions in neuropsychiatric diseases. Pharmacol. Ther. 123, 353–370.
Steinborn, R., and Schulz, S. (2018). Targeting multiple opioid receptors - 50. Che, T., Majumdar, S., Zaidi, S.A., Ondachi, P., McCorvy, J.D., Wang, S.,
improved analgesics with reduced side effects? Br. J. Pharmacol. 175, Mosier, P.D., Uprety, R., Vardy, E., Krumm, B.E., et al. (2018). Structure of
2857–2868. the nanobody-stabilized active state of the kappa Opioid receptor. Cell
31. Machelska, H., and Celik, M.Ö. (2018). Advances in achieving opioid anal- 172, 55–67.e15.
gesia without side effects. Front. Pharmacol. 9, 1388. 51. Dooley, C.T., and Houghten, R.A. (1996). Orphanin FQ: receptor binding
32. Azevedo Neto, J., Costanzini, A., De Giorgio, R., Lambert, D.G., Ruzza, C., and analog structure activity relationships in rat brain. Life Sci. 59,
and Calò, G. (2020). Biased versus Partial Agonism in the Search for Safer PL23–PL29.
opioid analgesics. Molecules 25, 3870.
52. Joshi, A.A., Murray, T.F., and Aldrich, J.V. (2017). Alanine scan of the
33. Davenport, A.P., Scully, C.C.G., de Graaf, C., Brown, A.J.H., and Maguire, opioid peptide dynorphin B amide. Biopolymers 108.
J.J. (2020). Advances in therapeutic peptides targeting G protein-coupled
53. Pacifico, S., Albanese, V., Illuminati, D., Marzola, E., Fabbri, M., Ferrari, F.,
receptors. Nat. Rev. Drug Discov. 19, 389–413.
Holanda, V.A.D., Sturaro, C., Malfacini, D., Ruzza, C., et al. (2021). Novel
34. Fosgerau, K., and Hoffmann, T. (2015). Peptide therapeutics: current sta- mixed NOP/opioid receptor peptide agonists. J. Med. Chem. 64,
tus and future directions. Drug Discov. Today 20, 122–128. 6656–6669.
35. Ding, H.P., Kiguchi, N., Yasuda, D., Daga, P.R., Polgar, W.E., Lu, J.J., 54. Orsini, M.J., Nesmelova, I., Young, H.C., Hargittai, B., Beavers, M.P., Liu,
Czoty, P.W., Kishioka, S., Zaveri, N.T., and Ko, M.C. (2018). A bifunctional J., Connolly, P.J., Middleton, S.A., and Mayo, K.H. (2005). The nociceptin
nociceptin and Mu Opioid receptor agonist is analgesic without opioid side pharmacophore site for opioid receptor binding derived from the NMR
effects in nonhuman primates. Sci. Transl. Med. 10, eaar3483. structure and bioactivity relationships. J. Biol. Chem. 280, 8134–8142.
36. Lin, A.P., and Ko, M.C. (2013). The therapeutic potential of nociceptin/or- 55. Daga, P.R., and Zaveri, N.T. (2012). Homology modeling and molecular
phanin FQ receptor agonists as analgesics without abuse liability. ACS dynamics simulations of the active state of the nociceptin receptor reveal
Chem. Neurosci. 4, 214–224. new insights into agonist binding and activation. Proteins 80, 1948–1961.
37. Huang, W., Manglik, A., Venkatakrishnan, A.J., Laeremans, T., Feinberg, 56. Guerrini, R., Calo, G., Rizzi, A., Bianchi, C., Lazarus, L.H., Salvadori, S., Te-
E.N., Sanborn, A.L., and Kobilka, B.K. (2015). Structural insights into mi- mussi, P.A., and Regoli, D. (1997). Address and message sequences for
cro-opioid receptor activation. Nature 524, 315–321. the nociceptin receptor: a structure-activity study of nociceptin-(1–13)-
38. Koehl, A., Hu, H., Maeda, S., Zhang, Y., Qu, Q., Paggi, J.M., and Kobilka, peptide amide. J. Med. Chem. 40, 1789–1793.
B.K. (2018). Structure of the micro-opioid receptor-Gi protein complex. 57. Smyth, D.G. (2016). 60 years OF POMC: lipotropin and beta-endorphin: a
Nature 558, 547–552. perspective. J. Mol. Endocrinol. 56, T13–T25.
39. Ballesteros, J.A., and Weinstein, H. (1995). [19] Integrated methods for the
58. Schwyzer, R. (1986). Molecular mechanism of opioid receptor selection.
construction of three-dimensional models and computational probing of
Biochemistry 25, 6335–6342.
structure-function relations in G protein-coupled receptors. Methods Neu-
rosci. 25, 366–428. 59. Kato, H.E., Zhang, Y., Hu, H., Suomivuori, C.M., Kadji, F.M.N., Aoki, J.,
Krishna Kumar, K., Fonseca, R., Hilger, D., Huang, W., et al. (2019).
40. Manglik, A., Kruse, A.C., Kobilka, T.S., Thian, F.S., Mathiesen, J.M., Suna-
Conformational transitions of a neurotensin receptor 1-Gi1 complex. Na-
hara, R.K., and Granier, S. (2012). Crystal structure of the micro-opioid re-
ture 572, 80–85.
ceptor bound to a morphinan antagonist. Nature 485, 321–326.
60. Liu, H., Sun, D., Myasnikov, A., Damian, M., Baneres, J.L., Sun, J., and
41. Granier, S., Manglik, A., Kruse, A.C., Kobilka, T.S., Thian, F.S., Weis, W.I.,
Zhang, C. (2021). Structural basis of human ghrelin receptor signaling by
and Kobilka, B.K. (2012). Structure of the delta-opioid receptor bound to
ghrelin and the synthetic agonist ibutamoren. Nat. Commun. 12, 6410.
naltrindole. Nature 485, 400–404.
61. Flock, T., Ravarani, C.N.J., Sun, D., Venkatakrishnan, A.J., Kayikci, M.,
42. Thompson, A.A., Liu, W., Chun, E., Katritch, V., Wu, H., Vardy, E., Huang,
Tate, C.G., Veprintsev, D.B., and Babu, M.M. (2015). Universal allosteric
X.P., Trapella, C., Guerrini, R., Calo, G., et al. (2012). Structure of the no-
mechanism for Galpha activation by GPCRs. Nature 524, 173–179.
ciceptin/orphanin FQ receptor in complex with a peptide mimetic. Nature
485, 395–399. 62. Connor, M., and Christie, M.D. (1999). Opioid receptor signalling mecha-
nisms. Clin. Exp. Pharmacol. Physiol. 26, 493–499.
43. Wu, H., Wacker, D., Mileni, M., Katritch, V., Han, G.W., Vardy, E., Liu, W.,
Thompson, A.A., Huang, X.P., Carroll, F.I., et al. (2012). Structure of the hu- 63. Manglik, A. (2020). Molecular basis of opioid action: from structures to new
man kappa-opioid receptor in complex with JDTic. Nature 485, 327–332. leads. Biol. Psychiatry 87, 6–14.
44. Shang, Y., and Filizola, M. (2015). Opioid receptors: structural and mech- 64. Zhuang, Y., Wang, L., Guo, J., Sun, D., Wang, Y., Liu, W., Xu, H.E., and
anistic insights into pharmacology and signaling. Eur. J. Pharmacol. 763, Zhang, C. (2022). Molecular recognition of formylpeptides and diverse ag-
206–213. onists by the formylpeptide receptors FPR1 and FPR2. Nat. Commun.
13, 1054.
45. Fenalti, G., Giguere, P.M., Katritch, V., Huang, X.P., Thompson, A.A.,
Cherezov, V., Roth, B.L., and Stevens, R.C. (2014). Molecular control of 65. Michino, M., Beuming, T., Donthamsetti, P., Newman, A.H., Javitch, J.A.,
delta-opioid receptor signalling. Nature 506, 191–196. and Shi, L. (2015). What can crystal structures of aminergic receptors tell
46. Fenalti, G., Zatsepin, N.A., Betti, C., Giguere, P., Han, G.W., Ishchenko, A., us about designing subtype-selective ligands? Pharmacol. Rev. 67,
Liu, W., Guillemyn, K., Zhang, H., James, D., et al. (2015). Structural basis 198–213.
for bifunctional peptide recognition at human delta-opioid receptor. Nat. 66. Granier, S., and Kobilka, B. (2012). A new era of GPCR structural and
Struct. Mol. Biol. 22, 265–268. chemical biology. Nat. Chem. Biol. 8, 670–673.
47. Erspamer, V., Melchiorri, P., Falconieri-Erspamer, G., Negri, L., Corsi, R., 67. Egyed, A., Kelemen, Á.A., Vass, M., Visegrády, A., Thee, S.A., Wang, Z., de
Severini, C., Barra, D., Simmaco, M., and Kreil, G. (1989). Deltorphins: a Graaf, C., Brea, J., Loza, M.I., Leurs, R., et al. (2021). Controlling the selec-
family of naturally occurring peptides with high affinity and selectivity for tivity of aminergic GPCR ligands from the extracellular vestibule. Bioorg.
delta opioid binding sites. Proc. Natl. Acad. Sci. USA 86, 5188–5192. Chem. 111, 104832.
48. Claff, T., Yu, J., Blais, V., Patel, N., Martin, C., Wu, L., Han, G.W., Holleran, 68. Zhuang, Y., Liu, H., Edward Zhou, X., Kumar Verma, R., de Waal, P.W.,
B.J., Van der Poorten, O., White, K.L., et al. (2019). Elucidating the active Jang, W., Xu, T.H., Wang, L., Meng, X., Zhao, G., et al. (2020). Structure

426 Cell 186, 413–427, January 19, 2023


ll
Article
of formylpeptide receptor 2-Gi complex reveals insights into ligand recog- 83. Zivanov, J., Nakane, T., Forsberg, B.O., Kimanius, D., Hagen, W.J., Lin-
nition and signaling. Nat. Commun. 11, 885. dahl, E., et al. (2018). New tools for automated high-resolution cryo-EM
69. Xing, C., Zhuang, Y., Xu, T.H., Feng, Z., Zhou, X.E., Chen, M., Xie, X.Q., structure determination in RELION-3. Elife 7.
Meng, X., Xue, Y., Wang, J., et al. (2020). Cryo-EM structure of the human 84. Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt,
cannabinoid receptor CB2-Gi signaling complex. Cell 180, 645–654.e13. D.M., Meng, E.C., et al. (2004). UCSF Chimera–a visualization system
70. Hua, T., Li, X., Wu, L., Iliopoulos-Tsoutsouvas, C., Wang, Y., Wu, M., Shen, for exploratory research and analysis. J Comput Chem 25, 1605–1612.
L., Brust, C.A., Nikas, S.P., Song, F., et al. (2020). Activation and signaling 85. Pettersen, E.F., Goddard, T.D., Huang, C.C., Meng, E.C., Couch, G.S.,
mechanism revealed by cannabinoid receptor-Gi complex structures. Cell Croll, T.I., et al. (2021). UCSF ChimeraX: Structure visualization for re-
180, 655–665.e18. searchers, educators, and developers. Protein Sci 30, 70–82.
71. Liu, Q., Yang, D., Zhuang, Y., Croll, T.I., Cai, X., Dai, A., He, X., Duan, 86. Adams, P.D., Afonine, P.V., Bunkoczi, G., Chen, V.B., Davis, I.W., Echols,
J., Yin, W., Ye, C., et al. (2021). Ligand recognition and G-protein N., et al. (2010). PHENIX: a comprehensive Python-based system for
coupling selectivity of cholecystokinin A receptor. Nat. Chem. Biol. macromolecular structure solution. Acta Crystallogr D Biol Crystallogr
17, 1238–1244. 66, 213–221.
72. Ma, S., Chen, Y., Dai, A., Yin, W., Guo, J., Yang, D., Zhou, F., Jiang, Y., 87. Chen, V.B., Arendall, W.B., 3rd, Headd, J.J., Keedy, D.A., Immormino,
Wang, M.W., and Xu, H.E. (2021). Structural mechanism of calcium-medi- R.M., Kapral, G.J., et al. (2010). MolProbity: all-atom structure validation
ated hormone recognition and Gbeta interaction by the human for macromolecular crystallography. Acta Crystallogr D Biol Crystallogr
melanocortin-1 receptor. Cell Res. 31, 1061–1071. 66, 12–21.
73. Xia, R., Wang, N., Xu, Z., Lu, Y., Song, J., Zhang, A., Guo, C., and He, Y. 88. Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molec-
(2021). Cryo-EM structure of the human histamine H1 receptor/Gq com- ular graphics. Acta Crystallogr D Biol Crystallogr 60, 2126–2132.
plex. Nat. Commun. 12, 2086. 89. Liu, P., Jia, M.Z., Zhou, X.E., De Waal, and Dickson, B.M. (2016). The
74. Zhuang, Y., Wang, Y., He, B., He, X., Zhou, X.E., Guo, S., Rao, Q., Yang, J., structural basis of the dominant negative phenotype of the Galphai1beta1-
Liu, J., Zhou, Q., et al. (2022). Molecular recognition of morphine and fen- gamma2 G203A/A326S heterotrimer. Acta Pharmacol Sin 37, 1259–1272.
tanyl by the human mu-opioid receptor. Cell 185, 4361–4375.e19. 90. Liang, Y.-L., Zhao, P., Draper-Joyce, C., Baltos, J.-A., Glukhova, A.,
75. Qu, Q., Huang, W., Aydin, D., Paggi, J.M., Seven, A.B., Wang, H., and Ski- Truong, T.T., et al. (2018). Dominant Negative G Proteins Enhance Forma-
niotis, G. (2022). Insights into distinct signaling profiles of the muOR acti- tion and Purification of Agonist-GPCR-G Protein Complexes for Structure
vated by diverse agonists. Nat. Chem. Biol. Determination. ACS Pharmacology & Translational Science 1, 12–20.
76. Cvejic, S., and Devi, L.A. (1997). Dimerization of the delta Opioid receptor: 91. Maeda, S., Koehl, A., Matile, H., Hu, H.L., Hilger, D., Schertler, G.F.X., et al.
implication for a role in receptor internalization. J. Biol. Chem. 272, (2018). Development of an antibody fragment that stabilizes GPCR/G-pro-
26959–26964. tein complexes. Nature communications 9.
77. Wang, D., Sun, X., Bohn, L.M., and Sadée, W. (2005). Opioid receptor 92. Rohou, A., and Grigorieff, N. (2015). CTFFIND4: Fast and accurate defocus
homo- and heterodimerization in living cells by quantitative biolumines- estimation from electron micrographs. J Struct Biol 192, 216–221.
cence resonance energy transfer. Mol. Pharmacol. 67, 2173–2184. 93. Kimanius, D., Dong, L., Sharov, G., Nakane, T., and Scheres, S.H.W.
78. Claff, T., Yu, J., Blais, V., Patel, N., Martin, C., Wu, L., et al. (2019). Eluci- (2021). New tools for automated cryo-EM single-particle analysis in
dating the active delta-opioid receptor crystal structure with peptide and RELION-4.0. The Biochemical journal 478, 4169–4185.
small-molecule agonists. Sci Adv 5, eaax9115. 94. Olsen, R.H.J., DiBerto, J.F., English, J.G., Glaudin, A.M., Krumm, B.E.,
79. Wang, D., Sun, X., Bohn, L.M., and Sadee, W. (2005). Opioid receptor Slocum, S.T., et al. (2020). TRUPATH, an open-source biosensor platform
homo- and heterodimerization in living cells by quantitative biolumines- for interrogating the GPCR transducerome. Nat Chem Biol 16, 841–849.
cence resonance energy transfer. Mol Pharmacol 67, 2173–2184. 95. DiBerto, J.F., Smart, K., Olsen, R.H.J., and Roth, B.L. (2022). Agonist and
80. Zhuang, Y., Liu, H., Edward Zhou, X., Kumar Verma, R., de Waal, P. W., antagonist TRUPATH assays for G protein-coupled receptors. STAR Pro-
Jang, W., Zhang, C. (2020). Structure of formylpeptide receptor 2-G(i) toc 3, 101259.
complex reveals insights into ligand recognition and signaling. Nat Com- 96. Chakraborty, S., DiBerto, J.F., Faouzi, A., Bernhard, S.M., Gutridge, A.M.,
mun 11, 885. Ramsey, S., et al. (2021). A Novel Mitragynine Analog with Low-Efficacy
81. Mastronarde, D.N. (2005). Automated electron microscope tomography Mu Opioid Receptor Agonism Displays Antinociception with Attenuated
using robust prediction of specimen movements. J Struct Biol 152, 36–51. Adverse Effects. J Med Chem 64, 13873–13892.
82. Zheng, S.Q., Palovcak, E., Armache, J.P., Verba, K.A., Cheng, Y., and 97. Qu, Q., Huang, W., Aydin, D., Paggi, J.M., Seven, A.B., Wang, H., et al.
Agard, D.A. (2017). MotionCor2: anisotropic correction of beam-induced (2022). Insights into distinct signaling profiles of the mOR activated by
motion for improved cryo-electron microscopy. Nat Methods 14, 331–332. diverse agonists. Nature Chemical Biology.

Cell 186, 413–427, January 19, 2023 427


ll
Article

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
GP64-PE antibody Expression systems Cat# 97-201; RRID: AB_2922960
Anti-HA-FITC antibody, Mouse monoclonal Sigma-Aldrich Cat# H7411; RRID: AB_439704
Monoclonal ANTI-FLAG M2-Peroxidase Sigma-Aldrich Cat# A8592; RRID: AB_439702
(HRP) antibody produced in mouse
Chemicals, Peptides, and Recombinant Proteins
b-endorphin1-31 Genscript Custom Synthesis
Endomorphin-1 Genscript Custom Synthesis
[D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) Med Chem Express Cat# HY-P0210B
[D-Ala2] Deltorphin TFA Med Chem Express Cat# HY-P1013A
Dynorphin A1-13 Genscript Custom Synthesis
Nociceptin/orphanin FQ (NFQ)1-17 Genscript Custom Synthesis
YGGF Genscript Custom Synthesis
Leu-Enkephalin Sigma-Aldrich Cat# E5008
Met-Enkephalin Sigma-Aldrich Cat# M6638
Lauryl maltose neopentyl glycol Anatrace Cat# NG310
Glyco-diosgenin (GDN) Anatrace Cat# GDN101
Cholesteryl Hemisuccinate Anatrace Cat# CH210
ClonExpress II One Step Cloning Kit Vazyme Biotech Co.,Ltd Cat# C112
PrimeSTAR Max Premix TAKARA Cat# R045Q
Protease Inhibitor Cocktail, EDTA-free Bimake Cat# B14003
Apyrase New England Biolabs Cat# M0398L
Tris-(2-carboxyethyl)-phosphine hydrochloride (TCEP) Sangon Biotech Cat# A600974
Nickel Sepharose resin GE healthcare Cat# 17526801
Anti-Flag resin Smart-Lifesciences Cat# C20042002
FLAG peptide Genscript Custom Synthesis
ESF921 culture medium Expression systems Cat# 96-001-01
Dulbecco’s Modified Eagle Medium (DMEM) Gibco Cat# 12800-017
Fetal Bovine Serum (FBS) Gibco Cat# 10099-141
Hanks’ Balanced Salt Solution (HBSS) Invitrogen Cat# 14065-056
Coelenterazine 400A Nanolight Cat# 340
Coelenterazine-h Promega Cat# S2011
[3H]DADLE PerkinElmer Cat# NET648250U
[3H]DAMGO PerkinElmer Cat# NET902250UC
[3H]Nociceptin PerkinElmer Cat# NET1130050UC
[3H]U-69,593 PerkinElmer Cat# NET952250UC
Critical Commercial Assays
Bac-to-Bac Baculovirus Expression System ThermoFisher Cat# 10359016
LANCE UltracAMP kit PerkinElmer Cat# 2952744
SuperSignal ELISA Pico Chemiluminescent Substrate ThermoFisher Cat# 37069
Deposited Data
mOR-b-endorphin1-31-Gi coordinate This paper PDB: 8F7Q
mOR-endomorphin-1-Gi coordinate This paper PDB: 8F7R
(Continued on next page)

e1 Cell 186, 413–427.e1–e7, January 19, 2023


ll
Article

Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
2
dOR-[D-Ala ]deltorphin -Gi coordinate This paper PDB: 8F7S
kOR-dynorphin A1-13-Gi coordinate This paper PDB: 8F7W
NOPR-NFQ-Gi coordinate This paper PDB: 8F7X
mOR-b-endorphin1-31-Gi EM map This paper EMDB: EMD-28907
mOR-endomorphin-1-Gi EM map This paper EMDB: EMD-28908
dOR-[D-Ala2]deltorphin-Gi EM map This paper EMDB: EMD-28909
kOR-dynorphin A1-13-Gi EM map This paper EMDB: EMD-28911
NOPR-NFQ-Gi EM map This paper EMDB: EMD-28912
Reagent or Resource
dOR-naltrindole Fenalti et al.45 PDB: 4N6H
dOR-KGCHM07 Claff et al.48 PDB: 6PT2
dOR-DPI-287 Claff et al.48 PDB: 6PT3
50
kOR-MP1104 Che et al. PDB: 6B73
kOR-JDTic Wu et al.43 PDB: 4DJH
mOR-fentanyl-Gi-scFv16 Zhuang et al.74 PDB: 8EF5
mOR-morphine-Gi-scFv16 Zhuang et al.74 PDB: 8EF6
mOR-BU72 Huang et al.37 PDB: 5C1M
mOR-b-FNA Manglik et al.40 PDB: 4DKL
NOPR-Compound-24 Thompson et al.42 PDB: 4EA3
PZM21-mOR-Gi-scFv16 EM map Zhuang et al.74 EMDB: EMD-28086
FPR2-WKYMVm-Gi-scFv16 EM map Zhuang et al.80 EMDB: EMD-20126
Experimental Models: Organisms/strains
E. coli strain OmniMAXTM Invitrogen homemade
TOP10 Competent E. coli TIANGEN Biotech Cat# CB104
Experimental Models: Cell Lines
Spodoptera frugiperda Sf9 cells Expression Systems Cat# 94-001F
HEK-293 cells ATCC Cat# CRL-1573
Recombinant DNA
pFastbac-prolactin-FLAG-mOR(1-388)-His8 This paper N/A
pFastbac-prolactin-FLAG-dOR(2-372, 7 mutations)-His8 This paper N/A
pFastbac-prolactin-FLAG-kOR(3-380)-His8 This paper N/A
pFastbac-prolactin-FLAG-NOPR(2-370)-His8 This paper N/A
pFastbac-DN_Gai(G203A, A326A) This paper N/A
pFastbac-DN_Gai(S47N, G203A, E245A, A326A) This paper N/A
pFastbac-H8-Gb1 This paper N/A
pFastbac-Gg2 This paper N/A
pFastbac-GP67-scFv16-Tev-His8 This paper N/A
pcDNA3.0-HA-FLAG-mOR This paper N/A
pcDNA3.0-HA-FLAG-dOR This paper N/A
pcDNA3.0-HA-FLAG-kOR This paper N/A
pcDNA3.0-HA-FLAG-NOPR This paper N/A
pcDNA3.0-HA-FLAG-mOR-RLuc8 This paper N/A
pcDNA3.0-HA-FLAG-dOR-RLuc8 This paper N/A
pcDNA3.0-HA-FLAG-kOR-RLuc8 This paper N/A
pcDNA3.0-HA-FLAG-NOPR-RLuc8 This paper N/A
pcDNA3.1-Beta3 Addgene Cat# 140988
pcDNA3.1-GGamma9-GFP2 Addgene Cat# 140991
pcDNA3.1-GRK2 Genscript CloneID OHu08965C
(Continued on next page)

Cell 186, 413–427.e1–e7, January 19, 2023 e2


ll
Article

Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
pcDNA5/FRT/TO-mVenus-Beta-Arrestin2 This paper N/A
pcDNA5/FRT/TO-GAlphai1-RLuc8 Addgene Cat# 140973
Software and Algorithms
Clonemanager Sci-Ed Software http://www.scied.com/pr_cmpro.htm
Prism 8 GraphPad https://www.graphpad.com/scientific-
software/prism/
SerialEM Mastronarde et al.81 http://bio3d.colorado.edu/SerialEM/
MotionCor2 Zheng et al.82 http://msg.ucsf.edu/em/software/motioncor2.html
Relion 3.1 Zivanov et al.83 https://relion.readthedocs.io/en/release-3.1/
Installation.html
UCSF Chimera Pettersen et al.84 https://www.cgl.ucsf.edu/chimera/
UCSF ChimeraX Pettersen et al.85 https://www.cgl.ucsf.edu/chimerax/
Phenix Adams et al.86 https://www.phenix-online.org/
MolProbity Chen et al.87 http://molprobity.biochem.duke.edu/
Coot Emsley et al.88 https://www2.mrc-lmb.cam.ac.uk/personal/
pemsley/coot/
PyMol software Schrödinger https://pymol.org/2/
Adobe Illustrator CC Adobe https://www.adobe.com
Other
100 kDa molecular weight cut-off Amicon Ultra Cat# UFC910024
Quantifoil R1.2/1.3 300-mesh Gold grids Quantifoil https://www.emsdiasum.com/microscopy/
products/grids/quantifoil.aspx
Superdex 200 10/300GL Increase column GE healthcare Cat#28990944
Superose 6 Increase 10/300GL column Cytiva Cat#29091596
PHERAstar FSX with BRET1 and BRET2 optic modules BMG Labtech www.bmglabtech.com

RESOURCE AVAILABILITY

Lead contact
Further information and requests for reagents may be directed and will be fulfilled by the lead contact, H. Eric Xu (eric.xu@simm.
ac.cn).

Materials availability
All stable reagents generated in this study are available from the lead contact without restriction. Plasmids and strains are available
from the authors upon request.

Data and code availability


d Cryo-EM density maps of b-endorphin-mOR-Gi, endomorphin-mOR-Gi, deltorphin-dOR-Gi, dynorphin-kOR-Gi, and NFQ-
NOPR-Gi complex have been deposited in the Electron Microscopy Data Bank (EMDB) under the accession numbers EMD-
28907, EMD-28908, EMD-28909, EMD-28911, and EMD-28912, respectively. Atomic coordinates for the atomic models of
b-endorphin-mOR-Gi, endomorphin-mOR-Gi, deltorphin-dOR-Gi, dynorphin-kOR-Gi, and NFQ-NOPR-Gi complex structures
have been deposited in the Protein Data Bank (PDB) under the accession numbers 8F7Q, 8F7R, 8F7S, 8F7W, and 8F7X,
respectively. These accession numbers are also listed in the key resources table.
d This paper does not report original code.
d Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Spodoptera frugiperda (Sf9, Expression systems) cells were used for recombinant protein expression. HEK293 cells (ATCC) were
used for functional studies. The Sf9 cells were grown in ESF 921 medium (Expression systems) at 27 C, 120 rpm. HEK293 cells

e3 Cell 186, 413–427.e1–e7, January 19, 2023


ll
Article

were grown in humidified 37 C incubator in condition of 5% CO2. The medium for human cell lines HEK293 was DMEM High-Glucose
(Gibco) containing 10% fetal bovine serum (FBS, Gibco).

METHOD DETAILS

Constructs design
To facilitate expression and receptor anchoring to the membrane, the N-terminus of human opioid receptors were inserted with the
prolactin precursor sequence as a signaling peptide. A FLAG tag was inserted between the signaling peptide and opioid receptors for
purification usage. In addition, the 83His tag was fused at the C-terminus of opioid receptors for two-step purification. For mOR, the
wild type mOR (1-388) truncation was used to obtain the endomorphin-bound mOR-Gi complex, while mOR (1-388) with thermostable
mutation F3.41W was used to prepare the b-endorphin-bound mOR-Gi signaling complex. As for dOR, a total of seven thermostable
mutations, including G731.56V, N902.45S, D952.50G, N1313.35S, S1433.47C, G2686.42V, and A3097.44I, were introduced into the dOR
sequence based on the previously reported dOR structure (PDB: 6PT2).46 The wild-type kOR and NOPR amino acid sequences
were used to prepare the kOR-Gi and NOPR-Gi complexes. These modified opioid receptors were constructed into the pFastBac
vector (ThermoFisher) for expression usage. Two dominant-negative mutations, G203A and A326S, were added to the human
Gai1 (Gai1_2M) to reduce GDP/GTP binding.89 Another two dominant-negative mutations, S47N and E245A, were further incorpo-
rated into Gai1_2M, leading to Gai1_4M.90 All the three components of Gi1 heterotrimer, human Gai1_2M or Gai1_4M, rat His8-Gb1
and bovine Gg2, were constructed into the pFastBac vector (ThermoFisher), respectively.

Preparation of scFv16 antibody


For the expression of scFv16, the GP67 signaling peptide was inserted into the N terminus of scFv16, while a TEV cleavage site-His8
was fused to its C terminus. The modified scFv16 was then subcloned into the pFastBac vector (ThermoFisher). The scFv16 stocks
were prepared by methods previously described.38,91 The purified scFv16 was fast frozen by liquid nitrogen and stored at -80 C for
further usage in GPCR-Gi signaling complex preparation.

Expression and purification of opioid receptor- Gi complexes


Opioid receptors, Gai1, Gb1, and Gg2 were co-expressed in sf9 insect cells using the Bac-to-Bac baculovirus expression system
(ThermoFisher). The sf9 cells were cultured in ESF 921 serum-free medium (Expression Systems). For expression of opioid re-
ceptor-Gi signaling complexes, baculoviruses of each opioid receptor, Gai1, Gb1, and Gg2 were added in equal proportions to
the cell culture when the cell density reached 43106 cells/mL. For mOR, dOR and NOPR, we used Gai1_2M for the expression of
protein complexes, while for kOR, we used the Gai1_4M instead of Gai1_2M to form the stable signaling complex. After being
cultured for 48 hours, the cells were harvested by centrifugation at 1300 g for 20 min and frozen at -80  C for further protein
purification.
The cell pellets were thawed at room temperature and resuspended in 20 mM HEPES pH 7.2, 50 mM NaCl, 10mM KCl, 5 mM
MgCl2, 0.3 mM TCEP, protease inhibitor cocktail (Bimake, 1mL/100mL suspension). After quick cell lysis by the French press, the
cell suspension was centrifugated at 100,000 3 g for 30 min at 4 C. The cell precipitates were then collected and resuspended in
buffer including 20 mM HEPES pH 7.2, 75 mM NaCl, 5 mM CaCl2, 5 mM MgCl2, 5% glycerol, 0.3 mM TCEP, protease inhibitor
cocktail (Bimake, 1mL/100mL suspension). The b-endorphin or endomorphin-bound mOR-Gi complexes were assembled on cell
membranes by the addition of 20mM b-endorphin or 10mM endomorphin. For the dOR-Gi, kOR-Gi, and NOPR-Gi complexes,
10mM deltorphin, 10mM dynorphin, and 10mM NFQ were added to stimulate the formation of signaling complexes, respectively.
The protein complexes were all assembled at room temperature. Half an hour after ligand addition, the cell suspension was subse-
quently treated with 25mU mL 1 apyrase to digest the nucleotides. The suspension was continually incubated for another one hour at
room temperature before being extracted from the membrane using 0.5% (w/v) lauryl maltose neopentylglycol (LMNG, Anatrace) and
0.1% (w/v) cholesteryl hemisuccinate TRIS salt (CHS, Anatrace). After membrane solubilization for 3 hours at 4 C, the solubilized
fractions were isolated by centrifugation at 100,000 3 g for 45 min and then incubated overnight at 4 C with pre-equilibrated
Nickel-NTA resin using distilled deionized water (dd H2O). After batch binding, the nickel resin with immobilized protein complex
was manually loaded onto a gravity flow column. The nickel resin was firstly washed with 20 column volumes of 20 mM HEPES,
pH 7.2, 100 mM NaCl, 25 mM imidazole, 0.3 mM TCEP, 0.05% LMNG (w/v), 0.01% CHS (w/v) and 10 mM corresponding ligands,
and then eluted with the same buffer containing 300 mM imidazole.
The Ni-NTA eluates were further incubated plus 2.5 mg scFv16 by batch binding with 2 mL FLAG resin (Smart-Lifesciences) for
2 hours at 4 C. The FLAG resin embedded with protein complexes was then washed by 10 column volumes of detergent buffer
containing 20 mM HEPES, pH 7.2, 100 mM NaCl, 0.3 mM TCEP, 0.0075% LMNG (w/v), 0.0025% GDN, 0.002% CHS (w/v),
10 mM ligands. Subsequently, the material bound to FLAG resin was then eluted with the same detergent buffer above containing
200mg/mL FLAG peptide. The complex was then concentrated using an Ultra Centrifugal Filter (Millipore, Sigma, 100 kDa molecular
weight cutoff) and subjected to Superdex 200 Increase 10/300 GL (GE Healthcare) or Superose 6 Increase 10/300 GL (GE Healthcare)
size exclusion chromatography columns pre-equilibrated with buffer containing 20 mM HEPES, pH 7.2, 100 mM NaCl, 0.00075%
LMNG (w/v), 0.00025% GDN, 0.0002% CHS (w/v), 0.3 mM TCEP and 5 mM ligands. Fractions of the main peak of the protein com-
plexes were collected and concentrated to approximately 10-13 mg/ml for electron microscopy experiments.

Cell 186, 413–427.e1–e7, January 19, 2023 e4


ll
Article

Cryo-EM grid preparation and data collection


Before grid making, the monodispersed b-endorphin-mOR-Gi complex, endomorphin-mOR-Gi complex, deltorphin-dOR-Gi complex,
dynorphin-kOR-Gi complex, and NFQ-NOPR-Gi complex were concentrated to about 7.5 mg mL-1, 6.8 mg mL-1, 13 mg mL-1, 12 mg
mL-1, and 10 mg mL-1, respectively. For cryo-EM grids preparation, about 3.0 mL of each protein complex sample was applied
individually to EM grids (Quantifoil R1.2/1.3 holey carbon films, 300 mesh Au) that were glow-discharged for 50s in a Vitrobot cham-
ber (FEI Vitrobot Mark IV). Vitrification was performed in 100% humidity at 4 C. The sample was blotted for 3s before plunge-freezing
into liquid ethane. The prepared cryo-EM grids were stored in liquid nitrogen for screening and data collection usage.
For the b-endorphin-mOR-Gi complex and dynorphin-kOR-Gi complex, automatic cryo-EM movie stacks were collected on an FEI
Titan Krios microscope operated at 300kV accelerating voltage in Cryo-Electron Microscopy Research Center, Shanghai Institute of
Materia Medica, Chinese Academy of Sciences (Shanghai, China). The microscope was operated with a nominal magnification of
81,0003 in super-resolution counting mode. A total of 4,959 movies for the dataset of b-endorphin-mOR-Gi-scFv16 complex and
4,254 movies for the dataset of dynorphin-kOR-Gi-scFv16 complex were collected individually by a Gatan K3 Summit direct electron
detector equipped with a Gatan energy filter (operated with a slit width of 20 eV) (GIF) using the SerialEM software.81 The images were
recorded at a dose rate of 23.3 e/Å2/s with a defocus ranging from -0.5 to -3.0 mm. The total exposure time was 3 s and intermediate
frames were recorded in 0.083 s intervals, resulting in a total of 36 frames per micrograph.
For the deltorphin-dOR-Gi-scFv16 complex and NFQ-NOPR-Gi-scFv16 complex, automatic cryo-EM movie stacks were collected
on an FEI Titan Krios microscope operated at 300kV in Advanced Center for Electron Microscopy, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences (Shanghai, China). The microscope was equipped with a Gatan Quantum energy filter.
The movie stacks were collected automatically using a Gatan K3 direct electron detector with a nominal magnification of
105,0003 in super-resolution counting mode at a pixel size of 0.42 Å. The energy filter was operated with a slit width of 20 eV.
Each movie stack was dose-fractionated in 36 frames with a dose of 1.39 electrons per frame and collected within a defocus ranging
from -0.8 to -1.8 mm. The total exposure time was 2.35 s. A total of 6,841 movies were collected for deltorphin-dOR-Gi-scFv16 com-
plex. A total of 7,125 movies were collected for NFQ-NOPR-Gi-scFv16 complex. Data collection was performed using EPU with one
exposure per hole on the grid squares.
For the endomorphin-mOR-Gi-scFv16 complex, automatic cryo-EM movie stacks were collected by a Titan Krios G4 at 300KV
accelerating voltage equipped with Falcon4 detector in Advanced Center for Electron Microscopy, Shanghai Institute of
Materia Medica, Chinese Academy of Sciences (Shanghai, China). The movie stacks were collected automatically with a nominal
magnification of 96,0003 in counting mode at a pixel size of 0.8 Å. Each movie stack was dose-fractionated in 160 frames with
50 total doses (e/ Å2) and collected within a defocus ranging from -1.0 to -2.0 mm. A total of 6,418 movies for the dataset of endo-
morphin-mOR-Gi-scFv16 complex were collected. Data collection was performed using EPU with one exposure per hole on the
grid squares.

Image processing and 3D reconstruction


Movie stacks were subjected to beam-induced motion correction using MotionCor 2.1.82 Contrast transfer function (CTF) parameters
for each non-dose-weighted micrograph were determined by Ctffind4.92 Automated particle selection and data processing were per-
formed using RELION-3.1 beta283 or RELION-4.0 beta.93
For the datasets of b-endorphin-mOR-Gi-scFv16 and dynorphin-kOR-Gi-scFv16 complex, the movie stack was aligned, dose
weighted, and binned by 2 to 1.071 Å per pixel. The micrographs with resolution worse than 4.0 Å and micrographs imaged within
the carbon area of grid squares were abandoned, producing 3,908 and 3,523 micrographs respectively to do further data processing.
For the b-endorphin-mOR-Gi-scFv16 complex, template-based particle selection yielded 3,095,570 particles which were subjected
to reference-free 2D classifications to discard bad particles. The cryo-EM density map of the FPR2-Gi complex78 (EMDB: EMD-
20126) low-pass filtered to 40 Å was used as a reference model for four rounds of maximum-likelihood-based 3D classifications.
Further 3D classification focusing on the anti-parallel dimer of receptors produced 320,047 particles. These particles were subse-
quently subjected to 3D refinement, CTF refinement, and Bayesian polishing, which generated a density map with an indicated global
resolution of 3.2 Å at a Fourier shell correlation of 0.143. For dynorphin-kOR-Gi-scFv16 complex, template-based particle selection
yielded 2,480,199 particles which were subjected to reference-free 2D classifications to discard bad particles. The map of the FPR2-
Gi complex (EMDB: EMD-20126) low-pass filtered to 40 Å was used as a reference model for four rounds of maximum-likelihood-
based 3D classifications, resulting in three well-defined subsets with 173,555 particles. These particles were subsequently subjected
to 3D refinement, CTF refinement, Bayesian polishing, and post-processing, which generated a density map with an indicated global
resolution of 3.3 Å at a Fourier shell correlation of 0.143.
For the datasets of endomorphin-mOR-Gi-scFv16 complex, the movie stack was aligned, dose weighted, and binned by 2 to 1.6 Å
per pixel. The micrographs with resolution worse than 4.0 Å and micrographs imaged within the carbon area of grid squares were
abandoned, producing 5,704 micrographs to do further data processing. Template-based particle selection yielded 2,012,939
particles which were subjected to reference-free 2D classifications to discard bad particles. The map of PZM21-mOR-Gi-scFv1674
low-pass filtered to 60 Å was used as a reference model for four rounds of maximum-likelihood-based 3D classifications. Further
3D classification focusing on the anti-parallel dimer of receptors produced 185,378 particles. These particles were subsequently sub-
jected to 3D refinement, CTF refinement, and Bayesian polishing, which generated a density map with an indicated global resolution
of 3.3 Å at a Fourier shell correlation of 0.143.

e5 Cell 186, 413–427.e1–e7, January 19, 2023


ll
Article

For the datasets of deltorphin-dOR-Gi- scFv16 complex and NFQ-NOPR-Gi-scFv16 complex, the movie stack was aligned, dose
weighted and binned by 2 to 0.84 Å per pixel. The micrographs with resolution worse than 4.0 Å and micrographs imaged within the
carbon area of grid squares were abandoned, producing 6,413 and 6,439 micrographs respectively to do further data processing
micrographs to do further data processing. For the deltorphin-dOR-Gi- scFv16 complex, template-based particle selection yielded
4,839,714 particles which were subjected to reference-free 2D classifications to discard bad particles. The map of PZM21-mOR-Gi-
scFv16 low-pass filtered to 40 Å was used as a reference model for three rounds of maximum-likelihood-based 3D classifications.
Rotary anti-parallel dimer conformations were observed. The further 3D classification was performed with a mask excluding one of
the G protein heterotrimers with poor density. The final 3D classifications lead to two well-defined subsets with 650,970 particles.
These particles were subsequently subjected to 3D refinement, CTF refinement, and Bayesian polishing, which generated a density
map with an indicated global resolution of 3.0 Å at a Fourier shell correlation of 0.143. For NFQ-NOPR-Gi-scFv16 complex, template-
based particle selection yielded 4,809,226 particles which were subjected to reference-free 2D classifications to discard bad parti-
cles. The map of dynorphin-kOR-Gi-scFv16 complex (this study) low-pass filtered to 60 Å was used as a reference model for four
rounds of maximum-likelihood-based 3D classifications resulting in one well-defined subset with 184,353 particles. These particles
were subsequently subjected to 3D refinement, CTF refinement, and Bayesian polishing, which generated a density map with an indi-
cated global resolution of 3.3 Å at a Fourier shell correlation of 0.143.

Model building, structure refinement, and figure preparation


The previously published structures of mOR-Gi complex (PDB: 8EF5), dOR (PDB: 6PT2), kOR (PDB: 6B73), and NOPR (PDB: 4EA3)
were used as the start models for model building and refinement of mOR-Gi, dOR-Gi, kOR-Gi, and NOPR-Gi, respectively. The struc-
tural model was firstly docked as a rigid body into the cryo-EM density maps using UCSF Chimera.84 Models were then manually
rebuilt and/or adjusted in COOT.88 Real space and Rosetta refinements were performed using Phenix.86 The model statistics
were validated using MolProbity.87 Structural figures were prepared in ChimeraX85 and PyMOL (https://pymol.org/2/). The final
refinement statistics are provided in Table S1. The maximum distance cutoffs for polar hydrogen-bond interactions and hydrophobic
interactions were set at 3.5 Å and 4.5 Å, respectively.

cAMP accumulation assays


The open reading frames of the full-length human opioid receptors (hORs) were cloned into the pcDNA3.0 vector. For detecting Gi
signaling of mOR, dOR, kOR, and NOPR, HEK293 cells expressing WT or mutant hORs were harvested and resuspended in HBSS
buffer (Gibco) containing 0.1% BSA, 5mM HEPES (Gibco) and 500 mM IBMX (Abcone) at a density of 6 3105 cells/ mL. Cells
were then plated onto 384-well assay plates at 3000 cells/ 5 mL/ well. Another 5 mL buffer containing 2 mM Forskolin and various con-
centrations of test ligands were added to the cells. After incubation at room temperature for 30 minutes, the intracellular cAMP level
was tested by a LANCE Ultra cAMP kit (PerkinElmer, TRF0264) and EnVision multiplate reader according to the manufacturer’s
instructions.

Radio-ligand binding assays


Binding assays were performed on crude membrane preparations made from HEK293T cells transiently transfected with dOR, kOR,
mOR, or NOPR. The following radioligands (final concentration in nM) were used to label dOR, kOR, mOR, and NOPR, respectively:
[3H]DADLE (0.5 nM), [3H]U-69,593 (0.5 nM), [3H]DAMGO (0.5 nM), [3H]nociceptin (0.5 nM). In brief, 75 mL of membrane were co-incu-
bated with 25 mL of 5x radioligand and 25 mL of 5x drug for 90 minutes in standard assay buffer (50 mM TRIS hydrochloride buffer,
pH 7.4, containing 10 mM magnesium chloride and 0.1 mM EDTA) supplemented with 1 mg/mL fatty-acid free bovine serum albumin
and 0.1 mg/mL ascorbic acid. Following incubation, membranes were collected onto GF/C glass fiber filter-bottom, polyethylenei-
mine-coated 96-well microplates by vacuum filtration (an initial vacuum followed by three subsequent vacuums separated by washes
with cold harvesting buffer (50 mM Tris-HCl, pH 7.4). Non-specific binding for dOR, kOR, mOR, and NOPR were determined by 10 mM
concentrations of deltorphin-II, dynorphin A, DAMGO, and NFQ, respectively. All data were analyzed using GraphPad Prism.

Bioluminescence resonance energy transfer assays


Bioluminescence resonance energy transfer (BRET) assays were performed to measure G protein dissociation and beta-arrestin
recruitment. For G protein dissociation measurements, the TRUPATH platform94 was used as experiments were performed as pre-
viously described.94,95 In brief, wildtype or mutant dOR kOR, mOR, or NOPR were co-transfected with Gai1-RLuc8, Gb3, and GFP2-
Gg9 in a 1:1:1:1 ratio in HEK293T cells maintained in Dulbecco’s Modified Eagle Media (DMEM) supplement with 10% fetal bovine
serum (FBS) and 1% penicillin-streptomycin (pen-strep). After a minimum of 12 hours, cells were plated in 96-well microplates in
DMEM supplemented with 1% dialyzed FBS and 1% pen-strep. After an additional minimum of 12 hours, plates were first vacuum
aspirated and 60 mL of assay buffer (1x Hank’s Balanced Salt Solution in phosphate buffered saline, 20 mM HEPES, pH 7.4) were
added to the wells. Next, 10 mL of coelenterazine 400a diluted in assay buffer (50 mM) were added to the wells, and allowed to incu-
bate for 5 minutes. Next, 30 mL of 3x drug diluted in drug buffer (assay buffer supplemented with 3 mg/mL fatty acid-free bovine serum
albumin and 0.3 mg/mL ascorbic acid) were added to the wells, and allowed to incubate for 5 minutes. Last, plates were read using a
BMG Labtech PHERAstar FSX with BRET2 plus optic module. For b-arrestin recruitment measurements, RLuc8 was cloned directly
into the end of the C-terminus of wildtype and mutant dOR kOR, mOR, or NOPR, and experiments were performed as previously

Cell 186, 413–427.e1–e7, January 19, 2023 e6


ll
Article

described96,97 and identically to BRET2 assays with the following exceptions: 1) Wildtype or mutant kOR-RLuc8, mOR-RLuc8, or
NOPR-RLuc8 were co-transfected with GRK2 and mVenus-b-arrestin2 in a 1:1:5 ratio and dOR-RLuc8 was co-transfected with
GRK2 and mVenus-b-arrestin2 in a 1:1:15 ratio, 2) 10 mL of coelenterazine h diluted in assay buffer (50 mM) were added to the wells
instead of coelenterazine 400a, and 3) plates were read using a BMG Labtech PHERAstar FSX with BRET1 plus optic module. All data
were analyzed using GraphPad Prism.

Enzyme-Linked Immunosorbent Assay


Cell surface expression of the opioid receptor-RLuc8 constructs used in the BRET-based b-arrestin2 recruitment assays were
measured using an enzyme-linked immunosorbent assay (ELISA). Cells were transfected and plated in 96-well microplates as
described for the BRET assays. For the ELISA, all steps were performed at room temperature, with care given to liquid addition
and aspiration to minimize cell loss. First, plates were vacuum aspirated and 50 mL of 4% paraformaldehyde (PFA) in phosphate buff-
ered saline (PBS) were added to the wells and allowed to incubate for 10 minutes. Next, the PFA was aspirated, and cells were
washed twice with 50 mL of PBS. After the second PBS wash, 50 mL of 5% bovine serum album in PBS (blocking buffer) were added
to the wells and allowed to incubate for 30 minutes. Following incubation, an additional 25 mL of blocking buffer containing mono-
clonal ANTI-FLAG M2-Peroxidase (HRP) antibody produced in mouse (1:10,000 final dilution) were added to the wells and allowed
to incubate for 1 hour. Following incubation, cells were washed twice with 50 mL of PBS. Finally, 50 ml of SuperSignal enzyme-linked
immunosorbent assay (ELISA) Pico Chemiluminescent Substrate were added to the wells, and luminescence was counted using a
BMG Labtech PHERAstar FSX.

QUANTIFICATION AND STATISTICAL ANALYSIS

All functional study data were analyzed using Prism 8 (GraphPad) and presented as means ± S.E.M. from at least three independent
experiments. Non-linear curve fit was performed using a three-parameter logistic equation [log (agonist vs response)] for cAMP and
BRET assays. Competitive binding was performed using a one site – Fite Ki equation.

e7 Cell 186, 413–427.e1–e7, January 19, 2023


ll
Article

Supplemental figures

Figure S1. Purification of ORs-Gi complexes bound to respective opioid peptides, related to Figure 1
(A) Sequence alignment of common opioid peptides. Conserved residues are indicated in red.
(B) Characterization of the activities of opioid peptide ligands on all four ORs by radio-ligand competition binding assays.
(C) G protein BRET assays of all four opioid receptors induced by peptide ligands.
(D–H) Size exclusion chromatography (SEC) profiles (left) and SDS-PAGE analysis (right) of purified opioid peptide-bound ORs-Gi complexes.
ll
Article

Figure S2. Structure determination of b-endorphin- and endomorphin- bound mOR-Gi complexes and dynorphin- bound kOR-Gi complex,
related to Figures 1, 2, and 4
(A) Representative cryo-EM raw image and 2D classification averages of the b-endorphin-mOR-Gi complex.
(legend continued on next page)
ll
Article

(B) The flow chart of cryo-EM data processing of the b-endorphin-mOR-Gi complex.
(C) The Fourier shell correlation (FSC) curve of the b-endorphin-mOR-Gi complex. The global resolution of the final 3D density map defined at the FSC = 0.143
is 3.22 Å.
(D) Local resolution and angle distribution maps of the b-endorphin-mOR-Gi complex.
(E) EM density of transmembrane helices of mOR, a5 helix of Gai1, and b-endorphin.
(F) Representative cryo-EM raw image and 2D classification averages of the endomorphin-mOR-Gi complex.
(G) The flow chart of cryo-EM data processing of the endomorphin-mOR-Gi complex.
(H) The FSC curve of the endomorphin-mOR-Gi complex. The overall resolution of the final 3D density map defined at the FSC = 0.143 is 3.28 Å.
(I) Local resolution and angle distribution map of the endomorphin-mOR-Gi complex.
(J) EM density of transmembrane helices of mOR, a5 helix of Gai1, and endomorphin.
(K) Representative cryo-EM image and 2D classification averages of the dynorphin-kOR-Gi complex.
(L) The flow chart of cryo-EM data processing of the dynorphin-kOR-Gi complex.
(M) The FSC curve of the dynorphin-kOR-Gi complex. The global resolution of the final 3D density map defined at the FSC = 0.143 is 3.34 Å.
(N) Local resolutions and angle distribution map of the dynorphin-kOR-Gi complex.
(O) EM density of transmembrane helices of kOR, a5 helix of Gai1, and dynorphin.
ll
Article

Figure S3. Structure determination of deltorphin-bound dOR-Gi and NFQ-bound NOPR-Gi complexes, related to Figures 1, 3, and 5
(A) Representative cryo-EM micrographs of deltorphin-bound dOR-Gi complex.
(B) The flow chart of cryo-EM data processing of the deltorphin-bound dOR-Gi complex. In the procedure of data processing, two types of complexes with good
densities on the receptor part were divided. One of the two classes contained well-resolved G protein density, but with fewer particles. The overall resolution of
the complete dOR-Gi map was determined at 3.03 Å. For the other type with more particles, the density on G protein part was poorly resolved and the overall
resolution was determined at 2.62 Å.
(C and D) 2D classification averages and the FSC curve of the complete deltorphin-bound dOR-Gi complex. At the FSC 0.143 cutoff, the overall resolution of the
final 3D density map is defined at 3.03 Å.
(legend continued on next page)
ll
Article

(E and F) 2D classification averages and the FSC curve of the incomplete deltorphin-bound dOR-Gi complex. At the FSC 0.143 cutoff, the overall resolution of the
final 3D density map is 2.62 Å.
(G) Local resolution and angle distribution maps of the complete deltorphin-bound dOR-Gi complex.
(H) EM density of transmembrane helices of dOR, a5 helix of Gai1, and deltorphin.
(I) Representative cryo-EM image and 2D classification averages of the NFQ-NOPR-Gi complex.
(J) The flow chart of cryo-EM data processing of the NFQ-bound NOPR-Gi complex.
(K) Local resolutions and angle distribution map of the NFQ-bound NOPR-Gi complex.
(L) The FSC curve of the NFQ-bound NOPR-Gi complex. At the FSC 0.143 cutoff, the overall resolution of the final 3D density map is defined at 3.28 Å.
(M) Local EM density of transmembrane helices of NOPR, a5 helix of Gai1, and NFQ.
ll
Article

Figure S4. Full activation of the dOR-Gi complex by deltorphin, related to Figure 3
(A) Different rotatory angles and interaction patterns of mOR and dOR anti-parallel homodimers. Upper panel: mOR-Gi complex; lower panel: dOR-Gi complex.
(B) The mOR anti-parallel dimer interacted mainly through the interface of TM4 and TM5, the cholesterol molecules at the dimer interface are shown in black stick
representation.
(C) dOR dimer rotated at about 159 . The dimer interface was mainly formed through the residues from TM5 and TM6.
(D) Superposition of deltorphin-bound dOR with previous reported crystal structures of active and inactive dOR (PDB: 4N6H; 6PT2; 6PT3).
(E) Different binding modes of three agonists in the dOR OBP.
(F) Superposition of deltorphin-dOR with KGCHM07-dOR showed closer interactions of deltorphin with the DQY motif in dOR. Red dashes indicate hydrogen
bonds of deltorphin with the DQY motif. Black dashes indicate hydrogen bonds of KGCHM07 with DQY motif. Color usage: deltorphin-bound dOR, pink;
KGCHM07 bound dOR, green; deltorphin: teal; KGCHM07: yellow.
(G) Structural comparison of deltorphin and KGCHM07 bound dOR in TM5, TM6, and TM7-H8 regions. Larger inward movement of TM5, outward movement of
TM6, and rotation of TM7 were observed in deltorphin-bound dOR structure relative to that bound to KGCHM07.
(H) Pronounced rearrangement of the DRY and NPxxY motifs in the deltorphin-bound dOR complex relative to the KGCHM07-dOR complex.
ll
Article

Figure S5. cAMP, Gi BRET, and b-arrestin2 recruitment assays of wild-type opioid receptors and mutants, related to Figures 2–6 and
Tables S4–S7
HEK293 cells were transfected with WT or mutant opioid receptors. Cell-based cAMP, Gi BRET, and b-arrestin2 recruitment assays were performed. Results of
each point were represented as mean ± SEM from at least three independent experiments and each in duplicate. Dose response curves were further analyzed
using the ‘‘log(agonist) vs. response—three parameter’’ function in Graphpad Prism.
ll
Article

Figure S6. Interactions between opioid receptors and Gai, related to Figure 7
(A–D) Representative interactions of four opioid receptors with Gai. The left panel displays interactions with a5. The middle panel shows ICL2 interactions with aN
and b2-b3 loop. The right panel shows ICL3 interactions with b6 and a5. The electrostatic surface of ICL3 is also displayed.
ll
Article

Figure S7. GPCRs coupled to different G protein subtypes, related to Figure 7


(A) Interactions of ICL3 of ORs with Gai showing the positive residues of ICL3 to form electrostatic interactions with the negative charge patch of Gai. Similar ionic
patches were also observed in Gi subtype-selective GPCRs such as FPR2 and CB2.

(legend continued on next page)


ll
Article

(B and C) ICL3 of most Gaq/11- or Gas-coupled receptors were missing in their structures.
(D) Structural comparisons of mOR bound to morphine (PDB: 8EF6), fentanyl (PDB: 8EF5), b-endorphin and endomorphin.
(E and F) cAMP response of mOR induced by morphine, fentanyl, b-endorphin and endomorphin, relative to DAMGO.
(G and H) Superposition of the binding modes of fentanyl with b-endorphin and endomorphin, showing the extra-interactions of fentanyl with mOR (circle).

You might also like